1
|
Zamanian M, Gumpricht E, Salehabadi S, Kesharwani P, Sahebkar A. The effects of selected phytochemicals on schizophrenia symptoms: A review. Tissue Cell 2025; 95:102911. [PMID: 40253798 DOI: 10.1016/j.tice.2025.102911] [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: 11/07/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
There are suggested treatment options for schizophrenia (SZ), including antipsychotic medications. Unfortunately, these drugs mostly ameliorate only the positive symptoms of SZ, and patients have less tendency for compliance due to the drug's side effects. Hence, there is a need for additional or adjunct therapeutic options. This review considers selected phytochemicals with anti-schizophrenic activity as an alternative therapy. We searched the scientific literature and reviewed the evidence from pre-clinical (animal) and clinical studies using some phytochemicals in SZ. The reviewed phytochemicals provided varying potential beneficial effects on SZ. Of particular interest, berberine may provide additional ameliorative advantages against the disorder. Although still nascent in scientific research, these studies suggest a potential adjunct therapeutic option against the pathophysiological pathways implicated in SZ. We recommend robust, carefully performed randomized controlled trials evaluating the role of these phytochemicals in SZ.
Collapse
Affiliation(s)
- Melika Zamanian
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Eric Gumpricht
- Department of Pharmacology, Isagenix International, LLC, Gilbert, Arizona, AZ 85297, USA
| | - Sepideh Salehabadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura, Punjab 140417, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Lu Y, Wang K, Hu L. Advancements in delivery systems for dietary polyphenols in enhancing radioprotection effects: challenges and opportunities. NPJ Sci Food 2025; 9:51. [PMID: 40229284 PMCID: PMC11997175 DOI: 10.1038/s41538-025-00419-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
Radiotherapy, a widely employed cancer treatment, often triggers diverse inflammatory responses such as radiation enteritis, pulmonary injury, pelvic inflammation, dermatitis, and osteitis. Dietary polyphenols have recently emerged as promising agents for mitigating radiation-induced inflammation. However, their clinical application faced challenges related to variable bioavailability, individual pharmacokinetics, optimal dosing, and limited clinical evidence. Current researches revealed the efficacy of bioactive small molecule polyphenols in addressing radiation-induced inflammation. In this review, along with a comprehensive examination of the etiology and categories of radiation-induced inflammatory conditions, the diversity of polyphenols and elucidating their anti-inflammatory mechanisms are explored. This study emphasizes the recent progresses in delivery systems for dietary polyphenols, aiming to enhance radioprotection effects. The optimized utilization of polyphenols, with a theoretical framework and reference guide, is of paramount relevance. Through diverse delivery mechanisms, the more effective and safer radioprotective strategies become achievable. This endeavor aspires to contribute to breakthroughs in the dietary polyphenols' application, significantly enhancing human health protection during radiotherapy. These comprehensive insights presented here also support (pre)-clinical practices in navigating the complexities of utilizing dietary polyphenols for radioprotection, fostering advancements in the field and improving patient outcomes.
Collapse
Affiliation(s)
- Yuxuan Lu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, China
| | - Kai Wang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China.
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
3
|
Liu X, Li Y, Wang J, Meng T, Song L, Yang L, Yu J, Ma C. Polysaccharides from Ganoderma lucidum attenuate cognitive impairment in 5xFAD mice by inhibiting oxidative stress and modulating mitochondrial dynamics via the Nrf2/antioxidative axis activation. Metab Brain Dis 2025; 40:180. [PMID: 40227285 DOI: 10.1007/s11011-025-01601-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/04/2025] [Indexed: 04/15/2025]
Abstract
Oxidative stress and mitochondrial dynamics imbalance are key contributors to AD pathogenesis. GLPS, an extract from Ganoderma lucidum spores, exhibits anti-inflammatory, antioxidant, and immunomodulatory properties. However, the roles of GLPS in regulating oxidative stress and mitochondrial dynamics in AD remain poorly understood. Here, the underlying mechanisms of neuroprotective effects on cognitive dysfunction in 5 × FAD mice were explored. C57BL/6 mice served as WT controls, while 5 × FAD mice were divided into an AD group and an AD + GLPS group. The mice in AD + GLPS group were administered daily GLPS (25 mg/kg) by i.p. injection for two months, while WT and AD mice received an equivalent volume of normal saline. The results indicated that GLPS markedly improved cognitive function and decreased p-tau and Aβ levels in 5 × FAD mice. Moreover, GLPS alleviated oxidative stress by increasing SOD levels and decreasing MDA concentrations. It also inhibited excessive mitochondrial fragmentation by decreasing the expression of p-Drp1 and Fis1, while increasing the levels of Mfn1, Mfn2, and OPA1 in 5 × FAD mice. Mechanistically, GLPS activated Nrf2, leading to a marked upregulation of antioxidant enzymes, including HO- 1, NQO1, and SOD2 in 5 × FAD mice. Collectively, these findings suggest that GLPS ameliorates cognitive deficits in 5 × FAD mice by reducing oxidative stress and modulating mitochondrial dynamics through Nrf2-mediated antioxidant enzyme activation.
Collapse
Affiliation(s)
- Xiaoqin Liu
- Institute of Brain Science, Medical School, Shanxi Datong University, Datong, 037009, China
| | - Yanbing Li
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple, Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Jiwei Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple, Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Tao Meng
- Institute of Brain Science, Medical School, Shanxi Datong University, Datong, 037009, China
| | - Lijuan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple, Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Lizhi Yang
- Shanxi Guorun Pharmaceutical Co., Ltd, Datong, 038100, China
| | - Jiezhong Yu
- Institute of Brain Science, Medical School, Shanxi Datong University, Datong, 037009, China.
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple, Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, 030619, China.
- Department of Neurology, The Fifth People's Hospital, Datong, 037009, China.
| | - Cungen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple, Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, 030619, China.
| |
Collapse
|
4
|
Chiang MC, Nicol CJB, Yang YP, Chiang T, Yen C. The α-MG exhibits neuroprotective potential by reducing amyloid beta peptide-induced inflammation, oxidative stress, and tau aggregation in human neural stem cells. Brain Res 2025; 1852:149506. [PMID: 39954799 DOI: 10.1016/j.brainres.2025.149506] [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: 06/27/2024] [Revised: 12/29/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Alzheimer's disease (AD) is the primary cause of dementia in older adults. Amyloid-beta (Aβ) and tau protein neurofibrillary tangles accumulate in the brain, leading to a progressive decline in memory, thinking, and behavior. Neuroinflammation and oxidative stress play a significant role in the development and progression of AD. Research has suggested that α-mangostin (α-MG), a compound found in mangosteen peels, may have anti-inflammatory, antioxidant, and neuroprotective properties, which could be beneficial in the context of AD. Further research is required to fully comprehend the therapeutic mechanisms of α-MG on AD and determine its potential as a treatment option. α-MG treatment significantly improves the viability of hNSCs exposed to Aβ and reduces caspase activity. Furthermore, this treatment is associated with a notable decrease in the expression of TNF-α and IL-1β. The treatment effectively restores alterations in the expression of IKK and NF-κB (p65) induced by Aβ, which are critical factors in the inflammatory response. Moreover, α-MG effectively reduces iNOS and COX-2 levels in Aβ-treated hNSCs, showcasing its potential therapeutic benefits. Treatment with α-MG protects hNSCs against Aβ-induced oxidative stress and effectively prevents the decrease in Nrf2 levels caused by Aβ. The treatment significantly enhances the activity and mRNA expression of Nrf2 downstream antioxidant target genes, including SOD-1, SOD-2, Gpx1, GSH, catalase, and HO-1, compared to Aβ-treated controls. α-MG significantly reduces tau and ubiquitin (Ub) aggregates, enhances proteasome activity, and increases the mRNA expression of HSF1, HSP27, HSP70, and HSP90 in Tau-GFP-expressed hNSCs. This study significantly improves our comprehension of the anti-inflammatory, antioxidative stress, and anti-aggregated effects of α-MG. These findings have potential therapeutic implications for developing treatments that could delay AD progression and promote healthy aging.
Collapse
Affiliation(s)
- Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Christopher J B Nicol
- Departments of Pathology & Molecular Medicine and Biomedical & Molecular Sciences, and Cancer Biology and Genetics Division, Sinclair Cancer Research Institute, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Yu-Ping Yang
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Tairui Chiang
- New Taipei Municipal Jinhe High School, New Taipei City 235, Taiwan; Ames Middle School, Ames, IA 50014, USA
| | - Chiahui Yen
- Department of International Business, Ming Chuan University, Taipei 111, Taiwan
| |
Collapse
|
5
|
Selvaraj NR, Nandan D, Nair BG, Nair VA, Venugopal P, Aradhya R. Oxidative Stress and Redox Imbalance: Common Mechanisms in Cancer Stem Cells and Neurodegenerative Diseases. Cells 2025; 14:511. [PMID: 40214466 PMCID: PMC11988017 DOI: 10.3390/cells14070511] [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: 12/24/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/14/2025] Open
Abstract
Oxidative stress (OS) is an established hallmark of cancer and neurodegenerative disorders (NDDs), which contributes to genomic instability and neuronal loss. This review explores the contrasting role of OS in cancer stem cells (CSCs) and NDDs. Elevated levels of reactive oxygen species (ROS) contribute to genomic instability and promote tumor initiation and progression in CSCs, while in NDDs such as Alzheimer's and Parkinson's disease, OS accelerates neuronal death and impairs cellular repair mechanisms. Both scenarios involve disruption of the delicate balance between pro-oxidant and antioxidant systems, which leads to chronic oxidative stress. Notably, CSCs and neurons display alterations in redox-sensitive signaling pathways, including Nrf2 and NF-κB, which influence cell survival, proliferation, and differentiation. Mitochondrial dynamics further illustrate these differences: enhanced function in CSCs supports adaptability and survival, whereas impairments in neurons heighten vulnerability. Understanding these common mechanisms of OS-induced redox imbalance may provide insights for developing interventions, addressing aging hallmarks, and potentially mitigating or preventing both cancer and NDDs.
Collapse
Affiliation(s)
| | | | | | | | - Parvathy Venugopal
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| | - Rajaguru Aradhya
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| |
Collapse
|
6
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial NOX2 as a therapeutic target in traumatic brain injury: Mechanisms, consequences, and potential for neuroprotection. Ageing Res Rev 2025; 108:102735. [PMID: 40122395 DOI: 10.1016/j.arr.2025.102735] [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: 01/24/2025] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability worldwide, with secondary injury mechanisms, including neuroinflammation and oxidative stress, driving much of its chronic pathology. While NADPH oxidase 2 (NOX2)-mediated reactive oxygen species (ROS) production is a recognized factor in TBI, the specific role of microglial NOX2 in perpetuating oxidative and inflammatory damage remains underexplored. Addressing this gap is critical, as current therapeutic approaches primarily target acute symptoms and fail to interrupt the persistent neuroinflammation that contributes to progressive neurodegeneration. Besides NOX, other ROS-generating enzymes, such as CYP1B1, COX2, and XO, also play crucial roles in triggering oxidative stress and neuroinflammatory conditions in TBI. However, this review highlights the pathophysiological role of microglial NOX2 in TBI, focusing on its activation following injury and its impact on ROS generation, neuroinflammatory signaling, and neuronal loss. These insights reveal NOX2 as a critical driver of secondary injury, linked to worsened outcomes, particularly in aged individuals where NOX2 activation is more pronounced. In addition, this review evaluates emerging therapeutic approaches targeting NOX2, such as GSK2795039 and other selective NOX2 inhibitors, which show potential in reducing ROS levels, limiting neuroinflammation, and preserving neurological functions. By highlighting the specific role of NOX2 in microglial ROS production and secondary neurodegeneration, this study advocates for NOX2 inhibition as a promising strategy to improve TBI outcomes by addressing the unmet need for therapies targeting long-term inflammation and neuroprotection. Our review highlights the potential of NOX2-targeted interventions to disrupt the cycle of oxidative stress and inflammation, ultimately offering a pathway to mitigate the chronic impact of TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, New Delhi, India.
| | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
7
|
Chaubey S, Singh L. Deciphering the mechanisms underlying the neuroprotective potential of kaempferol: a comprehensive investigation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2275-2292. [PMID: 39414700 DOI: 10.1007/s00210-024-03515-8] [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: 08/10/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024]
Abstract
Neurodegenerative disorders are characterized by neuronal degradation, dysfunction, or death within the CNS. Oxidative and inflammatory stress play crucial roles in the pathogenesis of various neurodegenerative diseases. The interplay between these stressors and dysregulated cellular signaling pathways contributes to neurodegeneration. Downregulation of NRF-2 compromises antioxidant defense, exacerbating neuronal damage, while increased TLR-4/MAPK and TLR-4/NF-κB signaling promotes neuroinflammation. Excessive ROS production by NADPH oxidase leads to oxidative damage and neuronal apoptosis. The strategies targeting NRF-2, TLR-4-mediated inflammatory stress, and NADPH oxidase activity promise to mitigate neuronal damage and halt the progression of the disease. Kaempferol is a flavonoid polyphenol antioxidant found abundantly in various fruits and vegetables, including apples, grapes, tomatoes, and broccoli. It is widely found in medicinal plants including Equisetum spp., Sophora japonica, Ginkgo biloba, and Euphorbia pekinensis (Rupr.). A substantial body of in vitro and in vivo evidences have demonstrated the neuroprotective potential of kaempferol against neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Kaempferol demonstrates multifaceted potential in mitigating neuroinflammation, apoptosis, and oxidative stress in different neurodegenerative diseases through the modulation of various pathways including NRF-2, NADPH oxidase, TLR-4/MAPK, and TLR-4/NF-κB. This review article was developed through a comprehensive analysis and interpretation of research published between 2009 and 2024, sourced from multiple scientific databases, including PubMed, Scopus, ScienceDirect, and Web of Science. This review aims to provide an in-depth overview of the neuroprotective effects of kaempferol, focusing on its underlying molecular mechanisms. A total of 24 research evidence were included to elucidate the molecular pathways by which kaempferol exerts its protective effects against neurodegenerative diseases.
Collapse
Affiliation(s)
- Satyam Chaubey
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Lovedeep Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| |
Collapse
|
8
|
Singh AA, Katiyar S, Song M. Phytochemicals Targeting BDNF Signaling for Treating Neurological Disorders. Brain Sci 2025; 15:252. [PMID: 40149774 PMCID: PMC11939912 DOI: 10.3390/brainsci15030252] [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: 01/23/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Neurological disorders are defined by a deterioration or disruption of the nervous system's structure and function. These diseases, which include multiple sclerosis, Alzheimer's disease, Parkinson's disease, Huntington's disease, and schizophrenia, are caused by intricate pathological processes that include excitotoxicity, neuroinflammation, oxidative stress, genetic mutations, and compromised neurotrophic signaling. Although current pharmaceutical treatments relieve symptoms, their long-term efficacy is limited due to adverse side effects and weak neuroprotective properties. However, when combined with other neuroprotective drugs or adjunct therapy, they may offer additional benefits and improve treatment outcomes. Phytochemicals have emerged as attractive therapeutic agents due to their ability to regulate essential neurotrophic pathways, especially the brain-derived neurotrophic factor (BDNF) signaling cascade. BDNF is an important target for neurodegenerative disease (ND) treatment since it regulates neuronal survival, synaptic plasticity, neurogenesis, and neuroprotection. This review emphasizes the molecular pathways through which various phytochemicals-such as flavonoids, terpenoids, alkaloids, and phenolic compounds-stimulate BDNF expression and modulate its downstream signaling pathways, including GSK-3β, MAPK/ERK, PI3K/Akt/mTOR, CREB, and Wnt/β-catenin. This paper also highlights how phytochemical combinations may interact to enhance BDNF activity, offering new therapeutic options for ND treatment. Despite their potential for neuroprotection, phytochemicals face challenges related to pharmacokinetics, blood-brain barrier (BBB) permeability, and absorption, highlighting the need for further research into combination therapies and improved formulations. Clinical assessment and mechanistic understanding of BDNF-targeted phytotherapy should be the main goals of future studies. The therapeutic efficacy of natural compounds in regulating neurotrophic signaling is highlighted in this review, providing a viable approach to the prevention and treatment of NDs.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Shweta Katiyar
- Department of Botany, SBN Government PG College, Barwani 451551, MP, India;
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
9
|
Mengoni B, Armeli F, Schifano E, Prencipe SA, Pompa L, Sciubba F, Brasili E, Giampaoli O, Mura F, Reverberi M, Beccaccioli M, Pinto A, De Giusti M, Uccelletti D, Businaro R, Vinci G. In Vitro and In Vivo Antioxidant and Immune Stimulation Activity of Wheat Product Extracts. Nutrients 2025; 17:302. [PMID: 39861432 PMCID: PMC11767776 DOI: 10.3390/nu17020302] [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: 12/19/2024] [Revised: 01/13/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Inflammation and oxidative stress are the main pathogenetic pathways involved in the development of several chronic degenerative diseases. Our study is aimed at assessing the antioxidant and anti-inflammatory activity of hydroalcoholic extracts obtained from wheat and its derivatives. METHODS The content of total phenolic and total flavonoid compounds and antioxidant activity were carried out by ABTS and DPPH assays. The ability of wheat extracts to promote microglia polarization towards an anti-inflammatory phenotype was evaluated analyzing the increased expression of anti-inflammatory markers by real-time qPCR and immunofluorescence assays. Antioxidant activity of all the extracts was evaluated in C. elegans by analyzing ROS levels and the expression of the antioxidant enzymes GST-4 and SOD-3 by real-time qPCR and fluorescence experiments. The expression of key genes involved in the innate immune response and stress resistance pathways-daf-16, sek-1, and pmk-1-was evaluated by real-time qPCR. RESULTS Wheat extracts showed the ability to polarize microglia cells towards an anti-inflammatory phenotype, even after the addition of LPS. An antioxidant response was detected both in microglia and in Caenorhabditis elegans nematode, where the extracts also implemented an anti-stress resilience response and stimulated the innate immunity. CONCLUSIONS The present study shows that wheat seeds, flour, chaff, and pasta present anti-inflammatory as well as antioxidant activities and may be considered as prospective positive health agents for the preparation of functional foods. Moreover, the valorization of by-products from agricultural and agro-industrial activities would also have significant implications in terms of circular economy.
Collapse
Affiliation(s)
- Beatrice Mengoni
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (B.M.); (F.A.)
| | - Federica Armeli
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (B.M.); (F.A.)
- Department of Human Sciences, European University of Rome, 00163 Rome, Italy
| | - Emily Schifano
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (E.S.); (L.P.); (D.U.)
| | | | - Laura Pompa
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (E.S.); (L.P.); (D.U.)
| | - Fabio Sciubba
- Department of Environmental Biology, Sapienza University of Rome, 00185 Rome, Italy; (F.S.); (E.B.); (F.M.); (M.R.); (M.B.)
- NMR-Based Metabolomics Laboratory (NMLab), Sapienza University of Rome, 00185 Rome, Italy
| | - Elisa Brasili
- Department of Environmental Biology, Sapienza University of Rome, 00185 Rome, Italy; (F.S.); (E.B.); (F.M.); (M.R.); (M.B.)
| | - Ottavia Giampaoli
- Department of Chemistry, Sapienza University of Rome, 00185 Rome, Italy;
| | - Francesco Mura
- Department of Environmental Biology, Sapienza University of Rome, 00185 Rome, Italy; (F.S.); (E.B.); (F.M.); (M.R.); (M.B.)
| | - Massimo Reverberi
- Department of Environmental Biology, Sapienza University of Rome, 00185 Rome, Italy; (F.S.); (E.B.); (F.M.); (M.R.); (M.B.)
| | - Marzia Beccaccioli
- Department of Environmental Biology, Sapienza University of Rome, 00185 Rome, Italy; (F.S.); (E.B.); (F.M.); (M.R.); (M.B.)
| | - Alessandro Pinto
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Maria De Giusti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy;
| | - Daniela Uccelletti
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (E.S.); (L.P.); (D.U.)
- NMR-Based Metabolomics Laboratory (NMLab), Sapienza University of Rome, 00185 Rome, Italy
| | - Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (B.M.); (F.A.)
| | - Giuliana Vinci
- Department of Management, Sapienza University of Rome, 00161 Rome, Italy; (S.A.P.); (G.V.)
| |
Collapse
|
10
|
Akhtar W, Muazzam Khan M, Kumar S, Ahmad U, Husen A, Avirmed S. Pathophysiology of cerebral ischemia-reperfusion injury: An overview of oxidative stress and plant-based therapeutic approaches. Brain Res 2025; 1847:149308. [PMID: 39491664 DOI: 10.1016/j.brainres.2024.149308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Stroke is a debilitating neurological disorder that causes substantial morbidity and mortality on a global scale. Ischemic stroke, the most common type, occurs when the brain's blood supply is interrupted. Oxidative stress is a key factor in stroke pathology, contributing to inflammation and neuronal cell death. As a result, there is increasing interest in the potential of plant extracts, which have been used in traditional medicine for centuries and are generally considered safe, to serve as alternative or complementary treatments for stroke. The plant extracts can target multiple pathological processes, including oxidative stress, offering neuroprotective effects. The development of highly efficient, low-toxicity, and cost-effective natural products is crucial for enhancing stroke treatment options. In this review, we examine 60 plant extracts that have been focused on the studies published from year 2000 to 2024 along with the studies' experimental models, dosages, and results. The plant extracts hold promise in modulating cerebral ischemia-reperfusion injury through counteraction of relevant pathophysiologic processes such as oxidative stress.
Collapse
Affiliation(s)
- Wasim Akhtar
- Hygia Institute of Pharmacy, Lucknow 226013, Uttar Pradesh, India
| | - Mohd Muazzam Khan
- Faculty of Pharmacy, Integral University, Lucknow 226020, Uttar Pradesh, India.
| | - Sanjay Kumar
- Hygia Institute of Pharmacy, Lucknow 226013, Uttar Pradesh, India
| | - Usama Ahmad
- Faculty of Pharmacy, Integral University, Lucknow 226020, Uttar Pradesh, India
| | - Ali Husen
- Hygia Institute of Pharmacy, Lucknow 226013, Uttar Pradesh, India
| | | |
Collapse
|
11
|
Gichuru V, Sbrocca I, Molinari M, Tonto TC, Locato V, Cimini S, De Gara L. Exploring the antioxidant and antimicrobial properties of five indigenous Kenyan plants used in traditional medicine. Sci Rep 2025; 15:1459. [PMID: 39789046 PMCID: PMC11718201 DOI: 10.1038/s41598-024-80883-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/22/2024] [Indexed: 01/12/2025] Open
Abstract
Defined by the World Health Organization (WHO) as indigenous knowledge and practices used for maintaining health and treating illnesses, traditional medicine (TM) represents a rich reservoir of ancient healing practices rooted in cultural traditions and accumulated wisdom over centuries. Five indigenous Kenyan plant species traditionally used in African TM, named Afzelia quanzensis, Azadirachta indica, Gigasiphon macrosiphon, Grewia bicolor, and Lannea schweinfurthii, represent a valuable resource in healing practices, yet their chemical composition and bioactivity remain understudied. To depict a primary bio-chemical characterization of these plants, their antioxidant and antimicrobial features have been evaluated by the use of methods validated in this context. G. bicolor, and G. macrosiphon were found to have great potential as sources of bioactive metabolites, such as chlorophyll a (1456.29 µg/ g DW; 1104.33 µg/ g DW), chlorophyll b (712.48 µg/ g DW; 443.31 µg/ g DW), and carotenoids (369.71 µg/ g DW; 300 µg/ g DW) as well as phenols (31.78 mg GAE/g DW; 27.54 GAE/g DW), and exhibiting high antioxidant activity, according to TEAC, DPPH and FRAP assays. Additionally, L. schweinfurthii and G. macrosiphon demonstrated antimicrobial activity against the Gram-negative bacteria E. coli, as well as against Gram-positive ones, S. aureus and B. subtilis.
Collapse
Affiliation(s)
- Virginia Gichuru
- Department of Biological Sciences, Pwani University, Kilifi, Kenya
| | - Irene Sbrocca
- Department of Science and Technology for Sustainable Development and One Health, Unit of Food Science and Nutrition, Università Campus Bio-Medico di Roma, Rome, 00128, Italy
| | - Michela Molinari
- Department of Science and Technology for Sustainable Development and One Health, Unit of Food Science and Nutrition, Università Campus Bio-Medico di Roma, Rome, 00128, Italy
| | - Teodora Chiara Tonto
- Department of Science and Technology for Sustainable Development and One Health, Unit of Food Science and Nutrition, Università Campus Bio-Medico di Roma, Rome, 00128, Italy
| | - Vittoria Locato
- Department of Science and Technology for Sustainable Development and One Health, Unit of Food Science and Nutrition, Università Campus Bio-Medico di Roma, Rome, 00128, Italy
- NBFC, National Biodiversity Future Center, Palermo, 90133, Italy
| | - Sara Cimini
- Department of Science and Technology for Sustainable Development and One Health, Unit of Food Science and Nutrition, Università Campus Bio-Medico di Roma, Rome, 00128, Italy.
- NBFC, National Biodiversity Future Center, Palermo, 90133, Italy.
| | - Laura De Gara
- Department of Science and Technology for Sustainable Development and One Health, Unit of Food Science and Nutrition, Università Campus Bio-Medico di Roma, Rome, 00128, Italy
- NBFC, National Biodiversity Future Center, Palermo, 90133, Italy
| |
Collapse
|
12
|
Deabes DAH, El-Abd EAW, Baraka SM, El-Gendy ZA, Korany RMS, Elbatanony MM. Metabolomics analyses and comparative insight to neuroprotective potential of unripe fruits and leaves of Citrus aurantium ethanolic extracts against cadmium-induced rat brain dysfunction: involvement of oxidative stress and akt-mediated CREB/BDNF and GSK3β/NF-κB signaling pathways. Metab Brain Dis 2025; 40:89. [PMID: 39760898 PMCID: PMC11703990 DOI: 10.1007/s11011-024-01513-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025]
Abstract
Serious neurological disorders were associated with cadmium toxicity. Hence, this research aimed to investigate the potential neuroprotective impacts of the ethanolic extracts of Citrus aurantium unripe fruits and leaves (CAF and CAL, respectively) at doses 100 and 200 mg/kg against cadmium chloride-provoked brain dysfunction in rats for 30 consecutive days. HPLC for natural pigment content revealed that CAF implied higher contents of Chlorophyll B, while the CAL has a high yield of chlorophyll A and total carotenoid. Fifty-seven chromatographic peaks were identified by UPLC/MS/MS; 49 and 29 were recognized from CAF or CAL, respectively. Four compounds were isolated from CAF: 3',4',7 -trihydroxyflavone, isorhainetin, vitexin, and apigenin. In vitro studies outlined the antioxidant capacity of studied extracts where CAF showed better scavenging radical DPPH activity. Results clarified that both extracts with a superior function of CAF at the high adopted dose significantly ameliorated CdCl2-induced neuro-oxidative stress and neuro-inflammatory response via restoring antioxidant status and hindering nuclear factor kappa B (NF-κB) stimulation. Moreover, it up-regulated the levels of phospho-protein kinase B (p-Akt), phospho- cAMP-response element binding protein (p-CREB), and brain-derived neurotropic factor (BDNF) levels, and elicited a marked decrease in the content of glycogen synthase kinase 3 beta (GSK3β), besides amending Caspase-3 and hyperphosphorylation of tau protein in brain tissues. Moreover, a significant improvement in the rats' behavioral tasks of the CAL and CAF-treated groups has been recorded, as indicated by marked preservation in locomotion, exploratory, and memory functions of the experimental rats. In conclusion, the reported neuroprotective impacts of C. aurantium extracts may be through modulating p-AKT/p-CREB/BDNF and / or p-Akt/ GSK3β/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Doaa A H Deabes
- Pharmacognosy Department, National Research Centre (NRC), El Behouth St., P.O. 12622, Cairo, Egypt
| | - Eman A W El-Abd
- Pharmacognosy Department, National Research Centre (NRC), El Behouth St., P.O. 12622, Cairo, Egypt
| | - Sara M Baraka
- Chemistry of Natural Compounds Department, National Research Centre, Giza, 12622, Egypt.
| | - Zeinab A El-Gendy
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Reda M S Korany
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Marwa M Elbatanony
- Pharmacognosy Department, National Research Centre (NRC), El Behouth St., P.O. 12622, Cairo, Egypt
| |
Collapse
|
13
|
Singh L. Daidzein's potential in halting neurodegeneration: unveiling mechanistic insights. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:243-259. [PMID: 39158734 DOI: 10.1007/s00210-024-03356-5] [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: 05/21/2024] [Accepted: 08/01/2024] [Indexed: 08/20/2024]
Abstract
Neurological conditions encompassing a wide range of disorders pose significant challenges globally. The complex interactions among signaling pathways and molecular elements play pivotal roles in the initiation and progression of neurodegenerative diseases. Isoflavones have emerged as a promising candidate to fight against neurodegenerative diseases. Daidzein, a 7-hydroxy-3-(4-hydroxyphenyl)-chromen-4-one, belongs to the isoflavone class and exhibits a diverse pharmacological profile. It is found primarily in soybeans and soy products, as well as in some other legumes and herbs. Investigations into daidzein have revealed that it confers neuroprotection by inhibiting oxidative stress, inflammation, and apoptosis, which are key contributors to neuronal damage and degeneration. Activating pathways like PI3K/Akt/mTOR and promoting neurotrophic factors like BDNF by daidzein underscore its potential in supporting neuronal function and combating neurodegeneration. Daidzein's effects on dopamine provide further avenues for intervention in conditions like Parkinson's disease. Additionally, the modulation of inflammatory and NRF-2-antioxidant signaling by daidzein reinforces its neuroprotective role. Moreover, daidzein's interaction with receptors and cellular processes like ER-β, GPR30, MAO, VEGF, and GnRH highlights its multifaceted effects across multiple pathways involved in neuroprotection and neuronal function. This review article delves into the mechanistic interplay of various mediators in mediating the neuroprotective effects of daidzein. The review article consolidates and analyzes research published over nearly two decades (2005-2024) from various databases, including PubMed, Scopus, ScienceDirect, and Web of Science, to provide a comprehensive understanding of daidzein's effects and mechanisms in neuroprotection.
Collapse
Affiliation(s)
- Lovedeep Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| |
Collapse
|
14
|
Desouky MA, Michel HE, Elsherbiny DA, George MY. Recent pharmacological insights on abating toxic protein species burden in neurological disorders: Emphasis on 26S proteasome activation. Life Sci 2024; 359:123206. [PMID: 39489397 DOI: 10.1016/j.lfs.2024.123206] [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: 06/08/2024] [Revised: 07/30/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Protein homeostasis (proteostasis) refers to the plethora of mechanisms that safeguard the proper folding of the newly synthesized proteins. It entails various intricately regulated cues that demolish the toxic protein species to prevent their aggregation. The ubiquitin-proteasome system (UPS) is recognized as a salient protein degradation system, with a substantial role in maintaining proteostasis. However, under certain circumstances the protein degradation capacity of the UPS is overwhelmed, leading to the accumulation of misfolded proteins. Several neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, Huntington disease, and amyotrophic lateral sclerosis are characterized with the presence of protein aggregates and proteinopathy. Accordingly, enhancing the 26S proteasome degradation activity might delineate a pioneering approach in targeting various proteotoxic disorders. Regrettably, the exact molecular approaches that enhance the proteasomal activity are still not fully understood. Therefore, this review aimed to underscore several signaling cascades that might restore the degradation capacity of this molecular machine. In this review, we discuss the different molecular components of the UPS and how 26S proteasomes are deleteriously affected in many neurodegenerative diseases. Moreover, we summarize different signaling pathways that can be utilized to renovate the 26S proteasome functional capacity, alongside currently known druggable targets in this circuit and various classes of proteasome activators.
Collapse
Affiliation(s)
- Mahmoud A Desouky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Doaa A Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt.
| |
Collapse
|
15
|
Elhemiely AA, Darwish A. Pharmacological and biochemical insights into lead-induced hepatotoxicity: Pathway interplay and the protective effects of arbutin via the oral and intraperitoneal routes in silico and in vivo. Int Immunopharmacol 2024; 142:112968. [PMID: 39226827 DOI: 10.1016/j.intimp.2024.112968] [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: 06/24/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024]
Abstract
INTRODUCTION Lead acetate (PbAc), a hazardous heavy metal, poses significant threats to human health and the environment because of widespread industrial exposure. PbAc exposure leads to liver injury primarily through oxidative stress and the disruption of key regulatory pathways. Understanding these mechanisms and exploring protective agents are vital for mitigating PbAc-induced hepatotoxicity. Therefore, we aimed to investigate the molecular pathways implicated in PbAc-induced liver damage, focusing on Sirt-1, Nrf2 (HO-1, NQO1, and SOD), Akt-1/GSK3β, m-TOR, and P53. Additionally, we aimed to assess the hepatoprotective effects of arbutin, which is administered orally and intraperitoneally, to determine the most effective delivery method. METHODOLOGY In silico analyses were conducted to identify relevant protein networks associated with Sirt-1 and AKT-1/GSK-3B pathways. The pharmacodynamic properties of arbutin were examined, followed by molecular docking studies to elucidate its interactions with the selected protein network. In vivo preclinical studies were carried out on adult male rats randomly assigned to 6 different treatment groups, including PbAc exposure and PbAc exposure treated with arbutin either orally or intraperitoneally. RESULTS PbAc exposure led to hepatic oxidative stress, as evidenced by elevated MDA levels and SIRT-1 inhibition, disrupting antioxidant pathways and activating antiautophagic and proapoptotic pathways, ultimately resulting in hepatocyte necrosis. Both oral and intraperitoneal arbutin administration effectively modifed these effects, with intraperitoneal delivery showing superior efficacy in mitigating PbAc-induced histological, immunological, and biochemical alterations. CONCLUSION This study provides insights into the molecular mechanisms underlying PbAc-induced liver injury and highlights the hepatoprotective potential of arbutin. These findings suggest that arbutin, particularly when administered intraperitoneally, holds promise as a therapeutic agent for combating PbAc-induced hepatotoxicity.
Collapse
Affiliation(s)
| | - Alshaymaa Darwish
- Department of Biochemistry, Faculty of Pharmacy, Sohag University, Sohag, Egypt.
| |
Collapse
|
16
|
Pal R, Mukherjee S, Khan A, Nathani M, Maji S, Tandey R, Das S, Patra A, Mandal V. A critical appraisal on the involvement of plant-based extracts as neuroprotective agents (2012-2022): an effort to ease out decision-making process for researchers. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9367-9415. [PMID: 38985312 DOI: 10.1007/s00210-024-03266-6] [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: 05/21/2024] [Accepted: 06/28/2024] [Indexed: 07/11/2024]
Abstract
The purpose of this review study is to provide a condensed compilation of 164 medicinal plants that have been investigated for their neuroprotective aspects by researchers between the years 2012 and 2022 which also includes a recent update of 2023-2024. After using certain keywords to retrieve the data from SCOPUS, it was manually sorted to eliminate any instances of duplication. The article is streamlined into three major segments. The first segment takes a dig into the current global trend and attempts to decrypt vital information related to plant names, families, plant parts used, and neurological disorders investigated. The second segment of the article makes an attempt to present a comprehensive insight into the various mechanistic pathways through which phytochemicals can intervene to exert neuroprotection. The final segment of the manuscript is a bibliometric appraisal of all researches conducted. The study is based on 256 handpicked articles based on decided inclusion criteria. Illustrative compilation of various pathways citing their activation and deactivation channels are also presented with possible hitting points of various phytochemicals. The present study employed Microsoft Excel 2019 and VOS viewer as data visualisation tools.
Collapse
Affiliation(s)
- Riya Pal
- Department of Pharmacy, Guru Ghasidas Central University, Bilaspur, C.G, 495009, India
| | - Souvik Mukherjee
- Department of Pharmacy, Guru Ghasidas Central University, Bilaspur, C.G, 495009, India
| | - Altamash Khan
- Department of Pharmacy, Guru Ghasidas Central University, Bilaspur, C.G, 495009, India
| | - Mansi Nathani
- Department of Pharmacy, Guru Ghasidas Central University, Bilaspur, C.G, 495009, India
| | - Sayani Maji
- Department of Pharmacy, Guru Ghasidas Central University, Bilaspur, C.G, 495009, India
| | - Roshni Tandey
- Department of B. Pharm Ayurveda, Delhi Pharmaceutical Sciences and Research University, Sector-3, MB Road, Pushp Vihar, New Delhi, 110017, India
| | - Sinchan Das
- Department of Pharmacy, Guru Ghasidas Central University, Bilaspur, C.G, 495009, India
| | - Arjun Patra
- Department of Pharmacy, Guru Ghasidas Central University, Bilaspur, C.G, 495009, India
| | - Vivekananda Mandal
- Department of Pharmacy, Guru Ghasidas Central University, Bilaspur, C.G, 495009, India.
| |
Collapse
|
17
|
Li W, Wang L, Qian Y, Wang M, Li F, Zeng M. True-solution-scale utilization of natural chlorophyll a in aqueous media through cooperative aggregation with phycocyanin. Food Chem 2024; 460:140678. [PMID: 39098190 DOI: 10.1016/j.foodchem.2024.140678] [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] [Revised: 07/13/2024] [Accepted: 07/25/2024] [Indexed: 08/06/2024]
Abstract
The challenge of applying chlorophyll(Chl) in aqueous media has been a significant obstacle to the diversified development of Chl a-related industries. This study presents the first report on the true-solution-scale utilization of Chl in aqueous media through the construction of chlorophyll a-phycocyanin (Chls-PC) composite nanoparticles. This study determined the optimal conditions for Chls-PC preparation: a composite ratio of 1:25, a solvent ratio of 1:4, and a stirring time of 1 h. Fluorescence spectroscopy, transmission electron microscope, and confocal microscopy confirmed Chl a and PC aggregation. Surface hydrophobicity and contact angle measurements showed that Chls-PC water solubility was similar to PC and much higher than Chl. Infrared spectroscopy, quantum chemical calculations, X-ray photoelectron spectroscopy, and molecular dynamics simulations elucidated the water solubilization mechanism of Chls-PC both experimentally and theoretically. This research provides theoretical guidance for the development and production of water-based products using Chl as a raw material.
Collapse
Affiliation(s)
- Wei Li
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266400, People's Republic of China; Sanya Institute of Oceanography, Ocean University of China, Sanya 572000, People's Republic of China
| | - Lijuan Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266400, People's Republic of China; Sanya Institute of Oceanography, Ocean University of China, Sanya 572000, People's Republic of China
| | - Yuemiao Qian
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266400, People's Republic of China; Sanya Institute of Oceanography, Ocean University of China, Sanya 572000, People's Republic of China
| | - Mengwei Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266400, People's Republic of China
| | - Fangwei Li
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266400, People's Republic of China; Sanya Institute of Oceanography, Ocean University of China, Sanya 572000, People's Republic of China.
| | - Mingyong Zeng
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266400, People's Republic of China; Sanya Institute of Oceanography, Ocean University of China, Sanya 572000, People's Republic of China.
| |
Collapse
|
18
|
Thuy PT, Ha NX. Theoretical studies on the antioxidant activity of potential marine xanthones. Free Radic Res 2024; 58:826-840. [PMID: 39676294 DOI: 10.1080/10715762.2024.2438918] [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: 09/10/2024] [Revised: 11/14/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
In this study, a quantum chemical exploration was conducted to assess the antioxidant activity of xanthones isolated from marine sources, focusing on thermodynamics and kinetics within simulated physiological environments. DFT analysis revealed that xanthones such as 1,4,7-trihydroxy-6-methylxanthone (1), 1,4,5-trihydroxy-2-methylxanthone (2), arthone C (3), 2,3,4,6,8-pentahydroxy-1-methylxanthone (4), sterigmatocystin (5), oxisterigmatocystin C (6), and oxisterigmatocystin D (7) favor the SPLET pathway in water and the FHT pathway in lipid environments. The kinetic study of these xanthones reacting with the hydroperoxyl radical (HOO•) was conducted using the formal hydrogen atom transfer (FHT) mechanism and the single electron transfer (SET) mechanism. The results showed that compounds 1-4 exhibited antioxidant activities in aqueous environments surpassing that of the reference compound Trolox, with rate constants ranging from 2.02 x 105 to 9.44 x 107 M-1·s-1. In lipid environments, compounds 1 and 2 also demonstrated higher rate constants than Trolox. Additionally, molecular docking and molecular dynamics analysis suggested that xanthones 1-7 potentially inhibit the pro-oxidant effect of the Keap1 enzyme, highlighting their promise as both antiradicals and enzyme inhibitors.
Collapse
Affiliation(s)
- Phan Thi Thuy
- Department of Chemistry, Vinh University, Vinh, Vietnam
| | - Nguyen Xuan Ha
- Graduate University of Science and Technology, VAST, Hanoi, Vietnam
| |
Collapse
|
19
|
Nguyen CD, Yoo J, Jeong SJ, Ha HA, Yang JH, Lee G, Shin JC, Kim JH. Melittin - the main component of bee venom: a promising therapeutic agent for neuroprotection through keap1/Nrf2/HO-1 pathway activation. Chin Med 2024; 19:166. [PMID: 39605070 PMCID: PMC11603938 DOI: 10.1186/s13020-024-01020-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/07/2024] [Indexed: 11/29/2024] Open
Abstract
The Nuclear factor erythroid 2-related factor (Nrf2)/ Heme oxygenase-1 (HO-1) pathway, known for its significant role in regulating innate antioxidant defense mechanisms, is increasingly being recognized for its potential in neuroprotection studies. Derived from bee venom, melittin's neuroprotective effects have raised interest. This study confirmed that melittin specificity upregulated the weakened Nrf2/HO-1 signaling in mice brain. Interestingly, we also revealed melittin's efficient tactic, as it was suggested to first restore redox balance and then gradually stabilized other regulations of the mouse hippocampus. Using a neuro-stress-induced scopolamine model, chromatography and mass spectrometry analysis revealed that melittin crossed the compromised blood-brain barrier and accumulated in the hippocampus, which provided the chance to interact directly to weakened neurons. A wide range of improvements of melittin action were observed from various tests from behavior Morris water maze, Y maze test to immune florescent staining, western blots. As we need to find out what is the focus of melittin action, we conducted a careful observation in mice which showed that: the first signs of changes, in the hippocampus, within 5 h after melittin administration were the restoration of the Nrf2/HO-1 system and suppression of oxidative stress. After this event, from 7 to 12.5 h after administration, a diversity of conditions was all ameliorated: inflammation, apoptosis, neurotrophic factors, cholinergic function, and tissue ATP level. This chain reaction underscores that melittin focus was on redox balance's role, which revived multiple neuronal functions. Evidence of enhancement in the mouse hippocampus led to further exploration with hippocampal cell line HT22 model. Immunofluorescence analysis showed melittin-induced Nrf2 translocation to the nucleus, which would initiating the translation of antioxidant genes like HO-1. Pathway inhibitors pinpointed melittin's direct influence on the Nrf2/HO-1 pathway. 3D docking models and pull-down assays suggested melittin's direct interaction with Keap1, the regulator of the Nrf2/HO-1 pathway. Overall, this study not only highlighted melittin specifically effect on Nrf2/HO-1, thus rebalancing cellular redox, and also showed that this is an effective multi-faceted therapeutic strategy against neurodegeneration.
Collapse
Affiliation(s)
- Cong Duc Nguyen
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Jaehee Yoo
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Sang Jun Jeong
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Hai-Anh Ha
- Faculty of Pharmacy, College of Medicine and Pharmacy, Duy Tan University, Da Nang, 550000, Vietnam
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Gihyun Lee
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Jeong Cheol Shin
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea.
- Department of Acupuncture and Moxibustion Medicine, Dongshin University Gwangju Korean Medicine Hospital, 141, Wolsan-ro, Nam-gu, Gwangju City 61619, Republic of Korea , 141 Wolsan-Ro Nam-Gu, Gwangju, 61619, Republic of Korea.
| | - Jae-Hong Kim
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea.
- Department of Acupuncture and Moxibustion Medicine, Dongshin University Gwangju Korean Medicine Hospital, 141, Wolsan-ro, Nam-gu, Gwangju City 61619, Republic of Korea , 141 Wolsan-Ro Nam-Gu, Gwangju, 61619, Republic of Korea.
| |
Collapse
|
20
|
Hijam AC, Tongbram YC, Nongthombam PD, Meitei HN, Koijam AS, Rajashekar Y, Haobam R. Traditionally used edible medicinal plants protect against rotenone induced toxicity in SH-SY5Y cells-a prospect for the development of herbal nutraceuticals. Neurochem Int 2024; 180:105855. [PMID: 39244037 DOI: 10.1016/j.neuint.2024.105855] [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: 06/30/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Plants are good sources of pharmacologically active compounds. The present study aimed to examine the neuroprotective potentials of the methanol extracts of Salix tetrasperma Roxb. leaf (STME) and Plantago asiatica L. (PAME), two edibles medicinal plants of Manipur, India against neurotoxicity induced by rotenone in SH-SY5Y cells. Free radical quenching activities were evaluated by ABTS and DPPH assays. The cytotoxicity of rotenone and the neuronal survival were assessed by MTT assay and MAP2 expression analysis. DCF-DA, Rhodamine 123 (Rh-123), and DAPI measured the intracellular reactive oxygen species (ROS) levels, mitochondrial membrane potential (MMP), and apoptotic nuclei, respectively. Superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT) activities were also assessed. LC-QTOF-MS analysis was performed for the identification of the compounds present in STME and PAME. The study showed that both the plant extracts (STME and PAME) showed antioxidant and neuroprotective capabilities in rotenone-induced neurotoxicity by preventing oxidative stress through the reduction of intracellular ROS levels and reversing the activities of GPx, SOD, and CAT caused by rotenone. Further, both plants prevented apoptotic cell death by normalizing the steady state of MMP and protecting nuclear DNA condensation. LC-QTOF-MS analysis shows the presence of known neuroprotective compounds like uridine and gabapentin in STME and PAME respectively. The two plants might be an important source of natural antioxidants and nutraceuticals with neuroprotective abilities. This could be investigated further to formulate herbal nutraceuticals for the treatment of neurodegenerative disease like Parkinson's disease.
Collapse
Affiliation(s)
- Aruna Chanu Hijam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India
| | | | - Pooja Devi Nongthombam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India
| | | | - Arunkumar Singh Koijam
- Insect Bioresources Laboratory, Animal Bioresources Programme, Institute of Bioresources & Sustainable Development, Department of Biotechnology, Govt. of India, Takyelpat, Imphal, 795001, Manipur, India
| | - Yallapa Rajashekar
- Insect Bioresources Laboratory, Animal Bioresources Programme, Institute of Bioresources & Sustainable Development, Department of Biotechnology, Govt. of India, Takyelpat, Imphal, 795001, Manipur, India
| | - Reena Haobam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India.
| |
Collapse
|
21
|
Chi F, Cheng C, Zhang M, Su B, Hou Y, Bai G. Resveratrol targeting NRF2 disrupts the binding between KEAP1 and NRF2-DLG motif to ameliorate oxidative stress damage in mice pulmonary infection. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118353. [PMID: 38762209 DOI: 10.1016/j.jep.2024.118353] [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: 03/27/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The root of Polygonum cuspidatum Sieb. et Zucc (PC), known as 'Huzhang' in the Chinese Pharmacopoeia, has been traditionally employed for its anti-inflammatory, antiviral, antimicrobial, and other biological activities. Polydatin (PD) and its aglycone, resveratrol (RES), are key pharmacologically active components responsible for exerting anti-inflammatory and antioxidant effects. However, its specific targets and action mechanisms remain unclear. AIM OF THE STUDY The equilibrium of the KEAP1-NRF2 system serves as the primary protective response to oxidative and electrophilic stresses within the body, particularly in cases of acute lung injury caused by pathogenic microbial infection. In this study, the precise mechanisms by which RES alleviates oxidative stress damage in conjunction with NRF2 activators are discussed. MATERIALS AND METHODS The active components from PC were screened to evaluate their potential to inhibit reactive oxygen species (ROS) and activate antioxidant activity dependent on antioxidant response elements (ARE). RES was evaluated for its potential to alleviate the oxidative stress caused by pathogenic microbial infection. Functional probes were designed to study the RES distribution and identify its targets. A lipopolysaccharide (LPS)-induced oxidative injury model was used to evaluate the effects of RES on the KEAP1-NRF2/ARE pathway in RAW 264.7 cells. The interaction between RES and NRF2 was elucidated using drug-affinity responsive target stability (DARTS), cellular thermal shift assays (CETSA), co-immunoprecipitation (Co-IP), and microscale thermophoresis (MST) techniques. The key binding sites were predicted using molecular docking and validated in NRF2-knockdownand reconstructed cells. Finally, protective effects against pulmonary stress were verified in a mouse model of pathogenic infection. RESULTS The accumulation of RES in lung macrophages disrupted the binding between KEAP1 and NRF2, thereby preventing the ubiquitination degradation of NRF2 through its interaction with Ile28 on the NRF2-DLG motif. The activation of NRF2 resulted in the upregulation of nuclear transcription, enhances the expression of antioxidant genes dependent on ARE, suppresses ROS generation, and ameliorates oxidative damage both in vivo and in vitro. CONCLUSION These findings shed light on the potential of RES to mitigate oxidative stress damage caused by pathogenic microorganism-induced lung infections and facilitate the discovery of novel small molecule modulators targeting the KEAP1-NRF2 DLG motif interaction.
Collapse
Affiliation(s)
- Fuyun Chi
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China
| | - Chuanjing Cheng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China
| | - Man Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China
| | - Bo Su
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China.
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China.
| |
Collapse
|
22
|
Abu-Elfotuh K, Abbas AN, Najm MAA, Qasim QA, Hamdan AME, Abdelrehim AB, Gowifel AMH, Al-Najjar AH, Atwa AM, Kozman MR, Khalil AS, Negm AM, Mousa SNM, Hamdan AM, Abd El-Rhman RH, Abdelmohsen SR, Tolba AMA, Aboelsoud HA, Salahuddin A, Darwish A. Neuroprotective effects of punicalagin and/or micronized zeolite clinoptilolite on manganese-induced Parkinson's disease in a rat model: Involvement of multiple pathways. CNS Neurosci Ther 2024; 30:e70008. [PMID: 39374157 PMCID: PMC11457879 DOI: 10.1111/cns.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/27/2024] [Accepted: 08/06/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Manganism, a central nervous system dysfunction correlated with neurological deficits such as Parkinsonism, is caused by the substantial collection of manganese chloride (MnCl2) in the brain. OBJECTIVES To explore the neuroprotective effects of natural compounds, namely, micronized zeolite clinoptilolite (ZC) and punicalagin (PUN), either individually or in combination, against MnCl2-induced Parkinson's disease (PD). METHODS Fifty male albino rats were divided into 5 groups (Gps). Gp I was used as the control group, and the remaining animals received MnCl2 (Gp II-Gp V). Rats in Gps III and IV were treated with ZC and PUN, respectively. Gp V received both ZC and PUN as previously reported for the solo-treated plants. RESULTS ZC and/or PUN reversed the depletion of monoamines in the brain and decreased acetyl choline esterase activity, which primarily adjusted the animals' behavior and motor coordination. ZC and PUN restored the balance between glutamate/γ-amino butyric acid content and markedly improved the brain levels of brain-derived neurotrophic factor and nuclear factor erythroid 2-related factor 2/heme oxygenase-1 and decreased glycogen synthase kinase-3 beta activity. ZC and PUN also inhibited inflammatory and oxidative markers, including nuclear factor kappa-light-chain-enhancer of activated B cells, Toll-like receptor 4, nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 and caspase-1. Bcl-2-associated X-protein and B-cell leukemia/lymphoma 2 protein (Bcl-2) can significantly modify caspase-3 expression. ZC and/or PUN ameliorated PD in rats by decreasing the levels of endoplasmic reticulum (ER) stress markers (p-protein kinase-like ER kinase (PERK), glucose-regulated protein 78, and C/EBP homologous protein (CHOP)) and enhancing the levels of an autophagy marker (Beclin-1). DISCUSSION AND CONCLUSION ZC and/or PUN mitigated the progression of PD through their potential neurotrophic, neurogenic, anti-inflammatory, antioxidant, and anti-apoptotic activities and by controlling ER stress through modulation of the PERK/CHOP/Bcl-2 pathway.
Collapse
Affiliation(s)
- Karema Abu-Elfotuh
- Clinical Pharmacy Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
- Al-Ayen Iraqi University, Thi-Qar, Iraq
| | - Ashwaq N Abbas
- College of Dentistry, University of Sulaimanyia, Kurdistan, Iraq
| | - Mazin A A Najm
- Department of Pharmacy, Mazaya University College, Thi-Qar, Alnasiriya, Iraq
| | - Qutaiba A Qasim
- Department of Clinical Laboratory Sciences, College of Pharmacy, Al-Ayen Iraqi University, Thi-Qar, Iraq
- Department of Clinical Laboratory Sciences, College of Pharmacy, University of Basrah, Basrah, Iraq
| | - Ahmed M E Hamdan
- Faculty of Pharmacy, Department of Pharmacy Practice, University of Tabuk, Tabuk, Saudi Arabia
| | - Amany B Abdelrehim
- Biochemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Ayah M H Gowifel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Aya H Al-Najjar
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Ahmed M Atwa
- Al-Ayen Iraqi University, Thi-Qar, Iraq
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Magy R Kozman
- Clinical Pharmacy Department, Faculty of Pharmacy, Misr University for Science and Technology, Giza, Egypt
| | - Azza S Khalil
- Physiology Department, Faculty of Medicine (Girls), Al-Azhar University, Cairo, Egypt
| | - Amira M Negm
- Physiology Department, Faculty of Medicine (Girls), Al-Azhar University, Cairo, Egypt
| | | | - Amira M Hamdan
- Oceanography Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Rana H Abd El-Rhman
- Department of pharmacology & Toxicology, Faculty of Pharmacy, Sinai University - Kantara Branch, Ismailia, Egypt
| | - Shaimaa R Abdelmohsen
- Anatomy and Embryology Department, Faculty of Medicine (Girls), Al-Azhar University, Cairo, Egypt
| | - Amina M A Tolba
- Anatomy and Embryology Department, Faculty of Medicine (Girls), Al-Azhar University, Cairo, Egypt
| | - Heba Abdelnaser Aboelsoud
- Anatomy and Embryology Department, Faculty of Medicine (Girls), Al-Azhar University, Cairo, Egypt
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Ahmad Salahuddin
- Biochemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
- Department of Biochemistry, College of Pharmacy, Al-Ayen Iraqi University, Thi-Qar, Iraq
| | - Alshaymaa Darwish
- Biochemistry Department, Faculty of Pharmacy, Sohag University, Sohag, Egypt
| |
Collapse
|
23
|
Kim Y, Lim J, Oh J. Taming neuroinflammation in Alzheimer's disease: The protective role of phytochemicals through the gut-brain axis. Biomed Pharmacother 2024; 178:117277. [PMID: 39126772 DOI: 10.1016/j.biopha.2024.117277] [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: 05/24/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive degenerative neurological condition characterized by cognitive decline, primarily affecting memory and logical thinking, attributed to amyloid-β plaques and tau protein tangles in the brain, leading to neuronal loss and brain atrophy. Neuroinflammation, a hallmark of AD, involves the activation of microglia and astrocytes in response to pathological changes, potentially exacerbating neuronal damage. The gut-brain axis is a bidirectional communication pathway between the gastrointestinal and central nervous systems, crucial for maintaining brain health. Phytochemicals, natural compounds found in plants with antioxidant and anti-inflammatory properties, such as flavonoids, curcumin, resveratrol, and quercetin, have emerged as potential modulators of this axis, suggesting implications for AD prevention. Intake of phytochemicals influences the gut microbial composition and its metabolites, thereby impacting neuroinflammation and oxidative stress in the brain. Consumption of phytochemical-rich foods may promote a healthy gut microbiota, fostering the production of anti-inflammatory and neuroprotective substances. Early dietary incorporation of phytochemicals offers a non-invasive strategy for modulating the gut-brain axis and potentially reducing AD risk or delaying its onset. The exploration of interventions targeting the gut-brain axis through phytochemical intake represents a promising avenue for the development of preventive or therapeutic strategies against AD initiation and progression.
Collapse
Affiliation(s)
- Yoonsu Kim
- Department of Integrative Biology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jinkyu Lim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea.
| | - Jisun Oh
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea.
| |
Collapse
|
24
|
Chencen L, Shuo Z, Zhiyu C, Xiaoyu F, Min Z, Pengjiao W, Xiuli G. (+)-catechin protects PC12 cells against CORT-induced oxidative stress and pyroptosis through the pathways of PI3K/AKT and Nrf2/HO-1/NF-κB. Front Pharmacol 2024; 15:1450211. [PMID: 39263574 PMCID: PMC11387166 DOI: 10.3389/fphar.2024.1450211] [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: 06/17/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024] Open
Abstract
Pyroptosis induced by oxidative stress is a significant contributor to mental health disorders, including depression (+)-Catechin (CA), a polyphenolic compound prevalent in various food sources, has been substantiated by prior research to exhibit potent antioxidant properties and potential antidepressant effects. Nonetheless, the precise antidepressive mechanisms and effects of CA remain incompletely elucidated. In this study, we employed corticosterone (CORT) and PC12 cells to develop a cellular model of depression, aiming to investigate the protective effects of CA against CORT-induced cellular damage. Our objective was to elucidate the underlying mechanisms of protective action. We utilized transcriptomic analysis to identify differentially expressed genes and employed bioinformatics approaches to predict the potential mechanisms of CA's protective effects in PC12 cells. These transcriptomic predictions were subsequently validated through western blot analysis. The findings indicated that CA possesses the capacity to mitigate oxidative stress and suppress pyroptosis in PC12 cells via the activation of the PI3K/AKT signaling pathway. This activation subsequently modulates the Nrf2/HO1/NF-κB pathways, thereby providing protection to PC12 cells against damage induced by CORT. Furthermore, we investigated the interaction between CA and the Keap1 protein employing molecular docking and protein thermal shift assays. We propose that CA can activate Nrf2 through two mechanisms to decrease reactive oxygen species (ROS) levels and inhibit pyroptosis: one mechanism involves the activation of the PI3K/AKT signaling pathway, and the other involves direct binding to Keap1, leading to an increase in p-Nrf2.
Collapse
Affiliation(s)
- Lai Chencen
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
- Department of Nosocomial Infection, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zhang Shuo
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Experimental Animal Center of Guizhou Medical University, Guiyang, China
| | - Chen Zhiyu
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| | - Fu Xiaoyu
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| | - Zhang Min
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| | - Wang Pengjiao
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guizhou Medical University, Guiyang, China
| | - Gao Xiuli
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| |
Collapse
|
25
|
Liu H, Zhang Y, Hou X, Zhu C, Yang Q, Li K, Fan L, Zhang X, Jiang X, Jin X, Lei H, Chen T, Zhang F, Zhang Z, Song J. CRHR1 antagonist alleviated depression-like behavior by downregulating p62 in a rat model of post-stroke depression. Exp Neurol 2024; 378:114822. [PMID: 38823676 DOI: 10.1016/j.expneurol.2024.114822] [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: 12/14/2023] [Revised: 05/08/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
Post-stroke depression (PSD) is a complication of cerebrovascular disease, which can increase mortality after stroke. CRH is one of the main signaling peptides released after activation of the hypothalamic-pituitary-adrenal (HPA) axis in response to stress. It affects synaptic plasticity by regulating inflammation, oxidative stress and autophagy in the central nervous system. And the loss of spines exacerbates depression-like behavior. Therefore, synaptic deficits induced by CRH may be related to post-stroke depression. However, the underlying mechanism remains unclear. The Keap1-Nrf2 complex is one of the core components of the antioxidant response. As an autophagy associated protein, p62 participates in the Keap1-NrF2 pathway through its Keap1 interaction domain. Oxidative stress is involved in the feedback regulation between Keap1-Nrf2 pathway and p62.However, whether the relationship between CRH and the Keap1-Nrf2-p62 pathway is involved in PSD remains unknown. This study found that serum levels of CRH in 22 patients with PSD were higher than those in healthy subjects. We used MCAO combined with CUMS single-cage SD rats to establish an animal model of PSD. Animal experiments showed that CRHR1 antagonist prevented synaptic loss in the hippocampus of PSD rats and alleviated depression-like behavior. CRH induced p62 accumulation in the prefrontal cortex of PSD rats through CRHR1. CRHR1 antagonist inhibited Keap1-Nrf2-p62 pathway by attenuating oxidative stress. In addition, we found that abnormal accumulation of p62 induces PSD. It alleviates depression-like behavior by inhibiting the expression of p62 and promoting the clearance of p62 in PSD rats. These findings can help explore the pathogenesis of PSD and design targeted treatments for PSD.
Collapse
Affiliation(s)
- Huanhuan Liu
- Henan Key Laboratory of Biological Psychiatry, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University
| | - Yunfei Zhang
- Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University; The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiaoli Hou
- General Hospital of Pingmei Shenma Group, Pingdingshan, Henan, China
| | - Chuanzhou Zhu
- Henan Key Laboratory of Biological Psychiatry, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University
| | - Qianling Yang
- Henan Key Laboratory of Biological Psychiatry, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University
| | - Kun Li
- Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University; The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Lifei Fan
- Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University; The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Xinyue Zhang
- Henan Key Laboratory of Biological Psychiatry, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University
| | - Xinhui Jiang
- The Third People's Hospital of Luoyang, Luoyang, Henan, China
| | - Xuejiao Jin
- Henan Key Laboratory of Biological Psychiatry, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University
| | - Hao Lei
- Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University; The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Tengfei Chen
- Henan Key Laboratory of Biological Psychiatry, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University
| | - Fuping Zhang
- Henan Key Laboratory of Biological Psychiatry, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University.
| | - Zhaohui Zhang
- Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University; The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.
| | - Jinggui Song
- Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University.
| |
Collapse
|
26
|
Singh AA, Yadav D, Khan F, Song M. Indole-3-Carbinol and Its Derivatives as Neuroprotective Modulators. Brain Sci 2024; 14:674. [PMID: 39061415 PMCID: PMC11274471 DOI: 10.3390/brainsci14070674] [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: 06/06/2024] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its downstream tropomyosin receptor kinase B (TrkB) signaling pathway play pivotal roles in the resilience and action of antidepressant drugs, making them prominent targets in psychiatric research. Oxidative stress (OS) contributes to various neurological disorders, including neurodegenerative diseases, stroke, and mental illnesses, and exacerbates the aging process. The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant responsive element (ARE) serves as the primary cellular defense mechanism against OS-induced brain damage. Thus, Nrf2 activation may confer endogenous neuroprotection against OS-related cellular damage; notably, the TrkB/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, stimulated by BDNF-dependent TrkB signaling, activates Nrf2 and promotes its nuclear translocation. However, insufficient neurotrophin support often leads to the downregulation of the TrkB signaling pathway in brain diseases. Thus, targeting TrkB activation and the Nrf2-ARE system is a promising therapeutic strategy for treating neurodegenerative diseases. Phytochemicals, including indole-3-carbinol (I3C) and its metabolite, diindolylmethane (DIM), exhibit neuroprotective effects through BDNF's mimetic activity; Akt phosphorylation is induced, and the antioxidant defense mechanism is activated by blocking the Nrf2-kelch-like ECH-associated protein 1 (Keap1) complex. This review emphasizes the therapeutic potential of I3C and its derivatives for concurrently activating neuronal defense mechanisms in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Dhananjay Yadav
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Fazlurrahman Khan
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea;
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| |
Collapse
|
27
|
Gonçalves M, Vale N, Silva P. Neuroprotective Effects of Olive Oil: A Comprehensive Review of Antioxidant Properties. Antioxidants (Basel) 2024; 13:762. [PMID: 39061831 PMCID: PMC11274152 DOI: 10.3390/antiox13070762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 06/15/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Neurodegenerative diseases are a significant challenge to global healthcare, and oxidative stress plays a crucial role in their development. This paper presents a comprehensive analysis of the neuroprotective potential of olive oil, with a primary focus on its antioxidant properties. The chemical composition of olive oil, including key antioxidants, such as oleuropein, hydroxytyrosol, and oleocanthal, is systematically examined. The mechanisms by which these compounds provide neuroprotection, including counteracting oxidative damage and modulating neuroprotective pathways, are explored. The neuroprotective efficacy of olive oil is evaluated by synthesizing findings from various sources, including in vitro studies, animal models, and clinical trials. The integration of olive oil into dietary patterns, particularly its role in the Mediterranean diet, and its broader implications in neurodegenerative disease prevention are also discussed. The challenges in translating preclinical findings to clinical applications are acknowledged and future research directions are proposed to better understand the potential of olive oil in mitigating the risk of neurodegenerative conditions. This review highlights olive oil not only as a dietary component, but also as a promising candidate in preventive neurology, advocating for further investigation in the context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Marta Gonçalves
- Laboratory of Histology and Embryology, Department of Microscopy, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (U.Porto), Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Paula Silva
- Laboratory of Histology and Embryology, Department of Microscopy, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (U.Porto), Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
- iNOVA Media Lab, ICNOVA-NOVA Institute of Communication, NOVA School of Social Sciences and Humanities, Universidade NOVA de Lisboa, 1069-061 Lisbon, Portugal
| |
Collapse
|
28
|
Nyulas KI, Simon-Szabó Z, Pál S, Fodor MA, Dénes L, Cseh MJ, Barabás-Hajdu E, Csipor B, Szakács J, Preg Z, Germán-Salló M, Nemes-Nagy E. Cardiovascular Effects of Herbal Products and Their Interaction with Antihypertensive Drugs-Comprehensive Review. Int J Mol Sci 2024; 25:6388. [PMID: 38928095 PMCID: PMC11203894 DOI: 10.3390/ijms25126388] [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: 04/29/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Hypertension is a highly prevalent population-level disease that represents an important risk factor for several cardiovascular complications and occupies a leading position in mortality statistics. Antihypertensive therapy includes a wide variety of drugs. Additionally, the potential antihypertensive and cardioprotective effects of several phytotherapy products have been evaluated, as these could also be a valuable therapeutic option for the prevention, improvement or treatment of hypertension and its complications. The present review includes an evaluation of the cardioprotective and antihypertensive effects of garlic, Aloe vera, green tea, Ginkgo biloba, berberine, ginseng, Nigella sativa, Apium graveolens, thyme, cinnamon and ginger, and their possible interactions with antihypertensive drugs. A literature search was undertaken via the PubMed, Google Scholar, Embase and Cochrane databases. Research articles, systematic reviews and meta-analyses published between 2010 and 2023, in the English, Hungarian, and Romanian languages were selected.
Collapse
Affiliation(s)
- Kinga-Ilona Nyulas
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540139 Târgu Mureș, Romania
| | - Zsuzsánna Simon-Szabó
- Department of Pathophysiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540139 Târgu Mureș, Romania
| | - Sándor Pál
- Department of Laboratory Medicine, Department of Transfusion Medicine, Medical School, University of Pécs, 7622 Pécs, Hungary
| | - Márta-Andrea Fodor
- Department of Laboratory Medicine, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540139 Târgu Mureș, Romania
| | - Lóránd Dénes
- Department of Anatomy and Embryology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540139 Târgu Mureș, Romania
| | - Margit Judit Cseh
- Master Program of Nutrition and Dietetics, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540139 Târgu Mureș, Romania
| | - Enikő Barabás-Hajdu
- Department of Cell Biology and Microbiology, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540139 Târgu Mureș, Romania
| | - Bernadett Csipor
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540139 Târgu Mureș, Romania
| | - Juliánna Szakács
- Department of Biophysics, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540139 Târgu Mureș, Romania
| | - Zoltán Preg
- Department of Family Medicine, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540139 Târgu Mureș, Romania
| | - Márta Germán-Salló
- Department of Internal Medicine III, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540139 Târgu Mureș, Romania
| | - Enikő Nemes-Nagy
- Department of Chemistry and Medical Biochemistry, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540139 Târgu Mureș, Romania;
| |
Collapse
|
29
|
Makvand M, Mirtorabi SD, Campbell A, Zali A, Ahangari G. Exploring neuroadaptive cellular pathways in chronic morphine exposure: An in-vitro analysis of cabergoline and Mdivi-1 co-treatment effects on the autophagy-apoptosis axis. J Cell Biochem 2024; 125:e30558. [PMID: 38577900 DOI: 10.1002/jcb.30558] [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: 09/20/2023] [Revised: 02/08/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024]
Abstract
The complex impacts of prolonged morphine exposure continue to be a significant focus in the expanding area of addiction studies. This research investigates the effectiveness of a combined treatment using Cabergoline and Mdivi-1 to counteract the neuroadaptive changes caused by in vitro morphine treatment. The impact of Methadone, Cabergoline, and a combination of Cabergoline and Mdivi-1 on the cellular and molecular responses associated with Morphine-induced changes was studied in human Neuroblastoma (SK-N-MC) and Glioblastoma (U87-MG) cell lines that were exposed to prolong Morphine treatment. Cabergoline and Mdivi-1 combined treatment effectively influenced the molecular alterations associated with neuroadaptation in chronic morphine-exposed neural cells. This combination therapy normalized autophagy and reduced oxidative stress by enhancing total-antioxidant capacity, mitigating apoptosis, restoring BDNF expression, and balancing apoptotic elements. Our research outlines morphine's dual role in modulating mitochondrial dynamics via the dysregulation of the autophagy-apoptosis axis. This emphasizes the significant involvement of DRP1 activity in neurological adaptation processes, as well as disturbances in the dopaminergic pathway during in vitro chronic exposure to morphine in neural cells. This study proposes a novel approach by recommending the potential effectiveness of combining Cabergoline and Mdivi-1 to modulate the neuroadaptations caused by morphine. Additionally, we identified BDNF and PCNA in neural cells as potential neuroprotective markers for assessing the effectiveness of drugs against opioid toxicity, emphasizing the need for further validation. The study uncovers diverse effects observed in pretreated morphine glioblastoma cells under treatment with Cabergoline and methadone. This highlights the potential for new treatments in the DRD2 pathway and underscores the importance of investigating the interplay between autophagy and apoptosis to advance research in managing cancer-related pain. The study necessitates an in-depth investigation into the relationship between autophagy and apoptosis, with a specific emphasis on protein interactions and the dynamics of cell signaling.
Collapse
Affiliation(s)
- Mina Makvand
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | | | - Arezoo Campbell
- Department of Pharmaceutical Sciences, Western University of Health Sciences, Pomona, California, USA
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghasem Ahangari
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
30
|
Golmohammadi M, Meibodi SAA, Al-Hawary SIS, Gupta J, Sapaev IB, Najm MAA, Alwave M, Nazifi M, Rahmani M, Zamanian MY, Moriasi G. Neuroprotective effects of resveratrol on retinal ganglion cells in glaucoma in rodents: A narrative review. Animal Model Exp Med 2024; 7:195-207. [PMID: 38808561 PMCID: PMC11228121 DOI: 10.1002/ame2.12438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 05/30/2024] Open
Abstract
Glaucoma, an irreversible optic neuropathy, primarily affects retinal ganglion cells (RGC) and causes vision loss and blindness. The damage to RGCs in glaucoma occurs by various mechanisms, including elevated intraocular pressure, oxidative stress, inflammation, and other neurodegenerative processes. As the disease progresses, the loss of RGCs leads to vision loss. Therefore, protecting RGCs from damage and promoting their survival are important goals in managing glaucoma. In this regard, resveratrol (RES), a polyphenolic phytoalexin, exerts antioxidant effects and slows down the evolution and progression of glaucoma. The present review shows that RES plays a protective role in RGCs in cases of ischemic injury and hypoxia as well as in ErbB2 protein expression in the retina. Additionally, RES plays protective roles in RGCs by promoting cell growth, reducing apoptosis, and decreasing oxidative stress in H2O2-exposed RGCs. RES was also found to inhibit oxidative stress damage in RGCs and suppress the activation of mitogen-activated protein kinase signaling pathways. RES could alleviate retinal function impairment by suppressing the hypoxia-inducible factor-1 alpha/vascular endothelial growth factor and p38/p53 axes while stimulating the PI3K/Akt pathway. Therefore, RES might exert potential therapeutic effects for managing glaucoma by protecting RGCs from damage and promoting their survival.
Collapse
Affiliation(s)
- Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Ibrohim B Sapaev
- Tashkent Institute of Irrigation and Agricultural Mechanization Engineers, Tashkent, Uzbekistan
- New Uzbekistan University, Tashkent, Uzbekistan
| | - Mazin A A Najm
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Marim Alwave
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Mozhgan Nazifi
- Department of Neurology, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammadreza Rahmani
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Yasin Zamanian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Gervason Moriasi
- Department of Medical Biochemistry, School of Medicine, College of Health Sciences, Mount Kenya University, Thika, Kenya
| |
Collapse
|
31
|
Ullah I, Zhao L, Uddin S, Zhou Y, Wang X, Li H. Nicotine-mediated therapy for Parkinson's disease in transgenic Caenorhabditis elegans model. Front Aging Neurosci 2024; 16:1358141. [PMID: 38813528 PMCID: PMC11135287 DOI: 10.3389/fnagi.2024.1358141] [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: 12/19/2023] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Parkinson's disease resultant in the degeneration of Dopaminergic neurons and accumulation of α-synuclein in the substantia nigra pars compacta. The synthetic therapeutics for Parkinson's disease have moderate symptomatic benefits but cannot prevent or delay disease progression. In this study, nicotine was employed by using transgenic Caenorhabditis elegans Parkinson's disease models to minimize the Parkinson's disease symptoms. The results showed that the nicotine at 100, 150, and 200 μM doses reduced degeneration of Dopaminergic neurons caused by 6-hydroxydopamine (14, 33, and 40%), lowered the aggregative toxicity of α-synuclein by 53, 56, and 78%, respectively. The reduction in food-sensing behavioral disabilities of BZ555 was observed to be 18, 49, and 86%, respectively, with nicotine concentrations of 100 μM, 150 μM, and 200 μM. Additionally, nicotine was found to enhance Daf-16 nuclear translocation by 14, 31, and 49%, and dose-dependently increased SOD-3 expression by 10, 19, and 23%. In summary, the nicotine might a promising therapy option for Parkinson's disease.
Collapse
Affiliation(s)
- Inam Ullah
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Longhe Zhao
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Shahab Uddin
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yangtao Zhou
- Department of Neurology, Clinical Center for Parkinson's Disease, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xin Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Hongyu Li
- School of Life Sciences, Lanzhou University, Lanzhou, China
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
32
|
Ducza L, Gaál B. The Neglected Sibling: NLRP2 Inflammasome in the Nervous System. Aging Dis 2024; 15:1006-1028. [PMID: 38722788 PMCID: PMC11081174 DOI: 10.14336/ad.2023.0926-1] [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: 08/01/2023] [Accepted: 09/26/2023] [Indexed: 05/13/2024] Open
Abstract
While classical NOD-like receptor pyrin domain containing protein 1 (NLRP1) and NLRP3 inflammasomal proteins have been extensively investigated, the contribution of NLRP2 is still ill-defined in the nervous system. Given the putative significance of NLRP2 in orchestrating neuroinflammation, further inquiry is needed to gain a better understanding of its connectome, hence its specific targeting may hold a promising therapeutic implication. Therefore, bioinformatical approach for extracting information, specifically in the context of neuropathologies, is also undoubtedly preferred. To the best of our knowledge, there is no review study selectively targeting only NLRP2. Increasing, but still fragmentary evidence should encourage researchers to thoroughly investigate this inflammasome in various animal- and human models. Taken together, herein we aimed to review the current literature focusing on the role of NLRP2 inflammasome in the nervous system and more importantly, we provide an algorithm-based protein network of human NLRP2 for elucidating potentially valuable molecular partnerships that can be the beginning of a new discourse and future therapeutic considerations.
Collapse
Affiliation(s)
- László Ducza
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Hungary, Hungary
| | - Botond Gaál
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Hungary, Hungary
| |
Collapse
|
33
|
Orisakwe OE, Ikpeama EU, Orish CN, Ezejiofor AN, Okolo KO, Cirovic A, Cirovic A, Nwaogazie IL, Onoyima CS. Prosopis africana exerts neuroprotective activity against quaternary metal mixture-induced memory impairment mediated by oxido-inflammatory response via Nrf2 pathway. AIMS Neurosci 2024; 11:118-143. [PMID: 38988888 PMCID: PMC11230863 DOI: 10.3934/neuroscience.2024008] [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: 02/19/2024] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 07/12/2024] Open
Abstract
The beneficial effects of Prosopis africana (PA) on human health have been demonstrated; however, its protective effects against heavy metals (HM) are not yet understood. This study evaluated the potential neuroprotective effects of PA in the cerebral cortex and cerebellum. To accomplish this, we divided 35 albino Sprague Dawley rats into five groups. Group I did not receive either heavy metal mixture (HMM) or PA. Group II received a HMM of PbCl2 (20 mg/kg), CdCl2 (1.61 mg/kg), HgCl2 (0.40 mg/kg), and NaAsO3 (10 mg/kg) orally for a period of two months. Groups III, IV, and V received HMM along with PA at doses of 500, 1000, and 1500 mg/kg, respectively. PA caused decreased levels of HM accumulation in the cerebral cortex and cerebellum and improved performance in the Barnes maze and rotarod tests. PA significantly reduced levels of IL-6 and TNF-α. PA increased concentrations of SOD, CAT, GSH, and Hmox-1 and decreased the activities of AChE and Nrf2. In addition, levels of MDA and NO decreased in groups III, IV, and V, along with an increase in the number of live neurons. In conclusion, PA demonstrates a complex neuroprotective effect with the potential to alleviate various aspects of HM-induced neurotoxicity.
Collapse
Affiliation(s)
- Orish E Orisakwe
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Choba, Nigeria
- Advanced Research Centre, European University of Lefke, Lefke, Northern Cyprus, TR-10 Mersin, Turkey
| | - Evelyn Utomoibor Ikpeama
- World Bank Africa Centre of Excellence in Oilfield Chemicals Research (ACE-CEFOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Choba, Nigeria
| | - Chinna N Orish
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Port Harcourt, PMB, 5323 Port Harcourt, Choba, Nigeria
| | - Anthonet N Ezejiofor
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Choba, Nigeria
| | - Kenneth O Okolo
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Enugu State, University of Science & Technology, Nigeria
| | - Aleksandar Cirovic
- University of Belgrade, Faculty of Medicine, Institute of Anatomy, Belgrade, Serbia
| | - Ana Cirovic
- University of Belgrade, Faculty of Medicine, Institute of Anatomy, Belgrade, Serbia
| | - Ify L Nwaogazie
- World Bank Africa Centre of Excellence in Oilfield Chemicals Research (ACE-CEFOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Choba, Nigeria
| | - Chinekwu Samson Onoyima
- Dept. of Biochemistry, Faculty of Biological Sciences, University of Nigeria Nsukka, Enugu State, Nigeria
| |
Collapse
|
34
|
Affortit C, Coyat C, Saidia AR, Ceccato JC, Charif M, Sarzi E, Flamant F, Guyot R, Cazevieille C, Puel JL, Lenaers G, Wang J. The human OPA1 delTTAG mutation induces adult onset and progressive auditory neuropathy in mice. Cell Mol Life Sci 2024; 81:80. [PMID: 38334784 PMCID: PMC10858076 DOI: 10.1007/s00018-024-05115-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 02/10/2024]
Abstract
Dominant optic atrophy (DOA) is one of the most prevalent forms of hereditary optic neuropathies and is mainly caused by heterozygous variants in OPA1, encoding a mitochondrial dynamin-related large GTPase. The clinical spectrum of DOA has been extended to a wide variety of syndromic presentations, called DOAplus, including deafness as the main secondary symptom associated to vision impairment. To date, the pathophysiological mechanisms underlying the deafness in DOA remain unknown. To gain insights into the process leading to hearing impairment, we have analyzed the Opa1delTTAG mouse model that recapitulates the DOAplus syndrome through complementary approaches combining morpho-physiology, biochemistry, and cellular and molecular biology. We found that Opa1delTTAG mutation leads an adult-onset progressive auditory neuropathy in mice, as attested by the auditory brainstem response threshold shift over time. However, the mutant mice harbored larger otoacoustic emissions in comparison to wild-type littermates, whereas the endocochlear potential, which is a proxy for the functional state of the stria vascularis, was comparable between both genotypes. Ultrastructural examination of the mutant mice revealed a selective loss of sensory inner hair cells, together with a progressive degeneration of the axons and myelin sheaths of the afferent terminals of the spiral ganglion neurons, supporting an auditory neuropathy spectrum disorder (ANSD). Molecular assessment of cochlea demonstrated a reduction of Opa1 mRNA level by greater than 40%, supporting haploinsufficiency as the disease mechanism. In addition, we evidenced an early increase in Sirtuin 3 level and in Beclin1 activity, and subsequently an age-related mtDNA depletion, increased oxidative stress, mitophagy as well as an impaired autophagic flux. Together, these results support a novel role for OPA1 in the maintenance of inner hair cells and auditory neural structures, addressing new challenges for the exploration and treatment of OPA1-linked ANSD in patients.
Collapse
Affiliation(s)
- Corentin Affortit
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, UMR 1298, 80 Rue Augustin Fliche, 34295, Montpellier, France
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology, Head and Neck Surgery, University of Iowa, Iowa City, IA, 52242, USA
| | - Carolanne Coyat
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, UMR 1298, 80 Rue Augustin Fliche, 34295, Montpellier, France
| | - Anissa Rym Saidia
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, UMR 1298, 80 Rue Augustin Fliche, 34295, Montpellier, France
| | - Jean-Charles Ceccato
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, UMR 1298, 80 Rue Augustin Fliche, 34295, Montpellier, France
| | - Majida Charif
- Genetics, and Immuno-Cell Therapy Team, Mohamed First University, 60000, Oujda, Morocco
| | - Emmanuelle Sarzi
- Institut NeuroMyoGène, Pathophysiology and Genetics of Neuron and Muscle (INMG-PGNM) UCBL-CNRS UMR5261, Inserm U1315, Université Claude Bernard, Lyon I, Faculty of Medicine and Pharmacy, Lyon, France
| | - Frédéric Flamant
- Institut de Génomique Fonctionnelle de Lyon (IGFL), INRAE USC1370, CNRS (UMR5242), ENS Lyon, Lyon, France
| | - Romain Guyot
- Institut de Génomique Fonctionnelle de Lyon (IGFL), INRAE USC1370, CNRS (UMR5242), ENS Lyon, Lyon, France
| | - Chantal Cazevieille
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, UMR 1298, 80 Rue Augustin Fliche, 34295, Montpellier, France
| | - Jean-Luc Puel
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, UMR 1298, 80 Rue Augustin Fliche, 34295, Montpellier, France
| | - Guy Lenaers
- Université Angers, MitoLab Team, Unité MitoVasc, UMR CNRS 6015, INSERM U1083, SFR ICAT, Angers, France
- Service de Neurologie, CHU d'Angers, Angers, France
| | - Jing Wang
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, UMR 1298, 80 Rue Augustin Fliche, 34295, Montpellier, France.
- Department of ENT and Head and Neck Surgery, University Hospital of Montpellier, Montpellier, France.
| |
Collapse
|
35
|
Singh L, Bhatti R. Signaling Pathways Involved in the Neuroprotective Effect of Osthole: Evidence and Mechanisms. Mol Neurobiol 2024; 61:1100-1118. [PMID: 37682453 DOI: 10.1007/s12035-023-03580-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/14/2023] [Indexed: 09/09/2023]
Abstract
Neurodegenerative diseases constitute a major threat to human health and are usually accompanied by progressive structural and functional loss of neurons. Abnormalities in synaptic plasticity are involved in neurodegenerative disorders. Aberrant cell signaling cascades play a predominant role in the initiation, progress as well as in the severity of these ailments. Notch signaling is a pivotal role in the maintenance of neural stem cells and also participates in neurogenesis. PI3k/Akt cascade regulates different biological processes including cell proliferation, apoptosis, and metabolism. It regulates neurotoxicity and mediates the survival of neurons. Moreover, the activated BDNF/TrkB cascade is involved in promoting the transcription of genes responsible for cell survival and neurogenesis. Despite significant progress made in delineating the underlying pathological mechanisms involved and derangements in cellular metabolic promenades implicated in these diseases, satisfactory strategies for the clinical management of these ailments are yet to be achieved. Therefore, the molecules targeting these cell signaling cascades may emerge as useful leads in developing newer management strategies. Osthole is an important ingredient of traditional Chinese medicinal plants, often found in various plants of the Apiaceae family and has been observed to target these aforementioned mediators. Until now, no review has been aimed to discuss the possible molecular signaling cascades involved in osthole-mediated neuroprotection at one platform. The current review aimed to explore the interplay of various mediators and the modulation of the different molecular signaling cascades in osthole-mediated neuroprotection. This review could open new insights into research involving diseases of neuronal origin, especially the effect on neurodegeneration, neurogenesis, and synaptic plasticity. The articles gathered to compose the current review were extracted by using the PubMed, Scopus, Science Direct, and Web of Science databases. A methodical approach was used to integrate and discuss all published original reports describing the modulation of different mediators by osthole to confer neuroprotection at one platform to provide possible molecular pathways. Based on the inclusion and exclusion criteria, 32 articles were included in the systematic review. Moreover, literature evidence was also used to construct the biosynthetic pathway of osthole. The current review reveals that osthole promotes neurogenesis and neuronal functioning via stimulation of Notch, BDNF/Trk, and P13k/Akt signaling pathways. It upregulates the expression of various proteins, such as BDNF, TrkB, CREB, Nrf-2, P13k, and Akt. Activation of Wnt by osthole, in turn, regulates downstream GSK-1β to inhibit tau phosphorylation and β-catenin degradation to prevent neuronal apoptosis. The activation of Wnt and inhibition of oxidative stress, Aβ, and GSK-3β mediated β-catenin degradation by osthole might also be involved in mediating the protection against neurodegenerative diseases. Furthermore, it also inhibits neuroinflammation by suppressing MAPK/NF-κB-mediated transcription of genes involved in the generation of inflammatory cytokines and NLRP-3 inflammasomes. This review delineates the various underlying signaling pathways involved in mediating the neuroprotective effect of osthole. Modulation of Notch, BDNF/Trk, MAPK/NF-κB, and P13k/Akt signaling pathways by osthole confers protection against neurodegenerative diseases. The preclinical effects of osthole suggest that it could be a valuable molecule in inspiring the development of new drugs for the management of neurodegenerative diseases and demands clinical studies to explore its potential. An effort has been made to unify the varied mechanisms and target sites involved in the neuroprotective effect of osthole. The comprehensive description of the molecular pathways in the present work reflects its originality and thoroughness. The reviewed literature findings may be extrapolated to suggest the role of othole as a "biological response modifier" which contributes to neuroprotection through kinase modulatory, immunomodulatory, and anti-oxidative activity, which is documented even at lower doses. The current review attempts to emphasize the gaps in the existing literature which can be explored in the future.
Collapse
Affiliation(s)
- Lovedeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
- University Institute of Pharma Sciences, Chandigarh University, Mohali, 140413, Punjab, India.
| | - Rajbir Bhatti
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| |
Collapse
|
36
|
Can B, Sanlier N. Alzheimer, Parkinson, dementia, and phytochemicals: insight review. Crit Rev Food Sci Nutr 2024; 65:1706-1728. [PMID: 38189347 DOI: 10.1080/10408398.2023.2299340] [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: 01/09/2024]
Abstract
Alzheimer's, Parkinson's, and dementia are the leading neurodegenerative diseases that threaten the world with the aging population. Although the pathophysiology of each disease is unique, the steps to be taken to prevent diseases are similar. One of the changes that a person can make alone is to gain the habit of an antioxidant-rich diet. Phytochemicals known for their antioxidant properties have been reported to prevent neurodegenerative diseases in various studies. Phytochemicals with similar chemical structures are grouped. Accordingly, there are two main groups of phytochemicals, flavonoid and non-flavonoid. Various in vitro and in vivo studies on phytochemicals have proven neuroprotective effects by increasing cognitive function with their anti-inflammatory and antioxidant mechanisms. The purpose of this review is to summarize the in vitro and in vivo studies on phytochemicals with neuroprotective effects and to provide insight.
Collapse
Affiliation(s)
- Basak Can
- Nutrition and Dietetics, School of Health Sciences, Istanbul Gelisim University, Istanbul, Turkey
| | - Nevin Sanlier
- School of Health Sciences, Nutrition and Dietetics, Ankara Medipol University, Ankara, Turkey
| |
Collapse
|
37
|
Yap RS, Kumar J, Teoh SL. Potential Neuroprotective Role of Neurotrophin in Traumatic Brain Injury. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1189-1202. [PMID: 38279761 DOI: 10.2174/0118715273289222231219094225] [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: 10/25/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 01/28/2024]
Abstract
Traumatic brain injury (TBI) is a major global health issue that affects millions of people every year. It is caused by any form of external force, resulting in temporary or permanent impairments in the brain. The pathophysiological process following TBI usually involves excitotoxicity, mitochondrial dysfunction, oxidative stress, inflammation, ischemia, and apoptotic cell death. It is challenging to find treatment for TBI due to its heterogeneous nature, and no therapeutic interventions have been approved thus far. Neurotrophins may represent an alternative approach for TBI treatment because they influence various functional activities in the brain. The present review highlights recent studies on neurotrophins shown to possess neuroprotective roles in TBI. Neurotrophins, specifically brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) have demonstrated reduced neuronal death, alleviated neuroinflammatory responses and improved neurological functions following TBI via their immunomodulatory, anti-inflammatory and antioxidant properties. Further studies are required to ensure the efficacy and safety of neurotrophins to be used as TBI treatment in clinical settings.
Collapse
Affiliation(s)
- Rei Shian Yap
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Sakai Y, Egawa D, Hattori J, Morikawa Y, Suenami K, Takayama T, Nagai A, Michiue T, Ikari A, Matsunaga T. α-Pyrrolidinononanophenone derivatives induce differentiated SH-SY5Y neuroblastoma cell apoptosis via reduction of antioxidant capacity: Involvement of NO depletion and inactivation of Nrf2/HO1 signaling pathway. Neurotoxicology 2024; 100:3-15. [PMID: 38040126 DOI: 10.1016/j.neuro.2023.11.010] [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: 09/06/2023] [Revised: 11/12/2023] [Accepted: 11/25/2023] [Indexed: 12/03/2023]
Abstract
α-Pyrrolidinononanophenone (α-PNP) derivatives are known to be one of the hazardous new psychoactive substances due to the most extended hydrocarbon chains of any pyrrolidinophenones on the illicit drug market. Our previous report showed that 4'-iodo-α-PNP (I-α-PNP) is the most potent cytotoxic compound among α-PNP derivatives and induces apoptosis due to mitochondrial dysfunction and suppression of nitric oxide (NO) production in differentiated human neuronal SH-SY5Y cells. In this study, to clarify the detailed action mechanisms by I-α-PNP, we investigated the mechanism of reactive oxygen species (ROS) -dependent apoptosis by I-α-PNP in differentiated SH-SY5Y with a focus on the antioxidant activities. Treatment with I-α-PNP elicits overproduction of ROS such as H2O2, hydroxyl radical, and 4-hydroxy-2-nonenal, and pretreatment with antioxidant N-acetyl-L-cysteine is attenuated the SH-SY5Y cells apoptosis by I-α-PNP. These results suggested that the overproduction of ROS is related to SH-SY5Y cell apoptosis by I-α-PNP. In addition, I-α-PNP markedly decreased antioxidant capacity in differentiated cells than in undifferentiated cells and inhibited the upregulation of hemeoxygenase 1 (HO1) and glutathione peroxidase 4 (GPX4) expression caused by induction of differentiation. Furthermore, the treatment with I-α-PNP increased the nuclear expression level of BTB Domain And CNC Homolog 1 (Bach1), a transcriptional repressor of Nrf2, only in differentiated cells, suggesting that the marked decrease in antioxidant capacity in differentiated cells was due to suppression of Nrf2/HO1 signaling by Bach1. Additionally, pretreatment with an NO donor suppresses the I-α-PNP-evoked ROS overproduction, HO1 down-regulation, increased nuclear Bach1 expression and reduced antioxidant activity in the differentiated cells. These findings suggest that the ROS-dependent apoptosis by I-α-PNP in differentiated cells is attributed to the inactivation of the Nrf2/HO1 signaling pathway triggered by NO depletion.
Collapse
Affiliation(s)
- Yuji Sakai
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu 500-8501, Japan.
| | - Daisuke Egawa
- Laboratory of Bioinformatics, Gifu Pharmaceutical University, Gifu 502-8585, Japan
| | - Junta Hattori
- Laboratory of Bioinformatics, Gifu Pharmaceutical University, Gifu 502-8585, Japan
| | - Yoshifumi Morikawa
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu 500-8501, Japan
| | - Koichi Suenami
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu 500-8501, Japan
| | - Tomohiro Takayama
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu 500-8501, Japan
| | - Atsushi Nagai
- Department of Legal Medicine, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | - Tomomi Michiue
- Department of Legal Medicine, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Toshiyuki Matsunaga
- Laboratory of Bioinformatics, Gifu Pharmaceutical University, Gifu 502-8585, Japan
| |
Collapse
|
39
|
Buemann B. Does activation of oxytocinergic reward circuits postpone the decline of the aging brain? Front Psychol 2023; 14:1250745. [PMID: 38222845 PMCID: PMC10786160 DOI: 10.3389/fpsyg.2023.1250745] [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: 06/30/2023] [Accepted: 10/20/2023] [Indexed: 01/16/2024] Open
Abstract
Oxytocin supports reproduction by promoting sexual- and nursing behavior. Moreover, it stimulates reproductive organs by different avenues. Oxytocin is released to the blood from terminals of oxytocinergic neurons which project from the hypothalamus to the pituitary gland. Concomitantly, the dendrites of these neurons discharge oxytocin into neighboring areas of the hypothalamus. At this location it affects other neuroendocrine systems by autocrine and paracrine mechanisms. Moreover, sensory processing, affective functions, and reward circuits are influenced by oxytocinergic neurons that reach different sites in the brain. In addition to its facilitating impact on various aspects of reproduction, oxytocin is revealed to possess significant anti-inflammatory, restoring, and tranquilizing properties. This has been demonstrated both in many in-vivo and in-vitro studies. The oxytocin system may therefore have the capacity to alleviate detrimental physiological- and mental stress reactions. Thus, high levels of endogenous oxytocin may counteract inadequate inflammation and malfunctioning of neurons and supportive cells in the brain. A persistent low-grade inflammation increasing with age-referred to as inflammaging-may lead to a cognitive decline but may also predispose to neurodegenerative diseases such as Alzheimer's and Parkinson. Interestingly, animal studies indicate that age-related destructive processes in the body can be postponed by techniques that preserve immune- and stem cell functions in the hypothalamus. It is argued in this article that sexual activity-by its stimulating impact on the oxytocinergic activity in many regions of the brain-has the capacity to delay the onset of age-related cerebral decay. This may also postpone frailty and age-associated diseases in the body. Finally, oxytocin possesses neuroplastic properties that may be applied to expand sexual reward. The release of oxytocin may therefore be further potentiated by learning processes that involves oxytocin itself. It may therefore be profitable to raise the consciousness about the potential health benefits of sexual activity particularly among the seniors.
Collapse
|
40
|
Abu-Elfotuh K, Darwish A, Elsanhory HMA, Alharthi HH, Hamdan AME, Hamdan AM, Masoud RAE, Abd El-Rhman RH, Reda E. In silico and in vivo analysis of the relationship between ADHD and social isolation in pups rat model: Implication of redox mechanisms, and the neuroprotective impact of Punicalagin. Life Sci 2023; 335:122252. [PMID: 37935275 DOI: 10.1016/j.lfs.2023.122252] [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: 09/10/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Attention deficit hyperactivity disorder (ADHD) has high incidence rate among children which may be due to excessive monosodium glutamate (MSG) consumption and social isolation (SI). AIM We aimed to explore the relationships between MSG, SI, and ADHD development and to evaluate the neuroprotective potential of Punicalagin (PUN). METHODS Eighty male rat pups randomly distributed into eight groups. Group I is the control, and Group II is socially engaged rats treated with PUN. Groups III to VII were exposed to ADHD-inducing factors: Group III to SI, Group IV to MSG, and Group V to both SI and MSG. Furthermore, Groups VI to VIII were the same Groups III to V but additionally received PUN treatment. KEY FINDINGS Exposure to MSG and/or SI led to pronounced behavioral anomalies, histological changes and indicative of ADHD-like symptoms in rat pups which is accompanied by inhibition of the nuclear factor erythroid 2-related factor 2 (Nrf2)/Heme-oxygenase 1 (HO-1)/Glutathione (GSH) pathway, decline of the brain-derived neurotrophic factor (BDNF) expression and activation of the Toll-like receptor 4 (TLR4)/Nuclear factor kappa B (NF-kB)/NLR Family Pyrin Domain Containing 3 (NLRP3) pathway. This resulted in elevated inflammatory biomarker levels, neuronal apoptosis, and disrupted neurotransmitter equilibrium. Meanwhile, pretreatment with PUN protected against all the previous alterations. SIGNIFICANCE We established compelling associations between MSG consumption, SI, and ADHD progression. Moreover, we proved that PUN is a promising neuroprotective agent against all risk factors of ADHD.
Collapse
Affiliation(s)
- Karema Abu-Elfotuh
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt; Al-Ayen University, Thi-Qar, 64001, Iraq.
| | - Alshaymaa Darwish
- Biochemistry Department, Faculty of Pharmacy, Sohag university, Sohag, Egypt.
| | - Heba M A Elsanhory
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University - Kantara Branch, Ismailia 41636, Egypt.
| | | | - Ahmed M E Hamdan
- Pharmacy Practice Department, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia.
| | - Amira M Hamdan
- Oceanography Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt.
| | - Rehab Ali Elsayed Masoud
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine for girls, Al-Azhar University, Cairo, Egypt.
| | - Rana H Abd El-Rhman
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University - Kantara Branch, Ismailia 41636, Egypt.
| | - Enji Reda
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University - Kantara Branch, Ismailia 41636, Egypt.
| |
Collapse
|
41
|
Chan SC, Tung CW, Lin CW, Tung YS, Wu PM, Cheng PH, Chen CM, Yang SH. miR-196a provides antioxidative neuroprotection via USP15/Nrf2 regulation in Huntington's disease. Free Radic Biol Med 2023; 209:292-300. [PMID: 37907121 DOI: 10.1016/j.freeradbiomed.2023.10.407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/02/2023]
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disorder characterized by the accumulation of mutant Huntingtin protein (mHTT) and oxidative stress-induced neuronal damage. Based on previous reports, microRNA-196a (miR-196a) has emerged as a potential therapeutic target due to its neuroprotective effects in various neurodegenerative diseases. However, whether miR-196a functions through antioxidative effects is still unknown. In this study, we demonstrated that HD models, both in vitro and in vivo, exhibit elevated levels of reactive oxygen species (ROS) and increased neuronal death, and miR-196a mitigates ROS levels and reduces cell death in HD cells. Moreover, we elucidated that miR-196a facilitates the translocation of nuclear factor erythroid 2 (Nrf2) into the nucleus, enhancing the transcription of antioxidant genes, including heme oxygenase-1 (HO-1). We further identified ubiquitin-specific peptidase 15 (USP15), a direct target of miR-196a related to the Nrf2 pathway, and USP15 exacerbates mHTT aggregate formation while partially counteracting miR-196a-induced reductions in mHTT levels. Taken together, these findings shed light on the multifaceted role of miR-196a in HD, highlighting its potential as a therapeutic avenue for ameliorating oxidative stress and neurodegeneration in this debilitating disease.
Collapse
Affiliation(s)
- Siew Chin Chan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Chih-Wei Tung
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Chia-Wei Lin
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yun-Shiuan Tung
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Po-Min Wu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Pei-Hsun Cheng
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Shang-Hsun Yang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
42
|
Amirkhosravi L, khaksari M, Amiresmaili S, Sanjari M, Khorasani P, Hashemian M. Evaluating the neuroprotective effects of progesterone receptors on experimental traumatic brain injury: The PI3K/Akt pathway. Brain Behav 2023; 13:e3244. [PMID: 37661235 PMCID: PMC10636406 DOI: 10.1002/brb3.3244] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/11/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND Studies have confirmed the salutary effects of progesterone (P4) on traumatic brain injury (TBI). This study investigated the beneficial effects of P4 via its receptors on TBI, and also whether progesterone receptors (PRs) can modulate TBI through PI3K/Akt pathway. MATERIAL AND METHODS Marmarou method was utilized to induce diffuse TBI in ovariectomized rats. P4 (1.7 mg/kg) or the vehicle (oil) was administered 30 min after TBI induction. Moreover, RU486 (PR antagonist) and its vehicle (DMSO) were injected before TBI induction and P4 injection. Brain Evans blue content, brain water content (WC), various oxidative stress parameters, IL-1β levels, tumor necrosis factor-α (TNF-α), histopathological alterations, and also phosphorylated Akt (p-Akt) and PI3K expressions in the brain were assessed 24 h after TBI. The veterinary comma scale (VCS) was measured before and after TBI at different times. RESULTS The findings revealed that P4 caused an increase in VCS and a decrease in brain WC, oxidative stress, TNF-α and IL-1β levels. RU486 inhibited the beneficial effects of P4 on these indices. Moreover, RU486 prevented the reduction of brain edema, inflammation, and apoptosis caused by P4. Moreover, P4 following TBI increased the expression of PI3K/p-Akt protein in the brain. RU486 eliminated the effects of P4 on PI3K/p-Akt expression. CONCLUSION According to these findings, PRs are acting as critical mediators for the neuroprotective properties of P4 on oxidative stress, pro-inflammatory cytokine levels, and neurological outcomes. PRs also play an important role in regulating the PI3K/p-Akt expression and nongenomic function of P4.
Collapse
Affiliation(s)
- Ladan Amirkhosravi
- Endocrinology and Metabolism Research CenterInstitute of Basic and Clinical Physiology SciencesKerman University of Medical SciencesKermanIran
| | - Mohammad khaksari
- Physiology Research CenterInstitute of NeuropharmacologyKerman University of Medical SciencesKermanIran
| | | | - Mojgan Sanjari
- Endocrinology and Metabolism Research CenterInstitute of Basic and Clinical Physiology SciencesKerman University of Medical SciencesKermanIran
| | - Parisa Khorasani
- Department of Pathology, Pathology, and Stem Cells Research Center, Afzalipour Medical FacultyKerman University of Medical SciencesKermanIran
| | - Morteza Hashemian
- Neuroscience Research Center, Institute of NeuropharmacologyKerman University of Medical SciencesKermanIran
| |
Collapse
|
43
|
Javanbakht P, Yazdi FR, Taghizadeh F, Khadivi F, Hamidabadi HG, Kashani IR, Zarini D, Mojaverrostami S. Quercetin as a possible complementary therapy in multiple sclerosis: Anti-oxidative, anti-inflammatory and remyelination potential properties. Heliyon 2023; 9:e21741. [PMID: 37954351 PMCID: PMC10638059 DOI: 10.1016/j.heliyon.2023.e21741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023] Open
Abstract
Multiple sclerosis (MS) is a complex autoimmune disorder of the central nervous system (CNS) which causes various symptoms such as fatigue, dyscoordination weakness and visual weakness. The intricacy of the immune system and obscure etiology are the main reasons for the lack of a definite treatment for MS. Oxidative stress is one of the most important key factors in MS pathogenesis. It can enhance inflammation, neurodegeneration and autoimmune-mediated processes, which can lead to excessive demyelination and axonal disruption. Recently, promising effects of Quercetin as a non-pharmacological anti-oxidant therapy have been reported in preclinical studies of MS disease. In this review, we provide a compendium of preclinical and clinical studies that have investigated the effects of Quercetin on MS disease to evaluate its potential utility as a complementary therapy in MS. Quercetin treatment in MS disease not only protects the CNS against oxidative stress and neuroinflammation, but it also declines the demyelination process and promotes remyelination potential. The present study clarifies the reported knowledge on the beneficial effects of Quercetin against MS, with future implication as a neuroprotective complementary therapy.
Collapse
Affiliation(s)
- Parinaz Javanbakht
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzane Rezaei Yazdi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Taghizadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Khadivi
- Department of Anatomy, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hatef Ghasemi Hamidabadi
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Zarini
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Mitra S, Dash R, Nishan AA, Habiba SU, Moon IS. Brain modulation by the gut microbiota: From disease to therapy. J Adv Res 2023; 53:153-173. [PMID: 36496175 PMCID: PMC10658262 DOI: 10.1016/j.jare.2022.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The gut microbiota (GM) and brain are strongly associated, which significantly affects neuronal development and disorders. GM-derived metabolites modulate neuronal function and influence many cascades in age-related neurodegenerative disorders (NDDs). Because of the dual role of GM in neuroprotection and neurodegeneration, understanding the balance between beneficial and harmful bacteria is crucial for applying this approach to clinical therapies. AIM OF THE REVIEW This review briefly discusses the role of the gut-brain relationship in promoting brain and cognitive function. Although a healthy gut environment is helpful for brain function, gut dysbiosis can disrupt the brain's environment and create a vicious cycle of degenerative cascades. The ways in which the GM population can affect brain function and the development of neurodegeneration are also discussed. In the treatment and management of NDDs, the beneficial effects of methods targeting GM populations and their derivatives, including probiotics, prebiotics, and fecal microbial transplantation (FMT) are also highlighted. KEY SCIENTIFIC CONCEPT OF THE REVIEW In this review, we aimed to provide a deeper understanding of the mechanisms of the gut microbe-brain relationship and their twin roles in neurodegeneration progression and therapeutic applications. Here, we attempted to highlight the different pathways connecting the brain and gut, together with the role of GM in neuroprotection and neuronal development. Furthermore, potential roles of GM metabolites in the pathogenesis of brain disorders and in strategies for its treatment are also investigated. By analyzing existing in vitro, in vivo and clinical studies, this review attempts to identify new and promising therapeutic strategies for central nervous system (CNS) disorders. As the connection between the gut microbe-brain relationship and responses to NDD treatments is less studied, this review will provide new insights into the global mechanisms of GM modulation in disease progression, and identify potential future perspectives for developing new therapies to treat NDDs.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Amena Al Nishan
- Department of Medicine, Chittagong Medical College, Chittagong 4203, Bangladesh
| | - Sarmin Ummey Habiba
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea.
| |
Collapse
|
45
|
Pak ME, Li W. Neuroprotective Effects of Sparassis crispa Ethanol Extract through the AKT/NRF2 and ERK/CREB Pathway in Mouse Hippocampal Cells. J Fungi (Basel) 2023; 9:910. [PMID: 37755018 PMCID: PMC10532724 DOI: 10.3390/jof9090910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
Sparassis crispa, known as the "Cauliflower mushroom", is an edible medicinal fungus found in Asia, Europe, and North America. Its fruiting bodies contain active biological and pharmacological ingredients with antitumor and anti-inflammatory properties. In this study, we investigated the neuroprotective effect of various Sparassis crispa extract against glutamate-induced toxicity and oxidative stress in hippocampal HT22 cells. Cell viability and reactive oxygen species (ROS) analyses served to evaluate the neuroprotective effects of Sparassis crispa ethanol extract (SCE) and their fractions partitioned with ethyl acetate (EtOAc; SCE-E) and water (SCE-W) in HT22 cells. SCE and SCE-E treatment reduced glutamate-induced cell death and ROS generation. SCE-E reduced apoptosis and ROS levels by regulating anti-apoptotic proteins. Under glutamate treatment, SCE-E activated nuclear factor erythroid-derived 2-related factor 2 (Nrf2) and regulated extracellular signal-regulated kinase (ERK) and AKT signals at late stages. SCE-E increased the protein expression of cAMP response element binding (CREB), brain-derived neurotrophic factor (BDNF), and Kelch-like ECH-associated protein 1 (Keap1), and decreased the Nrf2 protein expression. Moreover, co-treatment of SCE-E and wortmannin did not activate Nrf2 expression. Thus, the neuroprotective effect of SCE-E is likely due to Nrf2 and CREB activation through AKT and ERK phosphorylation, which effectively suppress glutamate-induced oxidative stress in HT22 cells. Accordingly, a daily supplement of SCE-E could become a potential treatment for oxidative-stress-related neurological diseases.
Collapse
Affiliation(s)
| | - Wei Li
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| |
Collapse
|
46
|
Zhu K, Zhang W, Wu Q, Yang Q, Gong Z, Shao S, Zhang W. Astragalin protects the liver from oxidative damage by modulating the lnc XIST/miR-155-5p/Nrf2 axis. J Funct Foods 2023; 108:105769. [DOI: 10.1016/j.jff.2023.105769] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
|
47
|
Yu M, Zhang M, Fu P, Wu M, Yin X, Chen Z. Research progress of mitophagy in chronic cerebral ischemia. Front Aging Neurosci 2023; 15:1224633. [PMID: 37600521 PMCID: PMC10434995 DOI: 10.3389/fnagi.2023.1224633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Chronic cerebral ischemia (CCI), a condition that can result in headaches, dizziness, cognitive decline, and stroke, is caused by a sustained decrease in cerebral blood flow. Statistics show that 70% of patients with CCI are aged > 80 years and approximately 30% are 45-50 years. The incidence of CCI tends to be lower, and treatment for CCI is urgent. Studies have confirmed that CCI can activate the corresponding mechanisms that lead to mitochondrial dysfunction, which, in turn, can induce mitophagy to maintain mitochondrial homeostasis. Simultaneously, mitochondrial dysfunction can aggravate the insufficient energy supply to cells and various diseases caused by CCI. Regulation of mitophagy has become a promising therapeutic target for the treatment of CCI. This article reviews the latest progress in the important role of mitophagy in CCI and discusses the induction pathways of mitophagy in CCI, including ATP synthesis disorder, oxidative stress injury, induction of reactive oxygen species, and Ca2+ homeostasis disorder, as well as the role of drugs in CCI by regulating mitophagy.
Collapse
Affiliation(s)
- Mayue Yu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Manqing Zhang
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi, China
| | - Peijie Fu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Moxin Wu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Xiaoping Yin
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Zhiying Chen
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| |
Collapse
|
48
|
Da Silva IO, Crespo-Lopez ME, Augusto-Oliveira M, Arrifano GDP, Ramos-Nunes NR, Gomes EB, da Silva FRP, de Sousa AA, Leal ALAB, Damasceno HC, de Oliveira ACA, Souza-Monteiro JR. What We Know about Euterpe Genus and Neuroprotection: A Scoping Review. Nutrients 2023; 15:3189. [PMID: 37513607 PMCID: PMC10384735 DOI: 10.3390/nu15143189] [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: 06/21/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
The Euterpe genus (mainly Euterpe oleracea Martius, Euterpe precatoria Martius, and Euterpe edulis Martius) has recently gained commercial and scientific notoriety due to the high nutritional value of its fruits, which are rich in polyphenols (phenolic acids and anthocyanins) and have potent antioxidant activity. These characteristics have contributed to the increased number of neuropharmacological evaluations of the three species over the last 10 years, especially açaí of the species Euterpe oleracea Martius. The fruits of the three species exert neuroprotective effects through the modulation of inflammatory and oxidative pathways and other mechanisms, including the inhibition of the mTOR pathway and protection of the blood-brain barrier, all of them intimately involved in several neuropathologies. Thus, a better understanding of the neuropharmacological properties of these three species may open new paths for the development of therapeutic tools aimed at preventing and treating a variety of neurological conditions.
Collapse
Affiliation(s)
- Ilano Oliveira Da Silva
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (M.E.C.-L.); (M.A.-O.); (G.d.P.A.)
| | - Marcus Augusto-Oliveira
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (M.E.C.-L.); (M.A.-O.); (G.d.P.A.)
| | - Gabriela de Paula Arrifano
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (M.E.C.-L.); (M.A.-O.); (G.d.P.A.)
| | - Natália Raphaela Ramos-Nunes
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Elielton Barreto Gomes
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Felipe Rodolfo Pereira da Silva
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Aline Andrade de Sousa
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Alessandro Luiz Araújo Bentes Leal
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Helane Conceição Damasceno
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Ana Carolina Alves de Oliveira
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - José Rogério Souza-Monteiro
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| |
Collapse
|
49
|
Martins T, Barros AN, Rosa E, Antunes L. Enhancing Health Benefits through Chlorophylls and Chlorophyll-Rich Agro-Food: A Comprehensive Review. Molecules 2023; 28:5344. [PMID: 37513218 PMCID: PMC10384064 DOI: 10.3390/molecules28145344] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Chlorophylls play a crucial role in photosynthesis and are abundantly found in green fruits and vegetables that form an integral part of our diet. Although limited, existing studies suggest that these photosynthetic pigments and their derivatives possess therapeutic properties. These bioactive molecules exhibit a wide range of beneficial effects, including antioxidant, antimutagenic, antigenotoxic, anti-cancer, and anti-obesogenic activities. However, it is unfortunate that leafy materials and fruit peels often go to waste in the food supply chain, contributing to the prevailing issue of food waste in modern societies. Nevertheless, these overlooked materials contain valuable bioactive compounds, including chlorophylls, which offer significant health benefits. Consequently, exploring the potential of these discarded resources, such as utilizing them as functional food ingredients, aligns with the principles of a circular economy and presents exciting opportunities for exploitation.
Collapse
Affiliation(s)
- Tânia Martins
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro (CITAB), 5000-801 Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), 5000-801 Vila Real, Portugal
| | - Ana Novo Barros
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro (CITAB), 5000-801 Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), 5000-801 Vila Real, Portugal
| | - Eduardo Rosa
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro (CITAB), 5000-801 Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), 5000-801 Vila Real, Portugal
| | - Luís Antunes
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro (CITAB), 5000-801 Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), 5000-801 Vila Real, Portugal
| |
Collapse
|
50
|
Jurcau A, Jurcau CM. Mitochondria in Huntington's disease: implications in pathogenesis and mitochondrial-targeted therapeutic strategies. Neural Regen Res 2023; 18:1472-1477. [PMID: 36571344 PMCID: PMC10075114 DOI: 10.4103/1673-5374.360289] [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] [Indexed: 11/19/2022] Open
Abstract
Huntington's disease is a genetic disease caused by expanded CAG repeats on exon 1 of the huntingtin gene located on chromosome 4. Compelling evidence implicates impaired mitochondrial energetics, altered mitochondrial biogenesis and quality control, disturbed mitochondrial trafficking, oxidative stress and mitochondrial calcium dyshomeostasis in the pathogenesis of the disorder. Unfortunately, conventional mitochondrial-targeted molecules, such as cysteamine, creatine, coenzyme Q10, or triheptanoin, yielded negative or inconclusive results. However, future therapeutic strategies, aiming to restore mitochondrial biogenesis, improving the fission/fusion balance, and improving mitochondrial trafficking, could prove useful tools in improving the phenotype of Huntington's disease and, used in combination with genome-editing methods, could lead to a cure for the disease.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea; Neurology 3 Ward, Clinical Emergency Hospital, Oradea, Romania
| | | |
Collapse
|