1
|
Begh MZA, Amin MA, Shatu MM, Sweilam SH, Puri S, Ramesh RB, Arjun UVNV, Shanmugarajan TS, Pommala N, Durairaj A, Ethiraj S, Shenbakadurai N, Ahmad I, Emran TB. Unraveling Berberine's Molecular Mechanisms in Neuroprotection Against Neurodegeneration. Chem Biodivers 2025:e202500170. [PMID: 40128128 DOI: 10.1002/cbdv.202500170] [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: 01/25/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 03/26/2025]
Abstract
Neurodegenerative diseases (NDs) exhibit significant global public health challenges due to the lack of effective treatments. Berberine (BBR), a natural alkaloid compound in various plants, has been recognized for its potential neuroprotective properties. This review explores the current understanding of BBR's mechanisms of action and its therapeutic potential in preventing and treating NDs such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. BBR's neuroprotective properties are attributed to its multifaceted actions, including anti-inflammatory, antioxidant, antiapoptotic, and neurotrophic effects. In addition, BBR can influence many signaling pathways involved in neurodegeneration, including AMP-activated protein kinase (AMPK), nuclear factor erythroid 2-related factor 2, and brain-derived neurotrophic factor pathways. Furthermore, BBR targets vital signaling pathways, including AMPK, PI3K/Akt, and MAPK, which are essential for developing NDs. In addition, BBR's efficacy in reducing neurodegenerative pathology and improving cognitive function has been demonstrated through preclinical studies using cellular and animal models. Clinical trials demonstrating BBR's therapeutic potential in NDs have yielded promising results, but further research is needed to confirm its safety and efficacy in humans.
Collapse
Affiliation(s)
- Md Zamshed Alam Begh
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md Al Amin
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mst Maharunnasa Shatu
- Department of Botany, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Badr City, Egypt
| | - Sachin Puri
- School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-University, Hyderabad, India
| | - Rathod Bhagyashri Ramesh
- School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-University, Hyderabad, India
| | - Uppuluri Varuna Naga Venkata Arjun
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology and Advanced Studies (VISTAS), Chennai, India
| | - Thukani Sathanantham Shanmugarajan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology and Advanced Studies (VISTAS), Chennai, India
| | - Nagaveni Pommala
- S. V. U. College of Pharmaceutical Sciences, Sri Venkateswara University, Tirupati, India
| | - Akiladevi Durairaj
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology and Advanced Studies (VISTAS), Chennai, India
| | - Susithra Ethiraj
- S. V. U. College of Pharmaceutical Sciences, Sri Venkateswara University, Tirupati, India
| | - Nagarajan Shenbakadurai
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology and Advanced Studies (VISTAS), Chennai, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, Bangladesh
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
2
|
Rajbongshi BL, Mukherjee AK. Drugs from poisonous plants: Ethnopharmacological relevance to modern perspectives. Toxicon X 2025; 25:100215. [PMID: 39990776 PMCID: PMC11847069 DOI: 10.1016/j.toxcx.2025.100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/18/2024] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
The world of plant diversity is endlessly fascinating and essential for life on Earth. Since the inception of early civilization, humans have utilized plants for several purposes, particularly for their medicinal value. While some plants are known for their toxicity, they also contain beneficial phytochemicals that are important for both plants and humans, indicating their dual nature. This study aims to explore and synthesize the existing knowledge on various poisonous plant species found worldwide. It primarily focuses on the therapeutic potential of specific types of phytochemicals responsible for treating multiple diseases. This review includes a list of 70 poisonous plants with medicinal properties for treating various ailments, as well as some of their traditional uses. A few of these plants are emphasized, which have been tremendously explored and studied, hold significant potential to contribute to modern drug discovery. Furthermore, it addresses the possible prospects and challenges of using poisonous plants and their phytochemicals as therapeutic agents. Although the therapeutic potential of poisonous plants is substantial, many toxins remain unexplored. This review accentuates the need for rigorous scientific investigations, prior to clinical trials to validate their traditional uses, which would reveal the pharmacological interventions that will eventually advance human health and well-being.
Collapse
Affiliation(s)
- Bhagya Lakhmi Rajbongshi
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Paschim Boragaon, Garchuk, Guwahati, Assam, 781035, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ashis K. Mukherjee
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Paschim Boragaon, Garchuk, Guwahati, Assam, 781035, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, 784028, India
| |
Collapse
|
3
|
Zhao L, Wang YF, Adamcakova-Dodd A, Thorne PS, Islam R, Liu KJ, Chen F, Luo J, Liu LZ. Nrf2/cyclooxygenase 2 signaling in Cr(VI)-induced carcinogenesis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117800. [PMID: 39923569 DOI: 10.1016/j.ecoenv.2025.117800] [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: 06/24/2024] [Revised: 10/29/2024] [Accepted: 01/22/2025] [Indexed: 02/11/2025]
Abstract
Long-term exposure to hexavalent chromium [Cr(VI)] has been linked to lung cancer, and cyclooxygenase-2 (COX-2) is a well-known inflammatory factor. However, the role and mechanism of COX-2 in Cr(VI)-induced carcinogenesis are not clear yet. To address this question, we employed a mouse model exposed to Cr(VI) through intranasal instillation of particulate zinc chromate (ZnCrO4) for 12 weeks. Metabolomics and RNA-seq assays revealed enhanced activity of the arachidonic acid (AA)/eicosanoid metabolism pathway in lung tissues from mice exposed to Cr(VI). COX-2, the key enzyme of the AA/eicosanoid pathway, was significantly upregulated in Cr(VI)-exposed lung tissues, as well as in the Cr(VI)-induced transformed (Cr-T) cells compared to parental BEAS-2B (B2B) cells. We then employed multidisciplinary in vitro and in vivo functional assays to characterize the role of COX-2 in Cr(VI)-induced lung cancer. The results indicated that COX-2 functioned as an oncogene to promote the malignant transformation of B2B cells and enhance the proliferation, migration, tumor growth, and angiogenesis of Cr-T cells. Nuclear factor E2-related factor-2 (Nrf2) was identified as a transcription factor for COX-2. Nrf2 was upregulated in response to Cr(VI) exposure and contributed to Cr(VI)-induced lung cancers, in part by upregulating COX-2 expression. Moreover, microRNA-379 (miR-379) was found to target COX-2 to inhibit its expression posttranscriptionally. MiR-379 was downregulated in Cr(VI)-exposed lung tissues and Cr-T cells, and ectopic miR-379 expression reduced Cr-T cell viability and migration, with partial reversal upon COX-2 restoration. In summary, our study revealed the oncogenic role of COX-2 and identified two novel regulatory mechanisms for COX-2 overexpression in Cr(VI)-induced carcinogenesis.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Pathology, University of Iowa, Iowa, IA 52242, USA; Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Yi-Fang Wang
- Department of Pathology, University of Iowa, Iowa, IA 52242, USA; Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrea Adamcakova-Dodd
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA, 52242, USA
| | - Peter S Thorne
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA, 52242, USA
| | - Ranakul Islam
- Department of Pathology, University of Iowa, Iowa, IA 52242, USA; Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ke Jian Liu
- Department of Pathology, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Fei Chen
- Department of Pathology, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jia Luo
- Department of Pathology, University of Iowa, Iowa, IA 52242, USA
| | - Ling-Zhi Liu
- Department of Pathology, University of Iowa, Iowa, IA 52242, USA; Department of Pathology, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
4
|
Sethi P, Mehan S, Khan Z, Maurya PK, Kumar N, Kumar A, Tiwari A, Sharma T, Das Gupta G, Narula AS, Kalfin R. The SIRT-1/Nrf2/HO-1 axis: Guardians of neuronal health in neurological disorders. Behav Brain Res 2025; 476:115280. [PMID: 39368713 DOI: 10.1016/j.bbr.2024.115280] [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: 06/21/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
SIRT1 (Sirtuin 1) is a NAD+-dependent deacetylase that functions through nucleoplasmic transfer and is present in nearly all mammalian tissues. SIRT1 is believed to deacetylate its protein substrates, resulting in neuroprotective actions, including reduced oxidative stress and inflammation, increased autophagy, increased nerve growth factors, and preserved neuronal integrity in aging or neurological disease. Nrf2 is a transcription factor that regulates the genes responsible for oxidative stress response and substance detoxification. The activation of Nrf2 guards cells against oxidative damage, inflammation, and carcinogenic stimuli. Several neurological abnormalities and inflammatory disorders have been associated with variations in Nrf2 activation caused by either pharmacological or genetic factors. Recent evidence indicates that Nrf2 is at the center of a complex cellular regulatory network, establishing it as a transcription factor with genuine pleiotropy. HO-1 is most likely a component of a defense mechanism in cells under stress, as it provides negative feedback for cell activation and mediator synthesis. This mediator is upregulated by Nrf2, nitric oxide (NO), and other factors in various inflammatory states. HO-1 or its metabolites, such as CO, may mitigate inflammation by modulating signal transduction pathways. Neurological diseases may be effectively treated by modulating the activity of HO-1. Multiple studies have demonstrated that SIRT1 and Nrf2 share an important connection. SIRT1 enhances Nrf2, activates HO-1, protects against oxidative injury, and decreases neuronal death. This has been associated with numerous neurodegenerative and neuropsychiatric disorders. Therefore, activating the SIRT1/Nrf2/HO-1 pathway may help treat various neurological disorders. This review focuses on the current understanding of the SIRT1 and Nrf2/HO-1 neuroprotective processes and the potential therapeutic applications of their target activators in neurodegenerative and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Pranshul Sethi
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Pankaj Kumar Maurya
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| | - Aakash Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Aarti Tiwari
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Tarun Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Block 23, Sofia 1113, Bulgaria; Department of Healthcare, South-West University "NeofitRilski", Ivan Mihailov St. 66, Blagoevgrad 2700, Bulgaria
| |
Collapse
|
5
|
Li L, Wang L, Ding W, Wu J, Liu F, Liu J, Zhang J, Wang J. the Improvement Effects of Sika Deer Antler Protein in an Alzheimer's Disease Mouse Model via the Microbe-Gut-Brain Axis. Food Sci Nutr 2025; 13:e4656. [PMID: 39803278 PMCID: PMC11717054 DOI: 10.1002/fsn3.4656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/24/2024] [Accepted: 11/23/2024] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder of the central nervous system. The interplay between the intestinal microbiota and metabolites is believed to influence brain function and the pathogenesis of neurodegenerative conditions through the microbe-gut-brain axis. Sika deer antler protein possesses neuroprotective properties; however, the precise mechanism by which it improves AD remains unclear. Sika deer antler protein ameliorated AD in vivo by activating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. The metabolome of brain and intestinal tissues and the microbiota of intestinal contents were tested and analyzed according to the microbe-gut-brain theory. Sika deer antler protein increased beneficial bacterial levels and decreased harmful bacterial levels. Correlation analyses using the gut flora-metabolomics pathway ultimately revealed that sika deer antler protein modulated the brain and intestinal tract bi-directionally via the tyrosine metabolism pathway, thereby establishing a connection within the microbe-gut-brain axis. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of the differential metabolite targets of the DAP4 group showed that the enriched pathways mainly included PI3K/AKT, which was consistent with the findings of the pharmacodynamic mechanisms observed in in vivo experiments. This suggests that antler protein may be involved in microbe-gut-brain interactions through tyrosine metabolism and may improve AD by activating the PI3K/AKT/Nrf2 signaling pathway. These findings add to our understanding of the microbe-gut-brain axis facilitated by sika deer antler protein and offer novel insights for further research on sika deer antler protein in alleviating AD.
Collapse
Affiliation(s)
- Lei Li
- College of Traditional Chinese Medicinal MaterialJilin Agricultural UniversityChangchunChina
| | - Lulu Wang
- School of MedicineChangchun Sci‐Tech UniversityChangchunChina
| | - Weixing Ding
- College of Traditional Chinese Medicinal MaterialJilin Agricultural UniversityChangchunChina
| | - Jianfa Wu
- College of Traditional Chinese Medicinal MaterialJilin Agricultural UniversityChangchunChina
| | - Fei Liu
- College of Traditional Chinese Medicinal MaterialJilin Agricultural UniversityChangchunChina
| | - Jiansong Liu
- College of Traditional Chinese Medicinal MaterialJilin Agricultural UniversityChangchunChina
| | - Jing Zhang
- College of Traditional Chinese Medicinal MaterialJilin Agricultural UniversityChangchunChina
- Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal MaterialsChangchunChina
| | - Jing Wang
- Jilin Province Faw General HospitalChangchunChina
| |
Collapse
|
6
|
Li Z, Xing J. Nuclear factor erythroid 2-related factor-mediated signaling alleviates ferroptosis during cerebral ischemia-reperfusion injury. Biomed Pharmacother 2024; 180:117513. [PMID: 39341075 DOI: 10.1016/j.biopha.2024.117513] [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: 07/25/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiac arrest (CA) is a significant challenge for emergency physicians worldwide and leads to increased morbidity and mortality rates. The poor prognosis of CA primarily stems from the complexity and irreversibility of cerebral ischemia-reperfusion injury (CIRI). Ferroptosis, a form of programmed cell death characterized by iron overload and lipid peroxidation, plays a crucial role in the progression and treatment of CIRI. In this review, we highlight the mechanisms of ferroptosis within the context of CIRI, focusing on its role as a key contributor to neuronal damage and dysfunction post-CA. We explore the crucial involvement of the nuclear factor erythroid 2-related factor (Nrf2)-mediated signaling pathway in modulating ferroptosis-associated processes during CIRI. Through comprehensive analysis of the regulatory role of Nrf2 in the cellular responses to oxidative stress, we highlight its potential as a therapeutic target for mitigating ferroptotic cell death and improving the neurological prognosis of patients experiencing CA. Furthermore, we discuss interventions targeting the Kelch-like ECH-associated protein 1/Nrf2/antioxidant response element pathway, including the use of traditional Chinese medicine and Western medicine, which demonstrate potential for attenuating ferroptosis and preserving neuronal function in CIRI. Owing to the limitations in the safety, specificity, and effectiveness of Nrf2-targeted drugs, as well as the technical difficulties and ethical constraints in obtaining the results related to the brain pathological examination of patients, most of the studies focusing on Nrf2-related regulation of ferroptosis in CIRI are still in the basic research stage. Overall, this review aims to provide a comprehensive understanding of the mechanisms underlying ferroptosis in CIRI, offering insights into novel therapeutics aimed at enhancing the clinical outcomes of patients with CA.
Collapse
Affiliation(s)
- Zheng Li
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
7
|
Kim Y, Kim J, Kim B, Kim R, Kim HJ, Lee EH, Kim J, Park J, Jeong Y, Park SI, Kim H, Kang M, Lee J, Bahn YS, Choi JW, Park JH, Park KD. Discovery and Optimization of a Series of Vinyl Sulfoximine-Based Analogues as Potent Nrf2 Activators for the Treatment of Multiple Sclerosis. J Med Chem 2024; 67:17866-17892. [PMID: 39323296 PMCID: PMC11472819 DOI: 10.1021/acs.jmedchem.4c01907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
Multiple sclerosis (MS) is an immune-mediated neurodegenerative disease of the central nervous system (CNS), which leads to demyelination, axonal loss, and neurodegeneration. Increased oxidative stress and neurodegeneration have been implicated in all stages of MS, making neuroprotective therapeutics a promising strategy for its treatment. We previously have reported vinyl sulfones with antioxidative and anti-inflammatory properties that activate nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor that induces the expression of cytoprotective genes against oxidative stress. In this study, we synthesized vinyl sulfoximine derivatives by modifying the core structure and determined therapeutic potential as Nrf2 activators. Among them, 10v effectively activated Nrf2 (EC50 = 83.5 nM) and exhibited favorable drug-like properties. 10v successfully induced expression of Nrf2-dependent antioxidant enzymes and suppressed lipopolysaccharide (LPS)-induced inflammatory responses in BV-2 microglial cells. We also confirmed that 10v effectively reversed disease progression and attenuated demyelination in an experimental autoimmune encephalitis (EAE) mouse model of MS.
Collapse
Affiliation(s)
- Yoowon Kim
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Department
of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jaehwan Kim
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Division
of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Byungeun Kim
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Division
of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Rium Kim
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Division
of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Hyeon Jeong Kim
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
| | - Elijah Hwejin Lee
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Division
of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Jushin Kim
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Department
of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jiwoo Park
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Division
of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Yeeun Jeong
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Division
of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Sang In Park
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Division
of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Hyemin Kim
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Division
of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Minsik Kang
- Doping
Control Center, KIST, Seoul 02792, Republic of Korea
| | - Jaeick Lee
- Doping
Control Center, KIST, Seoul 02792, Republic of Korea
| | - Yong-Sun Bahn
- Department
of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Ji Won Choi
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Cureverse
Co., Ltd., Seoul Biohub, Seoul 02455, Republic
of Korea
| | - Jong-Hyun Park
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Division
of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ki Duk Park
- Brain
Disorders Research Center, Brain Science Research Division, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
- Division
of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| |
Collapse
|
8
|
Khayatan D, Razavi SM, Arab ZN, Khanahmadi M, Samanian A, Momtaz S, Sukhorukov VN, Jamialahmadi T, Abdolghaffari AH, Barreto GE, Sahebkar A. Protective Effects of Plant-Derived Compounds Against Traumatic Brain Injury. Mol Neurobiol 2024; 61:7732-7750. [PMID: 38427213 DOI: 10.1007/s12035-024-04030-w] [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: 09/26/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Inflammation in the nervous system is one of the key features of many neurodegenerative diseases. It is increasingly being identified as a critical pathophysiological primitive mechanism associated with chronic neurodegenerative diseases following traumatic brain injury (TBI). Phytochemicals have a wide range of clinical properties due to their antioxidant and anti-inflammatory effects. Currently, there are few drugs available for the treatment of neurodegenerative diseases other than symptomatic relief. Numerous studies have shown that plant-derived compounds, in particular polyphenols, protect against various neurodegenerative diseases and are safe for consumption. Polyphenols exert protective effects on TBI via restoration of nuclear factor kappa B (NF-κB), toll-like receptor-4 (TLR4), and Nod-like receptor family proteins (NLRPs) pathways. In addition, these phytochemicals and their derivatives upregulate the phosphatidylinositol-3-Kinase/Protein Kinase B (PI3K/AKT) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways, which have critical functions in modulating TBI symptoms. There is supporting evidence that medicinal plants and phytochemicals are protective in different TBI models, though future clinical trials are needed to clarify the precise mechanisms and functions of different polyphenolic compounds in TBI.
Collapse
Affiliation(s)
- Danial Khayatan
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Seyed Mehrad Razavi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Zahra Najafi Arab
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Khanahmadi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amirreza Samanian
- Department of Neurology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeideh Momtaz
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Vasily N Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, Moscow, 121609, Russia
- Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| | - Amirhossein Sahebkar
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Bhardwaj S, Grewal AK, Singh S, Dhankar V, Jindal A. An insight into the concept of neuroinflammation and neurodegeneration in Alzheimer's disease: targeting molecular approach Nrf2, NF-κB, and CREB. Inflammopharmacology 2024; 32:2943-2960. [PMID: 38951436 DOI: 10.1007/s10787-024-01502-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024]
Abstract
Alzheimer's disease (AD) is a most prevalent neurologic disorder characterized by cognitive dysfunction, amyloid-β (Aβ) protein accumulation, and excessive neuroinflammation. It affects various life tasks and reduces thinking, memory, capability, reasoning and orientation ability, decision, and language. The major parts responsible for these abnormalities are the cerebral cortex, amygdala, and hippocampus. Excessive inflammatory markers release, and microglial activation affect post-synaptic neurotransmission. Various mechanisms of AD pathogenesis have been explored, but still, there is a need to debate the role of NF-κB, Nrf2, inflammatory markers, CREB signaling, etc. In this review, we have briefly discussed the signaling mechanisms and function of the NF-ĸB signaling pathway, inflammatory mediators, microglia activation, and alteration of autophagy. NF-κB inhibition is a current strategy to counter neuroinflammation and neurodegeneration in the brain of individuals with AD. In clinical trials, numbers of NF-κB modulators are being examined. Recent reports revealed that molecular and cellular pathways initiate complex pathological competencies that cause AD. Moreover, this review will provide extensive knowledge of the cAMP response element binding protein (CREB) and how these nuclear proteins affect neuronal plasticity.
Collapse
Affiliation(s)
- Shaveta Bhardwaj
- G.H.G. Khalsa College of Pharmacy, Gurusar Sudhar, Ludhiana, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Vaibhav Dhankar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Anu Jindal
- G.H.G. Khalsa College of Pharmacy, Gurusar Sudhar, Ludhiana, India
| |
Collapse
|
10
|
Chen M, He X, Fan Y, Xia L, Dong Z. Sofalcone attenuates neurodegeneration in MPTP-induced mouse model of Parkinson's disease by inhibiting oxidative stress and neuroinflammation. Mol Biol Rep 2024; 51:908. [PMID: 39141244 DOI: 10.1007/s11033-024-09852-4] [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: 05/14/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by oxidative stress and neuroinflammation. Sofalcone (SFC), a chalcone derivative known for its antioxidative and anti-inflammatory properties, is widely used clinically as a gastric mucosa protective agent. However, its therapeutic potential in PD remains to be fully explored. In this study, we investigated the neuroprotective effects of SFC in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model. METHODS AND RESULTS We found that SFC ameliorated MPTP-induced motor impairments in mice, as assessed by the rotarod and wire tests. Moreover, SFC administration prevented the loss of dopaminergic neurons and striatal degeneration induced by MPTP. Subsequent investigations revealed that SFC reversed MPTP-induced downregulation of NRF2, reduced elevated levels of reactive oxygen species (ROS) and malondialdehyde (MDA), and increased total antioxidant capacity (TAOC). Furthermore, SFC suppressed MPTP-induced activation of microglia and astrocytes, downregulated the pro-inflammatory cytokine TNF-α, and upregulated the anti-inflammatory cytokine IL-4. Additionally, SFC ameliorated the MPTP-induced downregulation of phosphorylation of Akt at Ser473. CONCLUSIONS This study provides evidence for the neuroprotective effects of SFC, highlighting its antioxidative and anti-inflammatory properties and its role in Akt activation in the PD model. These findings underscore SFC's potential as a promising therapeutic candidate for PD, warranting further clinical investigation.
Collapse
Affiliation(s)
- Mulan Chen
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Xin He
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yepeng Fan
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Lei Xia
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Zhifang Dong
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
11
|
Abd-Ellatieff HA, Georg K, Abourawash ARA, Ghazy EW, Samak DH, Goda WM. Aspergillus awamori: potential antioxidant, anti-inflammatory, and anti-apoptotic activities in acetic acid-induced ulcerative colitis in rats. Inflammopharmacology 2024; 32:2541-2553. [PMID: 38763983 PMCID: PMC11300502 DOI: 10.1007/s10787-024-01489-w] [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: 12/10/2023] [Accepted: 03/22/2024] [Indexed: 05/21/2024]
Abstract
Ulcerative colitis (UC) is a chronic colonic inflammation with a significant health hazard. Aspergillus awamori (A. awamori) is a microorganism with various bioactive compounds with natural antioxidant and anti-inflammatory properties. The present work aimed to elucidate the protective and therapeutic effects of varying concentrations of A. awamori against acetic acid (AA)-induced ulcerative colitis (UC) in rats. Nine groups of albino male rats were established: a control negative group (G1), a control positive group (G2,AA), and preventive protocol groups (including G3A, G4A, and G5A) that received 100 mg, 50 mg, and 25 mg/kg b.w, respectively, of A. awamori orally and daily from the 1st day of the experiment and for 7 consecutive days. Then, they were subjected to one dose of AA intrarectally on day 8th. G3B, G4B, and G5B were termed as curative protocol groups that received one dose of AA on day 8th and then administered 100 mg, 50 mg, and 25 mg/kg b.w. of A. awamori, respectively, on day 9th and continued receiving these doses daily until day 16th. Rats in the AA group exhibited marked histopathological alterations of the distal colon, with an exaggeration of the DAI. In addition, a remarkable increase in oxidative stress was represented by the elevation of MDA and NO levels with a decline in SOD and GPx activities. In addition, upregulation of TNF-α, IL-6, and IL-1β mRNA expressions and downregulation of Muc2 and Nrf2 levels were detected. Unambiguously, a remarkable anti-inflammatory effect was noticed either in A. awamori prevented or treated groups expounded by reducing and regulating TNF-α, IL-6, and IL-1β with improved pathological lesion scoring. The Muc2, Nrf2, and bcl-2 gene levels were upregulated and restored also. In summary, the findings in this work reveal that A. awamori supplementation successfully alleviated the UC induced by AA, which had a better effect when administered before colitis induction.
Collapse
Affiliation(s)
- Hoda A Abd-Ellatieff
- Pathology Department, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt.
| | - Kristen Georg
- Cure Lab Clinical Pathology, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | | | - Emad W Ghazy
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Dalia H Samak
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Wael M Goda
- Pathology Department, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
- Clinical Pathology Department, Faculty of Veterinary Medicine, Damanhour University, Damanhour-El-Beheira, Egypt
| |
Collapse
|
12
|
Chen Q, Wu B, Shi Z, Wang Y, Yuan Y, Chen X, Wang Y, Hu J, Mao L, Gao Y, Wu G. LncRNA H19 knockdown promotes neuropathologic and functional recovery via the Nrf2/HO-1 axis after traumatic brain injury. CNS Neurosci Ther 2024; 30:e14870. [PMID: 39049714 PMCID: PMC11269889 DOI: 10.1111/cns.14870] [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: 05/12/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
AIMS Traumatic brain injury (TBI) stands as a significant concern in public health, frequently leading to enduring neurological deficits. Long non-coding RNA H19 (lncRNA H19) exerts a potential regulator role in the pathology of brain injury. This study investigates the effects of lncRNA H19 knockdown (H19-KD) on the pathophysiology of TBI and its potential neuroprotective mechanisms. METHODS Controlled cortical impact was employed to establish a stable TBI mouse model. The expression levels of various genes in perilesional cortex and striatum tissue after TBI was detected by RT-qPCR. AAV9-shRNA-H19 was injected into the lateral ventricle of mice to knockdown the expression of lncRNA H19. Various behavioral tests were performed to evaluate sensorimotor and cognitive functions after TBI. Immunofluorescence and Nissl staining were performed to assess brain tissue damage and neuroinflammation. The Nrf2 and HO-1 expression was performed by Western blot. RESULTS After TBI, the expression of lncRNA H19 was elevated in perilesional tissue and gradually reverted to baseline. Behavioral tests demonstrated that H19-KD significantly promoted the recovery of sensorimotor and cognitive functions after TBI. Besides, H19-KD reduced brain tissue loss, preserved neuronal integrity, and ameliorated white matter damage at the histological level. In addition, H19-KD restrained the pro-inflammatory and facilitated anti-inflammatory phenotypes of microglia/macrophages, attenuating the neuroinflammatory response after TBI. Furthermore, H19-KD promoted activation of the Nrf2/HO-1 axis after TBI, while suppression of Nrf2 partially abolished the neuroprotective effect. CONCLUSION H19-KD exerts neuroprotective effects after TBI in mice, partially mediated by the activation of the Nrf2/HO-1 axis.
Collapse
Affiliation(s)
- Qiankang Chen
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| | - Biwu Wu
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| | - Ziyu Shi
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| | - Yana Wang
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| | - Yiwen Yuan
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| | - Xingdong Chen
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| | - Yuqing Wang
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| | - Jin Hu
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| | - Leilei Mao
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| | - Yanqin Gao
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| | - Gang Wu
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| |
Collapse
|
13
|
Rauf A, Khalil AA, Awadallah S, Khan SA, Abu‐Izneid T, Kamran M, Hemeg HA, Mubarak MS, Khalid A, Wilairatana P. Reactive oxygen species in biological systems: Pathways, associated diseases, and potential inhibitors-A review. Food Sci Nutr 2024; 12:675-693. [PMID: 38370049 PMCID: PMC10867483 DOI: 10.1002/fsn3.3784] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 02/20/2024] Open
Abstract
Reactive oxygen species (ROS) are produced under normal physiological conditions and may have beneficial and harmful effects on biological systems. ROS are involved in many physiological processes such as differentiation, proliferation, necrosis, autophagy, and apoptosis by acting as signaling molecules or regulators of transcription factors. In this case, maintaining proper cellular ROS levels is known as redox homeostasis. Oxidative stress occurs because of the imbalance between the production of ROS and antioxidant defenses. Sources of ROS include the mitochondria, auto-oxidation of glucose, and enzymatic pathways such as nicotinamide adenine dinucleotide phosphate reduced (NAD[P]H) oxidase. The possible ROS pathways are NF-κB, MAPKs, PI3K-Akt, and the Keap1-Nrf2-ARE signaling pathway. This review covers the literature pertaining to the possible ROS pathways and strategies to inhibit them. Additionally, this review summarizes the literature related to finding ROS inhibitors.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of ChemistryUniversity of SwabiAnbarPakistan
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahoreLahorePakistan
| | - Samir Awadallah
- Department of Medical Lab Sciences, Faculty of Allied Medical SciencesZarqa UniversityZarqaJordan
| | - Shahid Ali Khan
- Department of Chemistry, School of Natural SciencesNational University of Science and Technology (NUST)IslamabadPakistan
| | - Tareq Abu‐Izneid
- Pharmaceutical Sciences, College of PharmacyAl Ain UniversityAl Ain, Abu DhabiUAE
| | - Muhammad Kamran
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Hassan A. Hemeg
- Department of Medical Laboratory Technology, College of Applied Medical SciencesTaibah UniversityAl‐Medinah Al‐MonawaraSaudi Arabia
| | | | - Ahood Khalid
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahoreLahorePakistan
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical MedicineMahidol UniversityBangkokThailand
| |
Collapse
|
14
|
Aziel Alvarado-Ojeda Z, Zamilpa A, Costet-Mejia A, Méndez-Martínez M, Trejo-Moreno C, Jiménez-Ferrer JE, Salazar-Martínez AM, Cruz-Muñoz ME, Fragoso G, Rosas-Salgado G. Hydroalcoholic extract from Sechium edule (Jacq.) S.w. root reverses oleic acid-induced steatosis and insulin resistance in vitro. Heliyon 2024; 10:e24567. [PMID: 38312619 PMCID: PMC10835324 DOI: 10.1016/j.heliyon.2024.e24567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/17/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
Steatosis is characterized by fat accumulation and insulin resistance (IR) in hepatocytes, which triggers a pro-oxidant, pro-inflammatory environment that may eventually lead to cirrhosis or liver carcinoma. This work was aimed to assess the effect of Sechium edule root hydroalcoholic extract (rSe-HA) (rich in cinnamic and coumaric acid, among other phenolic compounds) on triglyceride esterification, lipid degradation, AMPK expression, and the phosphorylation of insulin receptor in a Ser312 residue, as well as on the redox status, malondialdehyde (MDA) production, and the production of proinflammatory cytokines in an in vitro model of steatosis induced by oleic acid, to help develop a phytomedicine that could reverse this pathology. rSe-HA reduced triglyceride levels in hepatocyte lysates, increased lipolysis by activating AMPK at Thr172, and improved the redox status, as evidenced by the concentration of glycerol and formazan, respectively. It also prevented insulin resistance (IR), as measured by glucose consumption and the phosphorylation of the insulin receptor at Ser312. It also prevented TNFα and IL6 production and decreased the levels of MDA and nitric oxide (ON). Our results indicate that rSe-HA reversed steatosis and controlled the proinflammatory and prooxidant environment in oleic acid-induced dysfunctional HepG2 hepatocytes, supporting its potential use to control this disorder.
Collapse
Affiliation(s)
- Zimri Aziel Alvarado-Ojeda
- Facultad de Medicina, Universidad Autónoma Del Estado de Morelos, Leñeros S/N, Cuernavaca, Morelos, 62350, Mexico
| | - Alejandro Zamilpa
- Laboratorio de Farmacología, Centro de Investigaciones Biomédicas Del Sur, Instituto Mexicano Del Seguro Social, Xochitepec, Morelos, 62790, Mexico
| | - Alejandro Costet-Mejia
- Laboratorio de Farmacología, Centro de Investigaciones Biomédicas Del Sur, Instituto Mexicano Del Seguro Social, Xochitepec, Morelos, 62790, Mexico
| | - Marisol Méndez-Martínez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana-Xochimilco, 04960, Mexico City, Mexico
| | - Celeste Trejo-Moreno
- Facultad de Medicina, Universidad Autónoma Del Estado de Morelos, Leñeros S/N, Cuernavaca, Morelos, 62350, Mexico
| | - Jesús Enrique Jiménez-Ferrer
- Laboratorio de Farmacología, Centro de Investigaciones Biomédicas Del Sur, Instituto Mexicano Del Seguro Social, Xochitepec, Morelos, 62790, Mexico
| | - Ana Maria Salazar-Martínez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Mario Ernesto Cruz-Muñoz
- Facultad de Medicina, Universidad Autónoma Del Estado de Morelos, Leñeros S/N, Cuernavaca, Morelos, 62350, Mexico
| | - Gladis Fragoso
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Gabriela Rosas-Salgado
- Facultad de Medicina, Universidad Autónoma Del Estado de Morelos, Leñeros S/N, Cuernavaca, Morelos, 62350, Mexico
| |
Collapse
|
15
|
Yuhan L, Khaleghi Ghadiri M, Gorji A. Impact of NQO1 dysregulation in CNS disorders. J Transl Med 2024; 22:4. [PMID: 38167027 PMCID: PMC10762857 DOI: 10.1186/s12967-023-04802-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
NAD(P)H Quinone Dehydrogenase 1 (NQO1) plays a pivotal role in the regulation of neuronal function and synaptic plasticity, cellular adaptation to oxidative stress, neuroinflammatory and degenerative processes, and tumorigenesis in the central nervous system (CNS). Impairment of the NQO1 activity in the CNS can result in abnormal neurotransmitter release and clearance, increased oxidative stress, and aggravated cellular injury/death. Furthermore, it can cause disturbances in neural circuit function and synaptic neurotransmission. The abnormalities of NQO1 enzyme activity have been linked to the pathophysiological mechanisms of multiple neurological disorders, including Parkinson's disease, Alzheimer's disease, epilepsy, multiple sclerosis, cerebrovascular disease, traumatic brain injury, and brain malignancy. NQO1 contributes to various dimensions of tumorigenesis and treatment response in various brain tumors. The precise mechanisms through which abnormalities in NQO1 function contribute to these neurological disorders continue to be a subject of ongoing research. Building upon the existing knowledge, the present study reviews current investigations describing the role of NQO1 dysregulations in various neurological disorders. This study emphasizes the potential of NQO1 as a biomarker in diagnostic and prognostic approaches, as well as its suitability as a target for drug development strategies in neurological disorders.
Collapse
Affiliation(s)
- Li Yuhan
- Epilepsy Research Center, Münster University, Münster, Germany
- Department of Breast Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Ali Gorji
- Epilepsy Research Center, Münster University, Münster, Germany.
- Department of Neurosurgery, Münster University, Münster, Germany.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
16
|
Kalantary-Charvadeh A, Nazari Soltan Ahmad S, Aslani S, Beyrami M, Mesgari-Abbasi M. β-lapachone protects against doxorubicin-induced hepatotoxicity through modulation of NAD + /SIRT-1/FXR/p-AMPK/NF-kB and Nrf2 signaling axis. J Biochem Mol Toxicol 2024; 38:e23564. [PMID: 37867446 DOI: 10.1002/jbt.23564] [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: 03/06/2023] [Revised: 07/05/2023] [Accepted: 10/09/2023] [Indexed: 10/24/2023]
Abstract
Doxorubicin (DOX) is a widely used antineoplastic drug, but its clinical use is limited by significant toxicities, such as hepatotoxicity. In this study, we evaluated the effects of β-lapachone (β-LAP), a natural quinone-containing compound, in a mouse model of DOX-induced hepatotoxicity. β-LAP was orally administered at 1.25, 2.5, and 5 mg/kg for 4 days, and a single dose of DOX (20 mg/kg) was injected intraperitoneally on the second day. Histopathological changes, liver function markers, antioxidant and inflammatory markers were assessed. β-LAP ameliorated liver injury and liver function markers evoked by DOX. β-LAP also downregulated the mRNA expression of nuclear factor-kB-corresponding genes including interleukin-6, interleukin-1β, and tumor necrosis factor-α. Moreover, β-LAP increased the nuclear factor erythroid 2-related factor 2 target genes heme oxygenase-1 and NAD(P)H: quinone oxidoreductase 1, along with antioxidant enzymes including reduced glutathione, catalase, and superoxide dismutase with simultaneous reduction in the lipid peroxidation product malondialdehyde. Meanwhile, it recovered NAD+ /NADH ratios and subsequently elevated the protein levels of sirtuin-1 (SIRT-1), farnesoid X receptor (FXR), and phosphorylated AMP-activated protein kinase (p-AMPK). Collectively, these findings suggest a protective role of β-LAP against DOX-induced hepatotoxicity by partly regulating the NAD+ /SIRT-1/FXR/p-AMPK axis.
Collapse
Affiliation(s)
- Ashkan Kalantary-Charvadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Somayeh Aslani
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehdi Beyrami
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | |
Collapse
|
17
|
Joung EJ, Lee MK, Lee M, Gwon M, Shin T, Ryu H, Jeong HH, Kim MJ, Van JY, Kim JI, Choi J, Jung WK, Kim HR, Lee B. Sargachromenol Attenuates Inflammatory Responses by Regulating NF-κB and Nrf2 Pathways in RAW 264.7 Cells and LPS-treated Mice. PLANTA MEDICA 2024; 90:25-37. [PMID: 37848042 DOI: 10.1055/a-2180-1338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
This study aims to explore the anti-inflammatory mechanisms of sargachromenol in both RAW 264.7 cells and lipopolysaccharide (LPS)-treated mice, as previous reports have suggested that sargachromenol possesses anti-aging, anti-inflammatory, antioxidant, and neuroprotective properties. Although the precise mechanism behind its anti-inflammatory activity remains unclear, pretreatment with sargachromenol effectively reduced the production of nitric oxide, prostaglandin E2, and interleukin (IL)-1β in LPS-stimulated RAW 264.7 cells by inhibiting cyclooxygenase-2. Moreover, sargachromenol inhibited the activation of nuclear factor-κB (NF-κB) by preventing the degradation of the inhibitor of κB-α (IκB-α) and inhibiting protein kinase B (Akt) phosphorylation in LPS-stimulated cells. We also found that sargachromenol induced the production of heme oxygenase-1 (HO-1) by activating the nuclear transcription factor erythroid-2-related factor 2 (Nrf2). In LPS-treated mice, oral administration of sargachromenol effectively reduced the levels of IL-1β, IL-6, and tumor necrosis factor-α (TNF-α) in the serum, suggesting its ability to suppress the production of inflammatory mediators by inhibiting the Akt/NF-κB pathway and upregulating the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Eun-Ji Joung
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Min-Kyeong Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Minsup Lee
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Louisiana, United States
| | - Misung Gwon
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Taisun Shin
- Division of Food and Nutrition, Chonnam National University, Gwangju, Republic of Korea
| | - Heeyeon Ryu
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Hyeon Hak Jeong
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Republic of Korea
| | - Myeong-Jin Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Ji Yun Van
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Republic of Korea
| | - Jae-Il Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Jinkyung Choi
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Won-Kyo Jung
- Division of Biomedical Engineering and Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
| | - Hyeung-Rak Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Republic of Korea
| | - Bonggi Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
18
|
Levine AJ, Thadani C, Soontornniyomkij V, Lopez-Aranda MF, Mesa YG, Kitchen S, Rezek V, Silva A, Kolson DL. Behavioral and histological assessment of a novel treatment of neuroHIV in humanized mice. RESEARCH SQUARE 2023:rs.3.rs-3678629. [PMID: 38168407 PMCID: PMC10760308 DOI: 10.21203/rs.3.rs-3678629/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Neurocognitive deficits are prevalent among people living with HIV, likely due to chronic inflammation and oxidative stress in the brain. To date, no pharmaceutical treatments beyond antiretroviral therapy (ARV) has been shown to reduce risk for, or severity of, HIV-associated neurocognitive disorder. Here we investigate a novel compound, CDDO-Me, with documented neuroprotective effects via activation of the nrf2 and inhibition of the NFkB pathways. Methods We conducted three studies to assess the efficacy of CDDO-Me alone or in combination with antiretroviral therapy in humanized mice infected with HIV; behavioral, histopathological, and immunohistochemical. Results CDDO-Me in combination with ARV rescued social interaction deficits; however, only ARV was associated with preserved functioning in other behaviors, and CDDO-Me may have attenuated those benefits. A modest neuroprotective effect was found for CDDO-Me when administered with ARV, via preservation of PSD-95 expression; however, ARV alone had a more consistent protective effect. No significant changes in antioxidant enzyme expression levels were observed in CDDO-Me-treated animals. Only ARV use seemed to affect some antioxidant levels, indicating that it is ARV rather than CDDO-Me that is the major factor providing neuroprotection in this animal model. Finally, immunohistochemical analysis found that several cellular markers in various brain regions varied due to ARV rather than CDDO-Me. Conclusion Limited benefit of CDDO-Me on behavior and neuroprotection were observed. Instead, ARV was shown to be the more beneficial treatment. These experiments support the future use of this chimeric mouse for behavioral experiments in neuroHIV research.
Collapse
Affiliation(s)
| | | | | | | | | | - Scott Kitchen
- UCLA Humanized Mouse Core Laboratory, University of California
| | - Valerie Rezek
- UCLA Humanized Mouse Core Laboratory, University of California
| | | | | |
Collapse
|
19
|
Tang X, Deng P, Jiang Y, Zhang L, He Y, Yang H. An Overview of Recent Advances in the Neuroprotective Potentials of Fisetin against Diverse Insults in Neurological Diseases and the Underlying Signaling Pathways. Biomedicines 2023; 11:2878. [PMID: 38001882 PMCID: PMC10669030 DOI: 10.3390/biomedicines11112878] [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: 09/06/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 11/26/2023] Open
Abstract
The nervous system plays a leading role in the regulation of physiological functions and activities in the body. However, a variety of diseases related to the nervous system have a serious impact on human health. It is increasingly clear that neurological diseases are multifactorial pathological processes involving multiple cellular systems, and the onset of these diseases usually involves a diverse array of molecular mechanisms. Unfortunately, no effective therapy exists to slow down the progression or prevent the development of diseases only through the regulation of a single factor. To this end, it is pivotal to seek an ideal therapeutic approach for challenging the complicated pathological process to achieve effective treatment. In recent years, fisetin, a kind of flavonoid widely existing in fruits, vegetables and other plants, has shown numerous interesting biological activities with clinical potentials including anti-inflammatory, antioxidant and neurotrophic effects. In addition, fisetin has been reported to have diverse pharmacological properties and neuroprotective potentials against various neurological diseases. The neuroprotective effects were ascribed to its unique biological properties and multiple clinical pharmacological activities associated with the treatment of different neurological disorders. In this review, we summarize recent research progress regarding the neuroprotective potential of fisetin and the underlying signaling pathways of the treatment of several neurological diseases.
Collapse
Affiliation(s)
- Xiangwen Tang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an 710054, China; (X.T.); (L.Z.)
- Basic Medical School Academy, Shaanxi University of Traditional Chinese Medicine, Xianyang 712046, China; (P.D.); (Y.J.)
| | - Peng Deng
- Basic Medical School Academy, Shaanxi University of Traditional Chinese Medicine, Xianyang 712046, China; (P.D.); (Y.J.)
| | - Yizhen Jiang
- Basic Medical School Academy, Shaanxi University of Traditional Chinese Medicine, Xianyang 712046, China; (P.D.); (Y.J.)
| | - Lingling Zhang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an 710054, China; (X.T.); (L.Z.)
| | - Yuqing He
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China;
| | - Hao Yang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an 710054, China; (X.T.); (L.Z.)
| |
Collapse
|
20
|
Escribano BM, Muñoz-Jurado A, Luque E, Galván A, LaTorre M, Caballero-Villarraso J, Giraldo AI, Agüera E, Túnez I. Effect of the Combination of Different Therapies on Oxidative Stress in the Experimental Model of Multiple Sclerosis. Neuroscience 2023; 529:116-128. [PMID: 37595941 DOI: 10.1016/j.neuroscience.2023.08.005] [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: 04/10/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/20/2023]
Abstract
Oxidative stress is heavily involved in several pathological features of Multiple Sclerosis (MS), such as myelin destruction, axonal degeneration, and inflammation. Different therapies have been shown to reduce the oxidative stress that occurs in the animal model of MS, experimental autoimmune encephalomyelitis (EAE). Some of these therapies are transcranial magnetic stimulation (TMS), extra virgin olive oil (EVOO) and S-allyl cysteine (SAC). This study aims to test the antioxidant effect of these three therapies, to compare the efficacy of SAC versus TMS and EVOO, and to analyze the effect of combining SAC + TMS and SAC and EVOO. Seventy Dark Agouti rats were used, which were divided into Control group; Vehicle group; Mock group; SAC; EVOO; TMS; SAC + EVOO; SAC + TMS; EAE; EAE + SAC; EAE + EVOO; EAE + TMS; EAE + SAC + EVOO; EAE + SAC + TMS. The TMS consisted of an oscillatory magnetic field in the form of a sine wave with a frequency of 60 Hz and an amplitude of 0.7mT (EL-EMF) applied for two hours in the morning, once a day, five days a week. SAC was administered at a dose of 50 mg/kg body weight, orally daily, five days a week. EVOO represented 10% of their calorie intake in the total standard daily diet of rats AIN-93G. All treatments were maintained for 51 days. TMS, EVOO and SAC, alone or in combination, reduce oxidative stress, increasing antioxidant defenses and also lowering the clinical score. Combination therapies do not appear to be more potent than individual therapies against the oxidative stress of EAE or its clinical symptoms.
Collapse
Affiliation(s)
- Begoña M Escribano
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Spain; Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain.
| | - Ana Muñoz-Jurado
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Spain
| | - Evelio Luque
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain; Department of Morphological Sciences, Histology Section, Faculty of Medicine and Nursing, University of Cordoba, Spain
| | - Alberto Galván
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Spain
| | - Manuel LaTorre
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain
| | - Javier Caballero-Villarraso
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Spain; Analysis Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Ana I Giraldo
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Spain
| | - Eduardo Agüera
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain; Neurology Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Isaac Túnez
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Spain; Cooperative Research Thematic Excellent Network on Brain Stimulation (REDESTIM), Spain.
| |
Collapse
|
21
|
Wang Y, Wei S, Lian H, Tong L, Yang L, Ren B, Guo D, Huang H. A Neutral Polysaccharide from Spores of Ophiocordyceps gracilis Regulates Oxidative Stress via NRF2/FNIP1 Pathway. Int J Mol Sci 2023; 24:14721. [PMID: 37834168 PMCID: PMC10572349 DOI: 10.3390/ijms241914721] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Ophiocordyceps gracilis (O. gracilis) is a parasitic fungus used in traditional Chinese medicine and functional foods. In this study, a neutral heteropolysaccharide (GSP-1a) was isolated from spores of O. gracilis, and its structure and antioxidant capacities were investigated. GSP-1a was found to have a molecular weight of 72.8 kDa and primarily consisted of mannose (42.28%), galactose (35.7%), and glucose (22.02%). The backbone of GSP-1a was composed of various sugar residues, including →6)-α-D-Manp-(1→, →2,6)-α-D-Manp-(1→, →2,4,6)-α-D-Manp-(1→, →6)-α-D-Glcp-(1→, and →3,6)-α-D-Glcp-(1→, with some branches consisting of →6)-α-D-Manp-(1→ and α-D-Gal-(1→. In vitro, antioxidant activity assays demonstrated that GSP-1a exhibited scavenging effects on hydroxyl radical (•OH), 2,2'-azino-bis-3-ethylbenzothiazoline-6-sulfonic acid radical cation (ABTS•+), and 2,2-diphenyl-1-picrylhydrazyl radical (DPPH•). Moreover, GSP-1a was found to alleviate H2O2-induced oxidative stress in HepG2 cells by reducing the levels of reactive oxygen species (ROS) and malondialdehyde (MDA), while enhancing the activities of superoxide dismutase (SOD). Furthermore, GSP-1a upregulated the mRNA expression of antioxidant enzymes such as Ho-1, Gclm, and Nqo1, and regulated the NRF2/KEAP1 and FNIP1/FEM1B pathways. The findings elucidated the structural types of GSP-1a and provided a reliable theoretical basis for its usage as a natural antioxidant in functional foods or medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dongsheng Guo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, NO 1, Wen Yuan Road, Nanjing 210023, China
| | - He Huang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, NO 1, Wen Yuan Road, Nanjing 210023, China
| |
Collapse
|
22
|
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
|
23
|
Lela L, Russo D, De Biasio F, Gorgoglione D, Ostuni A, Ponticelli M, Milella L. Solanum aethiopicum L. from the Basilicata Region Prevents Lipid Absorption, Fat Accumulation, Oxidative Stress, and Inflammation in OA-Treated HepG2 and Caco-2 Cell Lines. PLANTS (BASEL, SWITZERLAND) 2023; 12:2859. [PMID: 37571013 PMCID: PMC10421219 DOI: 10.3390/plants12152859] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
Obesity is widely associated with intestine barrier impairment, nonalcoholic fatty liver disease (NAFLD) outbreaks, oxidative stress, and inflammation. In a previous investigation, the Solanum aethiopicum L. growing in Basilicata Region has demonstrated to have antioxidant activity; hence this investigation was aimed to evaluate for the first time the antilipidemic and anti-inflammatory activity of the Lucanian S. aethiopicum L. peel extract in vitro on OA-treated HepG2 and Caco-2 cell lines. It was shown that the extract could reduce lipogenesis by down-regulating SREBP-1c and HMGCR expression and fatty acid β-oxidation by up-regulating PPARα, CPT1A, and UCP2 expression. In addition, the S. aethiopicum L. peel extract might also improve oxidative stress by reducing endoplasmic reticulum stress and regulating the Nrf2 and Nf-κB molecular pathways. Altogether, these results demonstrated for the first time the possible application of the Lucanian S. aethiopicum peel extract for preventing obesity and managing NAFLD.
Collapse
Affiliation(s)
- Ludovica Lela
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (L.L.); (D.R.); (A.O.)
| | - Daniela Russo
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (L.L.); (D.R.); (A.O.)
- Spinoff Bioactiplant s.r.l., Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | | | | | - Angela Ostuni
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (L.L.); (D.R.); (A.O.)
| | - Maria Ponticelli
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (L.L.); (D.R.); (A.O.)
| | - Luigi Milella
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (L.L.); (D.R.); (A.O.)
| |
Collapse
|
24
|
Muneer PMA. Nrf2 as a Potential Therapeutic Target for Traumatic Brain Injury. J Integr Neurosci 2023; 22:81. [PMID: 37519172 DOI: 10.31083/j.jin2204081] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 08/01/2023] Open
Abstract
In this review, we discuss the possibility and feasibility of nuclear factor erythroid 2-related factor 2 (Nrf2) as a therapeutic target to minimize the devastating effects of a brain injury. To complete this review, comprehensive literature searches were conducted in MEDLINE, PubMed, Embase, and PsycINFO databases for English scientific peer-reviewed articles through December 2022. This short review addressed the different sources of oxidative stress and its effects on blood-brain barrier (BBB) dysfunction, mitochondrial damage, and changes in a variety of inflammatory molecules associated with central nervous system (CNS) injury. At last, we explained the potential efficacy of the Nrf2 transcription factor in reducing oxidative stress-mediated secondary damages after a CNS injury. The role of CPUY192018, an inhibitor of Nrf2-Keap1 protein-protein interaction in protecting the injured brain cells is given as evidence of Nrf2's role in activating antioxidant genes. Overall, the scope of Nrf2 in developing therapeutic interventions for a variety of pathophysiological conditions associated with CNS injury-induced free radical/inflammatory signaling is acknowledged. Nrf2 has a widespread application in basic and clinical neuroscience for understanding and treating free radical/inflammatory signaling disorders, including neurological diseases. The development of innovative therapeutic strategies using Nrf2-inducing agents can be applied to reduce the complications of TBI before advancing it to posttraumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- P M Abdul Muneer
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian JFK University Medical Center, Edison, NJ 08820, USA
| |
Collapse
|
25
|
Park CH, Noh JS, Jeon JP, Yokozawa T. A systematic review on anti-diabetic action of 7-O-galloyl-D-sedoheptulose, a polyphenol from Corni Fructus, in type 2 diabetic mice with hepatic and pancreatic damage. Drug Discov Ther 2023:2022.01097. [PMID: 37245985 DOI: 10.5582/ddt.2022.01097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Traditional medicines are recently being focused on to treat diabetes and its complications because of their lack of toxic and/or side effects. This report describes the effects of 7-O-galloyl-D-sedoheptulose (GS), a polyphenolic compound isolated from Corni Fructus, on type 2 diabetic db/db mice with hepatic and pancreatic damage. We examined several biochemical factors and oxidative stress- and inflammation-related markers. In the serum, levels of glucose, leptin, insulin, C-peptide, resistin, tumor necrosis factor-α, and interleukin-6 were down-regulated, while adiponectin was augmented by GS treatment. In addition, GS suppressed the reactive oxygen species and lipid peroxidation in the serum, liver, and pancreas, but increased the pancreatic insulin and pancreatic C-peptide contents. These results were derived from attenuating the expression of nicotinamide adenine dinucleotide phosphate oxidase subunit proteins, Nox-4 and p22phox. Augmented nuclear factor (NF)-E2-related factor 2 and heme oxygenase-1 were reduced with a decrease in oxidative stress during GS treatment. NF-κB-related pro-inflammatory factors were also alleviated in hepatic tissue. Moreover, GS modulated the protein expressions of pro-inflammatory NF-κB, cyclooxygenase-2, inducible nitric oxide synthase, c-Jun N-terminal kinase (JNK), phosphor-JNK, activator protein-1, transforming growth factor-β1, and fibronectin. Based on these results, we demonstrated that the anti-diabetic action of GS may be due to its anti-oxidative stress property and anti-inflammatory action.
Collapse
Affiliation(s)
- Chan Hum Park
- Institute of New Frontier Research Team, Research Institute of Medical-Bio Convergence, Hallym University, Chuncheon, Republic of Korea
| | - Jeong Sook Noh
- Department of Food Science and Nutrition, Tongmyong University, Busan, Republic of Korea
| | - Jin Pyeong Jeon
- Department of Neurosurgery, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Takako Yokozawa
- Graduate School of Science and Engineering for Research, University of Toyama, Toyama, Japan
| |
Collapse
|
26
|
Alrouji M, Manouchehrinia A, Aram J, Alotaibi A, Alhajlah S, Almuhanna Y, Alomeir O, Shamsi A, Gran B, Constantinescu CS. Investigating the Effect of Cigarette Smoking on Serum Uric Acid Levels in Multiple Sclerosis Patients: A Cross Sectional Study. Brain Sci 2023; 13:brainsci13050800. [PMID: 37239272 DOI: 10.3390/brainsci13050800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
OBJECTIVES The present study is aimed at determining the effect of cigarette smoking (CS) on serum uric acid (UA) levels quantitatively before and after smoking cessation among people with MS (pwMS). Additionally, a possible correlation between UA levels and both disability progression and disease severity was also investigated. A retrospective cross-sectional study was conducted using the Nottingham University Hospitals MS Clinics database. It involves 127 people with definite MS recorded when reporting the latest smoking status and the clinical diagnosis. All necessary demographics and clinical characteristics were collected. We found that smoker pwMS had significantly lower serum UA levels than non-smoker pwMS (p-value = 0.0475), and this reduction was recovered after smoking cessation (p-value = 0.0216). However, the levels of disability or disease severity were not correlated with the levels of serum UA in current smoker pwMS, measured by the expanded disability status scale (EDSS; r = -0.24; p-value = 0.38), multiple sclerosis impact scale 29 (MSIS-29; r = 0.01; p-value = 0.97) and MS severity score (MSSS; r = -0.16; p-value = 0.58), respectively. Our result suggests that the reduction in UA levels is more likely a consequence of oxidative stress triggered by many risk factors, including CS, and could be considered a potential indicator of smoking cessation. In addition, the absence of a correlation between UA levels and disease severity and disability suggests that UA is not an optimal biomarker for disease severity and disability prediction among current smoker, ex-smoker or non-smoker pwMS.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Clinical Neurosciences Group, Shaqra University, Shaqra 11961, Saudi Arabia
- Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- Department of Clinical Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Sahqra 11961, Saudi Arabia
| | - Ali Manouchehrinia
- Department of Clinical Neuroscience, Karolinska Institute, 171 77 Solna, Sweden
| | - Jehan Aram
- Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| | - Abdulmajeed Alotaibi
- Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Sharif Alhajlah
- Clinical Neurosciences Group, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Yasir Almuhanna
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Othman Alomeir
- Clinical Neurosciences Group, Shaqra University, Shaqra 11961, Saudi Arabia
- Department of Pharmacy Practice, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Bruno Gran
- Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- Department of Neurology, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, UK
| | - Cris S Constantinescu
- Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- Department of Neurology, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, UK
- Cooper University Hospital, Cooper Neurological Institute, Camden, NJ 08103, USA
| |
Collapse
|
27
|
Kim B, Kim R, Kim HJ, Kim Y, Park SJ, Lee EH, Kim J, Kim J, Choi JW, Park JH, Park KD. Optimization and evaluation of pyridinyl vinyl sulfones as Nrf2 activator for the antioxidant and anti-inflammatory effects. Eur J Med Chem 2023; 256:115433. [PMID: 37187090 DOI: 10.1016/j.ejmech.2023.115433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/21/2023] [Accepted: 04/28/2023] [Indexed: 05/17/2023]
Abstract
Many studies have reported that chalcone-based compounds exhibit biological activities such as anticancer, antioxidant, anti-inflammatory and neuroprotective effects. Among the published chalcone derivatives, (E)-1-(3-methoxypyridin-2-yl)-3-(2-(trifluoromethyl)phenyl)prop-2-en-1-one (VEDA-1209), which is currently undergoing preclinical study, was selected as a starting compound for the development of new nuclear factor erythroid 2-related factor 2 (Nrf2) activators. Based on our previous knowledge, we attempted to redesign and synthesize VEDA-1209 derivatives by introducing the pyridine ring and sulfone moiety to ameliorate its Nrf2 efficacy and drug-like properties. Among the synthesized compounds, (E)-3-chloro-2-(2-((3-methoxypyridin-2-yl)sulfonyl)vinyl) pyridine (10e) was found to have approximately 16-folds higher Nrf2 activating effects than VEDA-1209 (10e: EC50 = 37.9 nM vs VEDA-1209: EC50 = 625 nM) in functional cell-based assay. In addition, 10e effectively improved drug-like properties such as CYP inhibition probability and metabolic stability. Finally, 10e demonstrated excellent antioxidant and anti-inflammatory effects in BV-2 microglial cells and significantly restored spatial memory deficits in lipopolysaccharide (LPS)-induced neuroinflammatory mouse models.
Collapse
Affiliation(s)
- Byungeun Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Rium Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Hyeon Jeong Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yoowon Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sun Jun Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Cureverse Co., Ltd., KIST, 1st Floor, H2 Building, Seoul, 02792, Republic of Korea
| | - Elijah Hwejin Lee
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Jushin Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jaehwan Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Ji Won Choi
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Cureverse Co., Ltd., KIST, 1st Floor, H2 Building, Seoul, 02792, Republic of Korea.
| | - Jong-Hyun Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Ki Duk Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
28
|
Namakin K, Moghaddam MH, Sadeghzadeh S, Mehranpour M, Vakili K, Fathi M, Golshan A, Bayat AH, Tajik AH, Eskandari N, Mohammadzadeh I, Benisi SZ, Aliaghaei A, Abdollahifar MA. Elderberry diet improves gut-brain axis dysfunction, neuroinflammation, and cognitive impairment in the rat model of irritable bowel syndrome. Metab Brain Dis 2023; 38:1555-1572. [PMID: 36877342 DOI: 10.1007/s11011-023-01187-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/13/2023] [Indexed: 03/07/2023]
Abstract
Irritable bowel syndrome (IBS) is related to a problem in the gut-brain axis. This experimental research aimed to shed light on the potential therapeutic application of elderberry (EB), which can work on the axis and get better the IBS symptoms. There were three groups (36 Sprague-Dawley rats) in this experiment, including control, IBS, and IBS with EB diet (IBS + EB). Making use of intracolonic instillation of 1 ml of 4% acetic acid for 30 s, IBS was induced. 7 days later, the EB extract (2%) was added to the diets of all animals for 8 weeks. Some histological, behavioral, and stereological techniques were used to detect the effects of EB on the gut and brain tissues. The findings showed that the EB diet improved locomotion and decreased anxiety-like behavior in the rat models of IBS. Moreover, the diet dropped the expression of TNF-α and increased mucosal layer thickness and the number of goblet and mast cells in colon tissue samples. In the hippocampal samples, administration of EB prevented astrogliosis and astrocyte reactivity. Although hippocampal and cortical neurons decreased markedly in the IBS group, EB prevented the drop in the number of neurons. Although lots of research is needed to elucidate the effectiveness of EB in IBS and its exact molecular mechanism, the result of this study showed that EB as an antioxidant and immune-modulatory agent could be a promising research target to prevent the impairment in the gut-brain axis, and could ameliorative classic IBS symptoms.
Collapse
Affiliation(s)
- Kosar Namakin
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Sara Sadeghzadeh
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Mehranpour
- Department of Genetics, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Golshan
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Hossein Bayat
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir-Hossein Tajik
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Eskandari
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Ibrahim Mohammadzadeh
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheila Zamanlui Benisi
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, 1385/768, Tehran, Iran
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad-Amin Abdollahifar
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Wan H, Cai Y, Xiao L, Ling Y, Ge L, Mo S, Xie Q, Peng S, Zhou B, Zeng X, Chen X. JFD, a Novel Natural Inhibitor of Keap1 Alkylation, Suppresses Intracellular Mycobacterium Tuberculosis Growth through Keap1/Nrf2/SOD2-Mediated ROS Accumulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:6726654. [PMID: 36819778 PMCID: PMC9937762 DOI: 10.1155/2023/6726654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 08/16/2022] [Accepted: 10/14/2022] [Indexed: 02/12/2023]
Abstract
It is an effective strategy to treat tuberculosis by enhancing reactive oxygen species- (ROS-) mediated killing of Mycobacterium tuberculosis in macrophages, but there are no current therapeutic agents targeting this pathway. Honeysuckle has been used as the traditional medicine for tuberculosis treatment for 1500 years. Japoflavone D (JFD) is a novel biflavonoid isolated from Honeysuckle promoting ROS accumulation by Nrf2 pathway in hepatocarcinoma cells. However, its activity to kill M. tuberculosis in macrophages and molecular mechanism has not been reported. Our results showed that JFD enhances the M. tuberculosis elimination by boosting ROS levels in THP-1 cells. Moreover, the massive ROS accumulation activates p38 to induce apoptosis. Notably, the mechanism revealed that JFD suppresses the nuclear transport of Nrf2, thereby inhibiting SOD2 transcription, leading to a large ROS accumulation. Further studies showed that JFD disrupts the Keap1 alkylation at specific residues Cys14, Cys257, and Cys319, which is crucial for Nrf2 activation, thereby interrupts the nuclear transport of Nrf2. In pharmacokinetic study, JFD can stay as the prototype for 24 h in mice and can be excreted in feces without any toxicity. Our data reveal for the first time that a novel biflavonoid JFD as a potent inhibitor of Keap1 alkylation can suppress the nuclear transport of Nrf2. And it is the first research of the inhibitor of Keap1 alkylation. Furthermore, JFD robustly promotes M. tuberculosis elimination from macrophages by inhibiting Keap1/Nrf2/SOD2 pathway, resulting in the ROS accumulation. This work identified Keap1 alkylation as a new drug target for tuberculosis and provides a preliminary basis for the development of antituberculosis lead compounds based on JFD.
Collapse
Affiliation(s)
- Haoqiang Wan
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
- Department of Pathology (Longhua Branch), Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, 518020 Guangdong Province, China
| | - Yi Cai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| | - Lingyun Xiao
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Yunzhi Ling
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Lanlan Ge
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
- Department of Pathology (Longhua Branch), Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, 518020 Guangdong Province, China
| | - Siwei Mo
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| | - Qiujie Xie
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Shusong Peng
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Boping Zhou
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Xiaobin Zeng
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
- Department of Pathology (Longhua Branch), Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, 518020 Guangdong Province, China
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| | - Xinchun Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| |
Collapse
|
30
|
Yang G, Yang L, Liu Q, Zhu Z, Yang Q, Liu J, Beta T. Protective effects of cyanidin-3-O-glucoside on BPA-induced neurodevelopmental toxicity in zebrafish embryo model. Comp Biochem Physiol C Toxicol Pharmacol 2023; 264:109525. [PMID: 36410639 DOI: 10.1016/j.cbpc.2022.109525] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 11/20/2022]
Abstract
Bisphenol A (BPA) is ubiquitous in the environment and poses a threat to wildlife and human health. It has been reported that BPA may cause the neurotoxicity during gestational and neonatal periods. Cyanidin-3-O-glucoside (C3G) is one of the most abundant anthocyanins that has shown multiple bio-functions. In this study, the protective effects and possible mechanism of C3G against BPA-induced neurodevelopment toxicity in zebrafish embryos/larvae were studied. The results showed that co-exposure of C3G (25 μg/mL) significantly attenuated BPA-induced deficit in locomotor behavior and restored the BPA-induced aberrant changes in brain morphology of zebrafish larvae. Further studies showed that the defects of central nervous development and the downregulated neurogenesis relative genes induced by BPA were significantly counteracted by co-exposure with 5 μg/mL of C3G. In addition, C3G (25 μg/mL) mitigated the decline of glutathione (GSH) content and enzymatic activities of superoxide dismutase (SOD), glutathione peroxidase (GPx) and catalase (CAT), attenuated oxidative stress and cell apoptosis induced by BPA in zebrafish. The enhancements of the expression of genes involved in the Nrf2-ARE pathway (Nrf2, HO-1, NQO1, GCLC, and GCLM) were also observed by co-exposure of C3G. The results indicate that C3G exerts protective effects on BPA-induced neurodevelopmental toxicity through improving transcription of neurogenesis related genes, enhancing antioxidative defense system and reducing cell apoptosis by regulation of apoptotic genes in zebrafish larvae. The results suggest that anthocyanins may play important role against the exogenous toxicity for vertebrates.
Collapse
Affiliation(s)
- Guangchao Yang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Lipin Yang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Qin Liu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Zhenzhu Zhu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Qian Yang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Jining Liu
- Research and Development Center for Watershed Environmental Eco-Engineering, Beijing Normal University, Zhuhai 519087, China.
| | - Trust Beta
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| |
Collapse
|
31
|
Protective Mechanisms of 3-Acetyl-11-keto-β-Boswellic Acid and Piperine in Fluid Percussion Rat Model of Traumatic Brain Injury Targeting Nrf2 and NFkB Signaling. Neurotox Res 2023; 41:57-84. [PMID: 36576717 DOI: 10.1007/s12640-022-00628-x] [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: 08/10/2022] [Revised: 10/18/2022] [Accepted: 12/16/2022] [Indexed: 12/29/2022]
Abstract
The current study aimed to investigate the neuroprotective effect of 3-acetyl-11-keto-β-boswellic acid (AKBA) in combination with bioenhancer piperine in lateral fluid percussion injury-induced TBI in experimental rats. Fluid percussion injury was introduced in the rat brain by delivering 50 mmHg of pressure for 3 min to the exposed brain. AKBA 25 mg/kg, 50 mg/kg orally, and AKBA (25 mg/kg, p.o.) in combination with piperine (2.5 mg/kg, p.o.) were administered from day 1 to day 14 to the assigned groups. On the 1st, 7th, and 14th day, behavioral parameters were checked. On the 15th day, animals were euthanized. In TBI rat model, AKBA-piperine combination significantly restored the altered performance of grip strength, rotarod test, open field task, narrow beam task (beam crossing time and no. of foot slips), and Morris water maze (escape latency and time spent in target quadrant) (p < 0.001 vs TBI control). Furthermore, the AKBA-piperine combination significantly reduced pro-inflammatory cytokine level in TBI rat model (&p < 0.001 vs TBI control). The combined effect of AKBA and piperine significantly restored oxidative stress parameters level, catecholamines level, and neurotransmitters level (p < 0.001 vs TBI control). Further findings showed that the AKBA-piperine combination prevented histopathological changes (p < 0.001), and the immunohistological study confirmed increased Nrf2-positive cells (p < 0.001 vs TBI control) and reduced nuclear factor kappa B (NFkB) expression (p < 0.001 vs TBI control, p < 0.01 vs TBI + AKBA 50 mg/kg) in the cortical region following AKBA-piperine administration. The present study concluded that AKBA along with piperine achieved anti-oxidant, and anti-inflammatory effects, and also prevented neuronal injury via targeting Nrf2 and NFkB expressions.
Collapse
|
32
|
Wang S, Zhao Y, Chan AWH, Yao M, Chen Z, Abbatt JPD. Organic Peroxides in Aerosol: Key Reactive Intermediates for Multiphase Processes in the Atmosphere. Chem Rev 2023; 123:1635-1679. [PMID: 36630720 DOI: 10.1021/acs.chemrev.2c00430] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Organic peroxides (POs) are organic molecules with one or more peroxide (-O-O-) functional groups. POs are commonly regarded as chemically labile termination products from gas-phase radical chemistry and therefore serve as temporary reservoirs for oxidative radicals (HOx and ROx) in the atmosphere. Owing to their ubiquity, active gas-particle partitioning behavior, and reactivity, POs are key reactive intermediates in atmospheric multiphase processes determining the life cycle (formation, growth, and aging), climate, and health impacts of aerosol. However, there remain substantial gaps in the origin, molecular diversity, and fate of POs due to their complex nature and dynamic behavior. Here, we summarize the current understanding on atmospheric POs, with a focus on their identification and quantification, state-of-the-art analytical developments, molecular-level formation mechanisms, multiphase chemical transformation pathways, as well as environmental and health impacts. We find that interactions with SO2 and transition metal ions are generally the fast PO transformation pathways in atmospheric liquid water, with lifetimes estimated to be minutes to hours, while hydrolysis is particularly important for α-substituted hydroperoxides. Meanwhile, photolysis and thermolysis are likely minor sinks for POs. These multiphase PO transformation pathways are distinctly different from their gas-phase fates, such as photolysis and reaction with OH radicals, which highlights the need to understand the multiphase partitioning of POs. By summarizing the current advances and remaining challenges for the investigation of POs, we propose future research priorities regarding their origin, fate, and impacts in the atmosphere.
Collapse
Affiliation(s)
- Shunyao Wang
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai200240, China
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai200444, China
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, OntarioM5S 3E5, Canada
| | - Yue Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai200240, China
| | - Arthur W H Chan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, OntarioM5S 3E5, Canada
- School of the Environment, University of Toronto, Toronto, OntarioM5S 3E8, Canada
| | - Min Yao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai200240, China
| | - Zhongming Chen
- State Key Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing100871, China
| | - Jonathan P D Abbatt
- Department of Chemistry, University of Toronto, Toronto, OntarioM5S 3H6, Canada
| |
Collapse
|
33
|
Deng Z, Zhang N, Jiang L, Liu H, Hu S, Zhang D, Chen B, Liu Q, Sun Y, Chen J, Zhang C. Influence of microplastics on microbial anaerobic detoxification of chlorophenols. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 316:120707. [PMID: 36427829 DOI: 10.1016/j.envpol.2022.120707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 06/16/2023]
Abstract
Microplastics (MPs) can absorb halogenated organic compounds and transport them into marine anaerobic zones. Microbial reductive dehalogenation is a major process that naturally attenuates organohalide pollutants in anaerobic environments. Here, we aimed to determine the mechanisms through which MPs affect the microbe-mediated marine halogen cycle by incubating 2,4,6-trichlorophenol (TCP) dechlorinating cultures with various types of MPs. We found that TCP was dechlorinated to 4-chlorophenol in biotic control and polypropylene (PP) cultures, but essentially terminated at 2,4-dichlorophenol in polyethylene (PE) and polyethylene terephthalate (PET) cultures after incubation for 20 days. Oxygen-containing functional groups such as peroxide and aldehyde were enriched on PE and PET after incubation and corresponded to elevated levels of intracellular reactive oxygen species (ROS) in the microorganisms. Adding PE or PET to the cultures exerted limited effects on hydrogenase and ATPase activities, but delayed the expression of the gene encoding reductive dehalogenase (RDase). Considering the limited changes in the microbial composition of the enriched cultures, these findings suggested that microbial dechlorination is probably affected by MPs through the ROS-induced inhibition of RDase synthesis and/or activity. Overall, our findings showed that extensive MP pollution is unfavorable to environmental xenobiotic detoxification.
Collapse
Affiliation(s)
- Zhaochao Deng
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China
| | - Ning Zhang
- Department of Environmental Engineering, School of Chemical Engineering and Pharmacy, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Lijia Jiang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China
| | - Hui Liu
- Shengzhou Bureau of Agriculture and Rural Affairs, Shaoxing, 312400, Zhejiang, China
| | - Songtao Hu
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China
| | - Dongdong Zhang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China
| | - Bairu Chen
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China
| | - Qing Liu
- The Guangxi Key Laboratory of Theory and Technology for Environmental Pollution Control, Guilin University of Technology, Guilin, 541006, Guangxi, China
| | - Yuxia Sun
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China
| | - Jiawang Chen
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China
| | - Chunfang Zhang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China.
| |
Collapse
|
34
|
Chakrovorty A, Bhattacharjee B, Saxena A, Samadder A, Nandi S. Current Naturopathy to Combat Alzheimer's Disease. Curr Neuropharmacol 2023; 21:808-841. [PMID: 36173068 PMCID: PMC10227918 DOI: 10.2174/1570159x20666220927121022] [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: 04/24/2022] [Revised: 06/13/2022] [Accepted: 07/18/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegeneration is the progressive loss of structure or function of neurons, which may ultimately involve cell death. The most common neurodegenerative disorder in the brain happens with Alzheimer's disease (AD), the most common cause of dementia. It ultimately leads to neuronal death, thereby impairing the normal functionality of the central or peripheral nervous system. The onset and prevalence of AD involve heterogeneous etiology, either in terms of genetic predisposition, neurometabolomic malfunctioning, or lifestyle. The worldwide relevancies are estimated to be over 45 million people. The rapid increase in AD has led to a concomitant increase in the research work directed towards discovering a lucrative cure for AD. The neuropathology of AD comprises the deficiency in the availability of neurotransmitters and important neurotrophic factors in the brain, extracellular betaamyloid plaque depositions, and intracellular neurofibrillary tangles of hyperphosphorylated tau protein. Current pharmaceutical interventions utilizing synthetic drugs have manifested resistance and toxicity problems. This has led to the quest for new pharmacotherapeutic candidates naturally prevalent in phytochemicals. This review aims to provide an elaborative description of promising Phyto component entities having activities against various potential AD targets. Therefore, naturopathy may combine with synthetic chemotherapeutics to longer the survival of the patients.
Collapse
Affiliation(s)
- Arnob Chakrovorty
- Department of Zoology, Cytogenetics and Molecular Biology Lab., University of Kalyani, Kalyani, 741235, India
| | - Banani Bhattacharjee
- Department of Zoology, Cytogenetics and Molecular Biology Lab., University of Kalyani, Kalyani, 741235, India
| | - Aaruni Saxena
- Department of Cardiovascular Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Asmita Samadder
- Department of Zoology, Cytogenetics and Molecular Biology Lab., University of Kalyani, Kalyani, 741235, India
| | - Sisir Nandi
- Department of Pharmaceutical Chemistry, Global Institute of Pharmaceutical Education and Research, Affiliated to Uttarakhand Technical University, Kashipur, 244713, India
| |
Collapse
|
35
|
Zhou J, Shen R, Makale EC, Zhong W, Chen Z, Huang Q. SS31 Confers Cerebral Protection by Reversing Mitochondrial Dysfunction in Early Brain Injury Following Subarachnoid Hemorrhage, via the Nrf2- and PGC-1α-Dependent Pathways. Neurochem Res 2022; 48:1580-1595. [PMID: 36574150 DOI: 10.1007/s11064-022-03850-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
In early brain injury (EBI), oxidative stress occurs following subarachnoid hemorrhage (SAH), and mitochondria are intricately linked to this process. SS31, a mitochondria-targeting antioxidative peptide, has been demonstrated to be beneficial for multiple diseases because of its powerful antioxidant and neuroprotective properties. Although our previous study revealed that SS31 was involved in the powerful antioxidant effect following SAH, the underlying molecular mechanisms remained unclear. Thus, our study aimed to investigate the neuroprotective effects of SS31 by reversing mitochondrial dysfunction in EBI following SAH, via activating the Nrf2 signaling and PGC-1α pathways. Our findings confirmed that SS31 ameliorated SAH-triggered oxidative insult. SS31 administration decreased redundant reactive oxygen species, alleviated lipid peroxidation, and elevated the activities of antioxidant enzymes. Concomitant with the inhibited oxidative insult, SS31 dramatically attenuated neurological deficits, cerebral edema, neural apoptosis, and blood-brain barrier disruption following SAH. Moreover, SS31 remarkably promoted nuclear factor-erythroid 2 related factor 2 (Nrf2) nuclear shuttle and upregulated the expression levels of heme oxygenase-1 and NADPH: quinine oxidoreductase1. Additionally, SS31 enhanced the expression levels of PGC-1α and its target genes, and increased the mtDNA copy number, promoting mitochondrial function. However, PGC-1α-specific inhibitor SR-18292 pretreatment dramatically suppressed SS31-induced Nrf2 expression and PGC-1α activation. Furthermore, pretreatment with SR-18292 reversed the neuroprotective and antioxidant roles of SS31. These significant beneficial effects were associated with the activation of the Nrf2 signaling and PGC-1α pathways and were antagonized by SR-18292 administration. Our findings reveal that SS31 exhibits its neuroprotective activity by reversing mitochondrial dysfunction via activating the Nrf2 signaling pathway, which could be mediated through PGC-1α activation.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Haikou, 570102, Hainan Province, China
| | - Ruiming Shen
- Department of Rheumatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Emmanuel C Makale
- Department of General Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Wangwang Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Haikou, 570102, Hainan Province, China
| | - Zhenggang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Haikou, 570102, Hainan Province, China
| | - Qiuhu Huang
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Haikou, 570102, Hainan Province, China.
| |
Collapse
|
36
|
Hassanein EHM, Althagafy HS, Atwa AM, Kozman MR, Kotb El-Sayed MI, Soubh AA. Taurine attenuated methotrexate-induced intestinal injury by regulating NF-κB/iNOS and Keap1/Nrf2/HO-1 signals. Life Sci 2022; 311:121180. [PMID: 36370869 DOI: 10.1016/j.lfs.2022.121180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/28/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
Methotrexate (MTX) is a well-known and widely used cytotoxic chemotherapeutic agent. However, intestinal mucosa damage is a serious adverse effect of MTX. Taurine (TUR) is a sulfur-containing free β-amino acid with antioxidant and therapeutic value against several diseases. The current study aimed to determine the protective effect of TUR against MTX-induced intestinal injury. Rats were allocated into four groups. The first group received vehicles only. The second group received TUR at a dose of 250 mg/kg i.p. For induction of intestinal injury, the rats in the third group were given MTX once at a dose of 20 mg/kg, i.p. The fourth group received TUR 7 days before and 7 days after MTX, as previously described. TUR significantly attenuated the cytokine release by suppressing NF-κB and iNOS expressions. Moreover, cotreatment with TUR attenuated the increased MDA level while it enhanced the antioxidant GSH and SOD levels mediated by effective downregulation of Keap1 expression, while the expression of Nrf2, HO-1, and cytoglobin were up-regulated. Additionally, TUR mitigated the apoptosis and proliferation indices by decreasing the elevated levels of intestinal PCNA and caspase-3. Finally, TUR potently increased the cytotoxic activity of MTX toward Caco-2, MCF-7, and A549 cancer cells. In conclusion, TUR was a promising agent for relieving MTX-mediated intestinal injury via various antioxidant, anti-inflammatory, and antiapoptotic mechanisms.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt.
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Magy R Kozman
- Department of Clinical Pharmacology, Faculty of Pharmacy, Misr University for Science and Technology, Cairo, Egypt
| | - Mohamed I Kotb El-Sayed
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Ain Helwan, Helwan, Cairo, Egypt
| | - Ayman A Soubh
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, Giza 12566, Egypt
| |
Collapse
|
37
|
ChemR23 signaling ameliorates cognitive impairments in diabetic mice via dampening oxidative stress and NLRP3 inflammasome activation. Redox Biol 2022; 58:102554. [PMID: 36446229 PMCID: PMC9703827 DOI: 10.1016/j.redox.2022.102554] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Diabetes mellitus is associated with cognitive impairment characterized by memory loss and cognitive inflexibility. Recent studies have revealed that ChemR23 is implicated in both diabetes mellitus and Alzheimer's disease. However, the impact of ChemR23 on diabetes-associated cognitive impairment remains elusive. In this study, we explored the longitudinal changes of ChemR23 expression and cognitive function in STZ-induced type 1 diabetic mice and leptin receptor knockout type 2 diabetic mice at different ages. We also treated diabetic mice with ChemR23 agonists RvE1 or chemerin-9 to explore whether ChemR23 activation could alleviate diabetes-associated cognitive impairment. The underlying mechanism was further investigated in diabetic mice with genetic deletion of ChemR23. The results showed that ChemR23 expression was decreased along with aging and the progression of diabetes, suggesting that abnormal ChemR23 signaling may be involved in diabetes-associated cognitive impairment. Administration of RvE1 or chemerin-9 ameliorated oxidative stress and inhibited NLRP3 inflammasome activation through Nrf2/TXNIP pathway, and ultimately alleviated cognitive impairment in diabetic mice. Depletion of ChemR23 in diabetic mice abolished the beneficial effects of RvE1 and chemerin-9, and exacerbated cognitive impairment via increasing oxidative stress and activating NLRP3 inflammasome. Collectively, our data highlight the crucial role of ChemR23 signaling in diabetes-associated cognitive impairment via regulating oxidative stress and NLRP3 inflammasome, and targeting ChemR23 may serve as a promising novel strategy for the treatment of diabetes-associated cognitive impairment.
Collapse
|
38
|
Hassan SSU, Samanta S, Dash R, Karpiński TM, Habibi E, Sadiq A, Ahmadi A, Bungau S. The neuroprotective effects of fisetin, a natural flavonoid in neurodegenerative diseases: Focus on the role of oxidative stress. Front Pharmacol 2022; 13:1015835. [PMID: 36299900 PMCID: PMC9589363 DOI: 10.3389/fphar.2022.1015835] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/08/2022] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress (OS) disrupts the chemical integrity of macromolecules and increases the risk of neurodegenerative diseases. Fisetin is a flavonoid that exhibits potent antioxidant properties and protects the cells against OS. We have viewed the NCBI database, PubMed, Science Direct (Elsevier), Springer-Nature, ResearchGate, and Google Scholar databases to search and collect relevant articles during the preparation of this review. The search keywords are OS, neurodegenerative diseases, fisetin, etc. High level of ROS in the brain tissue decreases ATP levels, and mitochondrial membrane potential and induces lipid peroxidation, chronic inflammation, DNA damage, and apoptosis. The subsequent results are various neuronal diseases. Fisetin is a polyphenolic compound, commonly present in dietary ingredients. The antioxidant properties of this flavonoid diminish oxidative stress, ROS production, neurotoxicity, neuro-inflammation, and neurological disorders. Moreover, it maintains the redox profiles, and mitochondrial functions and inhibits NO production. At the molecular level, fisetin regulates the activity of PI3K/Akt, Nrf2, NF-κB, protein kinase C, and MAPK pathways to prevent OS, inflammatory response, and cytotoxicity. The antioxidant properties of fisetin protect the neural cells from inflammation and apoptotic degeneration. Thus, it can be used in the prevention of neurodegenerative disorders.
Collapse
Affiliation(s)
- Syed Shams ul Hassan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- Department of Natural Product Chemistry, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Saptadip Samanta
- Department of Physiology, Midnapore College, Midnapore, West Bengal, India
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, South Korea
| | - Tomasz M. Karpiński
- Department of Medical Microbiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Emran Habibi
- Department of Pharmacognosy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdul Sadiq
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Centre, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
39
|
Khayatan D, Razavi SM, Arab ZN, Niknejad AH, Nouri K, Momtaz S, Gumpricht E, Jamialahmadi T, Abdolghaffari AH, Barreto GE, Sahebkar A. Protective effects of curcumin against traumatic brain injury. Biomed Pharmacother 2022; 154:113621. [PMID: 36055110 DOI: 10.1016/j.biopha.2022.113621] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 02/06/2023] Open
|
40
|
Peng S, Hou Y, Chen Z. Hispolon alleviates oxidative damage by stimulating the Nrf2 signaling pathway in PC12 cells. Arch Biochem Biophys 2022; 727:109303. [PMID: 35660410 DOI: 10.1016/j.abb.2022.109303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
Natural products derived from the daily diet are garnering increasing attention for neurodegenerative disease (ND) treatment. Hispolon (His), a small molecule from Phellinus linteus, has been reported to have various pharmacological activities. Here, we evaluated its protective effect on a neuron-like rat pheochromocytoma cell line (PC12). Results showed that His could restore cell death induced by oxidative damage. Nuclear factor-erythroid 2 (NF-E2)-related factor 2 (Nrf2) plays a significant role in maintaining cellular redox homeostasis. After treatment with His, some Nrf2-governed antioxidant genes were upregulated in a dose-dependent manner. However, the protective effect of His on PC12 cells was easily terminated by Nrf2 knockdown, demonstrating that Nrf2 is a critical component in this cytoprotective process. Taken together, our study showed that His was not only an effective activator of Nrf2 but also a promising candidate for ND treatment.
Collapse
Affiliation(s)
- Shoujiao Peng
- Department of General Surgery, Xiangya Hospital, Central South University (CSU), Changsha, Hunan, China; State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China; Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Yanan Hou
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Zihua Chen
- Department of General Surgery, Xiangya Hospital, Central South University (CSU), Changsha, Hunan, China.
| |
Collapse
|
41
|
Salem M, Shaheen M, Borjac J. Crocin suppresses inflammation-induced apoptosis in rmTBI mouse model via modulation of Nrf2 transcriptional activity. PHARMANUTRITION 2022. [DOI: 10.1016/j.phanu.2022.100308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Lin J, Niu Z, Xue Y, Gao J, Zhang M, Li M, Peng Y, Zhang S, Li W, Zhang Q, Li X. Chronic vitamin D 3 supplementation alleviates cognition impairment via inhibition of oxidative stress regulated by PI3K/AKT/Nrf2 in APP/PS1 transgenic mice. Neurosci Lett 2022; 783:136725. [PMID: 35697158 DOI: 10.1016/j.neulet.2022.136725] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/24/2022] [Accepted: 06/08/2022] [Indexed: 10/18/2022]
Abstract
Oxidative stress plays essential role in the pathogenesis of Alzheimer's disease, and vitamin D3 (VD3) is a nutrient with neuroprotective and antioxidant activities. The present study aimed to confirm the neuroprotective effect and the ameliorative effect of cortical oxidative stress of VD3 in APP/PS1 transgenic mice. APP/PS1 mice were treated with VD3 for 20 weeks. After treatment, Morris Water Maze test was used to evaluate cognitive level. Western blotting was used to determine APP, p-tau, tau and PI3K/AKT/Nrf2 pathway-related protein expression levels. Immunohistochemical staining was performed to determine the levels of β amyloid peptide (Aβ) deposition. Enzyme linked immunosorbent assay was used to determine the 25(OH)D3 levels and oxidative stress status. Our results showed that treatment with VD3 ameliorated behavioral deficits of APP/PS1 mice. In addition, the administration of VD3 significantly increased the cortical 25(OH)D3 levels, while reducing the levels of cortical Aβ deposition and decreasing the expression levels of cortical APP, tau and p-tau in APP/PS1 mice. Moreover, VD3 protected the cortex against oxidative stress by enhancing the levels of superoxide dismutase, glutathione and total antioxidant capacity, and downregulating the malondialdehyde levels. Furthermore, VD3 clearly activated the PI3K/AKT/Nrf2 pathway, thereby elevating the expression levels of HO1 and NQO1. We concluded that VD3 improved cognitive function and cortical Alzheimer-like pathology of APP/PS1 mice, which may be related to the inhibition of oxidative stress via activation the PI3K/AKT/Nrf2 pathway.
Collapse
Affiliation(s)
- Jisong Lin
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Zhuoya Niu
- Department of Occupational and Environmental Health Sciences, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yuan Xue
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Jiaojiao Gao
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Mimi Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Mengxin Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yangyang Peng
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Shenshen Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Wenjie Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Qian Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xing Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| |
Collapse
|
43
|
Vornoli A, Vizzarri F, Della Croce CM, Grande T, Palazzo M, Árvay J, Pucci L, Gabriele M, Matteucci M, Paolini M, Longo V, Pozzo L. The hypolipidemic, anti-inflammatory and antioxidant effect of Kavolì® aqueous extract, a mixture of Brassica oleracea leaves, in a rat model of NAFLD. Food Chem Toxicol 2022; 167:113261. [PMID: 35787436 DOI: 10.1016/j.fct.2022.113261] [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: 04/22/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/25/2022]
Abstract
Herein we characterized the bioactive metabolites of the aqueous extract of Kavolì®, a commercial product composed of a mixture of Brassica oleracea leaves, and assessed its potential ameliorating effects in a rat model of non-alcoholic fatty liver disease (NAFLD). Kavolì® extract showed high levels of bioactive compounds and strong in vitro antioxidant activities. Chlorogenic and neochlorogenic acids were identified as the most representative polyphenols. The administration of brassica extract to steatotic rats significantly ameliorated the levels of blood lipids and transaminases, and lipid content and inflammatory markers in liver. Oxidative stress parameters were significantly improved in both liver and brain of steatotic rats. Moreover, plasma and feces levels of short chain fatty acids (SCFAs) were bring back close to control values by Kavolì® treatment, in spite of high fat diet/streptozotocin (HFD/STZ)-induced alterations. The efficacy of Kavolì® in treating hypercholesterolemia, reducing the level of inflammation and cardiovascular disease biomarkers, steatosis and oxidative stress parameters, as well as the ability in modulating SCFAs levels is probably related to the bioactive compounds of the water extract administered to the rat model of NAFLD. In particular, the ameliorating effects are largely attributable to the high content in polyphenols observed in our study.
Collapse
Affiliation(s)
- Andrea Vornoli
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124, Pisa, Italy
| | - Francesco Vizzarri
- Department of Agro-Environmental and Territorial Sciences (Disaat), University of Bari, Campus - Via Orabona 4, 70125, Bari, Italy; National Agricultural and Food Centre Nitra, Hlohovecká 2, 95141, Lužianky, Slovak Republic
| | - Clara Maria Della Croce
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124, Pisa, Italy
| | - Teresa Grande
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124, Pisa, Italy
| | - Marisa Palazzo
- Department of Agricultural, Environmental and Food Sciences, University of Molise, Via De Sanctissnc, 86100, Campobasso, Italy
| | - Július Árvay
- Institute of Food Sciences, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra. Tr. A Hlinku 2, 949 76, Nitra, Slovak Republic
| | - Laura Pucci
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124, Pisa, Italy
| | - Morena Gabriele
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124, Pisa, Italy
| | - Marco Matteucci
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà, 33, 56127, Pisa, Italy
| | - Moreno Paolini
- Department of Pharmacology and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Zamboni, 33, 40126, Bologna, Italy
| | - Vincenzo Longo
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124, Pisa, Italy
| | - Luisa Pozzo
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124, Pisa, Italy.
| |
Collapse
|
44
|
Effects of Selen on the Antidepressant-like Activity of Agents Affecting the Adenosinergic Neurotransmission. Metabolites 2022; 12:metabo12070586. [PMID: 35888708 PMCID: PMC9316035 DOI: 10.3390/metabo12070586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 02/04/2023] Open
Abstract
The main goal of this study was to determine the antidepressant-like potential of the co-administration of sodium selenite (Se) and the selective adenosine A1 and A2A antagonists DPCPX and istradefylline (IST), respectively, in mice despair tests. Biochemical studies were performed to elucidate the action mechanisms of the investigated treatment strategies. The results confirmed that, when administered by itself, Se exerts an antidepressant-like effect in the FST and TST and that this activity is dose-dependent. Further experiments demonstrated that Se (0.25 mg/kg) significantly enhanced the activity of mice in both tests when co-administered with DPCPX (1 mg/kg) and IST (0.5 mg/kg) at doses which would be ineffective if administered individually. Our research revealed that neither DPCPX, IST, nor Se or combinations of the tested substances induced significant changes in the brain-derived neurotrophic factor (BDNF) levels in mice serum vs. the NaCl-treated group. However, we observed a decrease in the mRNA level of antioxidant defense enzymes. Molecular studies also showed changes in the expression of the Slc6a15, Comt, and Adora1 genes, particularly after exposure to the combination of Se and DPCPX, which indicates a beneficial effect and may help to explain the key mechanism of the antidepressant effect. The combination of Se with substances attenuating adenosine neurotransmission may become a new therapeutic strategy for patients with depression.
Collapse
|
45
|
Lupeol Treatment Attenuates Activation of Glial Cells and Oxidative-Stress-Mediated Neuropathology in Mouse Model of Traumatic Brain Injury. Int J Mol Sci 2022; 23:ijms23116086. [PMID: 35682768 PMCID: PMC9181489 DOI: 10.3390/ijms23116086] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 02/05/2023] Open
Abstract
Traumatic brain injury (TBI) signifies a major cause of death and disability. TBI causes central nervous system (CNS) damage under a variety of mechanisms, including protein aggregation, mitochondrial dysfunction, oxidative stress, and neuroinflammation. Astrocytes and microglia, cells of the CNS, are considered the key players in initiating an inflammatory response after injury. Several evidence suggests that activation of astrocytes/microglia and ROS/LPO have the potential to cause more harmful effects in the pathological processes following traumatic brain injury (TBI). Previous studies have established that lupeol provides neuroprotection through modulation of inflammation, oxidative stress, and apoptosis in Aβ and LPS model and neurodegenerative disease. However, the effects of lupeol on apoptosis caused by inflammation and oxidative stress in TBI have not yet been investigated. Therefore, we explored the role of Lupeol on antiapoptosis, anti-inflammatory, and antioxidative stress and its potential mechanism following TBI. In these experiments, adult male mice were randomly divided into four groups: control, TBI, TBI+ Lupeol, and Sham group. Western blotting, immunofluorescence staining, and ROS/LPO assays were performed to investigate the role of lupeol against neuroinflammation, oxidative stress, and apoptosis. Lupeol treatment reversed TBI-induced behavioral and memory disturbances. Lupeol attenuated TBI-induced generation of reactive oxygen species/lipid per oxidation (ROS/LPO) and improved the antioxidant protein level, such as nuclear factor erythroid 2-related factor 2 (Nrf2) and heme-oxygenase 1 (HO-1) in the mouse brain. Similarly, our results indicated that lupeol treatment inhibited glial cell activation, p-NF-κB, and downstream signaling molecules, such as TNF-α, COX-2, and IL-1β, in the mouse cortex and hippocampus. Moreover, lupeol treatment also inhibited mitochondrial apoptotic signaling molecules, such as caspase-3, Bax, cytochrome-C, and reversed deregulated Bcl2 in TBI-treated mice. Overall, our study demonstrated that lupeol inhibits the activation of astrocytes/microglia and ROS/LPO that lead to oxidative stress, neuroinflammation, and apoptosis followed by TBI.
Collapse
|
46
|
Therapeutic Potential of Phlorotannin-Rich Ecklonia cava Extract on Methylglyoxal-Induced Diabetic Nephropathy in In Vitro Model. Mar Drugs 2022; 20:md20060355. [PMID: 35736158 PMCID: PMC9229597 DOI: 10.3390/md20060355] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 11/22/2022] Open
Abstract
Advanced glycation end-products (AGEs) play a vital role in the pathogenesis of diabetic complications. Methylglyoxal (MGO), one of the major precursors of AGEs, is a highly reactive dicarbonyl compound that plays an important role in the pathogenesis of diabetic nephropathy. This study was designed to evaluate the therapeutic potential of phlorotannin-rich Ecklonia cava extract (ECE) on MGO-induced diabetic nephropathy in in vitro models using mouse glomerular mesangial cells. ECE showed anti-glycation activity via breaking of AGEs-collagen cross-links and inhibition of AGEs formation and AGE-collagen cross-linking formation. The renoprotective effects were determined by assessing intracellular reactive oxygen species (ROS) and MGO accumulation, cell apoptosis, and the Nrf-2/ARE signaling pathway. MGO-induced renal damage, intracellular ROS production level, and MGO-protein adduct accumulation were significantly decreased by pretreating ECE. Moreover, ECE pretreatment exhibited preventive properties against MGO-induced dicarbonyl stress via activation of the Nrf2/ARE signaling pathway and reduction of RAGE protein expression in mouse glomerular mesangial cells. Collectively, these results indicated potential anti-glycation properties and prominent preventive effects of ECE against MGO-induced renal damage. Additionally, ECE may be utilized for the management of AGE-related diabetic nephropathy.
Collapse
|
47
|
Neuroprotective Effects and Therapeutic Potential of the Citrus Flavonoid Hesperetin in Neurodegenerative Diseases. Nutrients 2022; 14:nu14112228. [PMID: 35684025 PMCID: PMC9183194 DOI: 10.3390/nu14112228] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative disorders affect more than fifty million Americans each year and represent serious health threats as the population ages. Neuroinflammation and oxidative stress are critical in the onset, progression, and pathogenesis of neurodegenerative diseases such as Alzheimer’s (AD), Parkinson’s (PD), and amyotrophic lateral sclerosis (ALS). A wide range of natural compounds has been investigated because of their antioxidant, anti-inflammatory, and neuroprotective properties. The citrus flavonoid hesperetin (HPT), an aglycone of hesperidin found in oranges, mandarins, and lemons, has been extensively reported to exert neuroprotective effects in experimental models of neurogenerative diseases. This review has compiled multiple studies on HPT in both in vivo and in vitro models to study neurodegeneration. We focused on the modulatory effects of hesperetin on the release of cellular anti-inflammatory and antioxidative stress mediators. Additionally, this review discusses the hesperetin effect in maintaining the levels of microRNA (miRNA) and modulating autophagy as it relates to hesperetin’s protective mechanisms against neurodegeneration. Moreover, this review is focused on providing experimental data for hesperetin’s potential as a neuroprotective compound and discusses reported evidence that HPT crosses the blood–brain barrier. In summary, this review shows the evidence available in the literature to indicate the efficacy of hesperetin in delaying the onset of neurodegenerative diseases.
Collapse
|
48
|
Yang S, Park SH, Oh SW, Kwon K, Yu E, Lee CW, Son YK, Kim C, Lee BH, Cho JY, Kim YJ, Lee J. Antioxidant Activities and Mechanisms of Tomentosin in Human Keratinocytes. Antioxidants (Basel) 2022; 11:antiox11050990. [PMID: 35624854 PMCID: PMC9137523 DOI: 10.3390/antiox11050990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 11/23/2022] Open
Abstract
Tomentosin, one of natural sesquiterpene lactones sourced from Inula viscosa L., exerts therapeutic effects in various cell types. Here, we investigated the antioxidant activities and the underlying action mechanisms of tomentosin in HaCaT cells (a human keratinocyte cell line). Specifically, we examined the involvement of tomentosin in aryl hydrocarbon receptor (AhR) and nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathways. Treatment with tomentosin for up to 60 min triggered the production of reactive oxygen species (ROS), whereas treatment for 4 h or longer decreased ROS production. Tomentosin treatment also induced the nuclear translocation of Nrf2 and upregulated the expression of Nrf2 and its target genes. These data indicate that tomentosin induces ROS production at an early stage which activates the Nrf2 pathway by disrupting the Nrf2–Keap1 complex. However, at a later stage, ROS levels were reduced by tomentosin-induced upregulation of antioxidant genes. In addition, tomentosin induced the phosphorylation of mitogen-activated protein kinases (MAPKs) including p38 MAPK and c-Jun N-terminal kinase (JNK). SB203580 (a p38 MAPK inhibitor) and SP600125 (a JNK inhibitor) attenuated the tomentosin-induced phosphorylation of Nrf2, suggesting that JNK and p38 MAPK signaling pathways can contribute to the tomentosin-induced Nrf2 activation through phosphorylation of Nrf2. Furthermore, N-acetyl-L-cysteine (NAC) treatment blocked both tomentosin-induced production of ROS and the nuclear translocation of Nrf2. These data suggest that tomentosin-induced Nrf2 signaling is mediated both by tomentosin-induced ROS production and the activation of p38 MAPK and JNK. Moreover, tomentosin inhibited the AhR signaling pathway, as evidenced by the suppression of xenobiotic-response element (XRE) reporter activity and the translocation of AhR into nucleus induced by urban pollutants, especially benzo[a]pyrene. These findings suggest that tomentosin can ameliorate skin damage induced by environmental pollutants.
Collapse
Affiliation(s)
- Seyoung Yang
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City 16419, Gyunggi Do, Korea; (S.Y.); (S.W.O.); (K.K.); (E.Y.)
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong City 30016, Korea;
| | - Sae Woong Oh
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City 16419, Gyunggi Do, Korea; (S.Y.); (S.W.O.); (K.K.); (E.Y.)
| | - Kitae Kwon
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City 16419, Gyunggi Do, Korea; (S.Y.); (S.W.O.); (K.K.); (E.Y.)
| | - Eunbi Yu
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City 16419, Gyunggi Do, Korea; (S.Y.); (S.W.O.); (K.K.); (E.Y.)
| | - Chae Won Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Korea; (C.W.L.); (Y.K.S.); (C.K.); (B.-H.L.)
| | - Youn Kyoung Son
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Korea; (C.W.L.); (Y.K.S.); (C.K.); (B.-H.L.)
| | - Changmu Kim
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Korea; (C.W.L.); (Y.K.S.); (C.K.); (B.-H.L.)
| | - Byoung-Hee Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Korea; (C.W.L.); (Y.K.S.); (C.K.); (B.-H.L.)
| | - Jae Youl Cho
- Molecular Immunology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City 16419, Gyunggi Do, Korea
- Correspondence: (J.Y.C.); (Y.-J.K.); (J.L.); Tel.: +82-31-290-7861 (J.L.)
| | - Youn-Jung Kim
- Department of Marine Sciences, Incheon National University, Incheon 22012, Korea
- Correspondence: (J.Y.C.); (Y.-J.K.); (J.L.); Tel.: +82-31-290-7861 (J.L.)
| | - Jongsung Lee
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City 16419, Gyunggi Do, Korea; (S.Y.); (S.W.O.); (K.K.); (E.Y.)
- Correspondence: (J.Y.C.); (Y.-J.K.); (J.L.); Tel.: +82-31-290-7861 (J.L.)
| |
Collapse
|
49
|
Bao TRG, Long GQ, Wang Y, Wang Q, Liu XL, Hu GS, Gao XX, Wang AH, Jia JM. New Lanostane-Type Triterpenes with Anti-Inflammatory Activity from the Epidermis of Wolfiporia cocos. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:4418-4433. [PMID: 35347990 DOI: 10.1021/acs.jafc.2c00823] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
A chemical study on the epidermis of cultivated edible mushroom Wolfiporia cocos resulted in the isolation and identification of 46 lanostane triterpenoids, containing 17 new compounds (1-17). An experimental determination of their anti-inflammatory activity showed that poricoic acid GM (39) most strongly inhibited NO production in LPS-induced RAW264.7 murine macrophages with an IC50 value at 9.73 μM. Furthermore, poricoic acid GM induced HO-1 protein expression and inhibited iNOS and COX2 protein expression as well as the release of PGE2, IL-1β, IL-6, TNF-α, and reactive oxygen species (ROS) in LPS-induced RAW264.7 cells. Mechanistically, poricoic acid GM suppressed the phosphorylation of the IκBα protein, which prevented NF-κB from entering the nucleus to lose transcriptional activity and inhibited the dissociation of Keap1 from Nrf2, thereby activating Nrf2 into the nucleus to regulate antioxidant genes. Furthermore, the MAPK signaling pathway may play a significant role in poricoic acid GM-induced elimination of inflammation. This work further confirms that lanostane triterpenoids are key ingredients responsible for the anti-inflammatory properties of the edible medicinal mushroom W. cocos.
Collapse
Affiliation(s)
- Te-Ri-Gen Bao
- Teaching and Research Department of Chinese Materia Medica Resources, College of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Guo-Qing Long
- Teaching and Research Department of Chinese Materia Medica Resources, College of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yong Wang
- Teaching and Research Department of Chinese Materia Medica Resources, College of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Qian Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Xuan-Li Liu
- Teaching and Research Department of Chinese Materia Medica Resources, College of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Gao-Sheng Hu
- Teaching and Research Department of Chinese Materia Medica Resources, College of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Xiao-Xu Gao
- Teaching and Research Department of Chinese Materia Medica Resources, College of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - An-Hua Wang
- Teaching and Research Department of Chinese Materia Medica Resources, College of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Jing-Ming Jia
- Teaching and Research Department of Chinese Materia Medica Resources, College of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| |
Collapse
|
50
|
Dai Z, An LY, Chen XY, Yang F, Zhao N, Li CC, Ren R, Li BY, Tao WY, Li P, Jiang C, Yan F, Jiang ZY, You QD, Di B, Xu LL. Target Fishing Reveals a Novel Mechanism of 1,2,4-Oxadiazole Derivatives Targeting Rpn6, a Subunit of 26S Proteasome. J Med Chem 2022; 65:5029-5043. [PMID: 35253427 DOI: 10.1021/acs.jmedchem.1c02210] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
1,2,4-Oxadiazole derivatives, a class of Nrf2-ARE activators, exert an extensive therapeutic effect on inflammation, cancer, neurodegeneration, and microbial infection. Among these analogues, DDO-7263 is the most potent Nrf2 activator and used as the core structure for bioactive probes to explore the precise mechanism. In this work, we obtained compound 7, a mimic of DDO-7263, and biotin-labeled and fluorescein-based probes, which exhibited homologous biological activities to DDO-7263, including activating Nrf2 and its downstream target genes, anti-oxidative stress, and anti-inflammatory effects. Affinity chromatography and mass analysis techniques revealed Rpn6 as the potential target protein regulating the Nrf2 signaling pathway. In vitro affinity experiments further confirmed that DDO-7263 upregulated Nrf2 through binding to Rpn6 to block the assembly of 26S proteasome and the subsequent degradation of ubiquitinated Nrf2. These results indicated that Rpn6 is a promising candidate target to activate the Nrf2 pathway for protecting cells and tissues from oxidative, electrophilic, and exogenous microbial stimulation.
Collapse
Affiliation(s)
- Zhen Dai
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Lu-Yan An
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Yi Chen
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Fan Yang
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Ni Zhao
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Cui-Cui Li
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Ren Ren
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Bing-Yan Li
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Wei-Yan Tao
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Pei Li
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng Jiang
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Fang Yan
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Zheng-Yu Jiang
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Qi-Dong You
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Bin Di
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Li-Li Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|