1
|
Gong P, Wang J, Long H, Yang W, Chen X, Li N, Chen F, Zhang J, Guo Y. Edible and Medicinal Fungi as Candidate Natural Antidepressants: Mechanisms and Nutritional Implications. Mol Nutr Food Res 2025:e70080. [PMID: 40289452 DOI: 10.1002/mnfr.70080] [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: 12/02/2024] [Revised: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
This systematic review examines the antidepressant potential of edible medicinal fungi (EMFs), focusing on their bioactive compounds and mechanisms of action. EMFs modulate neurotransmitter systems, including serotonin (5-HT) and dopamine (DA), alleviating depressive symptoms. Extracts from EMFs, such as Ganoderma lucidum, Hericium erinaceus, Poria cocos, and Cordyceps militaris, demonstrate significant antidepressant-like effects in preclinical studies. Their bioactive compounds influence the tryptophan-kynurenine (KYN) pathway, regulate the hypothalamus-pituitary-adrenal (HPA) axis, and reduce neuroinflammation, all of which are linked to stress response and mood regulation. The review also explores the gut-brain axis, highlighting how EMF-derived polysaccharides improve gut health by modulating microbiota, potentially mitigating depressive symptoms. Additionally, it discusses the use of EMFs in functional foods and dietary supplements, innovations like 3D food printing for depression-related issues, and synthetic biology for enhancing compound production. Artificial intelligence is used to model complex mechanisms. However, challenges remain, such as standardization and lack of clinical validation. Future research should address these gaps, emphasizing personalized interventions and advanced technologies for next-generation antidepressant foods.
Collapse
Affiliation(s)
- Pin Gong
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, China
- Key Laboratory of Precision Nutrition and Functional Product Development in Xi'an, Shaanxi University of Science and Technology, Xi'an, China
| | - Jiating Wang
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, China
- Key Laboratory of Precision Nutrition and Functional Product Development in Xi'an, Shaanxi University of Science and Technology, Xi'an, China
| | - Hui Long
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, China
- Key Laboratory of Precision Nutrition and Functional Product Development in Xi'an, Shaanxi University of Science and Technology, Xi'an, China
| | - Wenjuan Yang
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, China
- Key Laboratory of Precision Nutrition and Functional Product Development in Xi'an, Shaanxi University of Science and Technology, Xi'an, China
| | - Xuefeng Chen
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, China
- Key Laboratory of Precision Nutrition and Functional Product Development in Xi'an, Shaanxi University of Science and Technology, Xi'an, China
| | - Nan Li
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, China
- Key Laboratory of Precision Nutrition and Functional Product Development in Xi'an, Shaanxi University of Science and Technology, Xi'an, China
| | - Fuxin Chen
- School of Chemistry and Chemical Engineering, Xi'an University of Science and Technology, Xi'an, China
| | - Jie Zhang
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuxi Guo
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, China
- Key Laboratory of Precision Nutrition and Functional Product Development in Xi'an, Shaanxi University of Science and Technology, Xi'an, China
| |
Collapse
|
2
|
He L, Zhang Y, Li J, Chen D, Yue S, Liu Y, Guo Y, Wang Y, Xiu M, He J. Dunhuang Dabupi Decoction and its active components alleviate ulcerative colitis by activating glutathione metabolism and inhibiting JAK-STAT pathway in Drosophila and mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119717. [PMID: 40164365 DOI: 10.1016/j.jep.2025.119717] [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: 02/08/2025] [Revised: 03/22/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dabupi Decoction (DBPD) originates from the ancient Dunhuang medical literature "Fu Xing Jue Visceral to Drug law legend" for more than 1000 years, which has been extensively employed to treat various diseases related to the spleen and stomach. However, limited studies focus on the mechanism of DBPD against ulcerative colitis (UC). AIM OF THE STUDY The beneficial effect and mechanism of DBPD against UC were detected by adopting both Drosophila melanogaster and C57BL/6J mouse models. METHODS The protective effect of DBPD against DSS-induced intestinal damage in flies was investigated by utilizing survival rate, locomotion, excretion, smurf, intestinal length, intestinal acid-base homeostasis, and Tepan blue assay. In mice, HE staining and ELISA kit were employed to assess serum histopathological damage and inflammatory factor levels. Subsequently, the molecular mechanism of DBPD was subsequently detected via DHE staining, immunofluorescence, transmission electron microscopy (TEM), real-time PCR, and transcriptomic sequencing. Additionally, liquid chromatography-mass spectrometry (LC-MS) and phenotype experiments in UC flies were utilized to identify the bioactive components of DBPD against UC. RESULTS Oral administration of DBPD remarkably alleviated DSS-induced body damage in flies by improving survival rate, locomotion, and excretion. It also remarkably rescued intestinal morphological damage, repaired acid-base homeostatic imbalance, inhibited intestinal epithelial cells (IECs) death and excessive proliferation of intestinal stem cells (ISCs), and improved ultrastructural damage of IECs in flies treated with DSS. Consistently, DBPD attenuated colitis symptoms, alleviated intestinal histopathological damage, and restored the expression of inflammatory factors in DSS-induced UC mice. As suggested by an integration of transcriptome data with molecular biology experiments, DBPD not only dramatically alleviated oxidative damage by activating the glutathione metabolic pathway, but also lowered inflammatory reaction by inhibiting the JAK-STAT pathway. Additionally, four compounds of DBPD, rhein acid, isoquercitrin, curcumin, and zeaxanthin were identified to alleviate the DSS-induced intestinal injury. CONCLUSION DBPD demonstrate immense potential for intestinal injury predominantly by activating the glutathione metabolic pathway to alleviate oxidative damage, and inhibiting the JAK-STAT pathway to mitigate inflammatory response. Rhein acid, isoquercitrin, curcumin, and zeaxanthin were the bioactive compounds of DBPD against UC.
Collapse
Affiliation(s)
- Li He
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Yongxuan Zhang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Jiangnan Li
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Dandan Chen
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Shiqi Yue
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Yongqi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, 730000, China.
| | - Yaqiong Guo
- Second Provincial People's Hospital of Gansu, Lanzhou, 730000, China.
| | - Yan Wang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Minghui Xiu
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, 730000, China.
| | - Jianzheng He
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, 730000, China; Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| |
Collapse
|
3
|
Qiu B, Boudker O. Structural basis of excitatory amino acid transporter 3 substrate recognition. Proc Natl Acad Sci U S A 2025; 122:e2501627122. [PMID: 40249774 PMCID: PMC12036983 DOI: 10.1073/pnas.2501627122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/28/2025] [Indexed: 04/20/2025] Open
Abstract
Excitatory amino acid transporters (EAATs) reside on cell surfaces and uptake substrates, including L-glutamate, L-aspartate, and D-aspartate, using ion gradients. Among five EAATs, EAAT3 is the only isoform that can efficiently transport L-cysteine, a substrate for glutathione synthesis. Recent studies suggest that EAAT3 also transports the oncometabolite R-2-hydroxyglutarate (R-2HG). Here, we examined the structural basis of substrate recognition by determining the cryogenic electron microscopy (cryo-EM) structures of EAAT3 bound to different substrates. We found that L-cysteine binds to EAAT3 in thiolate form, and EAAT3 recognizes different substrates by fine-tuning local conformations of the coordinating residues. However, using purified human EAAT3, we could not observe R-2HG binding or transport. Imaging of EAAT3 bound to L-cysteine revealed several conformational states, including an outward-facing state with a semi-open gate and a disrupted sodium-binding site. These structures demonstrate that the full gate closure, coupled with the binding of the last sodium ion, occurs after substrate binding. Furthermore, we observed that different substrates affect how the transporter distributes between a fully outward-facing conformation and intermediate occluded states on a path to the inward-facing conformation, suggesting that translocation rates are substrate-dependent.
Collapse
Affiliation(s)
- Biao Qiu
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, NY10021
- HHMI, Weill Cornell Medicine, New York, NY10021
| | - Olga Boudker
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, NY10021
- HHMI, Weill Cornell Medicine, New York, NY10021
| |
Collapse
|
4
|
Fonseca-Fonseca LA, Taño Portuondo LR, Ramírez-Sánchez J, Pavón Fuentes N, Mondelo Rodríguez A, Amaral da Silva VD, Lima Costa S, Núñez-Figueredo Y. JM-20 administration to animals with lesion of the nigrostriatal dopamine pathway induced by 6-hydroxydopamine, partially reverses motor damage and oxidative stress. Neurol Res 2025:1-10. [PMID: 40217565 DOI: 10.1080/01616412.2025.2490089] [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: 07/30/2024] [Accepted: 03/31/2025] [Indexed: 04/23/2025]
Abstract
INTRODUCTION Previous studies have shown that JM-20, a new chemical hybrid molecule, protects against rotenone and 6-hydroxydopamine (6-OHDA) neurotoxicity. Also, we demonstrated that JM-20 inhibit the formation of toxic alpha-synuclein aggregated species and aminochrome cytotoxicity. OBJECTIVE The present study sought to determine the neuroprotective property of JM-20 in animals with a partial lesion of the nigrostriatal dopamine pathway induced by 6-OHDA. METHODS For in vivo studies, adult male Wistar rats were lesioned in the right substantia nigra pars compacta (SNpc) with a 6-OHDA administration. Fifteen days after surgery, the animal's asymmetry levels were assessed. Those with asymmetry values higher than 50% were divided into two groups: animals that did not receive any treatment and those that were administered with JM-20 (40 mg/kg, intragastric via gavage) for 27 days. Every 7 days, the asymmetry values of the animals were analyzed until day 42 after the surgery. At the end of the experiment, the animals were euthanized, and the SNpc and striatum were taken out for the analysis of oxidative stress. RESULTS Our results reveal a behavioral function progressively recovered in the JM-20-treated animals, diminishing the percentage of motor asymmetry. Also, it improves some oxidative stress markers in the SNpc and the striatum of these animals. CONCLUSION Our study provides the preclinical evidence to support the long-term neuroprotective potential of JM-20 in 6-OHDA hemiparkinson rat model, pointing out to its possible use as a disease-modifying agent in PD.
Collapse
Affiliation(s)
- Luis Arturo Fonseca-Fonseca
- Laboratory of Experimental Neuropharmacology, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), La Habana, Cuba
| | - Laura Reina Taño Portuondo
- Laboratory of Experimental Neuropharmacology, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), La Habana, Cuba
- Universidad Autónoma de Yucatán, Mérida, México
| | - Jeney Ramírez-Sánchez
- Laboratory of Experimental Neuropharmacology, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), La Habana, Cuba
| | | | - Abel Mondelo Rodríguez
- Laboratory of Experimental Neuropharmacology, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), La Habana, Cuba
| | - Víctor Diogenes Amaral da Silva
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia (UFBA), Salvador Bahia, Brazil
| | - Silvia Lima Costa
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia (UFBA), Salvador Bahia, Brazil
| | - Yanier Núñez-Figueredo
- Laboratory of Experimental Neuropharmacology, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), La Habana, Cuba
| |
Collapse
|
5
|
Song J, Wang T, Hong JS, Wang Y, Feng J. TFEB-dependent autophagy-lysosomal pathway is required for NRF2-driven antioxidative action in obstructive sleep apnea-induced neuronal injury. Cell Signal 2025; 128:111630. [PMID: 39875050 PMCID: PMC11913475 DOI: 10.1016/j.cellsig.2025.111630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025]
Abstract
Nearly one billion individuals worldwide suffer from obstructive sleep apnea (OSA) and are potentially impacted by related neurodegeneration. TFEB is considered a master regulator of autophagy and lysosomal biogenesis, but little is known about its role in neuronal oxidative stress and resultant injury induced by OSA. This study aimed to investigate these issues. Here, we demonstrated that neuronal TFEB induction is repressed in OSA mouse models. Activation of a TFEB-dependent autophagy-lysosomal pathway (ALP) reduces hippocampal neuronal cell death and mitigates OSA-related cognitive impairment. Neuronal NRF2 induction was also found to be defective in OSA mouse models. A series of staining assays for HO1, SOD3, ROS, GSH, 8-OHdG, MDA and PI revealed that enhancement of NRF2 expression restores neuronal redox balance and protects hippocampal neurons. We then identified a novel interplay between TFEB-dependent ALP and NRF2-mediated relief of oxidative stress. Inhibition of NRF2 hinders TFEB expression and lysosomal biogenesis. Conversely, knockdown of TFEB or blocking autophagy dampens the antioxidative effect of NRF2. Our findings highlight the unexpected and crucial role of TFEB-dependent ALP as a downstream event of NRF2 in NRF2-promoted redox balance. This study provides novel insights into the mechanism behind NRF2-driven antioxidative action and the regulation of TFEB-dependent ALP.
Collapse
Affiliation(s)
- Junxiu Song
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China
| | - Tian Wang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China; Respiratory Department, Cangzhou People's Hospital, 061000, Hebei, China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, Durham, NC 27709, USA
| | - Yubao Wang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China.
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China.
| |
Collapse
|
6
|
Wu X, Ruan C, Zhu X, Zou L, Wang R, Li G. Copper-doped Lanthanide Coordination Polymers as Luminescent Nanoenzyme for Ratiometric Sensing of H 2O 2 and Glutathione. J Fluoresc 2025; 35:2059-2069. [PMID: 38483693 DOI: 10.1007/s10895-024-03659-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/01/2024] [Indexed: 04/30/2025]
Abstract
Design and fabrication of integrated multifunctional probes with intrinsic catalytic and detection abilities is of great importance to simplify the operation in biosensing application with high sensitivity. Herein, dual-emitting lanthanide coordination polymers (Ln-CPs) were facilely prepared by self-assembly of guanine diphosphate (GDP), terephthalic acid (TA), Tb3+ and Cu2+ designated as Tb/Cu-GDP/TA CPs. The doped Cu2+ endowed CPs with obviously enhanced peroxidase mimicking activity compared with free Cu2+. In the presence of H2O2, the probe catalyzed the oxidation of TA generating a new blue fluorescent product, while the fluorescence of Tb3+ decreased simultaneously. Therefore, a new sensitive ratiometric fluorescent sensor for H2O2 has been developed with a good linear range from 0.01 to 300 μM and limit of 1.62 nM. Moreover, the proposed platform could be extended to GSH ratiometric assay in the presence of H2O2, and interestingly, the detection performance could be easily adjusted by adding different concentration of H2O2. This work will facilitate the development of luminescent nanoenzymes based on Ln-CPs to construct the simple ratiomatric sensing platform.
Collapse
Affiliation(s)
- Xinru Wu
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Chen Ruan
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Xinyue Zhu
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Lina Zou
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Rong Wang
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Gaiping Li
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, PR China.
| |
Collapse
|
7
|
Sarawi WS, Alhusaini AM, Barwaished GS, Altamimi MM, Hasan IH, Aljarboa AS, Algarzae NK, Bakheet SA, Alhabardi SA, Ahmad SF. Indole-3-acetic acid and chenodeoxycholic acid attenuate TLR4/NF-κB signaling and endoplasmic reticulum stress in valproic acid-induced neurotoxicity. Front Pharmacol 2025; 16:1570125. [PMID: 40196372 PMCID: PMC11973296 DOI: 10.3389/fphar.2025.1570125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Valproic acid (VA) is a commonly prescribed medication for epilepsy and other neurological conditions. Although effective, VA use can lead to neurotoxicity, especially with chronic use. This study aimed to investigate the potential neuroprotective properties of indole-3-acetic acid (IAA) and chenodeoxycholic acid (CDCA) in an animal model of VA-induced brain injury. Rats received intraperitoneal injections of VA at a dose of 500 mg/kg/day for 3 weeks. Concurrently, they were orally treated with IAA (40 mg/kg/day) and/or CDCA (90 mg/kg/day). The results showed significantly increased oxidative stress and inflammation markers in the VA-exposed group indicated by the reduced levels of glutathione (GSH, P < 0.0001) and superoxide dismutase (SOD, P < 0.01) and the elevated inflammatory cytokines Interleukin-6 (IL-6, P < 0.0001) and tumor necrosis factor-alpha (TNFα, P < 0.01). VA also induced nuclear factor kappa B (NF-κB, P < 0.01), toll-like receptor 4 (TLR4, P < 0.05), and endoplasmic reticulum (ER) stress markers, as evidenced by increased immunoreactivity of GRP78 (glucose-regulated protein 78, P < 0.0001), transcription factor 6 (ATF-6, P < 0.05) and CHOP (C/EBP homologous protein, P < 0.0001). Treatment with IAA or CDCA attenuated VA-induced neurotoxicity, to a variable extent, by improving oxidative, inflammatory, and ER stress markers. This study demonstrates that IAA and CDCA exert protective effects against VA-induced neurotoxicity by mitigating oxidative stress, inflammation, and ER stress. Further investigations are recommended to validate these findings in other neurotoxicity models.
Collapse
Affiliation(s)
- Wedad S. Sarawi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahlam M. Alhusaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | | | | - Iman H. Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Amjad S. Aljarboa
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Norah K. Algarzae
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A. Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Samiah A. Alhabardi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Pickering AJ, Lamson NG, Marand MH, Straehla JP, Hammond PT. Convection-Enhanced Delivery of Auristatin-Conjugated Layer-by-Layer Nanoparticles for Glioblastoma Treatment. J Am Chem Soc 2025; 147:9457-9471. [PMID: 40062779 DOI: 10.1021/jacs.4c16898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Glioblastoma (GBM) has limited treatment options, as the restrictive blood-brain barrier (BBB) prevents most therapeutics from accumulating at sufficient levels in the brain. Convection-enhanced delivery (CED) offers a method for administering therapeutics directly into brain tumor tissue, but free drugs can be cleared rapidly and may be toxic to off-target cells. Drug-loaded nanoparticles (NPs) are a promising platform to prolong the residence time and improve cellular targeting of therapeutics. We designed drug-conjugated NPs comprising a liposomal core modified with a layer-by-layer (LbL) polymer coating to promote tumor penetration, retention, and tumor-selective cellular association. Covalent conjugation of the potent microtubule inhibitor monomethyl auristatin-F (MMAF) to lipid headgroups resulted in striking potency against a range of patient-derived GBM cell lines compared to free MMAF and outperformed an EGFR-targeted antibody-drug conjugate of MMAF under clinical investigation. In vivo, a single CED infusion of LbL-functionalized MMAF NPs in orthotopic GBM-bearing mice displayed improved distribution and retention of both the NPs and the MMAF payload within the tumor. The LbL coating promotes selective uptake by GBM cells and prolongs drug retention, overcoming limitations of rapid clearance associated with traditional CED approaches. This treatment inhibited tumor progression and significantly extended survival compared to free MMAF, MMAF-conjugated liposomes, and an EGFR-MMAF antibody-drug conjugate. This NP platform offers a promising strategy for enhancing local GBM therapy by improving drug exposure within tumors while minimizing systemic toxicity.
Collapse
Affiliation(s)
- Andrew J Pickering
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Nicholas G Lamson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Michael H Marand
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Joelle P Straehla
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, United States
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts 02115, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Paula T Hammond
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
9
|
Mao X, Fei X, Cai T, Xu S, Zhang D, Pu S, Li Z. A turn-on mitochondria-targeted iridium (Ⅲ) Complex-Based probe for glutathione detection and photodynamic therapy of cancer cells. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 329:125579. [PMID: 39689545 DOI: 10.1016/j.saa.2024.125579] [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: 11/01/2024] [Revised: 12/04/2024] [Accepted: 12/08/2024] [Indexed: 12/19/2024]
Abstract
As one of the most abundant biothiols in cells, glutathione (GSH) usually exists in a dynamic equilibrium of oxidized glutathione (GSSG) and reduces glutathione redox, and plays an essential reducing substance to maintain the REDOX balance of the microenvironment. So, the development of a reliable GSH sensor will be important for living cells and organisms. We fabricated a mitochondria targeted "turn-on" fluorescent sensor based on Ir (III) complex and successfully detected endogenous and exogenous GSH in living cells and zebrafish. For the probe Ir-DINI, a robust electron-withdrawing group 2,4-dinitrobenzoyl was introduced to quench the fluorescence, which could be broken through electrostatic interaction with GSH, following exposing a strong fluorescent Ir (Ⅲ) complex Ir-OH. On the other hand, photodynamic therapy (PDT) has attracted much attention in recent years due to its minimally invasive treatment. We found that singlet oxygen yields of probe Ir-DINI displayed an enhancement before and after the detection of GSH. Additionally, photodynamic studies in living cells illustrated that after reacting with GSH, probe Ir-DINI exhibited more obvious phototoxicity than before the detection of GSH. So the probe Ir-DINI could be served as a GSH sensor and potential GSH-activated photosensitizer for photodynamic therapy.
Collapse
Affiliation(s)
- Xueting Mao
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Xiao Fei
- Department of Gastroenterology, First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Tangxuan Cai
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Sha Xu
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Daobin Zhang
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| | - Shouzhi Pu
- Institute of Carbon Neutral New Energy Research, Yuzhang Normal University, Nanchang 330031, China.
| | - Zhijian Li
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| |
Collapse
|
10
|
Santos-Silva T, Lopes CFB, Hazar Ülgen D, Guimarães DA, Guimarães FS, Alberici LC, Sandi C, Gomes FV. Adolescent Stress-Induced Ventral Hippocampus Redox Dysregulation Underlies Behavioral Deficits and Excitatory/Inhibitory Imbalance Related to Schizophrenia. Schizophr Bull 2025; 51:501-512. [PMID: 38525594 PMCID: PMC11908863 DOI: 10.1093/schbul/sbae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
BACKGROUND AND HYPOTHESIS Redox dysregulation has been proposed as a convergent point of childhood trauma and the emergence of psychiatric disorders, such as schizophrenia (SCZ). A critical region particularly vulnerable to environmental insults during adolescence is the ventral hippocampus (vHip). However, the impact of severe stress on vHip redox states and their functional consequences, including behavioral and electrophysiological changes related to SCZ, are not entirely understood. STUDY DESIGN After exposing adolescent animals to physical stress (postnatal day, PND31-40), we explored social and cognitive behaviors (PND47-49), the basal activity of pyramidal glutamate neurons, the number of parvalbumin (PV) interneurons, and the transcriptomic signature of the vHip (PND51). We also evaluated the impact of stress on the redox system, including mitochondrial respiratory function, reactive oxygen species (ROS) production, and glutathione (GSH) levels in the vHip and serum. STUDY RESULTS Adolescent-stressed animals exhibited loss of sociability, cognitive impairment, and vHip excitatory/inhibitory (E/I) imbalance. Genome-wide transcriptional profiling unveiled the impact of stress on redox system- and synaptic-related genes. Stress impacted mitochondrial respiratory function and changes in ROS levels in the vHip. GSH and glutathione disulfide (GSSG) levels were elevated in the serum of stressed animals, while GSSG was also increased in the vHip and negatively correlated with sociability. Additionally, PV interneuron deficits in the vHip caused by adolescent stress were associated with oxidative stress. CONCLUSIONS Our results highlight the negative impact of adolescent stress on vHip redox regulation and mitochondrial function, which are partially associated with E/I imbalance and behavioral abnormalities related to SCZ.
Collapse
Affiliation(s)
- Thamyris Santos-Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Caio Fábio Baeta Lopes
- Department of Biomolecular Sciences, Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, Ribeirão Preto, Brazil
| | - Doğukan Hazar Ülgen
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Danielle A Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Francisco S Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luciane Carla Alberici
- Department of Biomolecular Sciences, Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, Ribeirão Preto, Brazil
| | - Carmen Sandi
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
11
|
Chang J, Liu D, Xiao Y, Tan B, Deng J, Mei Z, Liao J. Disulfidptosis: a new target for central nervous system disease therapy. Front Neurosci 2025; 19:1514253. [PMID: 40109666 PMCID: PMC11920580 DOI: 10.3389/fnins.2025.1514253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/27/2025] [Indexed: 03/22/2025] Open
Abstract
Disulfidptosis is a pathologic process that occurs under conditions of NADPH deficiency and excess disulfide bonds in cells that express high levels of SLC7A11. This process is caused by glucose deprivation-induced disulfide stress and was first described by cancer researchers. Oxidative stress is a hypothesized mechanism underlying diseases of the central nervous system (CNS), and disulfide stress is a specific type of oxidative stress. Proteins linked to disulfidptosis and metabolic pathways involved in disulfidptosis are significantly associated with diseases of the CNS (neurodegenerative disease, neurogliomas and ischemic stroke). However, the specific mechanism responsible for this correlation remains unknown. This review provides a comprehensive overview of the current knowledge regarding the origin elements, genetic factors, and signaling proteins involved in the pathogenesis of disulfidptosis. It demonstrates that the disruption of thiometabolism and disulfide stress play critical roles in CNS diseases, which are associated with the potential role of disulfidptosis. We also summarize disulfidptosis-related drugs and highlight potential therapeutic strategies for treating CNS diseases. Additionally, this paper suggests a testable hypothesis that might be a promising target for treating CNS diseases.
Collapse
Affiliation(s)
- Jing Chang
- College of Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Danhong Liu
- Institute of Clinical Pharmacology of Chinese Materia Medica, Hunan Academy of Chinese Medicine, Changsha, China
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Chinese Medicine), Changsha, China
| | - Yuqi Xiao
- College of Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Boyao Tan
- College of Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Jun Deng
- Department of Neurology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jun Liao
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
- Vascular Biology Laboratory, Medical College, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
12
|
Cheng S, Zhou W, Ren Y, Gao X, Cai D, Li M, Zhou Z, Wang Z, Abliz Z. Spatial metabolic modulation in vascular dementia by Erigeron breviscapus injection using ambient mass spectrometry imaging. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156412. [PMID: 39889490 DOI: 10.1016/j.phymed.2025.156412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/16/2024] [Accepted: 01/19/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Vascular dementia (VaD), a significant cognitive disorder, is caused by reduced cerebral blood flow. Unraveling the metabolic heterogeneity and reprogramming in VaD is essential for understanding its molecular pathology and developing targeted therapies. However, the in situ metabolic regulation within the specific brain regions affected by VaD has not been thoroughly investigated, and the therapeutic mechanisms of Erigeron breviscapus injection (EBI), a traditional Chinese medicine, require further elucidation. PURPOSE To investigate the region-specific metabolic alterations in a VaD rat model, explore the therapeutic effects of EBI at a microregional level, identify the key metabolic pathways and metabolites involved in VaD, and elucidate how EBI modulates these pathways to exert its therapeutic effects. METHODS Air-flow-assisted desorption electrospray ionization mass spectrometry imaging (AFADESI-MSI), a novel technique, was employed to investigate the metabolic changes in the brain microregions. We used a bilateral common carotid artery occlusion model to induce VaD in rats. Network analysis and network pharmacology were used to assess the local metabolic effects of the EBI treatment (3.6 mL/kg/day for 2 weeks). RESULTS The EBI treatment significantly ameliorated the neurological deficits in VaD rats. AFADESI-MSI revealed 31 key metabolites with significant alterations in the VaD model, particularly within the pathways related to neurotransmitter metabolism, redox homeostasis, and osmoregulation. The metabolic disturbances were primarily observed in the striatum (ST), pyriform cortex (PCT), hippocampus (HP), and other critical brain regions. The EBI treatment effectively reversed these metabolic imbalances, especially in neurotransmitter metabolism, suggesting its potential in mitigating VaD-related cognitive decline. CONCLUSION Our findings not only shed light on the molecular underpinnings of VaD but also highlight the potential of EBI as a therapeutic agent in neurodegenerative disorders. Moreover, this study demonstrates the power of advanced mass spectrometry imaging techniques in phytomedicine, offering new insights into the spatial metabolic changes induced by botanical treatments.
Collapse
Affiliation(s)
- Shuohan Cheng
- School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Wenbin Zhou
- School of Pharmacy, Minzu University of China, Beijing, 100081, China; Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China; Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Yuhe Ren
- School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Xuemeng Gao
- School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Dalun Cai
- School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Mengyu Li
- School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Zhi Zhou
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China; Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China; Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Zhonghua Wang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China; Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China; Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing 100081, China.
| | - Zeper Abliz
- School of Pharmacy, Minzu University of China, Beijing, 100081, China; Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China; Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China; Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing 100081, China.
| |
Collapse
|
13
|
Sangeet S, Khan A. An in-silico approach to identify bioactive phytochemicals from Houttuynia cordata Thunb. As potential inhibitors of human glutathione reductase. J Biomol Struct Dyn 2025; 43:2300-2319. [PMID: 38109166 DOI: 10.1080/07391102.2023.2294181] [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: 08/17/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
Cellular infections are central to the etiology of various diseases, notably cancer and malaria. Counteracting cellular oxidative stress via the inhibition of glutathione reductase (GR) has emerged as a promising therapeutic strategy. Houttuynia cordata, a medicinal plant known for its potent antioxidant properties, has been the focus of our investigation. In this study, we conducted comprehensive in silico analyses involving the phytochemical constituents of H. cordata to identify potential natural GR inhibitors. Our methodological approach encompassed multiple in silico techniques, including molecular docking, molecular dynamics simulations, MMPBSA analysis, and dynamic cross-correlation analysis. Out of 13 docked phytochemicals, Quercetin, Quercitrin, and Sesamin emerged as particularly noteworthy due to their exceptional binding affinities for GR. Notably, our investigation demonstrated that Quercetin and Sesamin exhibited promising outcomes compared to the well-established pharmaceutical agent N-acetylcysteine (NAC). Molecular dynamics analyses provided insights into the ability of these phytochemicals to induce structural compaction and stabilization of the GR protein, as evidenced by changes in radius of gyration and solvent-accessible surface area. Moreover, MMPBSA analysis highlighted the crucial roles of specific residues, namely Gly27, Gly28, Ser51, His52, and Val61, in mediating essential interactions with these phytochemicals. Furthermore, an assessment of Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADME-Tox) profiles underscored the favourable drug-like attributes of these phytochemicals. Thus, the current findings underscore the immense potential of Houttuynia cordata phytochemicals as potent antioxidants with the capacity to combat a spectrum of maladies, including malaria and cancer. This study not only unveils novel therapeutic avenues but also underscores the distinctive outcomes and paramount significance of harnessing H. cordata phytochemicals for their efficacious antioxidant properties.
Collapse
Affiliation(s)
- Satyam Sangeet
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, India
- CompObelisk, Mirzapur, India
| | - Arshad Khan
- CompObelisk, Mirzapur, India
- Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, India
| |
Collapse
|
14
|
Soares JRP, dos Santos CC, de Oliveira LMG, Rocha Neto H, Victor MM, França EL, Costa MDFD, Costa SL, de Oliveira JVR. Synthesis of Naringenin and Senecioic Acid Ester Derivatives and Biological Evaluation of the Astrocyte Antioxidant Mechanism and Reactivity After Inflammatory Stimulus. Int J Mol Sci 2025; 26:2215. [PMID: 40076834 PMCID: PMC11900193 DOI: 10.3390/ijms26052215] [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: 02/07/2025] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
The imbalance between the overproduction of reactive species and antioxidant mechanisms can result in astrogliosis and oxidative stress associated with neurodegeneration. Based on the described antioxidant activity of naturally occurring flavonoids, this study evaluated the antioxidant mechanisms of the flavonoid naringenin and the senecioic acid ester derivatives in cortical astrocytes. Naringenin and (S)-naringenin were purified from Citrus paradisi, and from them 7,4-O-disenecioic ester naringenin, (S)-7,4-O-disenecioic ester naringenin, and 7-O-senecioic ester naringenin were synthesized and tested for antioxidant activity by the free-radical scavenging reaction with DPPH. The flavonoids' toxicity and glutathione (GS) depletion were determined in rat astrocyte cultures; the effects on the astrocytes' reactivity was determined by the expression of the glial fibrillary acidic protein (GFAP) and by measuring nitric oxide (NO) production in astrocytes treated with lipopolysaccharide (LPS, 1 µg/mL/24 h). The compounds (1-10 μM) presented antioxidant effects, and the (S)-7,4'-O-disenecioic ester naringenin was the most effective. The compounds (1-100 μM) were not toxic to the astrocytes, also promoting an antioxidant effect by increasing GSH. Moreover, naringenin, (S)-7,4'-O-disenecioic ester naringenin, and 7-O-senecioc ester naringenin mitigated the astrocyte reactivity induced by LPS, reducing GFAP expression and NO production. These findings indicate that naringenin and senecioic acid ester derivatives present a pharmacological potential as antioxidant and anti-inflammatory compounds for brain diseases via the modulation of astrocyte response.
Collapse
Affiliation(s)
- Janaína Ribeiro Pereira Soares
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
| | - Cleonice Creusa dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
| | - Lucas Matheus Gonçalves de Oliveira
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
| | - Heráclito Rocha Neto
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
| | - Maurício Moraes Victor
- Department of Organic Chemistry, Institute of Chemistry, Federal University of Bahia, Salvador 40170-115, Brazil;
| | - Elivana Lima França
- Federal Institute of Bahia, Campus Vitória da Conquista, Vitória da Conquista 45078-300, Brazil;
| | - Maria de Fátima Dias Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
- National Institute of Translational Neuroscience (INNT), Rio de Janeiro 21941-902, Brazil
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
- National Institute of Translational Neuroscience (INNT), Rio de Janeiro 21941-902, Brazil
| | - Juciele Valeria Ribeiro de Oliveira
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
| |
Collapse
|
15
|
Ruiter-Lopez L, Khan MAS, Wang X, Song BJ. Roles of Oxidative Stress and Autophagy in Alcohol-Mediated Brain Damage. Antioxidants (Basel) 2025; 14:302. [PMID: 40227291 PMCID: PMC11939343 DOI: 10.3390/antiox14030302] [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: 02/01/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 04/15/2025] Open
Abstract
Excessive alcohol consumption significantly impacts human health, particularly the brain, due to its susceptibility to oxidative stress, which contributes to neurodegenerative conditions. Alcohol metabolism in the brain occurs primarily via catalase, followed by CYP2E1 pathways. Excess alcohol metabolized by CYP2E1 generates reactive oxygen/nitrogen species (ROS/RNS), leading to cell injury via altering many different pathways. Elevated oxidative stress impairs autophagic processes, increasing post-translational modifications and further exacerbating mitochondrial dysfunction and ER stress, leading to cell death. The literature highlights that alcohol-induced oxidative stress disrupts autophagy and mitophagy, contributing to neuronal damage. Key mechanisms include mitochondrial dysfunction, ER stress, epigenetics, and the accumulation of oxidatively modified proteins, which lead to neuroinflammation and impaired cellular quality control. These processes are exacerbated by chronic alcohol exposure, resulting in the suppression of protective pathways like NRF2-mediated antioxidant responses and increased susceptibility to neurodegenerative changes in the brain. Alcohol-mediated neurotoxicity involves complex interactions between alcohol metabolism, oxidative stress, and autophagy regulation, which are influenced by various factors such as drinking patterns, nutritional status, and genetic/environmental factors, highlighting the need for further molecular studies to unravel these mechanisms and develop targeted interventions.
Collapse
Affiliation(s)
- Leon Ruiter-Lopez
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892, USA
| | - Mohammed A. S. Khan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.A.S.K.); (X.W.)
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.A.S.K.); (X.W.)
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
Cheng J, Ma X, Tao J, Jiang X, Chen P, Duan X. Neuroprotective effects of ethanol extraction from Rubia yunnanensis Diels on chronic cerebral hypoperfusion: modulation of the System Xc-/GSH/GPX4 axis to alleviate oxidative stress and ferroptosis. Front Pharmacol 2025; 16:1552228. [PMID: 40070574 PMCID: PMC11893507 DOI: 10.3389/fphar.2025.1552228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction Vascular dementia (VD) is a neurodegenerative disease caused by chronic cerebral hypoperfusion (CCH), which considerably impact patients' quality of life. Ethanol extraction from Rubia yunnanensis (RY-A) has gained attention for its potential neuroprotective effects, but its effects and mechanisms of action on CCH are unknown. Methods After 30 days of RY-A gavage treatment in a CCH rat model, its effects were evaluated using the Morris water maze test, cerebral blood flow measurements, and HE staining of the brain. These findings, combined with serum medicinal chemistry, RNA-seq, and metabolomics analyses, revealed the active compounds and mechanisms of RY-A in CCH rats. The results were further validated using assay kits and Western blot techniques. Results RY-A treatment significantly attenuated neurological damage and improved cognitive function in CCH rats. Ultra-high-performance liquid chromatography high-resolution mass spectrometry identified 511 blood-entry compounds of RY-A. RNA-seq and metabolomic analysis showed that RY-A might help to normalize changes in gene and metabolite expression caused by CCH. RY-A induced neuroprotective effects by increasing the production of key proteins involved in ferroptosis inhibition, such as SLC7A11, SLC3A2, GSS, and GPX4, while increasing antioxidant enzyme activities and alleviating oxidative stress. Conclusion RY-A inhibited oxidative stress and ferroptosis by activating the System Xc-/GSH/GPX4 pathway and balancing iron metabolism, thereby attenuating CCH-induced neurological damage and cognitive deficits.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaohua Duan
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
17
|
Kuźniar-Pałka A. The Role of Oxidative Stress in Autism Spectrum Disorder Pathophysiology, Diagnosis and Treatment. Biomedicines 2025; 13:388. [PMID: 40002801 PMCID: PMC11852718 DOI: 10.3390/biomedicines13020388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/20/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Autism spectrum disorder (ASD) is a significant health problem with no known single cause. There is a vast number of evidence to suggest that oxidative stress plays an important role in this disorder. The author of this article reviewed the current literature in order to summarise the knowledge on the subject. In this paper, the role of oxidative stress is investigated in the context of its influence on pathogenesis, the use of oxidative stress biomarkers as diagnostic tools and the use of antioxidants in ASD treatment. Given the heterogeneity of ASD aetiology and inadequate treatment approaches, the search for common metabolic traits is essential to find more efficient diagnostic tools and treatment methods. There are increasing data to suggest that oxidative stress is involved in the pathogenesis of ASD, both directly and through its interplay with inflammation and mitochondrial dysfunction. Oxidative stress biomarkers appear to have good potential to be used as diagnostic tools to aid early diagnosis of ASD. The results are most promising for glutathione and its derivatives and also for isoprostanses. Probably, complex dedicated multi-parametric metabolic panels may be used in the future. Antioxidants show good potential in ASD-supportive treatment. In all described fields, the data support the importance of oxidative stress but also a need for further research, especially in the context of sample size and, preferably, with a multicentre approach.
Collapse
Affiliation(s)
- Aleksandra Kuźniar-Pałka
- Clinic of Pediatric and Adolescent Neurology, Institute of Mother and Child, 01-211 Warsaw, Poland
| |
Collapse
|
18
|
Lee SW, Kim S, Chang Y, Cha H, Noeske R, Choi C, Lee SJ. Quantification of Glutathione and Its Associated Spontaneous Neuronal Activity in Major Depressive Disorder and Obsessive-Compulsive Disorder. Biol Psychiatry 2025; 97:279-289. [PMID: 39218137 DOI: 10.1016/j.biopsych.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Glutathione (GSH) is a crucial antioxidant in the human brain. Although proton magnetic resonance spectroscopy using the Mescher-Garwood point-resolved spectroscopy sequence is highly recommended, limited literature has measured cortical GSH using this method in major psychiatric disorders. METHODS By combining magnetic resonance spectroscopy and resting-state functional magnetic resonance imaging, we quantified brain GSH and glutamate in the medial prefrontal cortex and precuneus and explored relationships between GSH levels and intrinsic neuronal activity as well as clinical symptoms among healthy control (HC) participants (n = 30), people with major depressive disorder (MDD) (n = 28), and people with obsessive-compulsive disorder (OCD) (n = 28). RESULTS GSH concentrations were lower in the medial prefrontal cortex and precuneus in both the MDD and OCD groups than in the HC group. In the HC group, positive correlations were noted between GSH and glutamate levels and between GSH and fractional amplitude of low-frequency fluctuations in both regions. However, while these correlations were absent in both patient groups, there was a weak positive correlation between glutamate and fractional amplitude of low-frequency fluctuations. Moreover, GSH levels were negatively correlated with depressive and compulsive symptoms in MDD and OCD, respectively. CONCLUSIONS These findings suggest that reduced GSH levels and an imbalance between GSH and glutamate could increase oxidative stress and alter neurotransmitter signaling, thereby leading to disruptions in GSH-related neurochemical-neuronal coupling and psychopathologies across MDD and OCD. Understanding these mechanisms could provide valuable insights into the processes that underlie these disorders and potentially become a springboard for future directions and advancing our knowledge of their neurobiological foundations.
Collapse
Affiliation(s)
- Sang Won Lee
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu, Korea; Department of Psychiatry, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Seungho Kim
- Department of Medical & Biological Engineering, Kyungpook National University, Daegu, Korea
| | - Yongmin Chang
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu, Korea; Department of Radiology, Kyungpook National University Hospital, Daegu, Korea
| | - Hyunsil Cha
- Department of Medical & Biological Engineering, Kyungpook National University, Daegu, Korea
| | - Ralph Noeske
- Applied Science Laboratory Europe, GE HealthCare, Munich, Germany
| | - Changho Choi
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Seung Jae Lee
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu, Korea; Department of Psychiatry, Kyungpook National University Hospital, Daegu, Korea.
| |
Collapse
|
19
|
Kougias DG, Atillasoy E, Southall MD, Scialli AR, Ejaz S, Chu C, Jeminiwa BO, Massarsky A, Unice KM, Schaeffer TH, Kovochich M. A quantitative weight-of-evidence review of preclinical studies examining the potential developmental neurotoxicity of acetaminophen. Crit Rev Toxicol 2025; 55:124-178. [PMID: 39982125 DOI: 10.1080/10408444.2024.2442344] [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: 07/31/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 02/22/2025]
Abstract
Acetaminophen [paracetamol; N-acetyl-para-aminophenol (APAP)] is an antipyretic/analgesic commonly used in the treatment of fever and mild to moderate pain, headache, myalgia, and dysmenorrhea. Recent literature has questioned the safety of acetaminophen use during pregnancy, with an emphasis on whether exposure to the developing nervous system results in behavioral changes consistent with autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), and/or other cognitive deficits in the offspring. No previous review has used a fully detailed, quantitative weight-of-evidence (QWoE) approach to critically examine the preclinical acetaminophen data with regards to potential developmental neurotoxicity (DNT). Following regulatory guidance, a QWoE framework using prespecified scoring criteria was developed consistent with previous approaches to characterize potential adverse DNT outcomes with considerations for biological relevance of the response to adverse outcomes (outcome score) and the strength of methods and study design (methods score). Considerations for the methods score included (1) experimental design, (2) details/reliability of measurement(s), (3) data transparency, and (4) translational/methodological relevance. Considerations for the outcome score included response-related (1) statistical significance, (2) dose-response, (3) relevance/reliability/magnitude, (4) plausibility, and (5) translational relevance, including consideration of systemic toxicity/hepatotoxicity and therapeutic and/or non-systemically toxic doses and durations of use. Application of this QWoE framework to the 34 in vivo studies identified that assess the potential DNT of acetaminophen resulted in 188 QWoE entries documented across 11 DNT endpoints: social behavior, stereotypic behavior, behavioral rigidity, attention/impulsivity, hyperactivity, anxiety-like behavior, sensorimotor function, spatial learning/memory, nonspatial learning/memory, neuroanatomy, and neurotransmission. For each endpoint, the mean outcome score and methods score were calculated for total entries and for entries segregated by sex to assist in determining data quality and potential adversity. Informed by all 188 entries, the QWoE analysis demonstrated data of moderate quality showing no consistent evidence of DNT in male and female rodents following exposure to acetaminophen at therapeutic and/or nonsystemically toxic doses. Although some of the DNT endpoints (behavioral rigidity, attention/impulsivity, spatial learning/memory, neuroanatomy, and neurotransmission) generally displayed a more limited dataset and/or relatively lower data quality, similar conclusions were drawn based on results indicating a lack of biological relevance and reliability of reported adverse effects. Overall, this QWoE analysis on the preclinical in vivo data demonstrates no consistent evidence of adverse effects following developmental exposure to acetaminophen at therapeutic and/or non-systemically toxic doses on the structure and function of the nervous system, including neuroanatomical, neurotransmission, and behavioral endpoints.
Collapse
Affiliation(s)
| | - Evren Atillasoy
- Kenvue Medical Clinical and Safety Sciences, Fort Washington, PA, USA
| | | | - Anthony R Scialli
- Reproductive Toxicology Center, A Non-Profit Foundation, Washington, DC, USA
| | - Sadaff Ejaz
- Kenvue Medical Clinical and Safety Sciences, Skillman, NJ, USA
| | - Christopher Chu
- Kenvue Medical Clinical and Safety Sciences, Skillman, NJ, USA
| | | | | | | | | | | |
Collapse
|
20
|
Li H, Wang X, Chen Y, Leng H, Bai J, Lu Y, Wang N, Du C, Wang J, Chen M. Lanthanide-Assisted Function Tailoring of the HOF-Based Logic Gate Sensor Array for Biothiol Detection and Disease Discrimination. Anal Chem 2025; 97:1758-1766. [PMID: 39803691 DOI: 10.1021/acs.analchem.4c05296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
The advancement of lanthanide fingerprint sensors characterized by targeted emission responses and low self-fluorescence interference for the detection of biothiols is of considerable importance for the early diagnosis and treatment of cancer. Herein, the lanthanide "personality function tailoring" HOF composite sensor array is designed for the specific discrimination of biothiols (GSH, Cys, and Hcy) based on the activation of various luminescent molecules, such as r-AuNCs/luminol via HOF surface proximity. Lumi-HOF@Ce serves as a versatile platform for catalyzing the oxidation of o-phenylenediamine (OPD) to generate yellow fluorescent oligomers, accompanied by the fluorescence attenuation of luminol. HOF@Tb functions as a confinement interface that gathers gold nanoclusters (r-AuNCs) with red fluorescence, facilitating an aggregation-induced emission enhancement (AIEE). The fluorescence properties of AuNCs are subsequently impacted to varying degrees by the Au(I)-thiolate motifs from biothiol rooted in an enhanced ligand-metal charge transfer (LMCT) process. Additionally, the catalytic activity of Lumi-HOF@Ce, which exhibits oxidase-like properties, can be inhibited by different biothiols to varying extents. The five-channel fluorescent array demonstrates exceptional discrimination of biothiol fingerprints, aided by machine learning algorithms. Feature-tailored lanthanide HOF sensor arrays achieve sensitive identification with nearly 100% accuracy in classifying clinical liver cancer samples versus normal samples, using a logic gate strategy. The current strategy of lanthanide function tailoring boosts the suitability of biosensing applications.
Collapse
Affiliation(s)
- Haiyan Li
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Xin Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Yafei Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Han Leng
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Junjie Bai
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Yi Lu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Nan Wang
- Analytical and Testing Center, Northeastern University, Box 115, Shenyang 110819, China
| | - Cheng Du
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Jianhua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Mingli Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
- Analytical and Testing Center, Northeastern University, Box 115, Shenyang 110819, China
| |
Collapse
|
21
|
Quincozes-Santos A, Bobermin LD, Tramontina AC, Wartchow KM, Da Silva VF, Gayger-Dias V, Thomaz NK, de Moraes ADM, Schauren D, Nardin P, Gottfried C, Souza DO, Gonçalves CA. Glioprotective Effects of Resveratrol Against Glutamate-Induced Cellular Dysfunction: The Role of Heme Oxygenase 1 Pathway. Neurotox Res 2025; 43:7. [PMID: 39869271 DOI: 10.1007/s12640-025-00730-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: 06/12/2024] [Revised: 10/21/2024] [Accepted: 01/19/2025] [Indexed: 01/28/2025]
Abstract
Resveratrol, a natural polyphenol, has shown promising neuroprotective effects in several in vivo and in vitro experimental models. However, the mechanisms by which resveratrol mediates these effects are not fully understood. Glutamate is the major excitatory neurotransmitter in the brain; however, excessive extracellular glutamate levels can affect neural activity in several neurological diseases. Astrocytes are the glial cells that maintain brain homeostasis and can attenuate excitotoxicity by actively participating in glutamate neurotransmission. This study aimed to investigate the glioprotective effects of resveratrol against glutamate-induced cellular dysfunction in hippocampal slices and primary astrocyte cultures, with a focus on the role of heme-oxygenase 1 (HO-1). Glutamate impaired glutamate uptake activity through a glutamate receptor-dependent mechanism, in addition to altering other important astroglial parameters, including glutamine synthetase activity, glutathione levels and cystine uptake, which were normalized by resveratrol. Resveratrol also prevented glutamate-induced disruption in antioxidant defenses, as well as in trophic and inflammatory functions, including the nuclear factor κB (NFκB) transcriptional activity. Most of the effects of resveratrol, mainly in astrocytes, were dependent on the HO-1 signaling pathway, as they were abrogated when HO-1 was pharmacologically inhibited. Resveratrol also increased HO-1 mRNA expression and its transcriptional regulator, nuclear factor erythroid-derived 2-like 2 (Nrf2). Finally, resveratrol prevented glutamate-induced p21 senescence marker, indicating an anti-aging effect. Therefore, we demonstrated that the activation of the Nrf2/HO-1 system in astrocytes by resveratrol represents an astrocyte-targeted neuroprotective mechanism in neurodegeneration, with glutamate excitotoxicity, oxidative stress, and neuroinflammation as common neurochemical alterations.
Collapse
Affiliation(s)
- André Quincozes-Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600- Anexo Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Larissa Daniele Bobermin
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600- Anexo Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil.
| | - Ana Carolina Tramontina
- Programa de Pós-Graduação em Ambiente e Sustentabilidade, Universidade Estadual do Rio Grande do Sul, São Francisco de Paula, RS, Brazil
| | - Krista Minéia Wartchow
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York City, NY, USA
| | - Vanessa-Fernanda Da Silva
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Vitor Gayger-Dias
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Natalie K Thomaz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Aline Daniel Moreira de Moraes
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daniele Schauren
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Patrícia Nardin
- Escola de Saúde, Universidade do Vale do Rio dos Sinos (Unisinos), São Leopoldo, RS, Brazil
| | - Carmem Gottfried
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600- Anexo Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
- National Institute of Science and Technology in Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Diogo Onofre Souza
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600- Anexo Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Carlos-Alberto Gonçalves
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600- Anexo Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| |
Collapse
|
22
|
Ma X, Wang XM, Tang GZ, Wang Y, Liu XC, Wang SD, Peng P, Qi XH, Qin XY, Wang YJ, Wang CW, Zhou JN. Alterations of amino acids in older adults with Alzheimer's Disease and Vascular Dementia. Amino Acids 2025; 57:10. [PMID: 39825947 PMCID: PMC11742867 DOI: 10.1007/s00726-024-03442-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/31/2024] [Indexed: 01/20/2025]
Abstract
Metabolomics provide a promising tool for understanding dementia pathogenesis and identifying novel biomarkers. This study aimed to identify amino acid biomarkers for Alzheimer's Disease (AD) and Vascular Dementia (VD). By amino acid metabolomics, the concentrations of amino acids were determined in the serum of AD and VD patients as well as age-matched healthy controls. Several differences in the concentration of amino acids were observed in AD patients compared to both healthy controls and VD patients. However, no significant distinction was found between healthy controls and VD patients. Considering comorbidities, cystine levels were higher in AD than in VD among non-diabetic patients, but not in those with diabetes. Notably, creatine, spermidine, cystine, and tyrosine demonstrated favorable results in decision curve analyses and good discriminative performances, suggesting their potential for clinical application. These fundings give novel perspectives of serum amino acids for predicting metabolic pathways in AD and VD pathogenesis.
Collapse
Affiliation(s)
- Xin Ma
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
- Second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Xin-Meng Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
| | - Guo-Zhang Tang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
- Second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Yi Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
- First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Xue-Chun Liu
- Department of Neurology, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, P. R. China
| | - Shuai-Deng Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Peng Peng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
- First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Xiu-Hong Qi
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, 230026, P. R. China
| | - Xin-Ya Qin
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, 230026, P. R. China
- Institute of Brain Science, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
| | - Yue-Ju Wang
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, P. R. China.
| | - Chen-Wei Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China.
| | - Jiang-Ning Zhou
- Institute of Brain Science, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
| |
Collapse
|
23
|
Kuć J, Szarejko KD, Maciejczyk M, Dymicka-Piekarska V, Żendzian-Piotrowska M, Zalewska A. Oxidative imbalance as a co-player in jaw functional limitations and biopsychosocial profile in patients with temporomandibular disorder-myofascial pain with referral. Front Neurol 2025; 15:1509845. [PMID: 39830201 PMCID: PMC11739092 DOI: 10.3389/fneur.2024.1509845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/15/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Temporomandibular disorders have a multifactorial etiology including biological, biomechanical, neuromuscular, and biopsychosocial factors. Current research on temporomandibular disorders focuses on identifying clinically relevant biomarkers thus creating a new way of thinking about this dysfunction. The aim of the study was to determine the relationship between salivary/blood concentrations of oxidative/nitrosative stress biomarkers and biopsychosocial findings in patients with temporomandibular disorder-myofascial pain with referral. Methods The sample enrolled a total of 26 individuals with temporomandibular myofascial pain with referral (twenty women, six men). The procedure included clinical examination according to the Diagnostic Criteria for Temporomandibular Disorders, saliva and blood collection. Biochemical analysis concerned, among others, the content of reduced glutathione, uric acid, total antioxidant capacity, advanced glycation end products, malondialdehyde, total lipid hydroperoxides, kynurenine, N-formylkynurenine, and peroxynitrite. All determinations were considered with respect to the Patient Health Questionnaire-4 (PHQ-4), Patient Health Questionnaire-9 (PHQ-9), Patient Health Questionnaire-15 (PHQ-15), Generalized Anxiety Disorder-7 (GAD-7), Jaw Functional Limitation Scale-20 (JFLS-20), Perceived Stress Scale-10 (PSS-10), and Beck Depression Inventory (BDI). Results and discussion The average age of participants was 24.2 ± 1.23. High content of kynurenine and N-formylkynurenine in plasma was related to intensified psychological distress (PHQ-4) and anxiety (GAD-7). Low concentration of plasma malondialdehyde and total lipid hydroperoxides was linked with severe somatization (PHQ-15) and stress (PSS-10), respectively. Reduced levels of non-enzymatic antioxidants were associated with greater jaw functional mobility restrictions as well as limited mastication and communication factor with respect to JFLS-20. These findings indicate that oxidative stress biomarkers are significantly related to the biopsychosocial profile in patients with temporomandibular disorder.
Collapse
Affiliation(s)
- Joanna Kuć
- Department of Prosthodontics, Medical University of Białystok, Białystok, Poland
| | | | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology, and Ergonomics, Medical University of Białystok, Białystok, Poland
| | | | | | - Anna Zalewska
- Independent Laboratory of Experimental Dentistry, Medical University of Białystok, Białystok, Poland
- Restorative Dentistry Department, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
24
|
Sabra MS, Allam EAH, El-Aal MA, Hassan NH, Mostafa AHM, Ahmed AAN. A novel pharmacological strategy using nanoparticles with glutathione and virgin coconut oil to treat gentamicin-induced acute renal failure in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:933-950. [PMID: 39093465 PMCID: PMC11787276 DOI: 10.1007/s00210-024-03303-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
In acute renal failure (ARF), the glomerular filtration rate is reduced, and nitrogenous waste products accumulate persistently, which can last anywhere from a few hours to several days. There is hope for a reversal of the rapid loss of renal function caused by this condition. This study, with gentamicin-induced acute ARF as a prospective setting, sets out to examine the reno-protective benefits of virgin coconut oil (VCO) and GSH. Furthermore, the study evaluated the effect of medication nanoparticle compositions on several kidney function markers. The induction of ARF is achieved with the intraperitoneal injection of gentamicin. To assess renal function, rats underwent 24 h of dehydration and hunger before their deaths. The study examined various aspects, including kidney function tests, markers of oxidative stress, histology of kidney tissue, inflammatory cytokines, immunohistochemistry expression of nuclear factor-kappa B (NF-κB), and specific biomarkers for kidney tissue damage, such as kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL). The results of our study indicated that the combination of VCO and GSH, using both regular and nanoparticle formulations, had a better protective impact on the kidneys compared to using either drug alone. The recovery of renal tissue and serum markers, which are symptomatic of organ damage, indicates improvement. This was also demonstrated by the reduction in tubular expression of TNF-α, IL-1β, KIM-1, and NGAL. The immunohistochemical studies showed that the combination therapy, especially with the nanoforms, greatly improved the damaged cellular changes in the kidneys, as shown by higher levels of NF-κB. The study shows that VCO and GSH, when administered individually or combined, significantly improve ARF in a gentamicin-induced rat model, highlighting potential therapeutic implications. Notably, the combined nanoparticulate formulations exhibit substantial effectiveness.
Collapse
Affiliation(s)
- Mahmoud S Sabra
- Pharmacology Department, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
| | - Essmat A H Allam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Mohamed Abd El-Aal
- Chemistry Department, Faculty of Science, Assiut University, Assiut, 71516, Egypt
| | - Nessma H Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Al-Hassan Mohammed Mostafa
- Department of Pathology and Clinical Pathology, Agricultural Research Centre, Animal Health Research Institute, Assiut, 71526, Egypt
| | - Ahmed A N Ahmed
- Pharmacology Department, Faculty of Medicine, Al-Azhar University, Assiut Branch, , Assiut, 71526, Egypt
| |
Collapse
|
25
|
Glänzel NM, da Rosa-Junior NT, Signori MF, de Andrade Silveira J, Pinheiro CV, Marcuzzo MB, Campos-Carraro C, da Rosa Araujo AS, Schiöth HB, Wajner M, Leipnitz G. Increased ROS levels, antioxidant defense disturbances and bioenergetic disruption induced by thiosulfate administration in the brain of neonatal rats. Metab Brain Dis 2024; 40:73. [PMID: 39704910 DOI: 10.1007/s11011-024-01510-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
Sulfite oxidase deficiencies, either caused by deficiency of the apoenzyme or the molybdenum cofactor, and ethylmalonic encephalopathy are inherited disorders that impact sulfur metabolism. These patients present with severe neurodeterioration accompanied by cerebral cortex and cerebellum abnormalities, and high thiosulfate levels in plasma and tissues, including the brain. We aimed to clarify the mechanisms of such abnormalities, so we assessed the ex vivo effects of thiosulfate administration on energetic status and oxidative stress markers in cortical and cerebellar tissues of newborn rats. Thiosulfate (0.5 µmol/g) or PBS (vehicle) was injected into the fourth ventricle of rat pups. Thirty minutes after the injection, animals were euthanized and the brain structures were utilized for the experiments. Our data showed that thiosulfate decreased the reduced glutathione (GSH) concentrations, and superoxide dismutase (SOD), catalase (CAT) and glutathione S-transferase (GST) activities in the cortical structure. Thiosulfate also increased DCFH oxidation, hydrogen peroxide generation and glutathione reductase activity. In the cerebellum, thiosulfate reduced SOD and glutathione peroxidase activities but increased GST and CAT activities as well as DCFH oxidation. Regarding energy metabolism, thiosulfate specifically decreased complex IV activity in the cortex, whereas it increased cerebellar complex I and creatine kinase activities, indicating bioenergetic disturbances. The results suggest that the accumulation of thiosulfate causing redox disruption and bioenergetic alterations has a prominent role in the pathogenesis of sulfur metabolism deficiencies.
Collapse
Affiliation(s)
- Nícolas Manzke Glänzel
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, 90035-003, RS, Brazil
| | - Nevton Teixeira da Rosa-Junior
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, 90035-003, RS, Brazil
| | - Marian F Signori
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, 90035-003, RS, Brazil
| | - Josyane de Andrade Silveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, 90035-003, RS, Brazil
| | - Camila Vieira Pinheiro
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, 90035-003, RS, Brazil
| | - Manuela Bianchin Marcuzzo
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, 90035-003, RS, Brazil
| | - Cristina Campos-Carraro
- Laboratório de Fisiologia Cardiovascular, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Alex Sander da Rosa Araujo
- Laboratório de Fisiologia Cardiovascular, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Helgi B Schiöth
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, Uppsala, 75124, Sweden
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, 90035-003, RS, Brazil
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, Porto Alegre, 2350, 90035-903, RS, Brazil
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, 90035-003, RS, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, 90035-003, RS, Brazil.
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, Uppsala, 75124, Sweden.
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, 90035-003, RS, Brazil.
- Programa de Pós-Graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, 90035-003, RS, Brazil.
| |
Collapse
|
26
|
Xue Q, Wang R, Zhu-Ge R, Guo L. Research progresses on the effects of heavy metals on the circadian clock system. REVIEWS ON ENVIRONMENTAL HEALTH 2024; 39:721-727. [PMID: 37572029 DOI: 10.1515/reveh-2022-0104] [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: 07/04/2022] [Accepted: 06/12/2023] [Indexed: 08/14/2023]
Abstract
Environmental pollution with heavy metals is widespread, thus increasing attention has been paid to their toxic effects. Recent studies have suggested that heavy metals may influence the expression of circadian clock genes. Almost all organs and tissues exhibit circadian rhythms. The normal circadian rhythm of an organism is maintained by the central and peripheral circadian clock. Thus, circadian rhythm disorders perturb normal physiological processes. Here, we review the effects of heavy metals, including manganese, copper, cadmium, and lead, on four core circadian clock genes, i.e., ARNTL, CLOCK, PER, and CRY genes.
Collapse
Affiliation(s)
- Qian Xue
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Rui Wang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Ruijian Zhu-Ge
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Li Guo
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
27
|
Saraiva JT, Dos Santos FDS, Bona NP, da Silveira LM, Simões WS, da Silva GBDO, da Silva JA, Domingues WB, Nascimento MC, Campos VF, Spanevello RM, Pedra NS, Stefanello FM. Antitumor Effect of Butia odorata Hydroalcoholic Extract on C6 and U87MG Glioma Cell Lines: Impact on Redox Status and Inflammation Signaling. Neurochem Res 2024; 50:56. [PMID: 39671046 DOI: 10.1007/s11064-024-04305-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/14/2024]
Abstract
Among the spectrum of gliomas, glioblastoma stands out as the most aggressive brain tumor affecting the central nervous system. In addressing this urgent medical challenge, exploring therapeutic alternatives becomes imperative to enhance the patient's prognosis. In this regard, Butia odorata (BO) fruit emerges as a promising candidate due to its array of bioactive compounds, including flavonoids, phenolic acids, and carotenoids, known for their antioxidant, anti-inflammatory, and antitumor properties. Thus, this study aimed to investigate the impact of standardized hydroalcoholic extract of BO on rat C6 and human U87MG glioma cell lines. Cells were exposed to varying extract concentrations (125-2000 μg/mL) for intervals of 0, 2, 4, 6, 24, 48, or 72 h. Then, cell viability, proliferation, colony formation, redox equilibrium parameters, cell migration, and the relative mRNA expression of genes related to gliomagenesis were evaluated. Our findings revealed a reduction in viability, proliferation, colony formation, reactive oxygen species, and nitrite levels in both glioma cell lines upon exposure to the extract. Conversely, an increase in sulfhydryl content and the activity of superoxide dismutase and catalase were observed in both glioma cell lines. No significant changes in viability and proliferation were observed in astrocytes. Furthermore, in the C6 cells only, the BO extract reduced the migration and downregulated the relative mRNA expression of matrix metalloproteinase-2, O6-methylguanine-DNA methyltransferase, nuclear factor-kappa B, interleukin-6 genes, and upregulated caspase-3 gene. These results underscore the promising anti-glioma potential of BO extract, attributed to its diverse bioactive composition.
Collapse
Affiliation(s)
- Juliane Torchelsen Saraiva
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP 96010-900, Brazil
| | - Francieli da Silva Dos Santos
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP 96010-900, Brazil
| | - Natália Pontes Bona
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP 96010-900, Brazil
| | - Larissa Menezes da Silveira
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP 96010-900, Brazil
| | - William Sanabria Simões
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP 96010-900, Brazil
| | - Giulia Bueno de Oliveira da Silva
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP 96010-900, Brazil
| | - Júlia Araújo da Silva
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP 96010-900, Brazil
| | - William Borges Domingues
- Programa de Pós-Graduação em Biotecnologia - Laboratório de Genômica Estrutural, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Mariana Cavalcanti Nascimento
- Programa de Pós-Graduação em Biotecnologia - Laboratório de Genômica Estrutural, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Vinicius Farias Campos
- Programa de Pós-Graduação em Biotecnologia - Laboratório de Genômica Estrutural, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Roselia Maria Spanevello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Nathalia Stark Pedra
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Francieli Moro Stefanello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP 96010-900, Brazil.
| |
Collapse
|
28
|
Seo CR, Lee BK, Jee HJ, Yoo JR, Lee CK, Park JW, Jung YS. Ameliorating Effect of Fermented Perilla frutescens on Sleep Deprivation-Induced Cognitive Impairment Through Antioxidant and BDNF Signaling in Mice. Nutrients 2024; 16:4224. [PMID: 39683616 DOI: 10.3390/nu16234224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Adequate sleep is essential for maintaining cognitive function, as evidenced by literature. Perilla frutescens var. acuta Kudo (PF) is a traditional medicinal herb reported to improve vascular cognitive impairment and induce sedation. However, the effects of PF on cognitive impairment caused by sleep deprivation (SD) have not yet been evaluated. This study aims to evaluate the effects of fermented PF (FPF) and its underlying mechanisms in a model of SD-induced cognitive impairment. Methods: Mice were subjected to SD to establish cognitive impairment, and FPF was administered once daily for 3 days. Cognitive performance was assessed using Y-maze and passive avoidance tests, followed by molecular mechanisms analyses. Results: FPF treatment improved SD-induced cognitive impairment, as evidenced by increased spontaneous alternation and extended latency time. Histological analysis revealed that SD impaired the hippocampus, and this impairment was alleviated by FPF treatment. FPF demonstrated antioxidant activity by increasing glutathione levels and decreasing malondialdehyde levels. Furthermore, the decreased levels of brain-derived neurotrophic factor (BDNF) observed in sleep-deprived mice were restored with FPF treatment. FPF also enhanced the phosphorylation of tropomyosin receptor kinase B, extracellular signal-regulated kinase, and cAMP response element-binding protein. Conclusions: These results indicate that FPF may have beneficial effects on SD-induced cognitive impairment by protecting against oxidative stress and increasing BDNF expression.
Collapse
Affiliation(s)
- Chae-Ryeong Seo
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Bo Kyung Lee
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Hye Jin Jee
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Jae Ryeong Yoo
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Chul-Kyu Lee
- Headquarters of New Drug Development Support, Corestemchemon Inc., 15 F, Gyeonggi Bio Center, Suwon 16229, Republic of Korea
| | - Jin Wook Park
- Headquarters of New Drug Development Support, Corestemchemon Inc., 15 F, Gyeonggi Bio Center, Suwon 16229, Republic of Korea
| | - Yi-Sook Jung
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
29
|
Negi M, Amulya E, Phatale V, Abraham N, Hedaoo A, Srinivasarao DA, Srivastava S. Surface engineered nano architectonics: An evolving paradigm for tackling Alzheimer's disease. Life Sci 2024; 358:123155. [PMID: 39433085 DOI: 10.1016/j.lfs.2024.123155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/21/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
As per the World Health Organization (WHO) estimation, Alzheimer's disease (AD) will affect 100 million population across the globe by 2050. AD is an incurable neurodegenerative disease that remains a mystery for neurologists owing to its complex pathophysiology. Currently, available therapeutic regimens will only cause symptomatic relief by improving the cognitive and behavioral functions of AD. However, the major pitfalls in managing AD include tight junctions in the endothelial cells of the blood-brain barrier (BBB), diminished neuronal bioavailability, enzymatic degradation and reduced stability of the therapeutic moiety. In an effort to surmount the drawbacks mentioned above, researchers shifted their focus toward nanocarriers (NCs). Nevertheless, non-specific targeting of NCs imparts toxicity to the peripheral organs, thereby reducing the bioavailability of therapeutic moiety at the target site. To unravel this unmet clinical need, scientists came up with the idea of a novel intriguing strategy of surface engineering by targeting ligands. Surface-decorated NCs provide targeted drug delivery, controlled drug release, enhanced penetration and bioavailability. In this state-of-the-art review, we have highlighted in detail various molecular signalling pathways involved in AD pathogenesis. The significance of surface functionalization and its application in AD management have been deliberated. We have elaborated on the regulatory bottlenecks and clinical hurdles faced during lab-to-industrial scale translation along with possible solutions.
Collapse
Affiliation(s)
- Mansi Negi
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Etikala Amulya
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Noella Abraham
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Aachal Hedaoo
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dadi A Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
30
|
Michels L, O'Gorman-Tuura R, Bachmann D, Müller S, Studer S, Saake A, Gruber E, Rauen K, Buchmann A, Zuber I, Hock C, Gietl A, Treyer V. The links among age, sex, and glutathione: A cross-sectional magnetic resonance spectroscopy study. Neurobiol Aging 2024; 144:19-29. [PMID: 39255570 DOI: 10.1016/j.neurobiolaging.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024]
Abstract
Glutathione (GSH) is a brain marker for oxidative stress and has previously been associated with cerebral amyloid deposition and memory decline. However, to date, no study has examined the links among GSH, sex, age, amyloid, and Apolipoprotein E (APOE) genotype in a large non-clinical cohort of older adults. We performed APOE genotyping, magnetic resonance spectroscopy (MRS) as well as simultaneous positron emission tomography with the radiotracer Flutemetamol (Amyloid-PET), in a group of older adults. The final analysis set comprised 140 healthy older adults (mean age: 64.7 years) and 49 participants with mild cognitive impairment (mean age: 71.4 years). We recorded metabolites in the posterior cingulate cortex (PCC) by a GSH-edited MEGAPRESS sequence. Structural equation modeling revealed that higher GSH levels were associated with female sex, but neither APOE- epsilon 4 carrier status nor age showed significant associations with GSH. Conversely, older age and the presence of an APOE4 allele, but not sex, are linked to higher global amyloid load. Our results suggest that the PCC shows sex-specific GSH alterations in older adults.
Collapse
Affiliation(s)
- Lars Michels
- Department of Neuroradiology, Clinical Neuroscience Center (KNZ), University Hospital Zurich, Zurich, Switzerland.
| | | | - Dario Bachmann
- Institute for Regenerative Medicine, University of Zurich Campus Schlieren, Schlieren, Switzerland
| | - Susanne Müller
- Department of Neuroradiology, Clinical Neuroscience Center (KNZ), University Hospital Zurich, Zurich, Switzerland
| | - Sandro Studer
- Institute for Regenerative Medicine, University of Zurich Campus Schlieren, Schlieren, Switzerland
| | - Antje Saake
- Institute for Regenerative Medicine, University of Zurich Campus Schlieren, Schlieren, Switzerland
| | - Esmeralda Gruber
- Institute for Regenerative Medicine, University of Zurich Campus Schlieren, Schlieren, Switzerland
| | - Katrin Rauen
- Institute for Regenerative Medicine, University of Zurich Campus Schlieren, Schlieren, Switzerland; Department of Geriatric Psychiatry, Psychiatric Hospital Zurich, Zurich, Switzerland
| | - Andreas Buchmann
- Institute for Regenerative Medicine, University of Zurich Campus Schlieren, Schlieren, Switzerland
| | - Isabelle Zuber
- Institute for Regenerative Medicine, University of Zurich Campus Schlieren, Schlieren, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich Campus Schlieren, Schlieren, Switzerland; Neurimmune, Schlieren, Switzerland
| | - Anton Gietl
- Institute for Regenerative Medicine, University of Zurich Campus Schlieren, Schlieren, Switzerland; Department of Geriatric Psychiatry, Psychiatric Hospital Zurich, Zurich, Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland; Institute for Regenerative Medicine, University of Zurich Campus Schlieren, Schlieren, Switzerland
| |
Collapse
|
31
|
Fan Z, Yang C, Qu X, Zhang J, Wu H, Yang Y, Huang Y, Zeng P, Xiang Z, Yang J. Association of Oxidative Stress on Cognitive Function: A Bidirectional Mendelian Randomisation Study. Mol Neurobiol 2024; 61:10551-10560. [PMID: 38753129 DOI: 10.1007/s12035-024-04231-3] [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: 02/29/2024] [Accepted: 05/10/2024] [Indexed: 11/24/2024]
Abstract
The purpose of this study was to investigate the relationship between oxidative stress and cognitive function, encompassing cognitive performance, intelligence, memory, reaction time, speech and vision by a bidirectional Mendelian randomisation study. Independent genetic variants associated with glutathione S-transferase (GST), catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPX), peroxiredoxin (PRDX), sulfhydryl oxidase (SOX) and thyroid peroxidase (TPO) were explored using a genome-wide association study (GWAS). The inverse variance weighted (IVW) or Wald ratio method was employed to ascertain the relationship between antioxidant enzymes and cognitive function. The MR analyses indicated that the MR effect estimates of GST (β = 0.0352, P = 0.0047, FDR = 0.0164) and TPO (β = 0.0531, P = 0.0003, FDR = 0.0021) were significantly associated with cognitive performance elevation. Furthermore, genetically predicted GST (β = 0.0334, P = 0.0043, FDR = 0.0151) and TPO (β = 0.0496, P = 0.0031, FDR = 0.0151) were found to be associated with high intelligence. Additionally, there were also some associations of SOX (β = 0.0243, P = 0.0283, FDR = 0.066) on high cognitive performance, TPO (β = 0.1189, P = 0.0315, FDR = 0.2205) on larger maximum digits remembered correctly, and SOX (β = - 0.2435, P = 0.0395, FDR = 0.1185) on reaction time. Nevertheless, the associations between antioxidant enzymes and speech and linguistic disorders, as well as visual disturbances, were not significant. We did not find reverse causation between antioxidant enzymes and cognitive function traits. This study provides evidence of potential causal relationships between oxidative stress and cognitive function.
Collapse
Affiliation(s)
- Zhixing Fan
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
- Department of Medical Record Management, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
| | - Chaojun Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Xiaoling Qu
- Department of Medical Record Management, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
| | - Jing Zhang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Hui Wu
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Ying Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Yifan Huang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Ping Zeng
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Zujin Xiang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China.
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China.
| |
Collapse
|
32
|
Cesarini L, Grignaffini F, Alisi A, Pastore A. Alterations in Glutathione Redox Homeostasis in Metabolic Dysfunction-Associated Fatty Liver Disease: A Systematic Review. Antioxidants (Basel) 2024; 13:1461. [PMID: 39765791 PMCID: PMC11672975 DOI: 10.3390/antiox13121461] [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: 10/22/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Low molecular weight (LMW) thiols, particularly glutathione, play pathogenic roles in various multiorgan diseases. The liver is central for the production and systemic distribution of LMW thiols; thus, it is particularly susceptible to the imbalance of redox status that may determine increased oxidative stress and trigger the liver damage observed in metabolic dysfunction-associated steatotic liver disease (MASLD) models and humans. Indeed, increased LMW thiols at the cellular and extracellular levels may be associated with the severity of MASLD. Here, we present a systematic literature review of recent studies assessing the levels of LMW thiols in MASLD in in vivo and in vitro models and human subjects. Based on the PRISMA 2020 criteria, a search was conducted using PubMed and Scopus by applying inclusion/exclusion filters. The initial search returned 1012 documents, from which 165 eligible studies were selected, further described, and qualitatively analysed. Of these studies, most focused on animal and cellular models, while a minority used human fluids. The analysis of these studies revealed heterogeneity in the methods of sample processing and measurement of LMW thiol levels, which hinder cut-off values for diagnostic use. Standardisation of the analysis and measure of LMW thiol is necessary to facilitate future studies.
Collapse
Affiliation(s)
| | | | - Anna Alisi
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (L.C.); (F.G.); (A.P.)
| | | |
Collapse
|
33
|
Liu D, Guo P, Wang Y, Li W. Regulation of adult neurogenesis: the crucial role of astrocytic mitochondria. Front Mol Neurosci 2024; 17:1516119. [PMID: 39649104 PMCID: PMC11621070 DOI: 10.3389/fnmol.2024.1516119] [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: 10/23/2024] [Accepted: 11/08/2024] [Indexed: 12/10/2024] Open
Abstract
Neurogenesis has emerged as a promising therapeutic approach for central nervous system disorders. The role of neuronal mitochondria in neurogenesis is well-studied, however, recent evidence underscores the critical role of astrocytic mitochondrial function in regulating neurogenesis and the underlying mechanisms remain incompletely understood. This review highlights the regulatory effects of astrocyte mitochondria on neurogenesis, focusing on metabolic support, calcium homeostasis, and the secretion of neurotrophic factors. The effect of astrocytic mitochondrial dysfunction in the pathophysiology and treatment strategies of Alzheimer's disease and depression is discussed. Greater attention is needed to investigate the mitochondrial autophagy, dynamics, biogenesis, and energy metabolism in neurogenesis. Targeting astrocyte mitochondria presents a potential therapeutic strategy for enhancing neural regeneration.
Collapse
Affiliation(s)
| | | | | | - Weihong Li
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
34
|
Watermann P, Kalsi GK, Dringen R, Arend C. Differential Effects of Itaconate and its Esters on the Glutathione and Glucose Metabolism of Cultured Primary Rat Astrocytes. Neurochem Res 2024; 50:24. [PMID: 39562371 PMCID: PMC11576791 DOI: 10.1007/s11064-024-04263-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/09/2024] [Accepted: 10/07/2024] [Indexed: 11/21/2024]
Abstract
Itaconate is produced as endogenous metabolite by decarboxylation of the citric acid cycle intermediate cis-aconitate. As itaconate has anti-microbial and anti-inflammatory properties, this substance is considered as potential therapeutic drug for the treatment of inflammation in various diseases including traumatic brain injury and stroke. To test for potential adverse effects of itaconate on the viability and metabolism of brain cells, we investigated whether itaconate or its membrane permeable derivatives dimethyl itaconate (DI) and 4-octyl itaconate (OI) may affect the basal glucose and glutathione (GSH) metabolism of cultured primary astrocytes. Acute exposure of astrocytes to itaconate, DI or OI in concentrations of up to 300 µM for up to 6 h did not compromise cell viability. Of the tested substances, only OI stimulated aerobic glycolysis as shown by a time- and concentration-dependent increase in glucose-consumption and lactate release. None of the tested itaconates affected the pentose-phosphate pathway-dependent reduction of the water-soluble tetrazolium salt 1 (WST1). In contrast, both DI and OI, but not itaconate, depleted cellular GSH in a time- and concentration-dependent manner. For OI this depletion was accompanied by a matching increase in the extracellular GSH content that was completely prevented in the presence of the multidrug resistance protein 1 (Mrp1)-inhibitor MK571, while in DI-treated cultures GSH was depleted both in cells and medium. These data suggest that OI stimulates Mrp1-mediated astrocytic GSH export, while DI reacts with GSH to a conjugate that is not detectable by the GSH assay applied. The data presented demonstrate that itaconate, DI and OI differ strongly in their effects on the GSH and glucose metabolism of cultured astrocytes. Such results should be considered in the context of the discussed potential use of such compounds as therapeutic agents.
Collapse
Affiliation(s)
- Patrick Watermann
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, 28359, Bremen, Germany
- Centre for Environmental Research and Sustainable Technology, University of Bremen, 28359, Bremen, Germany
| | - Gurleen K Kalsi
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, 28359, Bremen, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, 28359, Bremen, Germany
- Centre for Environmental Research and Sustainable Technology, University of Bremen, 28359, Bremen, Germany
| | - Christian Arend
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, 28359, Bremen, Germany.
- Centre for Environmental Research and Sustainable Technology, University of Bremen, 28359, Bremen, Germany.
| |
Collapse
|
35
|
Moreno F, Méndez L, Fernández I, Miralles-Pérez B, Giralt M, Romeu M, Ramos-Romero S, Torres JL, Medina I. Influence of the Degree of Unsaturation in Fish Oil Supplements on Oxidative Stress and Protein Carbonylation in the Cerebral Cortex and Cerebellum of Healthy Rats. Antioxidants (Basel) 2024; 13:1408. [PMID: 39594550 PMCID: PMC11591239 DOI: 10.3390/antiox13111408] [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: 09/30/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
ω-3 polyunsaturated fatty acids (PUFAs) are crucial for brain structure and function, especially docosahexaenoic acid (DHA). However, an excess of DHA may increase lipid peroxidation due to its high degree of unsaturation, particularly in tissues highly susceptible to oxidative stress, such as the brain. Therefore, this study evaluated the effects of 10 weeks of dietary supplementation with fish oil containing 80% DHA on oxidative stress and the modulation of the carbonylated proteome in both the cerebral cortex and cerebellum of male Sprague Dawley rats. The results were compared with those induced by oils with a lower degree of fat unsaturation (fish oil containing 25% DHA and 25% eicosapentaenoic acid, soybean oil containing 50% linoleic acid and coconut oil containing 90% saturated fat). The results demonstrated that fish oil containing 80% DHA significantly increased the ω3/ω6 ratio in both the cortex and cerebellum while stimulating antioxidant defense by enhancing the reduced glutathione amount and decreasing the carbonylation of specific proteins, mainly those involved in glycolysis and neurotransmission. The majority of sensitive proteins in both brain regions followed this carbonylation trend (in decreasing order): soybean > EPA/DHA 1:1 > coconut > 80% DHA. The results also indicated that the cerebellum is more responsive than the cortex to changes in the cellular redox environment induced by varying degrees of fat unsaturation. In conclusion, under healthy conditions, dietary supplementation with fish oils containing high DHA levels makes the brain more resilient to potential oxidative insults compared to oils with lower DHA content and a lower degree of fatty acid unsaturation.
Collapse
Affiliation(s)
- Francisco Moreno
- Institute of Marine Research—Spanish National Research Council (IIM-CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (F.M.); (I.F.); (B.M.-P.); (I.M.)
- University of Vigo, Circunvalación ao Campus Universitario, 36310 Vigo, Spain
| | - Lucía Méndez
- Institute of Marine Research—Spanish National Research Council (IIM-CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (F.M.); (I.F.); (B.M.-P.); (I.M.)
| | - Ingrid Fernández
- Institute of Marine Research—Spanish National Research Council (IIM-CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (F.M.); (I.F.); (B.M.-P.); (I.M.)
| | - Bernat Miralles-Pérez
- Institute of Marine Research—Spanish National Research Council (IIM-CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (F.M.); (I.F.); (B.M.-P.); (I.M.)
- Pharmacology Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.G.); (M.R.)
| | - Montserrat Giralt
- Pharmacology Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.G.); (M.R.)
| | - Marta Romeu
- Pharmacology Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.G.); (M.R.)
| | - Sara Ramos-Romero
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain;
- Nutrition & Food Safety Research Institute (INSA-UB), Maria de Maeztu Unit of Excellence, Prat de la Riba 171, 08921 Santa Coloma de Gramenet, Spain;
- Institute of Advanced Chemistry of Catalonia—Spanish National Research Council (IQAC-CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Josep Lluís Torres
- Nutrition & Food Safety Research Institute (INSA-UB), Maria de Maeztu Unit of Excellence, Prat de la Riba 171, 08921 Santa Coloma de Gramenet, Spain;
- Institute of Advanced Chemistry of Catalonia—Spanish National Research Council (IQAC-CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Isabel Medina
- Institute of Marine Research—Spanish National Research Council (IIM-CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (F.M.); (I.F.); (B.M.-P.); (I.M.)
| |
Collapse
|
36
|
Arora Y, Samkaria A, Maroon JC, Mandal PK. Longitudinal Monitoring of Glutathione Stability in Different Microenvironments. Neurochem Res 2024; 50:9. [PMID: 39546088 PMCID: PMC11567985 DOI: 10.1007/s11064-024-04265-y] [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: 04/23/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 11/17/2024]
Abstract
Glutathione (GSH) is a master antioxidant which primarily protects cells from oxidative stress. Clinical studies have found significant depletion of GSH from the hippocampus in patients with mild cognitive impairment (MCI), a transitional stage before conversion to Alzheimer's disease (AD). Significant depletion of GSH is considered an early diagnostic biomarker of AD. Postmortem studies have confirmed significant GSH depletion in hippocampal tissue in MCI patients. The stability of GSH in different microenvironments is essential to validate GSH as a reliable biomarker for AD. Accordingly, we have conducted longitudinal monitoring of GSH from various brain regions (frontal cortex (FC), parietal cortex (PC), occipital cortex (OC), and cerebellum (CER)) from healthy subjects using MEshcher-GArwood Point RESolved Spectroscopy (MEGA-PRESS) pulse sequence on a 3T scanner. Additionally, in vitro magnetic resonance spectroscopy (MRS) assessments were conducted longitudinally using the same study protocol involving GSH supplement in a physiologically relevant phosphate buffer solution (PBS). We report that GSH within the brain microenvironment of a healthy person remains stable over time. GSH, however, is susceptible to oxidation over time in a phosphate buffer environment. The stability of GSH in a longitudinal study in the brains of healthy individuals supports the consideration of GSH as a candidate for stable biomarker for AD.
Collapse
Affiliation(s)
- Yashika Arora
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Center Research Centre, Gurgaon, Haryana, 122052, India
| | - Avantika Samkaria
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Center Research Centre, Gurgaon, Haryana, 122052, India
| | - Joseph C Maroon
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Pravat K Mandal
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Center Research Centre, Gurgaon, Haryana, 122052, India.
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA.
- Florey Institute of Neuroscience and Mental Health, Melbourne School of Medicine Campus, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
37
|
Swanberg KM, Prinsen H, Averill CL, Campos L, Kurada AV, Krystal JH, Petrakis IL, Averill LA, Rothman DL, Abdallah CG, Juchem C. Medial prefrontal cortex neurotransmitter abnormalities in posttraumatic stress disorder with and without comorbidity to major depression. NMR IN BIOMEDICINE 2024; 37:e5220. [PMID: 39054694 DOI: 10.1002/nbm.5220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/23/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
Posttraumatic stress disorder (PTSD) is a chronic psychiatric condition that follows exposure to a traumatic stressor. Though previous in vivo proton (1H) MRS) research conducted at 4 T or lower has identified alterations in glutamate metabolism associated with PTSD predisposition and/or progression, no prior investigations have been conducted at higher field strength. In addition, earlier studies have not extensively addressed the impact of psychiatric comorbidities such as major depressive disorder (MDD) on PTSD-associated 1H-MRS-visible brain metabolite abnormalities. Here we employ 7 T 1H MRS to examine concentrations of glutamate, glutamine, GABA, and glutathione in the medial prefrontal cortex (mPFC) of PTSD patients with MDD (PTSD+MDD+; N = 6) or without MDD (PTSD+MDD-; N = 5), as well as trauma-unmatched controls without PTSD but with MDD (PTSD-MDD+; N = 9) or without MDD (PTSD-MDD-; N = 18). Participants with PTSD demonstrated decreased ratios of GABA to glutamine relative to healthy PTSD-MDD- controls but no single-metabolite abnormalities. When comorbid MDD was considered, however, MDD but not PTSD diagnosis was significantly associated with increased mPFC glutamine concentration and decreased glutamate:glutamine ratio. In addition, all participants with PTSD and/or MDD collectively demonstrated decreased glutathione relative to healthy PTSD-MDD- controls. Despite limited findings in single metabolites, patterns of abnormality in prefrontal metabolite concentrations among individuals with PTSD and/or MDD enabled supervised classification to separate them from healthy controls with 80+% sensitivity and specificity, with glutathione, glutamine, and myoinositol consistently among the most informative metabolites for this classification. Our findings indicate that MDD can be an important factor in mPFC glutamate metabolism abnormalities observed using 1H MRS in cohorts with PTSD.
Collapse
Affiliation(s)
- Kelley M Swanberg
- Biomedical Engineering, Columbia University School of Engineering and Applied Science, New York, NY, USA
- Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Hetty Prinsen
- Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Christopher L Averill
- Clinical Neuroscience Division, Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
- US Department of Veterans Affairs Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Leonardo Campos
- Biomedical Engineering, Columbia University School of Engineering and Applied Science, New York, NY, USA
| | - Abhinav V Kurada
- Biomedical Engineering, Columbia University School of Engineering and Applied Science, New York, NY, USA
| | - John H Krystal
- Clinical Neuroscience Division, Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Ismene L Petrakis
- Clinical Neuroscience Division, Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Lynnette A Averill
- Clinical Neuroscience Division, Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
- US Department of Veterans Affairs Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Douglas L Rothman
- Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Chadi G Abdallah
- Clinical Neuroscience Division, Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
- US Department of Veterans Affairs Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Christoph Juchem
- Biomedical Engineering, Columbia University School of Engineering and Applied Science, New York, NY, USA
- Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Radiology, Columbia University Medical Center, New York, NY, USA
- Neurology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
38
|
Yang L, Lai X, Jin S, Wang H, Lin F, Jin X, Chen Y, Wang R, Huang Y, Zhang Y, Tian S, Fang X, Duan X, Zhang Q. Exploring the anti-ovarian aging mechanism of He's Yangchao formula: Insights from multi-omics analysis in naturally aged mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155961. [PMID: 39178679 DOI: 10.1016/j.phymed.2024.155961] [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: 06/05/2024] [Revised: 07/31/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND The rapid acceleration of female reproductive aging has become a major public health concern. He's Yangchao formula (HSYC), a compound comprising eight herbs, has demonstrated efficacy in enhancing ovarian function. Thus, an in-depth study of its anti-ovarian aging mechanism is required. PURPOSE To evaluate the anti-ovarian aging effect of HSYC in naturally aged mice and investigate the underlying mechanism by analyzing the gut microbiota (GM), metabolome, and transcriptome. METHODS Young and advanced maternal age (AMA) mice were selected for this study. Hematoxylin and eosin staining, fluorescence staining, western blotting, and qPCR analyses were used to detect the phenotypes associated with ovarian aging. Subsequently, analyses of the GM, transcriptome, and metabolome analyses were performed to explore the potential mechanisms of action of HSYC. Finally, in vivo and in vitro experiments were performed to verify potential therapeutic mechanisms. RESULTS HSYC promoted follicular development in AMA mice and ameliorated age-related mitochondrial dysfunction, apoptosis, and defects in DNA damage repair. GM analysis revealed that HSYC treatment significantly increased the abundance of Akkermansia and Turicibacter. Transcriptome and metabolome analyses showed that HSYC might mitigate ovarian aging by regulating metabolic pathways, amino acid metabolism, glutathione metabolism, and the synthesis of pantothenic acid and coenzyme A. Combined transcriptomic and metabolomic analyses identified the glutathione metabolic pathway as the key pathway through which HSYC counteracts ovarian aging. Additional experimental verification confirmed that HSYC upregulated the glutathione metabolic genes GPX8, GSTA1, and GSTA4, increased glutathione-related products (GSH), and reduced ROS levels. CONCLUSIONS HSYC exerts beneficial therapeutic effects on ovarian aging by regulating multiple endogenous metabolites, targets, and metabolic pathways, with an emphasis on its anti-ovarian aging effects through the glutathione metabolic pathway. These findings underscore the innovative potential of HSYC in addressing ovarian aging and offer a novel therapeutic approach that targets multiple biological pathways to improve the reproductive health of women with AMA..
Collapse
Affiliation(s)
- Liuqing Yang
- Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310000, PR China; Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou 310053, PR China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, PR China
| | - Xinle Lai
- Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310000, PR China; Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou 310053, PR China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, PR China
| | - Shuo Jin
- Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Heng Wang
- Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Fangxuan Lin
- Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Xin Jin
- Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Yun Chen
- Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310000, PR China; Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou 310053, PR China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, PR China
| | - Ruye Wang
- Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310000, PR China; Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou 310053, PR China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, PR China
| | - Yun Huang
- Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310000, PR China; Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou 310053, PR China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, PR China
| | - Yiqun Zhang
- Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310000, PR China; Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou 310053, PR China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, PR China
| | - Saisai Tian
- Department of Phytochemistry, School of Pharmacy, The Second Military Medical University, Shanghai 200433, PR China
| | - Xiaohong Fang
- Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310000, PR China; Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou 310053, PR China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, PR China
| | - Xing Duan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China; Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, PR China.
| | - Qin Zhang
- Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310000, PR China; Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou 310053, PR China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, PR China.
| |
Collapse
|
39
|
Park SY, Kim KY, Gwak DS, Shin SY, Jun DY, Kim YH. L-Cysteine mitigates ROS-induced apoptosis and neurocognitive deficits by protecting against endoplasmic reticulum stress and mitochondrial dysfunction in mouse neuronal cells. Biomed Pharmacother 2024; 180:117538. [PMID: 39393330 DOI: 10.1016/j.biopha.2024.117538] [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: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/13/2024] Open
Abstract
Oxidative stress and mitochondrial dysfunction play critical roles in neurodegenerative diseases. Glutathione (GSH), a key brain antioxidant, helps to neutralize reactive oxygen species (ROS) and maintain redox balance. We investigated the effectiveness of L-cysteine (L-Cys) in preventing apoptosis induced by the ROS generator 2,3-dimethoxy-1,4-naphthoquinone (DMNQ) in mouse hippocampal neuronal HT22 cells, as well as alleviating memory and cognitive impairments caused by the GSH synthesis inhibitor L-buthionine sulfoximine (BSO) in mice. DMNQ-induced apoptotic events in HT22 cells, including elevated cytosolic and mitochondrial ROS levels, DNA fragmentation, endoplasmic reticulum stress, and mitochondrial damage-mediated apoptotic pathways were dose-dependently abrogated by L-Cys (0.5-2 mM). The reduced intracellular GSH level, caused by DMNQ treatment, was restored by L-Cys cotreatment. Although L-Cys did not significantly restore GSH in the presence of BSO, it prevented DMNQ-induced ROS elevation, mitochondrial damage, and apoptosis. Furthermore, compared to N-acetylcysteine and GSH, L-Cys had higher 2,2-diphenyl-1-picrylhydrazyl and 2,2-azino-bis-3-ethylbenzothiazoline-6-sulphonic acid radical-scavenging activity. L-Cys also restored mitochondrial respiration capacity in DMNQ-treated HT22 cells by reversing mitochondrial fission-fusion dynamic balance. BSO administration (500 mg/kg/day) in mice led to neuronal deficits, including memory and cognitive impairments, which were effectively mitigated by oral L-Cys (15 or 30 mg/kg/day). L-Cys also reduced BSO-induced ROS levels in the mice hippocampus and cortex. These findings suggest that even though it does not contribute to intracellular GSH synthesis, exogenous L-Cys protects neuronal cells against oxidative stress-induced mitochondrial damage and apoptosis, by acting as a ROS scavenger, which is beneficial in ameliorating neurocognitive deficits caused by oxidative stress.
Collapse
Affiliation(s)
- Shin Young Park
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Ki Yun Kim
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Dong Seol Gwak
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Soon Young Shin
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Do Youn Jun
- AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Young Ho Kim
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea.
| |
Collapse
|
40
|
Firdous SM, Khan SA, Maity A. Oxidative stress-mediated neuroinflammation in Alzheimer's disease. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8189-8209. [PMID: 38832985 DOI: 10.1007/s00210-024-03188-3] [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: 02/20/2024] [Accepted: 05/24/2024] [Indexed: 06/06/2024]
Abstract
Reactive oxygen species (ROS) are metabolic by-products that constitute an indispensable component of physiological processes, albeit their heightened presence may proffer substantial perils to biological entities. Such a proliferation gives rise to a gradual escalation of oxidative stress within the organism, thereby compromising mitochondrial functionality and inflicting harm upon various bodily systems, with a particular predilection for the central nervous system. In its nascent stages, it is plausible that inflammation has been a facilitator in the progression of the malady. The precise role of inflammation in Alzheimer's disease (AD) remains somewhat enigmatic, although it is conceivable that activated microglia and astrocytes might be implicated in the removal of amyloid-β (Aβ) deposits. Nonetheless, prolonged microglial activation is associated with Tau phosphorylation and Aβ aggregation. Research studies have indicated that AD brains upregulate complementary molecules, inflammatory cytokines, acute phase reacting agents, and other inflammatory mediators that may cause neurodegeneration. In this review, oxidative damage products will be discussed as potential peripheral biomarkers for AD and its early stages. The disordered excretion of pro-inflammatory cytokines, chemokines, oxygen, and nitrogen-reactive species, along with the stimulation of the complement system by glial cells, has the potential to disrupt the functionality of neuronal termini. This perturbation, in turn, culminates in compromised synaptic function, a phenomenon empirically linked to the manifestation of cognitive impairments. The management of neurodegenerative conditions in the context of dementia necessitates therapeutic interventions that specifically target the excessive production of inflammatory and oxidative agents. Furthermore, we shall deliberate upon the function of microglia and oxidative injury in the etiology of AD and the ensuing neurodegenerative processes.
Collapse
Affiliation(s)
- Sayed Mohammed Firdous
- Department of Pharmacology, Calcutta Institute of Pharmaceutical Technology & AHS, Uluberia, Howrah, 711316, West Bengal, India.
| | - Sahabaj Ali Khan
- Department of Pharmacology, Calcutta Institute of Pharmaceutical Technology & AHS, Uluberia, Howrah, 711316, West Bengal, India
| | - Amritangshu Maity
- Department of Pharmacology, Calcutta Institute of Pharmaceutical Technology & AHS, Uluberia, Howrah, 711316, West Bengal, India
| |
Collapse
|
41
|
Rizoli C, Dos Santos NM, Maróstica Júnior MR, da Cruz-Höfling MA, Mendonça MCP, de Jesus MB. The therapeutic potential of reduced graphene oxide in attenuating cuprizone-induced demyelination in mice. NANOTECHNOLOGY 2024; 36:025102. [PMID: 39389086 DOI: 10.1088/1361-6528/ad857e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/10/2024] [Indexed: 10/12/2024]
Abstract
Reduced graphene oxide (rGO) has unique physicochemical properties that make it suitable for therapeutic applications in neurodegenerative scenarios. This study investigates the therapeutic potential of rGO in a cuprizone-induced demyelination model in mice through histomorphological techniques and analysis of biochemical parameters. We demonstrate that daily intraperitoneal administration of rGO (1 mg ml-1) for 21 days tends to reduce demyelination in theCorpus callosumby decreasing glial cell recruitment during the repair mechanism. Additionally, rGO interferes with oxidative stress markers in the brain and liver indicating potential neuroprotective effects in the central nervous system. No significant damage to vital organs was observed, suggesting that multiple doses could be used safely. However, further long-term investigations are needed to understand rGO distribution, metabolism, routes of action and associated challenges in central neurodegenerative therapies. Overall, these findings contribute to the comprehension of rGO effectsin vivo, paving the way for possible future clinical research.
Collapse
Affiliation(s)
- Cintia Rizoli
- Departmento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | | | | | - Maria Alice da Cruz-Höfling
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | | | - Marcelo Bispo de Jesus
- Departmento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
42
|
Zhang Y, Zhang C, Dai Q, Ma R. Continuous Theta Burst Stimulation Inhibits Oxidative Stress-Induced Inflammation and Autophagy in Hippocampal Neurons by Activating Glutathione Synthesis Pathway, Improving Cognitive Impairment in Sleep-Deprived Mice. Neuromolecular Med 2024; 26:40. [PMID: 39388015 DOI: 10.1007/s12017-024-08807-z] [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: 05/08/2024] [Accepted: 09/29/2024] [Indexed: 10/12/2024]
Abstract
Sleep deprivation (SD) has been reported to have a negative impact on cognitive function. Continuous theta burst stimulation (cTBS) shows certain effects in improving sleep and neurological diseases, and its molecular or cellular role in SD-induced cognition impairment still need further exploration. In this study, C57BL/6 mice were subjected to 48 h of SD and cTBS treatment, and cTBS treatment significantly improved SD-triggered impairment of spatial learning and memory abilities in mice. Additionally, cTBS reduced malondialdehyde levels, increased superoxide dismutase activities, and inhibited the production of inflammatory cytokines, alleviating oxidative stress and inflammation levels in hippocampal tissues of SD model mice. cTBS decreased LC3II/LC3I ratio, Beclin1 protein levels, and LC3B puncta intensity, and elevated p62 protein levels to suppress excessive autophagy in hippocampal tissues of SD-stimulated mice. Then, we proved that inhibiting oxidative stress alleviated inflammation, autophagy, and death of hippocampal neuron cells through an in vitro cellular model for oxidative stress, and cTBS treatment promoted the production of glutathione (GSH), the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and the mRNA expression of GSH synthesis-related genes to enhance antioxidant capacity in hippocampal tissues of SD mice. An Nrf2 inhibitor ML385 or a GSH synthesis inhibitor BSO reversed the alleviating effects of cTBS treatment on oxidative stress-associated damage of hippocampal tissues and cognitive impairment in SD model mice. Altogether, our study demonstrated that cTBS mitigates oxidative stress-associated inflammation and autophagy through activating the Nrf2-mediated GSH synthesis pathway, improving cognitive impairment in SD mice.
Collapse
Affiliation(s)
- Yi Zhang
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China
| | - Cheng Zhang
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China
| | - Qing Dai
- Anesthesiology Department, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Rui Ma
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China.
| |
Collapse
|
43
|
Xie C, Chen Y, Wang L, Liao K, Xue B, Han Y, Li L, Jiang Q. Recent research of peptide-based hydrogel in nervous regeneration. Bioact Mater 2024; 40:503-523. [PMID: 39040568 PMCID: PMC11261279 DOI: 10.1016/j.bioactmat.2024.06.013] [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: 04/03/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 07/24/2024] Open
Abstract
Neurological disorders exert significantly affect the quality of life for patients, necessitating effective strategies for nerve regeneration. Both traditional autologous nerve transplantation and emerging therapeutic approaches encounter scientific challenges due to the complex nature of the nervous system and the unsuitability of the surrounding environment for cell transplantation. Tissue engineering techniques offer a promising path for neurotherapy. Successful neural tissue engineering relies on modulating cell differentiation behavior and tissue repair by developing biomaterials that mimic the natural extracellular matrix (ECM) and establish a three-dimensional microenvironment. Peptide-based hydrogels have emerged as a potent option among these biomaterials due to their ability to replicate the structure and complexity of the ECM. This review aims to explore the diverse range of peptide-based hydrogels used in nerve regeneration with a specific focus on dipeptide hydrogels, tripeptide hydrogels, oligopeptide hydrogels, multidomain peptides (MDPs), and amphiphilic peptide hydrogels (PAs). Peptide-based hydrogels offer numerous advantages, including biocompatibility, structural diversity, adjustable mechanical properties, and degradation without adverse effects. Notably, hydrogels formed from self-assembled polypeptide nanofibers, derived from amino acids, show promising potential in engineering neural tissues, outperforming conventional materials like alginate, poly(ε-caprolactone), and polyaniline. Additionally, the simple design and cost-effectiveness of dipeptide-based hydrogels have enabled the creation of various functional supramolecular structures, with significant implications for nervous system regeneration. These hydrogels are expected to play a crucial role in future neural tissue engineering research. This review aims to highlight the benefits and potential applications of peptide-based hydrogels, contributing to the advancement of neural tissue engineering.
Collapse
Affiliation(s)
- Chunmei Xie
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Yueyang Chen
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Lang Wang
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Kin Liao
- Advanced Digital and Additive Manufacturing Center, Khalifa University of Science and Technology, Po Box 127788, Abu Dhabi, United Arab Emirates
| | - Bin Xue
- National Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing, China
| | - Yulong Han
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Lan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, China
| |
Collapse
|
44
|
Zhou Z, Fang C, Yu F, Shen Y, Xu H, Li H, Zhang Y. Visualization of cysteine in AD mouse with a high-quantum yield NIR fluorescent probe. Talanta 2024; 278:126482. [PMID: 38950502 DOI: 10.1016/j.talanta.2024.126482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
Alzheimer's disease (AD) has gradually received enthusiastic attention with the aging process, and studying its biological relevance is expected. Excitingly, fluorescence probes were considered to be powerful tools for exploring biological correlations. Therefore, a highly selective near-infrared (NIR) fluorescent probe (DCM-Cl-Acr) for imaging cysteine (Cys) in AD was designed and synthesized. Through structural optimization, the probe exhibited high fluorescence quantum yield and low detection limit (20 nM) towards Cys. Meanwhile, based on the high selectivity and high sensitivity response exhibited by the probe to Cys, it was successfully applied to visualize endogenous and exogenous Cys in living cells and zebrafish, and showed good discrimination from homocysteine (Hcy) and glutathione (GSH). Further, the correlation between AD and Cys concentration was clarified by imaging studies in hippocampus tissue of AD mouse, and the abnormal accumulation of Cys in the hippocampus of AD brain was demonstrated.
Collapse
Affiliation(s)
- Zile Zhou
- Key Laboratory of Water Treatment Functional Materials (Hunan Provincial), Engineering Research Center of Hunan Province for Recycling Technology of Electroplating Wastewater, College of Chemistry and Materials Engineering, Hunan University of Arts and Science, Changde, 415000, PR China
| | - Cong Fang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Feiju Yu
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Youming Shen
- Key Laboratory of Water Treatment Functional Materials (Hunan Provincial), Engineering Research Center of Hunan Province for Recycling Technology of Electroplating Wastewater, College of Chemistry and Materials Engineering, Hunan University of Arts and Science, Changde, 415000, PR China
| | - Hai Xu
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Haitao Li
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Youyu Zhang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China.
| |
Collapse
|
45
|
Kolobova E, Petrushanko I, Mitkevich V, Makarov AA, Grigorova IL. β-Amyloids and Immune Responses Associated with Alzheimer's Disease. Cells 2024; 13:1624. [PMID: 39404388 PMCID: PMC11475064 DOI: 10.3390/cells13191624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is associated with the accumulation of β-amyloids (Aβs) and the formation of Aβ plaques in the brain. Various structural forms and isoforms of Aβs that have variable propensities for oligomerization and toxicity and may differentially affect the development of AD have been identified. In addition, there is evidence that β-amyloids are engaged in complex interactions with the innate and adaptive immune systems, both of which may also play a role in the regulation of AD onset and progression. In this review, we discuss what is currently known about the intricate interplay between β-amyloids and the immune response to Aβs with a more in-depth focus on the possible roles of B cells in the pathogenesis of AD.
Collapse
Affiliation(s)
- Elizaveta Kolobova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Irina Petrushanko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
| | - Vladimir Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
| | - Irina L Grigorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| |
Collapse
|
46
|
Coden KM, Nguyen DKK, Moorhead R, Stix-Brunell BE, Baker JN, Parker KJ, Garner JP. Making bloodwork work: the impact of sample collection, processing, and storage on plasma glutathione measurement, and implications for translation. Transl Psychiatry 2024; 14:385. [PMID: 39313523 PMCID: PMC11420238 DOI: 10.1038/s41398-024-03086-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Psychiatry has traditionally focused on the study of neurons and neurotransmitter physiology in the pathophysiology and treatment of psychiatric disorders. A growing literature highlights REDOX imbalance (a state in which demand for antioxidants surpasses their bioavailability) as a common pathophysiology for a diverse array of brain conditions (e.g., trichotillomania, schizophrenia, autism, Parkinson's disease). REDOX imbalance is typically measured via plasma glutathione, as glutathione is critical to the adaptive antioxidant response in the brain. Accordingly, glutathione, its precursors, and/or metabolites serve as biomarkers of disease risk, therapeutic targets, and measures of treatment response. However, as with any emerging field, there are currently several different methods for collection, processing, storage, and calculation of summary measures of plasma glutathione metabolism, within and between preclinical and clinical research. The lack of evidence-based best-practice methodology hampers reproducibility (preclinical or clinical), and translation (between preclinical and clinical work). To address this methodological need, here we used a repeated measures within-subject design to investigate how sample preparation (type of anticoagulant used during blood collection, deproteinization status, and storage temperature) affects plasma glutathione levels. Accordingly, we collected whole blood from mice (N = 13), and then, using a commercially available kit, quantified glutathione in plasma stored in four different ways. Presuming that these preparation conditions and post-processing calculations are unimportant, we would expect to see no difference in glutathione levels and summary measures from the same sample. However, we found each of these variables to significantly alter quantified glutathione levels. Accordingly, we propose a vital, gold-standard methodology for both sample collection, processing, and storage of plasma used for glutathione quantification and for summary calculations of glutathione that can be used preclinically and clinically, thus yielding more streamlined translation.
Collapse
Affiliation(s)
- Kendall M Coden
- Department of Comparative Medicine, Stanford University, Stanford, CA, 94305, USA.
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Duyen K K Nguyen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Roberta Moorhead
- Department of Comparative Medicine, Stanford University, Stanford, CA, 94305, USA
| | | | - Joanna N Baker
- Department of Comparative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Karen J Parker
- Department of Comparative Medicine, Stanford University, Stanford, CA, 94305, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Joseph P Garner
- Department of Comparative Medicine, Stanford University, Stanford, CA, 94305, USA.
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
47
|
Bouchab H, Ishaq A, Limami Y, Saretzki G, Nasser B, El Kebbaj R. Antioxidant Effects of Cactus Seed Oil against Iron-Induced Oxidative Stress in Mouse Liver, Brain and Kidney. Molecules 2024; 29:4463. [PMID: 39339457 PMCID: PMC11433720 DOI: 10.3390/molecules29184463] [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: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
In recent times, exploring the protective potential of medicinal plants has attracted increasing attention. To fight reactive oxygen species (ROS), which are key players in hepatic, cerebral and renal diseases, scientists have directed their efforts towards identifying novel compounds with antioxidant effects. Due to its unique composition, significant attention has been given to Cactus Seed Oil (CSO). Iron, as a metal, can be a potent generator of reactive oxygen species, especially hydroxyl radicals, via the Fenton and Haber-Weiss reactions. Here, we employed ferrous sulfate (FeSO4) to induce oxidative stress and DNA damage in mice. Then, we used CSO and Colza oil (CO) and evaluated the levels of the antioxidants (superoxide dismutase [SOD], glutathione peroxidase [GPx] and glutathione [GSH]) as well as a metabolite marker for lipid peroxidation (malondialdehyde [MDA]) relating to the antioxidant balance in the liver, brain and kidney. In addition, we measured DNA damage levels in hepatic tissue and the effects of CSO on it. Our study found that iron-dependent GPx activity decreases in the liver and the kidney tissues. Additionally, while iron decreased SOD activity in the liver, it increased it in the kidney. Interestingly, iron treatment resulted in a significant increase in hepatic MDA levels. In contrast, in brain tissue, there was a significant decrease under iron treatment. In addition, we found varying protective effects of CSO in alleviating oxidative stress in the different tissues with ameliorating DNA damage after iron overload in a mouse liver model, adding compelling evidence to the protective potential of CSO.
Collapse
Affiliation(s)
- Habiba Bouchab
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco; (H.B.); (Y.L.)
- Higher Institute of Nursing Professions and Technical Health (ISPITS), Errachidia 52000, Morocco
- Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Science and Technology, Hassan First University of Settat, Settat 26000, Morocco;
- Biosciences Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE2 4HH, UK; (A.I.); (G.S.)
| | - Abbas Ishaq
- Biosciences Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE2 4HH, UK; (A.I.); (G.S.)
| | - Youness Limami
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco; (H.B.); (Y.L.)
| | - Gabriele Saretzki
- Biosciences Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE2 4HH, UK; (A.I.); (G.S.)
| | - Boubker Nasser
- Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Science and Technology, Hassan First University of Settat, Settat 26000, Morocco;
| | - Riad El Kebbaj
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco; (H.B.); (Y.L.)
| |
Collapse
|
48
|
Abramov AY, Myers I, Angelova PR. Carbon Monoxide: A Pleiotropic Redox Regulator of Life and Death. Antioxidants (Basel) 2024; 13:1121. [PMID: 39334780 PMCID: PMC11428877 DOI: 10.3390/antiox13091121] [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: 07/31/2024] [Revised: 09/01/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Despite recent technological progress, carbon monoxide poisoning is still one of the leading causes of domestic and industrial morbidity and mortality. The brain is particularly vulnerable to CO toxicity, and thus the majority of survivors develop delayed movement and cognitive complications. CO binds to haemoglobin in erythrocytes, preventing oxygen delivery to tissues, and additionally inhibits mitochondrial respiration. This renders the effect of CO to be closely related to hypoxia reperfusion injury. Oxygen deprivation, as well as CO poisoning and re-oxygenation, are shown to be able to activate the production of reactive oxygen species and to induce oxidative stress. Here, we review the role of reactive oxygen species production and oxidative stress in the mechanism of neuronal cell death induced by carbon monoxide and re-oxygenation. We discuss possible protective mechanisms used by brain cells with a specific focus on the inhibition of CO-induced ROS production and oxidative stress.
Collapse
Affiliation(s)
| | | | - Plamena R. Angelova
- UCL Queen Square Institute of Neurology, Department of Clinical and Movement Neurosciences, Queen Square, London WC1N3BG, UK; (A.Y.A.); (I.M.)
| |
Collapse
|
49
|
Park JM, Park JE, Park JS, Leem YH, Kim DY, Hyun JW, Kim HS. Anti-inflammatory and antioxidant mechanisms of coniferaldehyde in lipopolysaccharide-induced neuroinflammation: Involvement of AMPK/Nrf2 and TAK1/MAPK/NF-κB signaling pathways. Eur J Pharmacol 2024; 979:176850. [PMID: 39059571 DOI: 10.1016/j.ejphar.2024.176850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/09/2024] [Accepted: 07/24/2024] [Indexed: 07/28/2024]
Abstract
Microglia are primarily involved in inflammatory reactions and oxidative stress in the brain; as such reducing microglial activation has been proposed as a potential therapeutic strategy for neurodegenerative disorders. Herein, we investigated the anti-inflammatory and antioxidant activities of coniferaldehyde (CFA), a naturally occurring cinnamaldehyde derivative, on activated microglia to evaluate its therapeutic potential. CFA inhibited the production of nitric oxide (NO) and proinflammatory cytokines, such as tumor necrosis factor-α, interleukin (IL)-1β, and IL-6, in lipopolysaccharide (LPS)-stimulated BV2 microglial cells. CFA also inhibited intracellular reactive oxygen species levels and oxidative stress markers such as 4-HNE and 8-OHdG. Detailed mechanistic studies showed that CFA exerted anti-inflammatory effects by inhibiting TAK1-mediated MAP kinase/NF-κB activation and upregulating AMPK signaling pathways. In addition, CFA exerted antioxidant effects by inhibiting the NADPH oxidase subunits and by increasing the expression of antioxidant enzymes such as HO-1, NQO1, and catalase by upregulating Nrf2 signaling. Finally, we confirmed the effects of CFA on the brains of the LPS-injected mice. CFA inhibited microglial activation and the expression of proinflammatory markers and increased Nrf2-driven antioxidant enzymes. Furthermore, CFA inhibited the production of 4-HNE and 8-OHdG in the brains of LPS-injected mice. As a result, CFA's significant anti-inflammatory and antioxidant properties may have therapeutic applications in neuroinflammatory disorders related with oxidative stress and microglial activation.
Collapse
Affiliation(s)
- Jae-Min Park
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Jung-Eun Park
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Jin-Sun Park
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Yea-Hyun Leem
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Do-Yeon Kim
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Jin-Won Hyun
- Department of Biochemistry, College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
50
|
Xiao T, Yu X, Tao J, Yang L, Duan X. Metabolomics-Based Study of the Protective Effect of 4-Hydroxybenzyl Alcohol on Ischemic Astrocytes. Int J Mol Sci 2024; 25:9907. [PMID: 39337395 PMCID: PMC11432256 DOI: 10.3390/ijms25189907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Ischemic stroke is a common and dangerous disease in clinical practice. Astrocytes (ASs) are essential for maintaining the metabolic balance of the affected regions during the disease process. 4-Hydroxybenzyl alcohol (4HBA) from Gastrodia elata Bl. has potential neuroprotective properties due to its ability to cross the blood-brain barrier. In an in vitro experiment, we replicated the oxygen-glucose deprivation/reoxygenation model, and used methyl thiazoly tertrazolium, flow cytometry, kits, and other technical means to clarify the protective effect of 4HBA on primary ASs. In in vivo experiments, the 2VO model was replicated, and immunofluorescence and immunohistochemistry techniques were used to clarify the protective effect of 4HBA on ASs and the maintenance of the blood-brain barrier. Differential metabolites and related pathways were screened and verified using metabolomics analysis and western blot. 4HBA noticeably amplified AS cell survival, reduced mitochondrial dysfunction, and mitigated oxidative stress. It demonstrated a protective effect on ASs in both environments and was instrumental in stabilizing the blood-brain barrier. Metabolomic data indicated that 4HBA regulated nucleic acid and glutathione metabolism, influencing purines, pyrimidines, and amino acids, and it activated the N-methyl-D-aspartate/p-cAMP-response element binding protein/brain-derived neurotrophic factor signaling pathway via N-methyl-D-aspartate R1/N-methyl-D-aspartate 2C receptors. Our findings suggest that 4HBA is a potent neuroprotective agent against ischemic stroke, enhancing AS cell survival and function while stabilizing the blood-brain barrier. The N-methyl-D-aspartate/p-cAMP-response element binding protein/brain-derived neurotrophic factor signaling pathway is activated by 4HBA.
Collapse
Affiliation(s)
- Tian Xiao
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xingzhi Yu
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Jie Tao
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Liping Yang
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xiaohua Duan
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| |
Collapse
|