1
|
Feng J, Yang Q, Chen M, Wang Y, Luo D, Hu D, Cheng J, Song X, Zhou X, Meng Q, Lin Q, He F. Protective effects of 4-HBd on blood-brain barrier integrity in MCAO/R model rats based on brain pharmacokinetic characteristics. Front Pharmacol 2025; 16:1528839. [PMID: 40264675 PMCID: PMC12012380 DOI: 10.3389/fphar.2025.1528839] [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: 11/15/2024] [Accepted: 03/03/2025] [Indexed: 04/24/2025] Open
Abstract
Objectives This study explored the brain-targeting properties and mechanisms of 4-hydroxybenzaldehyde (4-HBd), the primary active component of Gastrodia elata, in mitigating ischemic stroke (IS)-induced injury by preserving blood-brain barrier (BBB) integrity, based on brain pharmacokinetic characteristics. Methods The anti-IS effects of the G. elata extract were assessed using a rat middle cerebral artery occlusion/reperfusion (MCAO/R) model, leading to the identification of 4-HBd as the principal active ingredient. BBB protection was evaluated through neurological scoring, Evans Blue (EB) extravasation, cerebral infarct volume, and ultrastructural integrity. Oxidative stress markers, including superoxide dismutase (SOD), malondialdehyde (MDA), nitric oxide (NO), and inducible nitric oxide synthase (iNOS), were quantified in ischemic brain tissue via biochemical assays. The expression levels of tight junction (TJ) proteins claudin-5 and occludin, as well as matrix metalloproteinase MMP-2/9 and aquaporin-4 (AQP-4), were analyzed by Western blotting. Microdialysis, combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS), was employed to determine the temporal distribution of 4-HBd in the brains of both normal and MCAO/R model rats. The ability of 4-HBd to scavenge intracellular reactive oxygen species (ROS) in brain endothelial cells (bEnd.3) was evaluated using a single-cell biochemical analyzer. Results G. elata ethanol extract exhibited significant anti-IS effects. When compared with the model group, 4-HBd treatment markedly alleviated BBB disruption and neurological deficits, suppressed oxidative stress in ischemic brain tissue, reduced MDA and NO levels, and enhanced SOD activity. The expressions of claudin-5, occludin, MMP-2/9, and AQP-4 were significantly upregulated in the 4-HBd group relative to the model group. Additionally, 4-HBd selectively eliminated nuclear-derived ROS. Pharmacokinetic analysis demonstrated that 4-HBd preferentially accumulated in the striatum and cortex of both normal and MCAO/R model rats. Under ischemic conditions, 4-HBd exhibited accelerated cortical penetration, increased exposure, and prolonged retention. Conclusion These findings indicate that 4-HBd exerts a pronounced brain-targeting effect and preserves BBB integrity via the RNS/ROS-MMP-TJ signaling pathway, highlighting its potential as a therapeutic agent for IS.
Collapse
Affiliation(s)
- Jin Feng
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Qian Yang
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Ming Chen
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Yan Wang
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Dan Luo
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Dongxiong Hu
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
- School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Jianjun Cheng
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Xuelan Song
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiaonan Zhou
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Qingting Meng
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Qing Lin
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Fangyan He
- Department of Pharmacology, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
2
|
Jahan I, Shuvo AUH, Alimullah M, Rahman ASMN, Siddiqua S, Rafia S, Khan F, Ahmed KS, Hossain H, Akramuddaula K, Alam MA, Subhan N. Purple potato extract modulates fat metabolizing genes expression, prevents oxidative stress, hepatic steatosis, and attenuates high-fat diet-induced obesity in male rats. PLoS One 2025; 20:e0318162. [PMID: 40168333 PMCID: PMC11960900 DOI: 10.1371/journal.pone.0318162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 01/03/2025] [Indexed: 04/03/2025] Open
Abstract
OBJECTIVE In this investigation, the significance of purple potato (Solanum tuberosum L.) extract treatment was assessed against oxidative stress and fat metabolizing transcription factors in the liver of high-fat (HF) diet-fed rats. METHODS Wistar (male) rats were arranged into several groups and provided with a control and HF diet along with the purple potato extract. Body weights, oral glucose tolerance test (OGTT), insulin, plasma lipids, and oxidative stress-related indicators were analyzed in plasma and tissue samples. Additionally, real-time PCR was performed to evaluate the gene expression for oxidative stress and fat metabolism in the liver. Histological staining was also performed on pancreatic and hepatic tissues. RESULTS Purple potato extract lowered body weights and improved glucose utilization in the OGTT test in HF diet-fed rats. Purple potato extract also suppressed HF-diet-induced oxidative stress in plasma and hepatic tissues. Purple potato extract also restored the Nrf-2 expression in the liver, followed by the improved expression of HO-1, HO-2, and other antioxidant genes in HF diet-fed rats. In addition, genes involved in lipid metabolism were also positively modulated due to purple potato extract treatment. Furthermore, histological examination revealed the reduction of lipid accumulation and amelioration of inflammation due to the consumption of purple potato extract. CONCLUSION This investigation revealed that antioxidant-rich purple potato extract can modulate the antioxidant and fat metabolizing genes expression, ameliorated oxidative stress and glucose intolerance as well as lowered blood lipids in male rats.
Collapse
Affiliation(s)
- Ishrat Jahan
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Asif Ul Haque Shuvo
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Mirza Alimullah
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | | | | | - Shatil Rafia
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Ferdous Khan
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Khondoker Shahin Ahmed
- Chemical Research Division, BCSIR Laboratories, Dhaka, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Hemayet Hossain
- Chemical Research Division, BCSIR Laboratories, Dhaka, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | | | - Md Ashraful Alam
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Nusrat Subhan
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| |
Collapse
|
3
|
Zhao Y, Liu H, Lin K, Chen C, Chi Q, Zhang M, Li J, Pan W, Lou H. Chemical components of Salvia cavaleriei and their anti-inflammatory effect via NF-κB signaling pathway. Fitoterapia 2025; 182:106406. [PMID: 39909357 DOI: 10.1016/j.fitote.2025.106406] [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: 08/19/2024] [Revised: 01/14/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
In this study, with the aimed to explore the anti-inflammatory ingredients of Salvia cavaleriei, a phytochemical study with bioactive guided isolation for this plant have been taken. As a result, two pairs of new phenylacetic acid enantiomers, (±) cavalvia A (±1) and B (±2), and one previously undescribed germacrane-type sesquiterpenoid, salviesquiten A (3), along with ten known ones (4-13) were isolated from the ethyl acetate portion of 95 % ethanol extract. Their chemical structures were established based on UV, IR, MS and NMR spectroscopic analyses, as well as PD4+ probability analysis and electronic circular dichroism (ECD) methods. Anti-inflammatory bioassay showed that compound 6 exhibited significant effect against LPS-induced NO production in the RAW264.7 macrophages model, with the IC50 value of 3.40 ± 0.24 μM. Furthermore, compound 6 significantly suppressed the production of pro-inflammatory cytokines (IL-6, IL-1β, and TNF-α) and the protein expression of the enzymes iNOS and COX-2 in a concentration-dependent manner. And further investigation showed that 6 exerts anti-inflammatory effect via NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yumin Zhao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 550014, PR China; Luoyang Branch of Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Luoyang Hospital of TCM, Luoyang 471000, PR China
| | - Hanfei Liu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang 550014, PR China
| | - Kaiqin Lin
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 550014, PR China
| | - Chao Chen
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang 550014, PR China
| | - Qin Chi
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang 550014, PR China
| | - Ming Zhang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang 550014, PR China
| | - Jinyu Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang 550014, PR China
| | - Weidong Pan
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 550014, PR China; School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China.
| | - Huayong Lou
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang 550014, PR China.
| |
Collapse
|
4
|
Bashandy SAE, Elbaset MA, Ibrahim FAA, Abdelrahman SS, Moussa SAA, El-Seidy AMA. Management of cardiovascular disease by cerium oxide nanoparticles via alleviating oxidative stress and adipokine abnormalities. Sci Rep 2025; 15:5709. [PMID: 39962072 PMCID: PMC11833101 DOI: 10.1038/s41598-025-85794-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
The current study aimed to evaluate the role of cerium oxide nanoparticles (C-1), a potent antioxidant, in the medication of cardiovascular disease in obese animal model. C-1 was prepared using a modified sonication sol-gel method. Thirty-two adult male rats were equally divided into 4 groups (n=8/each). The first (control) and second (obese) groups are not treated while the obese rats in the third and fourth groups were given 15 and 30 mg/kg C-1(IP), respectively, for 8 weeks. Parameters of insulin resistance, adipocyte hormones, inflammatory markers, lipid profile, cardiac enzymes and cardiac iron content (C-Fe) were estimated. Moreover, histological study and immunohistochemical stain for inducible nitric oxide synthase (INOS) for cardiac and aortic tissues were performed. The XRD patterns of C-1 showed narrow symmetric diffraction peaks. The particle diameters were calculated from the TEM histogram (21.09 nm) and the Debye-Scherrer Method (20.74 nm) which were very similar. Using the most intense peak ( 28 . 47 ∘ ), structural parameters were calculated including nano-crystallite size, Micro-strain, Lorentz factor, Thomson polarization parameter, and Lorentz polarization parameter. BET was used to calculate The total surface area (ST ), and specific surface area (SBET ). The XPS survey spectrum of C-1 showed peaks for C-1s, O-1s and Ce-3d. The treatment of obese rats with C-1 led to a significant decrease in body weight, C-Fe , plasma leptin, tumor necrosis factor-alpha (TNF α ), interleukin-6 (IL6), C-reactive protein (CRP), resistin, cholesterol, triglycerides, low-density lipoprotein (LDL), Troponin, Creatinine Kinase-MB (CK-MB), lactate dehydrogenase (LDH), and malondialdehyde (MDA) in cardiac tissue or in plasma. Also, C-1 lowered plasma monocyte chemoattractant protein-1 (MCP-1), Epithelial Neutrophil-Activating Peptide (ENA-78), and insulin and glucose levels in obese rats. Furthermore, C-1 alleviated the increase of cardiac iNOS. Moreover, C-1 mitigated pathological changes of cardiac muscle and aorta observed in obese rats. On the other hand, C-1 enhanced adiponectin, cardiac glutathione (GSH) and superoxide dismutase (SOD) in obese rats. The effect of C-1 is dose-dependent ( 30 mg/kg of C-1 is more evident than 15 mg/kg). The modified synthesis method may lead to a smaller particle size than that reported in our previously reported work. The XRD patterns of C-1 indicate its cubic structure with space group F m -3 m (225) which was matched by code id 4343161 from COD. The Raman spectrum of C-1 indicates the absence of rearrangement oxygen atoms, the presence of oxygen in its fluorite lattice positions, and the oxygen vacancies in C-1 and the Ce vibration model (F2g). The presence of ten peaks in the high-resolution Ce-3d XP spectrum indicates the existence of both Ce3+ and Ce4+. C-1 showed therapeutic efficacy in atherosclerosis and cardiac muscle abnormalities associated with obese rats, probably because of their antioxidant and anti-inflammatory properties, which lead to lowering oxidative stress.
Collapse
Affiliation(s)
- Samir A E Bashandy
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, El-bohouth St., P.O. 12622, Dokki, Cairo, Egypt
| | - Marawan A Elbaset
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, El-bohouth St., P.O. 12622, Dokki, Cairo, Egypt.
| | - Fatma A A Ibrahim
- Department of Biochemistry, Biotechnology Research Institute, National Research Centre, El-bohouth St., P.O. 12622, Dokki, Cairo, Egypt
| | - Sahar S Abdelrahman
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Sherif A Abdelmottaleb Moussa
- Department of Biochemistry, Biotechnology Research Institute, National Research Centre, El-bohouth St., P.O. 12622, Dokki, Cairo, Egypt
| | - Ahmed M A El-Seidy
- Inorganic Chemistry Department, Advanced Materials Technology & Mineral Resources Research Institute, National Research Centre, El-bohouth St., P.O. 12622, Dokki, Cairo, Egypt.
| |
Collapse
|
5
|
Ahmedy GS, Selim HM, El-Aasr M, Ibrahim SM, El-Sherbeni SA. Phytochemicals of Vitis vinifera L. var. King Ruby protect mice from benzo(a)pyrene-induced lung injury. Sci Rep 2025; 15:4536. [PMID: 39915503 PMCID: PMC11802910 DOI: 10.1038/s41598-025-86173-x] [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: 09/12/2024] [Accepted: 01/08/2025] [Indexed: 02/09/2025] Open
Abstract
The world's concern about smoking hazards, chronic obstructive pulmonary disease (COPD) and lung cancer was the motivation to investigate plants as a source of new drugs with lung protective effect. The phytochemical profile of Vitis vinifera L. var. King Ruby leaves methanol extract (VLME) was tentatively recognized by liquid chromatography electrospray ionization tandem mass spectrometric (LC-ESI-MS/MS). Fifty-two and forty-seven compounds were identified by negative and positive ESI modes, respectively. Taraxerol (1), β-sitosterol (2), daucosterol (3), quercetin-3-O-β-D-glucuronoide-6″-methyl ester (4) and isoquercetin (5) were isolated from VLME. The sulforhodamine B (SRB) assay of the different fractions against A-549 cell line revealed that the methylene chloride fraction (MCF) had the lowest cell viability at 300 µg/mL (4.54 ± 0.19%). Mice of 10 groups (n = 6) was treated as follows: Group I (negative control group), group II (disease control, mice received B(a)P 125 mg/kg, orally), groups III-V (mice received 100, 200, and 300 mg/kg of VLME, followed by B(a)P), group VI (mice received only 300 mg/kg of VLME), groups VII-XI (mice received 100, 200, and 300 mg/kg of MCF, followed by B(a)P), group X (mice received only 300 mg/kg of MCF). On the seventh day, all groups received a single oral dose of B(a)P 125 mg/kg body, except group I, VI and X. In vivo studies showed VLME and MCF (300 mg/kg body weight) effectively mitigated benzo(a)pyrene-induced lung injuries in mice. The anti-inflammatory effects were confirmed by the downregulation of cyclooxygenase-2 (COX-2) and CD34, alongside reduced nuclear factor-kappa B (NF-κB) expression. Antioxidant activity was indicated by decreased malondialdehyde (MDA) levels and inducible nitric oxide synthase (iNOS) expression with the remarkable increase in glutathione (GSH). Histological improvements further support the potential of Vitis vinifera L. leaves as a natural lung protectant. Further pre-clinical and clinical investigations will be required to deliver a new drug with promising protection effect.
Collapse
Affiliation(s)
- Gehad S Ahmedy
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Hend M Selim
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Mona El-Aasr
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Souzan M Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Suzy A El-Sherbeni
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| |
Collapse
|
6
|
Akyuz E, Arulsamy A, Aslan FS, Sarisözen B, Guney B, Hekimoglu A, Yilmaz BN, Retinasamy T, Shaikh MF. An Expanded Narrative Review of Neurotransmitters on Alzheimer's Disease: The Role of Therapeutic Interventions on Neurotransmission. Mol Neurobiol 2025; 62:1631-1674. [PMID: 39012443 PMCID: PMC11772559 DOI: 10.1007/s12035-024-04333-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: 07/06/2023] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. The accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles are the key players responsible for the pathogenesis of the disease. The accumulation of Aβ plaques and tau affect the balance in chemical neurotransmitters in the brain. Thus, the current review examined the role of neurotransmitters in the pathogenesis of Alzheimer's disease and discusses the alterations in the neurochemical activity and cross talk with their receptors and transporters. In the presence of Aβ plaques and neurofibrillary tangles, changes may occur in the expression of neuronal receptors which in turn triggers excessive release of glutamate into the synaptic cleft contributing to cell death and neuronal damage. The GABAergic system may also be affected by AD pathology in a similar way. In addition, decreased receptors in the cholinergic system and dysfunction in the dopamine neurotransmission of AD pathology may also contribute to the damage to cognitive function. Moreover, the presence of deficiencies in noradrenergic neurons within the locus coeruleus in AD suggests that noradrenergic stimulation could be useful in addressing its pathophysiology. The regulation of melatonin, known for its effectiveness in enhancing cognitive function and preventing Aβ accumulation, along with the involvement of the serotonergic system and histaminergic system in cognition and memory, becomes remarkable for promoting neurotransmission in AD. Additionally, nitric oxide and adenosine-based therapeutic approaches play a protective role in AD by preventing neuroinflammation. Overall, neurotransmitter-based therapeutic strategies emerge as pivotal for addressing neurotransmitter homeostasis and neurotransmission in the context of AD. This review discussed the potential for neurotransmitter-based drugs to be effective in slowing and correcting the neurodegenerative processes in AD by targeting the neurochemical imbalance in the brain. Therefore, neurotransmitter-based drugs could serve as a future therapeutic strategy to tackle AD.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, International School of Medicine, University of Health Sciences, Istanbul, Turkey
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
| | | | - Bugra Sarisözen
- School of Medicine, Tekirdağ Namık Kemal University, Tekirdağ, Turkey
| | - Beyzanur Guney
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | | | - Beyza Nur Yilmaz
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, New South Wales, 2800, Australia.
| |
Collapse
|
7
|
Li Z, Fan X, Liu Y, Yue M, Wu T, Wang X, Jiang W, Fan K. Engineering Mild-Photothermal Responsive and NO Donor Prussian Blue Nanozymes Using Mild Synthesis for Inflammation Regulation and Bacterial Eradication in Periodontal Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2409840. [PMID: 39690880 DOI: 10.1002/adma.202409840] [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: 07/08/2024] [Revised: 11/26/2024] [Indexed: 12/19/2024]
Abstract
Periodontitis, an infectious disease of periodontal tissues caused by oral bacterial biofilms, is characterized by reactive oxygen species (ROS) accumulation and immune microenvironment imbalance. Multifunctional nanozymes, leveraging their physiochemical properties and enzymatic activities, offer promising antibacterial and anti-inflammatory strategies for managing periodontitis. In particular, Prussian blue nanozymes (PBzymes) exhibit exceptional ROS control due to their robust catalytic activity, diverse antioxidant functions, and high biocompatibility. However, the practical application of traditional high-temperature synthesis methods is limited. This study introduces a class of metal-engineered PBzymes synthesized at room temperature, identified for their potent antioxidative activity and excellent photothermal performance at mild temperatures. Nitric oxide (NO) gas therapy offers promising strategies for targeting deep infections in periodontal tissues. Thus, sodium nitroprusside is introduced into PBzyme to create SPBzyme via an in situ loading method. NO release by SPBzyme enhances antibacterial effects and overcomes resistance linked to bacterial biofilms, resulting in mild-photothermal antibacterial properties and synergistic antioxidant effects. In vitro antibacterial assays demonstrate the superior efficacy of SPBzyme under mild temperature conditions and near-infrared light exposure. Furthermore, SPBzyme effectively reduces inflammation and has positive therapeutic effects in periodontal animal models. Overall, mild-temperature photothermal NO release nanozyme therapy represents a novel approach for treating periodontitis.
Collapse
Affiliation(s)
- Zheng Li
- Department of Prosthodontics, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, Haidian District, 100081, P. R. China
| | - Xiaowan Fan
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, P. R. China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, P. R. China
| | - Ying Liu
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Muxin Yue
- Institute of Medical Technology, Peking University Health Science Center, 38 Xueyuan Road, Beijing, Haidian District, 100191, P. R. China
| | - Tingting Wu
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Xing Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, P. R. China
| | - Wei Jiang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, P. R. China
| |
Collapse
|
8
|
Videtta G, Sasia C, Galeotti N. High Rosmarinic Acid Content Melissa officinalis L. Phytocomplex Modulates Microglia Neuroinflammation Induced by High Glucose. Antioxidants (Basel) 2025; 14:161. [PMID: 40002348 PMCID: PMC11851730 DOI: 10.3390/antiox14020161] [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: 12/29/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Diabetic patients experience hyperglycemia, which can affect multiple organs, including brain function, leading to disabling neurological complications. Hyperglycemia plays a key role in promoting neuroinflammation, the most common complication in diabetic individuals, through the activation of microglia. Attenuating hyperglycemia-related neuroinflammation in microglia may reduce diabetes-associated neurological comorbidities. Natural remedies containing phenolic compounds have shown efficacy in mitigating microglia-mediated neuroinflammation. The aim of this study was to investigate the potential of a Melissa officinalis L. (MO) phytocomplex, obtained from plant cell cultures and enriched in its main polyphenolic constituent, rosmarinic acid (RA), in attenuating hyperglycemia-induced neuroinflammation in microglia. A time-course morphological analysis of BV2 microglial cells exposed to high glucose (HG) levels showed a shift towards a proinflammatory phenotype, peaking after 48 h, which was reversed by pretreatment with MO. Biochemical assays revealed increased expression of the microglial marker CD11b (187%), activation of the NF-κB pathway (179%), expression of iNOS (225%), enhanced phosphorylation of ERK1/2 (180%), and increased expression of the proinflammatory cytokine IL-6 (173%). Pretreatment with MO prevented the aberrant expression of these proinflammatory mediators and restored SIRT1 levels. Exposure of neuronal SH-SY5Y cells to the conditioned medium from HG-exposed microglia significantly reduced cell viability. MO counteracted this effect, exhibiting neuroprotective activity. RA showed efficacy comparable to that of MO. In conclusion, MO and RA attenuated microglia-mediated oxidative imbalance and neuroinflammation under HG exposure by inhibiting the morphological shift toward a proinflammatory phenotype induced by HG and abrogating the subsequent activation of the downstream ERK1/2-NF-κB-iNOS pathway.
Collapse
Affiliation(s)
| | | | - Nicoletta Galeotti
- Department of Neurosciences, Psychology, Drug Research and Child Health (Neurofarba), Section of Pharmacology and Toxicology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy; (G.V.); (C.S.)
| |
Collapse
|
9
|
Rahman AFMT, Benko A, Bulbule S, Gottschalk CG, Arnold LA, Roy A. Tetrahydrobiopterin in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Friend or Foe? Biomolecules 2025; 15:102. [PMID: 39858496 PMCID: PMC11763651 DOI: 10.3390/biom15010102] [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: 11/05/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Myalgic Encephalomyelitis or Chronic Fatigue Syndrome (ME/CFS) is a chronic multisystem disease characterized by severe muscle fatigue, pain, dizziness, and brain fog. The two most common symptoms are post-exertional malaise (PEM) and orthostatic intolerance (OI). ME/CFS patients with OI (ME+OI) suffer from dizziness or faintness due to a sudden drop in blood pressure while maintaining an upright posture. Clinical research has demonstrated that patients with OI display severe cardiovascular abnormalities resulting in reduced effective blood flow in the cerebral blood vessels. However, despite intense investigation, it is not known why the effective cerebral blood flow is reduced in OI patients. Based on our recent findings, we observed that tetrahydrobiopterin (BH4) metabolism was highly dysregulated in ME+OI patients. In the current review article, we attempted to summarize our recent findings on BH4 metabolism to shed light on the molecular mechanisms of OI.
Collapse
Affiliation(s)
- A. F. M. Towheedur Rahman
- Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA; (A.F.M.T.R.); (A.B.); (L.A.A.)
| | - Anna Benko
- Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA; (A.F.M.T.R.); (A.B.); (L.A.A.)
| | - Sarojini Bulbule
- Research and Development Laboratory, Chemistry Building, 2000 E Kenwood Blvd, Suite #320, Milwaukee, WI 53211, USA; (S.B.); (C.G.G.)
| | - Carl Gunnar Gottschalk
- Research and Development Laboratory, Chemistry Building, 2000 E Kenwood Blvd, Suite #320, Milwaukee, WI 53211, USA; (S.B.); (C.G.G.)
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, USA
| | - Leggy A. Arnold
- Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA; (A.F.M.T.R.); (A.B.); (L.A.A.)
| | - Avik Roy
- Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA; (A.F.M.T.R.); (A.B.); (L.A.A.)
- Research and Development Laboratory, Chemistry Building, 2000 E Kenwood Blvd, Suite #320, Milwaukee, WI 53211, USA; (S.B.); (C.G.G.)
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, USA
| |
Collapse
|
10
|
Liu J, Gao J, Xiong A, Li G, Zhong K, Bai L, Li X, Ran Q, Xiong Y, He X. Exploring Cistanche's therapeutic potential and molecular mechanisms in asthma treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156265. [PMID: 39612890 DOI: 10.1016/j.phymed.2024.156265] [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: 04/28/2024] [Revised: 08/08/2024] [Accepted: 11/14/2024] [Indexed: 12/01/2024]
Abstract
Asthma, a prevalent chronic respiratory disease, is characterized by airway inflammation and hyperresponsiveness, significantly impacting global health. Current treatments, including corticosteroids, face limitations due to variable patient responses and side effects. This study explores the therapeutic potential of Cistanche, a traditional Chinese medicinal herb, in an asthmatic mouse model induced by house dust mites (HDM). We employed a multidisciplinary approach, including transcriptomics, mass spectrometry, Weighted Gene Co-expression Network Analysis (WGCNA), and experimental validation in animal and cellular models. Our findings demonstrate that Cistanche significantly attenuates airway hyperresponsiveness, cellular infiltration, and mucus hypersecretion in asthmatic mice. Transcriptomic analysis identified 643 differentially expressed genes (DEGs), with Cistanche treatment disrupting pathways associated with IL-4, IL-5, and eosinophils. Gene Set Enrichment Analysis (GSEA) revealed Cistanche's involvement in glucocorticoid processes, suggesting its potential as an alternative or complementary therapy to conventional glucocorticoids. Liquid Chromatography-High Resolution Mass Spectrometry (LCHRMS) analysis identified Pinoresinol 4-O-Beta-D-Glucopyranoside (PG) as a key active component of Cistanche, with targets predominantly associated with the PI3K-AKT pathway. WGCNA analysis of an asthma dataset identified the "skyblue" module as highly associated with asthma, with inducible nitric oxide synthase (iNOS) emerging as a core gene. Molecular docking confirmed a strong binding affinity between PG and iNOS, with Cistanche treatment down-regulating iNOS expression, suggesting a potential mechanism for its anti-inflammatory effects. In conclusion, Cistanche exhibits therapeutic effects in asthma by modulating immune responses and targeting key asthma-related genes and proteins. Our study provides novel insights into the treatment of asthma with natural products and highlights the potential of Cistanche as an alternative or complementary therapeutic agent. Further research is warranted to validate these findings and explore the integration of Cistanche into clinical asthma management.
Collapse
Affiliation(s)
- Jiliu Liu
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, Affiliated Hospital of Southwest Jiaotong University, the Third People's Hospital of Chengdu, Chengdu 610031, China; Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Jie Gao
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, Affiliated Hospital of Southwest Jiaotong University, the Third People's Hospital of Chengdu, Chengdu 610031, China; Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Anying Xiong
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, Affiliated Hospital of Southwest Jiaotong University, the Third People's Hospital of Chengdu, Chengdu 610031, China; Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Guoping Li
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, Affiliated Hospital of Southwest Jiaotong University, the Third People's Hospital of Chengdu, Chengdu 610031, China; Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Kai Zhong
- College of Biomass Science and Engineering, Sichuan University, Chengdu, China
| | - Lingling Bai
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, Affiliated Hospital of Southwest Jiaotong University, the Third People's Hospital of Chengdu, Chengdu 610031, China; Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Xiaolan Li
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, Affiliated Hospital of Southwest Jiaotong University, the Third People's Hospital of Chengdu, Chengdu 610031, China; Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China; National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Qin Ran
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, Affiliated Hospital of Southwest Jiaotong University, the Third People's Hospital of Chengdu, Chengdu 610031, China; Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Ying Xiong
- Department of Pulmonary and Critical Care Medicine, Sichuan Friendship Hospital, Chengdu 610000, China
| | - Xiang He
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, Affiliated Hospital of Southwest Jiaotong University, the Third People's Hospital of Chengdu, Chengdu 610031, China; Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China; National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China.
| |
Collapse
|
11
|
Huang Z, Zhang W, Shu Q, Guo XC, Zheng X, Lu YJ. Synergistic Anti-Inflammatory Effects of Dibenzoylmethane and Silibinin: Insights From LPS-Induced RAW 264.7 Cells and TPA-Induced Mouse Model. Chem Biodivers 2025:e202402567. [PMID: 39743480 DOI: 10.1002/cbdv.202402567] [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: 10/08/2024] [Revised: 12/11/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Inflammation is an important predisposing factor for many chronic diseases. The dietary flavonoid silibinin (SB) has excellent anti-inflammatory properties in cells, but its low bioavailability in the blood compromises its therapeutic potential. This study aims to investigate the potential of dibenzoylmethane (DBM) to synergistically enhance the anti-inflammatory benefits of SB. The synergistic effects of DBM and SB in combination were evaluated in lipopolysaccharide (LPS)-induced RAW264.7 cells and 12-O-tetradecanoylphorbol 13-acetate (TPA)-induced mice. In addition, a network pharmacology approach and molecular docking were used to explore the key targets and signaling pathways of DBM and SB in combination. The results showed that DBM and SB synergistically inhibited the production of nitric oxide (NO), reactive oxygen species (ROS), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α) in a 1:1 concentration ratio. These two compounds may exert their synergistic effects by modulating the nuclear factor kappa-B (NF-κB) and HIF-1 signaling pathways, among others. Molecular docking revealed that both compounds exhibited high binding affinities to inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Compared with single-compound use, the two compounds in combination significantly reduced ear edema and inflammatory cell infiltration and inhibited the protein expression of iNOS and COX-2 in TPA-induced mice. This research provides a rationale for the combination of DBM and SB as an effective anti-inflammatory agent.
Collapse
Affiliation(s)
- Zebin Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Wanying Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Qi Shu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Xiao-Chun Guo
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Xi Zheng
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Yu-Jing Lu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
- Guangdong Xianlingtong Biopharmaceutical Technology Co., Ltd, Meizhou, China
| |
Collapse
|
12
|
Xiang H, Lan Y, Hu L, Qin R, Li H, Weng T, Zou Y, Liu Y, Hu X, Ge W, Zhang H, Pan HL, Yang NN, Liu W, Cai G, Li M. AMPK activation mitigates inflammatory pain by modulating STAT3 phosphorylation in inflamed tissue macrophages of adult male mice. Mol Pain 2025; 21:17448069251321339. [PMID: 39921559 PMCID: PMC11843706 DOI: 10.1177/17448069251321339] [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: 11/21/2024] [Revised: 01/08/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
Inflammatory pain presents a significant clinical challenge. AMP-activated protein kinase (AMPK) is recognized for its capacity to alleviate inflammation by inhibiting transcription factors such as nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription (STAT). Our prior research demonstrated that AMPK reduces inflammatory pain by inhibiting NF-κB activation and interleukin-1 beta (IL-1β) expression. However, the role of AMPK in regulating reactive oxygen species (ROS) and inducible nitric oxide synthase (iNOS) by modulating STAT3 phosphorylation in inflammatory pain remains inadequately understood. This study aims to investigate the role of AMPK in modulating STAT3 phosphorylation in the macrophages of inflamed tissues to mitigate inflammatory pain. A Complete Freund's Adjuvant (CFA)-induced inflammatory pain model was established by subcutaneous injection into the plantar surface of the left hindpaw of adult male mice. Behavioral tests of mechanical allodynia and thermal latency were used to determine nociceptive behavior. Immunoblotting quantified p-AMPK and iNOS expression levels. Nuclear translocation of p-STAT3(Ser727) and STAT3 in macrophages was assessed by western blot and immunofluorescence. ROS accumulation and mitochondrial damage in NR8383 macrophages were detected by flow cytometry. Lentivirus infection cells experiment was performed to transfect vectors encoding the STAT3 S727D mutants. Treatment with the AMPK activator AICAR alleviated CFA-induced inflammatory pain, enhanced AMPK phosphorylation, and reduced iNOS expression in inflamed skin tissues. AICAR effectively prevented STAT3 nuclear translocation while promoting the phosphorylation of STAT3 (Ser727) in the cytoplasm. In vitro studies with CFA-stimulated NR8383 macrophages revealed that AICAR increased STAT3(Ser727) phosphorylation, curtailed iNOS expression, and attenuated ROS accumulation and mitochondrial damage. Furthermore, the S727D mutation, which enhances STAT3 phosphorylation, replicated the protective effects of AICAR against CFA-induced oxidative stress and mitochondrial dysfunction. Our study shows that the AMPK acitvation downregulates iNOS expression by inhibiting the STAT3 nuclear translocation and promotes cytoplasmic STAT3(Ser727) phosphorylation, which reduces ROS expression and mitochondrial dysfunction, thereby alleviating inflammatory pain. These findings underscore the therapeutic potential of targeting AMPK and STAT3 pathways in inflammatory pain management.
Collapse
Affiliation(s)
- Hongchun Xiang
- Department of Acupuncture-Moxibustion, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
- School of Basic Medicine, Tongji Medical College, Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Yuye Lan
- School of Basic Medicine, Tongji Medical College, Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Hu
- Department of Pharmacology, School of Basic Medicine, Nanjing Medical University, Nanjing, China
| | - Renjie Qin
- School of Basic Medicine, Tongji Medical College, Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Hongping Li
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture- Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Tao Weng
- School of Basic Medicine, Tongji Medical College, Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zou
- School of Basic Medicine, Tongji Medical College, Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Yongmin Liu
- School of Basic Medicine, Tongji Medical College, Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Xuefei Hu
- School of Basic Medicine, Tongji Medical College, Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Wenqiang Ge
- School of Basic Medicine, Tongji Medical College, Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhang
- School of Basic Medicine, Tongji Medical College, Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Hui-Lin Pan
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Na-na Yang
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture- Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Wentao Liu
- Department of Pharmacology, School of Basic Medicine, Nanjing Medical University, Nanjing, China
| | - Guowei Cai
- Department of Acupuncture-Moxibustion, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Man Li
- School of Basic Medicine, Tongji Medical College, Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Li Y, Liao H, Zeng M, Gao D, Kong C, Liu W, Zheng Y, Zheng Q, Wang J. Exposure to polystyrene nanoplastics causes immune damage, oxidative stress and intestinal flora disruption in salamander (Andrias davidianus) larvae. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 949:175169. [PMID: 39094663 DOI: 10.1016/j.scitotenv.2024.175169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
The toxic effects of nanoparticles have been increasingly investigated, but there has been limited research on amphibians, especially those of conservation value. This study examined the effects of different concentrations (0, 0.04, 0.2, 1, 5 mg/L) of polystyrene nanoplastics (PS-NPs, 80 nm) on the short-term exposure (7 d) of Andrias davidianus. Results demonstrated the concentration-dependent enrichment of PS-NPs in the intestine. Histological lesions displayed increased hepatic macrophages with cellular rupture, broken intestinal villi, decreased cuprocytes and crypt depression. Antioxidant- and inflammation-related enzyme activities were analysed, and it was found that hepatic and intestinal MDA content and CAT activity were highest in the N-1 group and SOD activity was highest in the N-0.2 group (p < 0.05). AKP activity continued to decline, and iNOS activity was highest in the N-0.2 group (p < 0.05). il-10, tgf-β, bcl-w and txnl1 were significantly downregulated in the N-0.2 group, while il-6 and il-8 were markedly upregulated in the N-0.2 group (p < 0.05). Exposing to PS-NPs decreased probiotic bacteria (Cetobacterium, Akkermansia) and increased pathogenic bacteria (Lachnoclostridium). Our results suggest that NPs exposure can have deleterious effects on salamanders, which predicts that NPs contamination may lead to continued amphibian declines. Therefore, we strongly recommend that attention be paid to amphibians, especially endangered species, in the field of NPs.
Collapse
Affiliation(s)
- Ye Li
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Hongping Liao
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Min Zeng
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Dandan Gao
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Chunmiao Kong
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Wanjing Liu
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Yufeng Zheng
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Qingzhi Zheng
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Jun Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; Institute of Eco-Environmental Research, Guangxi Academy of Sciences, Nanning 530007, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangzhou 510006, China.
| |
Collapse
|
14
|
Zhang X, Sun L, Wu M, Yu C, Zhao D, Wang L, Zhang Z, Yi D, Hou Y, Wu T. Effect of supplementation with Lactobacillus rhamnosus GG powder on intestinal and liver damage in broiler chickens challenged by lipopolysaccharide. Front Microbiol 2024; 15:1466274. [PMID: 39534507 PMCID: PMC11555397 DOI: 10.3389/fmicb.2024.1466274] [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: 07/17/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
This study explores the effect of dietary along with Lactobacillus rhamnosus GG (LGG) powder on intestinal and liver damage in broiler chickens challenged by lipopolysaccharide (LPS). A total of 100 healthy 1-day-old Ross 308 broiler chickens were selected and randomly divided into two treatments: the control group and the LGG treatment group. There were five replicates for each group, with 10 chickens per replicate. The chickens in the control group were fed a basal diet, while LGG treatment was supplemented with 1,000 mg/kg LGG along with the basal diet. The experiment lasted 29 days, and the trial included two phases. During the first 27 days, the animals were weighed on the 14th and 27th days to calculate growth performance. Then, on day 29, 2 animals from each replicate were intraperitoneally injected with 1 mg/kg BW LPS, and another 2 animals were treated with an equal volume of saline. The chickens were slaughtered 3 h later for sampling and further analysis. (1) LGG addition to the diet did not affect growth performance, including average daily gain (ADG), average daily feed intake (ADFI), and feed-to-weight ratio (F/G) of broiler chickens; (2) LPS stimulation decreased villus height (VH), and caused oxidative stress and increased the amount of diamine oxidase (DAO) in plasma, and the relative expression of intestinal inflammation genes (interleukin-8 [IL-8], interleukin 1β [IL-1β], inducible nitric oxide synthase [iNOS], and tumor necrosis factor-α [TNF-α]) and the relative expression of liver injury genes (b-cell lymphoma 2 [BCL2], heat shock protein70 [HSP70], and matrix metallopeptidase 13 [MMP13]). (3) Supplementation of LGG increased VH and the relative expression of intestinal barrier genes (mucins 2 [Mucin2] and occludin [Occludin]) and decreased the amount of DAO in plasma and the relative expression of intestinal inflammatory factors (IL-8, iNOS, and IL-1β). LGG supplementation also increased the expression of liver injury-related genes (MMP13 and MMP9). In conclusion, LGG enhanced intestinal barrier function, improved intestinal morphology, and alleviated the intestines' inflammatory response in LPS-stimulated broiler chicken, and it has a slightly protective effect on liver damage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tao Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
15
|
Nxumalo MB, Ntanzi N, Kumalo HM, Khan RB. Mitigating Hyperglycaemic Oxidative Stress in HepG2 Cells: The Role of Carica papaya Leaf and Root Extracts in Promoting Glucose Uptake and Antioxidant Defence. Nutrients 2024; 16:3496. [PMID: 39458491 PMCID: PMC11510471 DOI: 10.3390/nu16203496] [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: 09/05/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Diabetes often goes undiagnosed, with 60% of people in Africa unaware of their condition. Type 2 diabetes mellitus (T2DM) is associated with insulin resistance and is treated with metformin, despite the undesirable side effects. Medicinal plants with therapeutic potential, such as Carica papaya, have shown promising anti-diabetic properties. This study explored the role of C. papaya leaf and root extracts compared to metformin in reducing hyperglycaemia-induced oxidative stress and their impact on liver function using HepG2 as a reference. Methods: The cytotoxicity was assessed through the MTT assay. At the same time, glucose uptake and metabolism (ATP and ∆Ψm) in HepG2 cells treated with C. papaya aqueous leaf and root extract were evaluated using a luminometry assay. Additionally, antioxidant properties (SOD2, GPx1, GSH, and Nrf2) were measured using qPCR and Western blot following the detection of MDA, NO, and iNOS, indicators of free radicals. Results: The MTT assay showed that C. papaya extracts did not exhibit toxicity in HepG2 cells and enhanced glucose uptake compared to the hyperglycaemic control (HGC) and metformin. The glucose levels in C. papaya-treated cells increased ATP production (p < 0.05), while the ∆Ψm was significantly increased in HGR1000-treated cells (p < 0.05). Furthermore, C. papaya leaf extract upregulated GPx1 (p < 0.05), GSH, and Nrf2 gene (p < 0.05), while SOD2 and Nrf2 proteins were reduced (p > 0.05), ultimately lowering ROS (p > 0.05). Contrarily, the root extract stimulated SOD2 (p > 0.05), GPx1 (p < 0.05), and GSH levels (p < 0.05), reducing Nrf2 gene and protein expression (p < 0.05) and resulting in high MDA levels (p < 0.05). Additionally, the extracts elevated NO levels and iNOS expression (p < 0.05), suggesting potential RNS activation. Conclusion: Taken together, the leaf extract stimulated glucose metabolism and triggered ROS production, producing a strong antioxidant response that was more effective than the root extract and metformin. However, the root extract, particularly at high concentrations, was less effective at neutralising free radicals as it did not stimulate Nrf2 production, but it did maintain elevated levels of SOD2, GSH, and GPx1 antioxidants.
Collapse
Affiliation(s)
- Mthokozisi Bongani Nxumalo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (N.N.); (H.M.K.); (R.B.K.)
| | | | | | | |
Collapse
|
16
|
Liu J, Luo R, Zhang Y, Li X. Current status and perspective on molecular targets and therapeutic intervention strategy in hepatic ischemia-reperfusion injury. Clin Mol Hepatol 2024; 30:585-619. [PMID: 38946464 PMCID: PMC11540405 DOI: 10.3350/cmh.2024.0222] [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: 04/03/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024] Open
Abstract
Hepatic ischemia‒reperfusion injury (HIRI) is a common and inevitable complication of hepatic trauma, liver resection, or liver transplantation. It contributes to postoperative organ failure or tissue rejection, eventually affecting patient prognosis and overall survival. The pathological mechanism of HIRI is highly complex and has not yet been fully elucidated. The proposed underlying mechanisms include mitochondrial damage, oxidative stress imbalance, abnormal cell death, immune cell hyperactivation, intracellular inflammatory disorders and other complex events. In addition to serious clinical limitations, available antagonistic drugs and specific treatment regimens are still lacking. Therefore, there is an urgent need to not only clarify the exact etiology of HIRI but also reveal the possible reactions and bottlenecks of existing drugs, helping to reduce morbidity and shorten hospitalizations. We analyzed the possible underlying mechanism of HIRI, discussed various outcomes among different animal models and explored neglected potential therapeutic strategies for HIRI treatment. By thoroughly reviewing and analyzing the literature on HIRI, we gained a comprehensive understanding of the current research status in related fields and identified valuable references for future clinical and scientific investigations.
Collapse
Affiliation(s)
- Jia Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Ranyi Luo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yinhao Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
17
|
Duailibe JBB, Viau CM, Saffi J, Fernandes SA, Porawski M. Protective effect of long-chain polyunsaturated fatty acids on hepatorenal syndrome in rats. World J Nephrol 2024; 13:95627. [PMID: 39351184 PMCID: PMC11439093 DOI: 10.5527/wjn.v13.i3.95627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/07/2024] [Accepted: 07/25/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Hepatorenal syndrome (HRS) is the most prevalent form of acute kidney injury in cirrhotic patients. It is characterized by reduced renal blood flow and represents the most severe complication in cirrhotic patients with advanced disease. Previous research has indicated that antioxidants can delay the onset of a hyperdynamic circulatory state in cirrhosis and improve renal function in HRS patients. Regular omega-3 supplementation has significantly reduced the risk of liver disease. This supplementation could represent an additional therapy for individuals with HRS. AIM To evaluated the antioxidant effect of omega-3 polyunsaturated fatty acid supplementation on the kidneys of cirrhotic rats. METHODS Secondary biliary cirrhosis was induced in rats by biliary duct ligation (BDL) for 28 d. We used 24 male Wistar rats divided into the following groups: I (control); II (treated with omega-3, 1 g/kg of body weight); III (BDL treated with omega-3, 1 g/kg of body weight); and IV (BDL without treatment). The animals were killed by overdose of anesthetic; the kidneys were dissected, removed, frozen in liquid nitrogen, and stored in a freezer at -80℃ for later analysis. We evaluated oxidative stress, nitric oxide (NO) metabolites, DNA damage by the comet assay, cell viability test, and apoptosis in the kidneys. Data were analyzed by one-way analysis of variance, and means were compared using the Tukey test, with P ≤ 0.05. RESULTS Omega-3 significantly decreased the production of reactive oxygen species (P < 0.001) and lipoperoxidation in the kidneys of cirrhotic rats treated with omega-3 (P < 0.001). The activity of the antioxidant enzymes superoxide dismutase and catalase increased in the BDL+omega-3 group compared to the BDL group (P < 0.01). NO production, DNA damage, and caspase-9 cleavage decreased significantly in the omega-3-treated BDL group. There was an increase in mitochondrial electrochemical potential (P < 0.001) in BDL treated with omega-3 compared to BDL. No changes in the cell survival index in HRS with omega-3 compared to the control group (P > 0.05) were observed. CONCLUSION The study demonstrates that omega-3 can protect cellular integrity and function by increasing antioxidant enzymes, inhibiting the formation of free radicals, and reducing apoptosis.
Collapse
Affiliation(s)
- João Bruno Beretta Duailibe
- Department of Hepatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Brazil
| | - Cassiana Macagnan Viau
- Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Brazil
| | - Jenifer Saffi
- Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Brazil
| | - Sabrina Alves Fernandes
- Department of Hepatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Brazil
| | - Marilene Porawski
- Department of Hepatology and Basic Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Brazil
| |
Collapse
|
18
|
Laviano HD, Gómez G, Núñez Y, García-Casco JM, Benítez RM, de Las Heras-Molina A, Gómez F, Sánchez-Esquiliche F, Martínez-Fernández B, González-Bulnes A, Rey AI, López-Bote CJ, Muñoz M, Óvilo C. Maternal dietary antioxidant supplementation regulates weaned piglets' adipose tissue transcriptome and morphology. PLoS One 2024; 19:e0310399. [PMID: 39264906 PMCID: PMC11392410 DOI: 10.1371/journal.pone.0310399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/30/2024] [Indexed: 09/14/2024] Open
Abstract
Antioxidant supplementation in critical periods may be useful for improvement of piglet early viability and development. We have evaluated the effects of maternal perinatal diet inclusion of a high vitamin E level (VE, 100 mg all-rac-α-tocopheryl acetate /kg), hydroxytyrosol (HT, 1.5 mg/kg), or their combination (VEHT), in comparison to a control diet (C, 30 mg all-rac-α-tocopheryl acetate /kg), on the offspring homeostasis and metabolism, analysing the weaned piglets' adipose tissue transcriptome and adipocyte morphology. Diets were provided to pregnant Iberian sows (n = 48, 12 per treatment) from gestation day 85 to weaning (28 days postpartum) and 48 piglets (n = 12 per treatment) were sampled 5 days postweaning for dorsal subcutaneous adipose tissue analyses. RNA obtained from 6 animals for each diet was used for paired-end RNA sequencing. Results show that supplementation of sows' diet with either vitamin E or hydroxytyrosol had substantial effects on weaned piglet adipose transcriptome, with 664 and 587 genes being differentially expressed, in comparison to C, respectively (q-value<0.10, Fold Change>1.5). Genes upregulated in C were mainly involved in inflammatory and immune response, as well as oxidative stress, and relevant canonical pathways and upstream regulators involved in these processes were predicted as activated, such as TNF, IFNB or NFKB. Vitamin E, when supplemented alone at high dose, activated lipid biosynthesis functions, pathways and regulators, this finding being accompanied by increased adipocyte size. Results suggest an improved metabolic and antioxidant status of adipose tissue in animals born from sows supplemented with individual antioxidants, while the combined supplementation barely affected gene expression, with VEHT showing a prooxidant/proinflamatory functional profile similar to C animals. Different hypothesis are proposed to explain this unexpected result. Findings allow a deeper understanding of the processes taking place in adipose tissue of genetically fat animals and the role of antioxidants in the regulation of fat cells function.
Collapse
Affiliation(s)
- Hernán D Laviano
- Departamento Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Gerardo Gómez
- Instituto Regional de Investigación y Desarrollo Agroalimentario y Forestal de Castilla-La Mancha (IRIAF), Toledo, Spain
| | - Yolanda Núñez
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, INIA, CSIC, Madrid, Spain
| | | | - Rita M Benítez
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, INIA, CSIC, Madrid, Spain
| | - Ana de Las Heras-Molina
- Departamento Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | | | | | | | - Antonio González-Bulnes
- Departamento de Producción y Sanidad Animal, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, Valencia, Spain
| | - Ana I Rey
- Departamento Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Clemente J López-Bote
- Departamento Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - María Muñoz
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, INIA, CSIC, Madrid, Spain
| | - Cristina Óvilo
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, INIA, CSIC, Madrid, Spain
| |
Collapse
|
19
|
Yang X, Xia L, Shen C, Li J, Dong X, Liu J. Curcumin alleviates heatstroke-induced liver injury in dry-heat environments by inhibiting the expression of NF-κB, iNOS, and ICAM-1 in rats. PLoS One 2024; 19:e0309598. [PMID: 39240880 PMCID: PMC11379272 DOI: 10.1371/journal.pone.0309598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 08/14/2024] [Indexed: 09/08/2024] Open
Abstract
we aimed to monitor liver injury in rat model during heat stress and heatstroke in dry-heat environment and investigate the effects of curcumin on heatstroke-induced liver injury and the underlying mechanisms. Sprague-Dawley (SD) rats were randomly divided into four groups: normal saline (NS), and 50 (50-cur), 100 (100-cur), and 200 mg/kg curcumin (200-cur) groups. They were administered the indicated doses of curcumin by gavage once daily for 7 days. On day 8, the rats were transferred to a simulated climate cabin, At 0, 50, 100, and 150 min, the core temperature (Tc) was measured respectively. After sacrificing the rats, tissue samples were collected, measure histology indices, serum enzymes, lipopolysaccharides (LPSs), cytokines, nuclear factor-kappa B (NF-κB), inducible nitric oxide synthase (iNOS), and intercellular adhesion molecule-1 (ICAM-1). The Tc increased with time in all groups. Curcumin alleviation of symptoms and improvement in pathological scores. The level of enzymes, LPS, and cytokines increased during heatstroke in the NS group, but curcumin decreased the levels of these indicators. The differences of the indicators between NS and 200-cur groups at 150 min were significant (P < 0.05). The expression of NF-κB p65, iNOS, and ICAM-1 was upregulated in the NS group at 150 min, but their expression was relatively lower in the curcumin groups (P < 0.05). Thus, our findings indicate acute liver injury during heat stress and heatstroke. The mechanism involves cascade-amplification inflammatory response induced by the gut endotoxin. Furthermore, curcumin alleviated heatstroke-induced liver injury in a dose-dependent manner by downregulating NF-κB, iNOS, and ICAM-1.
Collapse
Affiliation(s)
- Xinyue Yang
- Key Laboratory of Special Environmental Medicine of Xinjiang, Urumqi, China
- Graduate School of Xinjiang Medical University, Urumqi, China
| | - Liang Xia
- Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Caifu Shen
- Key Laboratory of Special Environmental Medicine of Xinjiang, Urumqi, China
| | - Jiajia Li
- Key Laboratory of Special Environmental Medicine of Xinjiang, Urumqi, China
| | - Xiang Dong
- Key Laboratory of Special Environmental Medicine of Xinjiang, Urumqi, China
| | - Jiangwei Liu
- Key Laboratory of Special Environmental Medicine of Xinjiang, Urumqi, China
| |
Collapse
|
20
|
Dimou A, Zachou K, Kostara C, Azariadis K, Giannoulis G, Lyberopoulou A, Bairaktari E, Dalekos GN. NMR-based metabolomic signature: An important tool for the diagnosis and study of pathogenesis of autoimmune hepatitis. Hepatology 2024; 80:266-277. [PMID: 38305739 DOI: 10.1097/hep.0000000000000767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 12/08/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND AND AIMS Metabolomics is used to predict, diagnose, and monitor metabolic disorders but altered metabolomic signatures have also been reported in diverse diseases, including autoimmune disorders. However, the metabolomic profile in autoimmune hepatitis (AIH) has not been investigated in depth. Therefore, we investigated the metabolomic signature of AIH and its significance as a diagnostic and pathogenetic tool. APPROACH AND RESULTS Metabolites in plasma samples from 50 patients with AIH at diagnosis, 43 healthy controls, 72 patients with primary biliary cholangitis (PBC), 26 patients with metabolic dysfunction-associated liver disease, and 101 patients with chronic viral hepatitis were determined by 1 H NMR (nuclear magnetic resonance) spectroscopy. Fifty-two metabolites were quantified, and metabolic pathway analysis was performed. Multivariate analysis revealed that AIH could be differentiated from healthy controls and each of the disease controls ( p <0.001). Fifteen metabolites differentiated AIH from disease controls (PBC+chronic viral hepatitis+metabolic dysfunction-associated liver disease) (95% sensitivity and 92% specificity). Ten distinct metabolic pathways were altered in AIH compared to disease controls. The metabolic pathway of branched-chain amino acids (lower valine, leucine, and isoleucine levels and their catabolic intermediates in PBC), methionine (lower methionine, 2-aminobutyrate, and 2-hydroxybutyrate levels in PBC), alanine-aspartate-glutamate (lower metabolites in PBC), and that of metabolites associated with gut microbiota (lower choline, betaine, and dimethylamine levels in PBC) were significantly different between AIH and PBC ( p <0.01). CONCLUSIONS 1 H NMR spectroscopy could be a promising novel tool to diagnose and study AIH pathogenesis as there is no need for much sample handling, is highly reproducible with high sensitivity and specificity, and low cost.
Collapse
Affiliation(s)
- Aikaterini Dimou
- Department of Biochemistry, Laboratory of Clinical Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Kalliopi Zachou
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, General University Hospital of Larissa, Larissa, Greece
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa, Greece
| | - Christina Kostara
- Department of Biochemistry, Laboratory of Clinical Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Kalliopi Azariadis
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, General University Hospital of Larissa, Larissa, Greece
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa, Greece
| | - George Giannoulis
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, General University Hospital of Larissa, Larissa, Greece
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa, Greece
| | - Aggeliki Lyberopoulou
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, General University Hospital of Larissa, Larissa, Greece
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa, Greece
| | - Eleni Bairaktari
- Department of Biochemistry, Laboratory of Clinical Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - George N Dalekos
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, General University Hospital of Larissa, Larissa, Greece
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa, Greece
| |
Collapse
|
21
|
Lee HW, Jung HS, Lee NK, Paik HD. Anti-Inflammatory Effects of Paraprobiotic Lactiplantibacillus plantarum KU15122 in LPS-Induced RAW 264.7 Cells. J Microbiol Biotechnol 2024; 34:1491-1500. [PMID: 38960876 PMCID: PMC11294648 DOI: 10.4014/jmb.2404.04052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/14/2024] [Accepted: 05/19/2024] [Indexed: 07/05/2024]
Abstract
Inflammation is a biodefense mechanism that provides protection against painful conditions such as inflammatory bowel disease, other gastrointestinal problems, and irritable bowel syndrome. Paraprobiotics have probiotic characteristics of intestinal modulation along with merits of safety and stability. In this study, heat-killed Lactiplantibacillus plantarum KU15122 (KU15122) was investigated for its anti-inflammatory properties. KU15122 was subjected to heat-killed treatment for enhancement of its safety, and its concentration was set at 8 log CFU/mL for conducting different experiments. Nitric oxide production was most remarkably reduced in the KU15122 group, whereas it was increased in the LPS-treated group. In RAW 264.7 cells, KU15122 inhibited the expression of inducible nitric oxide synthase, cyclooxygenase-2, interleukin (IL)-1β, IL-6, and tumor necrosis factor-α. ELISA revealed that among the tested strains, KU15122 exhibited the most significant reduction in PGE2, IL-1β, and IL-6. Moreover, KU15122 inhibited various factors involved in the nuclear factor-kappa B, activator protein-1, and mitogen-activated protein kinase pathways. In addition, KU15122 reduced the generation of reactive oxygen species. The anti-inflammatory effect of KU15122 was likely attributable to the bacterial exopolysaccharides. Conclusively, KU15122 exhibits anti-inflammatory potential against inflammatory diseases.
Collapse
Affiliation(s)
- Hye-Won Lee
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Hee-Su Jung
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Na-Kyoung Lee
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyun-Dong Paik
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
22
|
Gao L, Li YJ, Zhao JM, Liao YX, Qin MC, Li JJ, Shi H, Wong NK, Lyu ZP, Shen JG. Mechanism of Reactive Oxygen/Nitrogen Species in Liver Ischemia-Reperfusion Injury and Preventive Effect of Chinese Medicine. Chin J Integr Med 2024:10.1007/s11655-024-3810-9. [PMID: 38941044 DOI: 10.1007/s11655-024-3810-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 06/29/2024]
Abstract
Liver ischemia-reperfusion injury (LIRI) is a pathological process involving multiple injury factors and cell types, with different stages. Currently, protective drugs targeting a single condition are limited in efficacy, and interventions on immune cells will also be accompanied by a series of side effects. In the current bottleneck research stage, the multi-target and obvious clinical efficacy of Chinese medicine (CM) is expected to become a breakthrough point in the research and development of new drugs. In this review, we summarize the roles of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in various stages of hepatic ischemia-reperfusion and on various types of cells. Combined with the current research progress in reducing ROS/RNS with CM, new therapies and mechanisms for the treatment of hepatic ischemia-reperfusion are discussed.
Collapse
Affiliation(s)
- Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yun-Jia Li
- The First Affiliated Hospital/the First Clinical Medicine School of Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jia-Min Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yu-Xin Liao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Meng-Chen Qin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jun-Jie Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Hao Shi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Nai-Kei Wong
- State Key Discipline of Infectious Diseases, Shenzhen Third People's Hospital, the Second Affiliated Hospital, Shenzhen University, Shenzhen, Guangdong Province, 518112, China
| | - Zhi-Ping Lyu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Gang Shen
- School of Chinese Medicine, the University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
23
|
Chong YK, Ong YS, Yeong KY. Unveiling sultam in drug discovery: spotlight on the underexplored scaffold. RSC Med Chem 2024; 15:1798-1827. [PMID: 38911171 PMCID: PMC11187559 DOI: 10.1039/d3md00653k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/14/2024] [Indexed: 06/25/2024] Open
Abstract
Decades ago, the application of cyclic sulfonamide (sultam) and its derivatives primarily focused on their antibacterial properties. However, recent years have seen a shift in research attention towards exploring their potential as anticancer, anti-inflammatory, antidiabetic, and antiviral agents. Despite this broadening scope, only a few sultam drugs have made it to the commercial market, as much of the research on sultams remains in the discovery phase. This class of compounds holds significant promise and remains pertinent in pharmaceutical research. Due to sultam's relevance and growing importance in drug discovery, this review paper aims to consolidate and examine the biological activities of sultam derivatives ranging from 4 to 8-membered ring structures.
Collapse
Affiliation(s)
- Yie Kie Chong
- School of Science, Monash University Malaysia Campus Jalan Lagoon Selatan 47500 Bandar Sunway Selangor Malaysia
| | - Yee Swen Ong
- School of Science, Monash University Malaysia Campus Jalan Lagoon Selatan 47500 Bandar Sunway Selangor Malaysia
| | - Keng Yoon Yeong
- School of Science, Monash University Malaysia Campus Jalan Lagoon Selatan 47500 Bandar Sunway Selangor Malaysia
| |
Collapse
|
24
|
Ji W, Xie X, Bai G, He Y, Li L, Zhang L, Qiang D. Metabolomic approaches to dissect dysregulated metabolism in the progression of pre-diabetes to T2DM. Mol Omics 2024; 20:333-347. [PMID: 38686662 DOI: 10.1039/d3mo00130j] [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: 05/02/2024]
Abstract
Many individuals with pre-diabetes eventually develop diabetes. Therefore, profiling of prediabetic metabolic disorders may be an effective targeted preventive measure. We aimed to elucidate the metabolic mechanism of progression of pre-diabetes to type 2 diabetes mellitus (T2DM) from a metabolic perspective. Four sets of plasma samples (20 subjects per group) collected according to fasting blood glucose (FBG) concentration were subjected to metabolomic analysis. An integrative approach of metabolome and WGCNA was employed to explore candidate metabolites. Compared with the healthy group (FBG < 5.6 mmol L-1), 113 metabolites were differentially expressed in the early stage of pre-diabetes (5.6 mmol L-1 ⩽ FBG < 6.1 mmol L-1), 237 in the late stage of pre-diabetes (6.1 mmol L-1 ⩽ FBG < 7.0 mmol L-1), and 245 in the T2DM group (FBG ⩾ 7.0 mmol L-1). A total of 27 differentially expressed metabolites (DEMs) were shared in all comparisons. Among them, L-norleucine was downregulated, whereas ethionamide, oxidized glutathione, 5-methylcytosine, and alpha-D-glucopyranoside beta-D-fructofuranosyl were increased with the rising levels of FBG. Surprisingly, 15 (11 lyso-phosphatidylcholines, L-norleucine, oxidized glutathione, arachidonic acid, and 5-oxoproline) of the 27 DEMs were ferroptosis-associated metabolites. WGCNA clustered all metabolites into 8 modules and the pathway enrichment analysis of DEMs showed a significant annotation to the insulin resistance-related pathway. Integrated analysis of DEMs, ROC and WGCNA modules determined 12 potential biomarkers for pre-diabetes and T2DM, including L-norleucine, 8 of which were L-arginine or its metabolites. L-Norleucine and L-arginine could serve as biomarkers for pre-diabetes. The inventory of metabolites provided by our plasma metabolome offers insights into T2DM physiology metabolism.
Collapse
Affiliation(s)
- Wenrui Ji
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Xiaomin Xie
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Guirong Bai
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Yanting He
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Ling Li
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Li Zhang
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Dan Qiang
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| |
Collapse
|
25
|
Sun Z, Lu K, He Q, Tang Y, Li H, Pazo EE, Hu L, Wei R. INOS ablation promotes corneal wound healing via activation of Akt signaling. Exp Eye Res 2024; 243:109886. [PMID: 38583755 DOI: 10.1016/j.exer.2024.109886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/24/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Corneal injury leads to impaired normal structure of the cornea. Improving the wound healing process in epithelial cells significantly contributes to ocular damage treatments. Here, we aimed to investigate the potential mechanisms of nitric oxide (NO) and its mediator, inducible nitric oxide synthase (iNOS), in the process of corneal wound healing. We established a corneal injury model of iNOS-/- mice, and treated human corneal epithelial cell lines (HCE-2) with the iNOS inhibitor L-INL, with or without NO replenishment by supplying sodium nitroferricyanide dihydrate (SNP). Our findings showed that inhibition of NO/iNOS accelerated corneal repair, enhanced uPAR (a receptor protein indicating the migration ability), and improved epithelial cell migration. Furthermore, NO/iNOS ablation activated Akt phosphorylation, reduced neutrophil marker protein MPO expression, and downregulated the transcription of inflammation cytokines CXCL-1, CXCL-2, IL-1β, IL-6, and TNF-α. However, the protective effects of NO/iNOS inhibition are significantly reduced by NO replenishment when treated with SNP. Therefore, we confirmed that inhibiting NO/iNOS improved the corneal wound healing by facilitating epithelial cell migration and reducing inflammatory reactions, which might be related to the activation of the Akt signaling pathway.
Collapse
Affiliation(s)
- Ziwen Sun
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300070, Tianjin, China
| | - Kunpeng Lu
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, 300070, Tianjin, China
| | - Qing He
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300070, Tianjin, China
| | - Yang Tang
- Qingdao State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, 266071, Qingdao, China
| | - Haoru Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300070, Tianjin, China
| | - Emmanuel Eric Pazo
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300070, Tianjin, China
| | - Lizhi Hu
- Basic Medical College, Immunology Department, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, 300070, China.
| | - Ruihua Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300070, Tianjin, China.
| |
Collapse
|
26
|
Huang Y, Chen Y, Xie H, Feng Y, Chen S, Bao B. Effects of Inducible Nitric Oxide Synthase (iNOS) Gene Knockout on the Diversity, Composition, and Function of Gut Microbiota in Adult Zebrafish. BIOLOGY 2024; 13:372. [PMID: 38927252 PMCID: PMC11201229 DOI: 10.3390/biology13060372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024]
Abstract
The gut microbiota constitutes a complex ecosystem that has an important impact on host health. In this study, genetically engineered zebrafish with inducible nitric oxide synthase (iNOS or NOS2) knockout were used as a model to investigate the effects of nos2a/nos2b gene single knockout and nos2 gene double knockout on intestinal microbiome composition and function. Extensive 16S rRNA sequencing revealed substantial changes in microbial diversity and specific taxonomic abundances, yet it did not affect the functional structure of the intestinal tissues. Notably, iNOS-deficient zebrafish demonstrated a decrease in Vibrio species and an increase in Aeromonas species, with more pronounced effects observed in double knockouts. Further transcriptomic analysis of the gut in double iNOS knockout zebrafish indicated significant alterations in immune-related and metabolic pathways, including the complement and PPAR signaling pathways. These findings underscore the crucial interplay between host genetics and gut microbiota, indicating that iNOS plays a key role in modulating the gut microbial ecology, host immune system, and metabolic responses.
Collapse
Affiliation(s)
- Yajuan Huang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; (Y.H.); (Y.C.); (H.X.); (Y.F.)
| | - Yadong Chen
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; (Y.H.); (Y.C.); (H.X.); (Y.F.)
- National Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Haisheng Xie
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; (Y.H.); (Y.C.); (H.X.); (Y.F.)
| | - Yidong Feng
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; (Y.H.); (Y.C.); (H.X.); (Y.F.)
| | - Songlin Chen
- National Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Baolong Bao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; (Y.H.); (Y.C.); (H.X.); (Y.F.)
| |
Collapse
|
27
|
Jiao ZW, Liu HF, Lin KQ, Xie GT, Lou HY, Pan WD, Zhang MS. Synthesis and in vitro Anti-Inflammatory Activity of Novel Dendrobine Amide/Sulfonamide Derivatives. Chem Biodivers 2024; 21:e202400030. [PMID: 38511964 DOI: 10.1002/cbdv.202400030] [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: 01/06/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 03/22/2024]
Abstract
A traditional Chinese medicine ingredient, dendrobine, has been demonstrated to have anti-inflammatory properties. However, due to its poor anti-inflammatory properties, its clinical use is limited. Consequently, we have designed and synthesized 32 new amide/sulfonamide dendrobine derivatives and screened their anti-inflammatory activities in vitro. Experiments showed that nitric oxide (NO) generation in lipopolysaccharide (LPS)-induced RAW264.7 cells was strongly reduced by derivative 14, with an IC50 of 2.96 μM. Western blot research revealed that 14 decreased the concentration-dependent expression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (INOS). Molecular docking was used to predict the binding of the inflammation-associated proteins COX-2 and INOS to compound 14.
Collapse
Affiliation(s)
- Zi-Wei Jiao
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, PR China
| | - Han-Fei Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Kai-Qin Lin
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Guang-Tong Xie
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Hua-Yong Lou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Wei-Dong Pan
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, PR China
| | - Mao-Sheng Zhang
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, PR China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China
| |
Collapse
|
28
|
Kamenshchyk A, Belenichev I, Oksenych V, Kamyshnyi O. Combined Pharmacological Modulation of Translational and Transcriptional Activity Signaling Pathways as a Promising Therapeutic Approach in Children with Myocardial Changes. Biomolecules 2024; 14:477. [PMID: 38672493 PMCID: PMC11047929 DOI: 10.3390/biom14040477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/29/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Myocardial hypertrophy is the most common condition that accompanies heart development in children. Transcriptional gene expression regulating pathways play a critical role both in cardiac embryogenesis and in the pathogenesis of congenital hypertrophic cardiomyopathy, neonatal posthypoxic myocardial hypertrophy, and congenital heart diseases. This paper describes the state of cardiac gene expression and potential pharmacological modulators at different transcriptional levels. An experimental model of perinatal cardiac hypoxia showed the downregulated expression of genes responsible for cardiac muscle integrity and overexpressed genes associated with energy metabolism and apoptosis, which may provide a basis for a therapeutic approach. Current evidence suggests that RNA drugs, theaflavin, neuraminidase, proton pumps, and histone deacetylase inhibitors are promising pharmacological agents in progressive cardiac hypertrophy. The different points of application of the above drugs make combined use possible, potentiating the effects of inhibition in specific signaling pathways. The special role of N-acetyl cysteine in both the inhibition of several signaling pathways and the reduction of oxidative stress was emphasized.
Collapse
Affiliation(s)
- Andrii Kamenshchyk
- Department of Hospital Pediatrics, Zaporizhzhya State Medical and Pharmaceutical University, 69035 Zaporizhzhya, Ukraine
| | - Igor Belenichev
- Department of Pharmacology, Zaporizhzhya State Medical and Pharmaceutical University, 69035 Zaporizhzhya, Ukraine;
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology and Immunology, I. Horbachevsky Ternopil State Medical University, 46001 Ternopil, Ukraine;
| |
Collapse
|
29
|
Choi S, Han S, Lee S, Kim J, Kim J, Kang DK. Synergistic Antioxidant and Anti-Inflammatory Effects of Phenolic Acid-Conjugated Glutamine-Histidine-Glycine-Valine (QHGV) Peptides Derived from Oysters ( Crassostrea talienwhanensis). Antioxidants (Basel) 2024; 13:447. [PMID: 38671896 PMCID: PMC11047712 DOI: 10.3390/antiox13040447] [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: 03/05/2024] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The glutamine-histidine-glycine-valine (QHGV), a peptide derived from oysters, exhibits antioxidant activity and is being actively researched as a potential pharmaceutical and functional cosmetic ingredient. In this study, we synthesized the QHGV peptide and explored the hitherto unknown anti-inflammatory effects of QHGV. The antioxidant property was also characterized by conjugating with various naturally derived phenolic acids, such as caffeic, gallic, ferulic, sinapinic, and vanillic acids. Conjugation with phenolic acids not only enhanced the antioxidant activity of QHGV but also diminished the lipopolysaccharide-induced generation of reactive oxygen species (ROS) in the murine macrophage cell line, RAW 264.7. The reduction in the levels of reactive oxygen species led to the reduced mRNA expression of inducible nitric oxide synthase (iNos) and cyclooxygenase 2 (Cox-2), resulting in an anti-inflammatory effect through the inhibition of the phosphorylation of mitogen-activated protein kinase, including extracellular signal-activated protein kinase, c-Jun NH2-terminal kinase, and p38. Furthermore, the phenolic acid-conjugated peptides increased the mRNA and protein levels of collagen type I, indicative of a wrinkle-improvement effect. The phenolic acid conjugates of the peptide were not cytotoxic to human keratinocytes such as HaCaT cells. These results suggest that phenolic acid conjugation can enhance the potential of peptides as drug and cosmetic resources.
Collapse
Affiliation(s)
- Soyun Choi
- Department of Chemistry, Incheon National University, Incheon 22012, Republic of Korea; (S.C.); (S.L.); (J.K.)
| | - Sohee Han
- WellPep Co., Ltd., Incheon 22012, Republic of Korea; (S.H.); (J.K.)
| | - Seungmi Lee
- Department of Chemistry, Incheon National University, Incheon 22012, Republic of Korea; (S.C.); (S.L.); (J.K.)
| | - Jongmin Kim
- WellPep Co., Ltd., Incheon 22012, Republic of Korea; (S.H.); (J.K.)
| | - Jinho Kim
- Department of Chemistry, Incheon National University, Incheon 22012, Republic of Korea; (S.C.); (S.L.); (J.K.)
| | - Dong-Ku Kang
- Department of Chemistry, Incheon National University, Incheon 22012, Republic of Korea; (S.C.); (S.L.); (J.K.)
- Bioplastic Research Center, Incheon National University, Incheon 22012, Republic of Korea
- Research Institute of Basic Sciences, Core Research Institute, Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
30
|
Zhang LS, Zhang ZS, Wu YZ, Guo B, Li J, Huang XQ, Zhang FM, Li MY, Yang PC, Zheng XB. Activation of free fatty acid receptors, FFAR1 and FFAR4, ameliorates ulcerative colitis by promote fatty acid metabolism and mediate macrophage polarization. Int Immunopharmacol 2024; 130:111778. [PMID: 38432147 DOI: 10.1016/j.intimp.2024.111778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE To investigate the mechanism of action of fatty acid receptors, FFAR1 and FFAR4, on ulcerative colitis (UC) through fatty acid metabolism and macrophage polarization. METHODS Dextran sulfate sodium (DSS)-induced mouse model of UC mice was used to evaluate the efficacy of FFAR1 (GW9508) and FFAR4 (GSK137647) agonists by analyzing body weight, colon length, disease activity index (DAI), and histological scores. Real-time PCR and immunofluorescence analysis were performed to quantify the levels of fatty acid metabolizing enzymes and macrophage makers. FFA-induced lipid accumulation in RAW264.7 cells was visualized by Oil Red O staining analysis, and cells were collected to detect macrophage polarization by flow cytometry. RESULTS The combination of GW9508 and GSK137647 significantly improved DSS-induced UC symptoms, caused recovery in colon length, and decreased histological injury. GW9508 + GSK137647 treatment upregulated the expressions of CD206, lipid oxidation enzyme (CPT-1α) and anti-inflammatory cytokines (IL-4, IL-10, IL-13) but downregulated those of CD86, lipogenic enzymes (ACC1, FASN, SCD1), and pro-inflammatory cytokines (IL-1β, IL-6, TNF-α). Combining the two agonists decreased FFA-induced lipid accumulation and increased CD206 expression in cell-based experiments. CONCLUSION Activated FFAR1 and FFAR4 ameliorates DSS-induced UC by promoting fatty acid metabolism to reduce lipid accumulation and mediate M2 macrophage polarization.
Collapse
Affiliation(s)
- Lin-Sheng Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhi-Shou Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu-Zhu Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Botang Guo
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jing Li
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiao-Qi Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Feng-Min Zhang
- Dongguan Hospital of Traditional Chinese Medicine, Dongguan, China
| | - Min-Yao Li
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China; Institute of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China.
| | - Ping-Chang Yang
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China; Institute of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China.
| | - Xue-Bao Zheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
31
|
Li RY, Guo L. Exercise in Diabetic Nephropathy: Protective Effects and Molecular Mechanism. Int J Mol Sci 2024; 25:3605. [PMID: 38612417 PMCID: PMC11012151 DOI: 10.3390/ijms25073605] [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: 02/15/2024] [Revised: 03/15/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Diabetic nephropathy (DN) is a serious complication of diabetes, and its progression is influenced by factors like oxidative stress, inflammation, cell death, and fibrosis. Compared to drug treatment, exercise offers a cost-effective and low-risk approach to slowing down DN progression. Through multiple ways and mechanisms, exercise helps to control blood sugar and blood pressure and reduce serum creatinine and albuminuria, thereby alleviating kidney damage. This review explores the beneficial effects of exercise on DN improvement and highlights its potential mechanisms for ameliorating DN. In-depth understanding of the role and mechanism of exercise in improving DN would pave the way for formulating safe and effective exercise programs for the treatment and prevention of DN.
Collapse
Affiliation(s)
- Ruo-Ying Li
- School of Exercise and Health, Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China;
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Liang Guo
- School of Exercise and Health, Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China;
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
32
|
Zhou B, Sh G, Xie D, Zhao X, Hao B, Liu D, Wang M, Wu L, Lin L, Qian X. Ginsenoside Rb1 prevents age-related endothelial senescence by modulating SIRT1/caveolin-1/enos signaling pathway. Heliyon 2024; 10:e24586. [PMID: 38322899 PMCID: PMC10844051 DOI: 10.1016/j.heliyon.2024.e24586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/08/2024] Open
Abstract
Background Advancing age is one of the independent risk factors for cardiovascular disorders. The Compendium of Materia Medica, a classic book on traditional Chinese medicine, states that ginseng "harmonizes the five internal organs, calming the spirit and prolonging the years of life." Considered one of the primary bioactive compounds derived from Panax ginseng, ginsenoside Rb1 (g-Rb1) has been scientifically suggested to possess anti-senescence efficacy. More research is needed to explore the vascular pharmacological activity and potential clinical application value of g-Rb1. Aims of the study Our previous study demonstrated that g-Rb1 could mitigate cellular senescence via the SIRT1/eNOS pathway. This study was performed to explore the exact mechanisms by which g-Rb1 modulates the SIRT1/eNOS pathway. Materials and methods We used human primary umbilical vein endothelial cells (HUVECs) to establish a replicative ageing model. Real-time (RT‒PCR), western blotting, small interfering RNA (siRNA), and immunoprecipitation were conducted to detect the effect of g-Rb1 on the SIRT1/caveolin-1/eNOS axis. Results G-Rb1 increased NO production and alleviated replicative senescence of HUVECs. The application of g-Rb1 elevated the mRNA and protein abundance of both SIRT1 and eNOS while concomitantly suppressing the expression of caveolin-1. Inhibition of SIRT1 and eNOS by siRNAs suppressed the anti-senescence function of g-Rb1, while caveolin-1 siRNA could enhance it. G-Rb1 decreased the acetylation level of caveolin-1 and increased NO production, which was suppressed by SIRT1 siRNA. Both g-Rb1 and caveolin-1 siRNA could reduce the acetylation level of eNOS and increase NO production. Conclusion G-Rb1 prevents age-related endothelial senescence by modulating the SIRT1/caveolin-1/eNOS signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | - Baoshun Hao
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dinhui Liu
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Min Wang
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lin Wu
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liangying Lin
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoxian Qian
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
33
|
Qin Q, Chen M, Yu N, Yao K, Liu X, Zhang Q, Wang Y, Ji J, Wang K, Jia F. Macromolecular carrier with long retention and body-temperature triggered nitric oxide release for corneal alkali burn therapy via leptin-related signaling. NANO TODAY 2024; 54:102108. [DOI: 10.1016/j.nantod.2023.102108] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
|
34
|
Zhang Y, Guo Z, Wang J, Yue Y, Yang Y, Wen Y, Luo Y, Zhang X. Qinlian hongqu decoction ameliorates hyperlipidemia via the IRE1-α/IKKB-β/NF-κb signaling pathway: Network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116856. [PMID: 37406747 DOI: 10.1016/j.jep.2023.116856] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/18/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qinlian Hongqu decoction (QLHQD) is a traditional Chinese medicine (TCM) formula. It has previously been found to mitigate hyperlipidemia, although its mechanism requires further clarification. AIM OF THE STUDY This study explored QLHQD's mechanism in treating hyperlipidemia based on network pharmacology and experimental validation. MATERIALS AND METHODS The components of QLHQD were analyzed by means of ultrahigh performanceliquid chromatography-quadrupole/orbitrapmass spectrometry (UHPLC-Q-Orbitrap-HRMS) and the targets of hyperlipidemia were predicted using the Swiss ADME, GeneCards, OMIM, DrugBank, TTD, and PharmGKB databases. A drug-component-target-disease network was constructed using Cytoscape v3.7.1. Moreover, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses were performed using the Bioinformatics platform. Based on the KEGG results, the non-alcoholic fatty liver disease signaling pathways were selected for experimental validation in an animal model. RESULTS We identified 34 components of QLHQD, 94 targets of hyperlipidemia, and 18 lipid metabolism-related pathways from the KEGG analysis. The results of the animal experiment revealed that QLHQD alleviated lipid metabolism disorders, obesity, insulin resistance, and inflammation in rats with hyperlipidemia induced by high-fat diets. Additionally, it reduced the expression of IRE1-α, TRAF2, IKKB-β, and NF-κB proteins in the liver of hyperlipidemic rats. CONCLUSION QLHQD is able to significantly mitigate hyperlipidemia induced via high-fat diets in rats. The mechanism of action in this regard might involve regulating the IRE1-α/IKKB-β/NF-κB signaling pathway in the liver, thereby attenuating inflammatory responses and insulin resistance.
Collapse
Affiliation(s)
- Yong Zhang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Zhiqing Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Jin Wang
- College of Computer Science, Chengdu University, Chengdu, Sichuan Province, China
| | - Yuanyuan Yue
- Department of Ultrasound, Chengdu First People's Hospital, Chengdu, Sichuan Province, China
| | - Yang Yang
- Institute of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan Province, China
| | - Yueqiang Wen
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yaqi Luo
- Institute of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan Province, China.
| | - Xiaobo Zhang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China.
| |
Collapse
|
35
|
Wang J, Qiu F, Zhang Z, Liu Y, Zhou Q, Dai S, Xiang S, Wei C. Clostridium butyricum Alleviates DEHP Plasticizer-Induced Learning and Memory Impairment in Mice via Gut-Brain Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:18524-18537. [PMID: 37963287 DOI: 10.1021/acs.jafc.3c03533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) plasticizer, a well-known environmental and food pollutant, has neurotoxicity. However, it is unknown whether DEHP leads to learning and memory impairment through gut-brain axis and whether Clostridium butyricum can alleviate this impairment. Here, C57BL/6 mice were exposed to DEHP and treated with C. butyricum. Learning and memory abilities were evaluated through the Morris water maze. The levels of synaptic proteins, inflammatory cytokines, and 5-hydroxytryptamine (5-HT) were detected by immunohistochemistry or ELISA. Gut microbiota were analyzed through 16S rRNA sequencing. C. butyricum alleviated DEHP-induced learning and memory impairment and restored synaptic proteins. It significantly relieved DEHP-induced inflammation and recovered 5-HT levels. C. butyricum recovered the richness of the gut microbiota decreased by DEHP, with the Bifidobacterium genus increasing the most. Overall, C. butyricum alleviated DEHP-induced learning and memory impairment due to reduced inflammation and increased 5-HT secretion, which was partly attributed to the recovery of gut microbiota.
Collapse
Affiliation(s)
- Jin Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Feng Qiu
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Zilong Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Yu Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Qian Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Siyu Dai
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Shuanglin Xiang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Chenxi Wei
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| |
Collapse
|
36
|
Li Z, Zhang W, Wang QR, Yang YJ, Liu XH, Cheng G, Chang FJ. Effect of Thrombolysis on Circulating Microparticles in Patients with ST-Segment Elevation Myocardial Infarction. Cardiovasc Ther 2023; 2023:5559368. [PMID: 38024103 PMCID: PMC10676276 DOI: 10.1155/2023/5559368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/11/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Objective We demonstrated that circulating microparticles (MPs) are increased in patients with coronary heart disease (both chronic coronary syndrome (CCS) and acute coronary syndrome). Whether thrombolysis affects MPs in patients with ST-segment elevation myocardial infarction (STEMI) with or without percutaneous coronary intervention (PCI) is unknown. Methods This study was divided into three groups: STEMI patients with thrombolysis (n = 18) were group T, patients with chronic coronary syndrome (n = 20) were group CCS, and healthy volunteers (n = 20) were the control group. Fasting venous blood was extracted from patients in the CCS and control groups, and venous blood was extracted from patients in the T group before (pre-T) and 2 hours after (post-T) thrombolysis. MPs from each group were obtained by centrifugation. After determining the concentration, the effects of MPs on endothelial nitric oxide synthase (eNOS) and inducible nitric oxide synthase (iNOS) in rat myocardial tissue in vitro were detected by immunohistochemistry and western blotting. Changes in nitric oxide (NO) and oxygen free radicals (O2•-) were also detected. The effect of MPs on vasodilation in isolated rat thoracic aortae was detected. Results Compared with that in the control group (2.60 ± 0.38 mg/ml), the concentration of MPs was increased in patients with CCS (3.49 ± 0.72 mg/ml) and in STEMI patients before thrombolysis (4.17 ± 0.58 mg/ml). However, thrombolysis did not further increase MP levels (post-T, 4.23 ± 1.01 mg/ml) compared with those in STEMI patients before thrombolysis. Compared with those in the control group, MPs in both CCS and STEMI patients before thrombolysis inhibited the expression of eNOS (both immunohistochemistry and western blot analysis of phosphorylation at Ser1177), NO production in the isolated myocardium and vasodilation in vitro and stimulated the expression of iNOS (immunohistochemistry and western blot analysis of phosphorylation at Thr495), and the generation of O2•- in the isolated myocardium. The effects of MPs were further enhanced by MPs from STEMI patients 2 hours after thrombolysis. Conclusion Changes in MP function after thrombolysis may be one of the mechanisms leading to ischemia-reperfusion after thrombolysis.
Collapse
Affiliation(s)
- Zhe Li
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Wei Zhang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Qun-Rang Wang
- Department of Cardiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xian'yang, China
| | - Yu-juan Yang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xin-Hong Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Gong Cheng
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Feng-Jun Chang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
37
|
Li C, Xu J, Abdurehim A, Sun Q, Xie J, Zhang Y. TRPA1: A promising target for pulmonary fibrosis? Eur J Pharmacol 2023; 959:176088. [PMID: 37777106 DOI: 10.1016/j.ejphar.2023.176088] [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: 07/02/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023]
Abstract
Pulmonary fibrosis is a disease characterized by progressive scar formation and the ultimate manifestation of numerous lung diseases. It is known as "cancer that is not cancer" and has attracted widespread attention. However, its formation process is very complex, and the mechanism of occurrence has not been fully elucidated. Current research has found that TRPA1 may be a promising target in the pathogenesis of pulmonary fibrosis. The TRPA1 channel was first successfully isolated in human lung fibroblasts, and it was found to have a relatively concentrated distribution in the lungs and respiratory tract. It is also involved in various acute and chronic inflammatory processes of lung diseases and may even play a core role in the progression and/or prevention of pulmonary fibrosis. Natural ligands targeting TRPA1 could offer a promising alternative treatment for pulmonary diseases. Therefore, this review delves into the current understanding of pulmonary fibrogenesis, analyzes TRPA1 biological properties and regulation of lung disease with a focus on pulmonary fibrosis, summarizes the TRPA1 molecular structure and its biological function, and summarizes TRPA1 natural ligand sources, anti-pulmonary fibrosis activity and potential mechanisms. The aim is to decipher the exact role of TRPA1 channels in the pathophysiology of pulmonary fibrosis and to consider their potential in the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Chao Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Jiawen Xu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Aliya Abdurehim
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Qing Sun
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Junbo Xie
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yanqing Zhang
- Biotechnology & Food Science College, Tianjin University of Commerce, Tianjin, 300134, China.
| |
Collapse
|
38
|
Wang W, Wang SK, Wang Q, Zhang Z, Li B, Zhou ZD, Zhang JF, Lin C, Chen TX, Jin Z, Tang YZ. Diclofenac and eugenol hybrid with enhanced anti-inflammatory activity through activating HO-1 and inhibiting NF-κB pathway in vitro and in vivo. Eur J Med Chem 2023; 259:115669. [PMID: 37517204 DOI: 10.1016/j.ejmech.2023.115669] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/18/2023] [Accepted: 07/18/2023] [Indexed: 08/01/2023]
Abstract
A series of diclofenac hybrid molecules were synthesized and evaluated for their NO-inhibitory ability in LPS-induced RAW 264.7 macrophage cells. Among them, compound 1 showed the highest NO-inhibitory ability (approximately 66%) and no significant cytotoxicity. Compound 1 exhibited superior NF-κB-inhibitory ability compared to diclofenac through the activation of Nrf2/HO-1 signaling pathway in RAW 264.7. 20 mg/kg compound 1 resulted in remarkable colitis improvement in dextran sulfate sodium (DSS)-induced mice model by up-regulating HO-1 and down-regulating phosphorylation level of NF-κB p65. Moreover, 50 mg/kg dose of compound 1 showed a lower ulcerogenic potential compared to diclofenac in rats. The diclofenac-eugenol hybrid (compound 1) may serve as a novel anti-inflammatory agent based on its role in inhibiting the NF-κB signaling pathway and activating HO-1 expression with no toxicity in vitro and in vivo.
Collapse
Affiliation(s)
- Wei Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Shou-Kai Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Qi Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Zhe Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Bo Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Zi-Dan Zhou
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Jian-Feng Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Chao Lin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Ting-Xiao Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Zhen Jin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - You-Zhi Tang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China.
| |
Collapse
|
39
|
Xie W, Xing N, Qu J, Liu D, Pang Q. The Physiological Function of nNOS-Associated CAPON Proteins and the Roles of CAPON in Diseases. Int J Mol Sci 2023; 24:15808. [PMID: 37958792 PMCID: PMC10647562 DOI: 10.3390/ijms242115808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
In this review, the structure, isoform, and physiological role of the carboxy-terminal PDZ ligand of neuronal nitric oxide synthase (CAPON) are summarized. There are three isoforms of CAPON in humans, including long CAPON protein (CAPON-L), short CAPON protein (CAPON-S), and CAPON-S' protein. CAPON-L includes three functional regions: a C-terminal PDZ-binding motif, carboxypeptidase (CPE)-binding region, and N-terminal phosphotyrosine (PTB) structural domain. Both CAPON-S and CAPON-S' only contain the C-terminal PDZ-binding motif. The C-terminal PDZ-binding motif of CAPON can bind with neuronal nitric oxide synthase (nNOS) and participates in regulating NO production and neuronal development. An overview is given on the relationship between CAPON and heart diseases, diabetes, psychiatric disorders, and tumors. This review will clarify future research directions on the signal pathways related to CAPON, which will be helpful for studying the regulatory mechanism of CAPON. CAPON may be used as a drug target, which will provide new ideas and solutions for treating human diseases.
Collapse
Affiliation(s)
| | | | | | - Dongwu Liu
- Anti-Aging & Regenerative Medicine Research Institution, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China; (W.X.); (N.X.)
| | - Qiuxiang Pang
- Anti-Aging & Regenerative Medicine Research Institution, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China; (W.X.); (N.X.)
| |
Collapse
|
40
|
Harvest CK, Abele TJ, Yu C, Beatty CJ, Amason ME, Billman ZP, DePrizio MA, Souza FW, Lacey CA, Maltez VI, Larson HN, McGlaughon BD, Saban DR, Montgomery SA, Miao EA. An innate granuloma eradicates an environmental pathogen using Gsdmd and Nos2. Nat Commun 2023; 14:6686. [PMID: 37865673 PMCID: PMC10590453 DOI: 10.1038/s41467-023-42218-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/21/2023] [Indexed: 10/23/2023] Open
Abstract
Granulomas often form around pathogens that cause chronic infections. Here, we discover an innate granuloma model in mice with an environmental bacterium called Chromobacterium violaceum. Granuloma formation not only successfully walls off, but also clears, the infection. The infected lesion can arise from a single bacterium that replicates despite the presence of a neutrophil swarm. Bacterial replication ceases when macrophages organize around the infection and form a granuloma. This granuloma response is accomplished independently of adaptive immunity that is typically required to organize granulomas. The C. violaceum-induced granuloma requires at least two separate defense pathways, gasdermin D and iNOS, to maintain the integrity of the granuloma architecture. This innate granuloma successfully eradicates C. violaceum infection. Therefore, this C. violaceum-induced granuloma model demonstrates that innate immune cells successfully organize a granuloma and thereby resolve infection by an environmental pathogen.
Collapse
Affiliation(s)
- Carissa K Harvest
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Taylor J Abele
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Chen Yu
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Cole J Beatty
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Megan E Amason
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zachary P Billman
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Morgan A DePrizio
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Fernando W Souza
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Carolyn A Lacey
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Vivien I Maltez
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Heather N Larson
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Benjamin D McGlaughon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel R Saban
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Stephanie A Montgomery
- Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Edward A Miao
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA.
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA.
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
41
|
Ferreira LO, Vasconcelos VW, Lima JDS, Vieira Neto JR, da Costa GE, Esteves JDC, de Sousa SC, Moura JA, Santos FRS, Leitão Filho JM, Protásio MR, Araújo PS, Lemos CJDS, Resende KD, Lopes DCF. Biochemical Changes in Cardiopulmonary Bypass in Cardiac Surgery: New Insights. J Pers Med 2023; 13:1506. [PMID: 37888117 PMCID: PMC10608001 DOI: 10.3390/jpm13101506] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 10/28/2023] Open
Abstract
Patients undergoing coronary revascularization with extracorporeal circulation or cardiopulmonary bypass (CPB) may develop several biochemical changes in the microcirculation that lead to a systemic inflammatory response. Surgical incision, post-CPB reperfusion injury and blood contact with non-endothelial membranes can activate inflammatory signaling pathways that lead to the production and activation of inflammatory cells, with cytokine production and oxidative stress. This inflammatory storm can cause damage to vital organs, especially the heart, and thus lead to complications in the postoperative period. In addition to the organic pathophysiology during and after the period of exposure to extracorporeal circulation, this review addresses new perspectives for intraoperative treatment and management that may lead to a reduction in this inflammatory storm and thereby improve the prognosis and possibly reduce the mortality of these patients.
Collapse
Affiliation(s)
- Luan Oliveira Ferreira
- Residency Program in Anesthesiology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (V.W.V.); (J.d.S.L.); (J.R.V.N.); (G.E.d.C.); (J.d.C.E.); (S.C.d.S.); (J.A.M.); (F.R.S.S.); (J.M.L.F.); (K.D.R.)
- Laboratory of Experimental Neuropathology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil
| | - Victoria Winkler Vasconcelos
- Residency Program in Anesthesiology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (V.W.V.); (J.d.S.L.); (J.R.V.N.); (G.E.d.C.); (J.d.C.E.); (S.C.d.S.); (J.A.M.); (F.R.S.S.); (J.M.L.F.); (K.D.R.)
| | - Janielle de Sousa Lima
- Residency Program in Anesthesiology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (V.W.V.); (J.d.S.L.); (J.R.V.N.); (G.E.d.C.); (J.d.C.E.); (S.C.d.S.); (J.A.M.); (F.R.S.S.); (J.M.L.F.); (K.D.R.)
| | - Jaime Rodrigues Vieira Neto
- Residency Program in Anesthesiology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (V.W.V.); (J.d.S.L.); (J.R.V.N.); (G.E.d.C.); (J.d.C.E.); (S.C.d.S.); (J.A.M.); (F.R.S.S.); (J.M.L.F.); (K.D.R.)
| | - Giovana Escribano da Costa
- Residency Program in Anesthesiology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (V.W.V.); (J.d.S.L.); (J.R.V.N.); (G.E.d.C.); (J.d.C.E.); (S.C.d.S.); (J.A.M.); (F.R.S.S.); (J.M.L.F.); (K.D.R.)
| | - Jordana de Castro Esteves
- Residency Program in Anesthesiology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (V.W.V.); (J.d.S.L.); (J.R.V.N.); (G.E.d.C.); (J.d.C.E.); (S.C.d.S.); (J.A.M.); (F.R.S.S.); (J.M.L.F.); (K.D.R.)
| | - Sallatiel Cabral de Sousa
- Residency Program in Anesthesiology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (V.W.V.); (J.d.S.L.); (J.R.V.N.); (G.E.d.C.); (J.d.C.E.); (S.C.d.S.); (J.A.M.); (F.R.S.S.); (J.M.L.F.); (K.D.R.)
| | - Jonathan Almeida Moura
- Residency Program in Anesthesiology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (V.W.V.); (J.d.S.L.); (J.R.V.N.); (G.E.d.C.); (J.d.C.E.); (S.C.d.S.); (J.A.M.); (F.R.S.S.); (J.M.L.F.); (K.D.R.)
| | - Felipe Ruda Silva Santos
- Residency Program in Anesthesiology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (V.W.V.); (J.d.S.L.); (J.R.V.N.); (G.E.d.C.); (J.d.C.E.); (S.C.d.S.); (J.A.M.); (F.R.S.S.); (J.M.L.F.); (K.D.R.)
| | - João Monteiro Leitão Filho
- Residency Program in Anesthesiology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (V.W.V.); (J.d.S.L.); (J.R.V.N.); (G.E.d.C.); (J.d.C.E.); (S.C.d.S.); (J.A.M.); (F.R.S.S.); (J.M.L.F.); (K.D.R.)
| | | | - Pollyana Sousa Araújo
- Department of Cardiovascular Anesthesiology, Hospital Clínicas Gaspar Vianna, Belém 66083-106, Brazil; (P.S.A.); (C.J.d.S.L.)
| | - Cláudio José da Silva Lemos
- Department of Cardiovascular Anesthesiology, Hospital Clínicas Gaspar Vianna, Belém 66083-106, Brazil; (P.S.A.); (C.J.d.S.L.)
| | - Karina Dias Resende
- Residency Program in Anesthesiology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (V.W.V.); (J.d.S.L.); (J.R.V.N.); (G.E.d.C.); (J.d.C.E.); (S.C.d.S.); (J.A.M.); (F.R.S.S.); (J.M.L.F.); (K.D.R.)
| | - Dielly Catrina Favacho Lopes
- Laboratory of Experimental Neuropathology, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil
| |
Collapse
|
42
|
Huang J, Xie M, He L, Song X, Cao T. Chlorogenic acid: a review on its mechanisms of anti-inflammation, disease treatment, and related delivery systems. Front Pharmacol 2023; 14:1218015. [PMID: 37781708 PMCID: PMC10534970 DOI: 10.3389/fphar.2023.1218015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023] Open
Abstract
Chlorogenic acid is a bioactive compound ubiquitously present in the natural realm, lauded for its salient anti-inflammatory and antioxidant attributes. It executes its anti-inflammatory function by moderating the synthesis and secretion of inflammatory mediators, namely, TNF-α, IL-1β, IL-6, IL-8, NO, and PGE2. Concurrently, it modulates key signaling pathways and associated factors, including NF-κB, MAPK, Nrf2, and others, bestowing protection upon cells and tissues against afflictions such as cardio-cerebrovascular and diabetes mellitus. Nevertheless, the inherent low bioavailability of chlorogenic acid poses challenges in practical deployments. To surmount this limitation, sophisticated delivery systems, encompassing liposomes, micelles, and nanoparticles, have been devised, accentuating their stability, release mechanisms, and bioactivity. Given its innate anti-inflammatory prowess and safety profile, chlorogenic acid stands as a promising contender for advanced biomedical investigations and translational clinical endeavors.
Collapse
Affiliation(s)
- Jianhuan Huang
- Breast Surgery, Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
- The Graduate School, Guilin Medical University, Guilin, Guangxi, China
| | - Mingxiang Xie
- Breast Surgery, Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Liang He
- Department of Anesthesiology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoping Song
- Breast Surgery, Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tianze Cao
- Breast Surgery, Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
43
|
Koop MA, Sleijser-Koehorst MLS, Hooijmans CR, Tdlohreg PQ, Lutke Schipholt IJ, Scholten-Peeters GGM, Coppieters MW. The potential protective effects of pre-injury exercise on neuroimmune responses following experimentally-induced traumatic neuropathy: a systematic review with meta-analysis. Front Immunol 2023; 14:1215566. [PMID: 37767095 PMCID: PMC10520553 DOI: 10.3389/fimmu.2023.1215566] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Pre-clinical evidence shows that neuropathy is associated with complex neuroimmune responses, which in turn are associated with increased intensity and persistence of neuropathic pain. Routine exercise has the potential to mitigate complications of future nerve damage and persistence of pain through neuroimmune regulation. This systematic review aimed to explore the effect of pre-injury exercise on neuroimmune responses, and other physiological and behavioural reactions following peripheral neuropathy in animals. Three electronic databases were searched from inception to July 2022. All controlled animal studies assessing the influence of an active exercise program prior to experimentally-induced traumatic peripheral neuropathy compared to a non-exercise control group on neuroimmune, physiological and behavioural outcomes were selected. The search identified 17,431 records. After screening, 11 articles were included. Meta-analyses showed that pre-injury exercise significantly reduced levels of IL-1β (SMD: -1.06, 95% CI: -1.99 to -0.13, n=40), but not iNOS (SMD: -0.71 95% CI: -1.66 to 0.25, n=82). From 72 comparisons of different neuroimmune outcomes at different anatomical locations, vote counting revealed reductions in 23 pro-inflammatory and increases in 6 anti-inflammatory neuroimmune outcomes. For physiological outcomes, meta-analyses revealed that pre-injury exercise improved one out of six nerve morphometric related outcomes (G-ratio; SMD: 1.95, 95%CI: 0.77 to 3.12, n=20) and one out of two muscle morphometric outcomes (muscle fibre cross-sectional area; SMD: 0.91, 95%CI: 0.27 to 1.54, n=48). For behavioural outcomes, mechanical allodynia was significantly less in the pre-injury exercise group (SMD -1.24, 95%CI: -1.87 to -0.61) whereas no overall effect was seen for sciatic function index. Post hoc subgroup analysis suggests that timing of outcome measurement may influence the effect of pre-injury exercise on mechanical allodynia. Risk of bias was unclear in most studies, as the design and conduct of the included experiments were poorly reported. Preventative exercise may have potential neuroprotective and immunoregulatory effects limiting the sequalae of nerve injury, but more research in this field is urgently needed.
Collapse
Affiliation(s)
- Meghan A. Koop
- Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands
| | - Marije L. S. Sleijser-Koehorst
- Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands
| | - Carlijn R. Hooijmans
- Meta Research Team, Department of Anaesthesiology, Pain and Palliative Care, Radboud University Medical Center, Nijmegen, Netherlands
| | - Paul Q. Tdlohreg
- Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands
| | - Ivo J. Lutke Schipholt
- Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands
- Department of Clinical Chemistry, Laboratory Medical Immunology, Amsterdam University Medical Centre, Location VUmc, Amsterdam, Netherlands
| | | | - Michel W. Coppieters
- Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands
- Menzies Health Institute Queensland, Griffith University, Brisbane, QLD, Australia
| |
Collapse
|
44
|
Chang H, Yang F, Bai H, Lu Z, Xing C, Dai X, Wan W, Liao S, Cao H. Molybdenum and/or cadmium induce NLRP3 inflammasome production by causing mitochondria-associated endoplasmic reticulum membrane dysfunction in sheep hepatocytes. Chem Biol Interact 2023; 382:110617. [PMID: 37385403 DOI: 10.1016/j.cbi.2023.110617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/01/2023]
Abstract
Accumulation of the heavy metals molybdenum (Mo) and cadmium (Cd) in the liver can induce organelle damage and inflammation, resulting in hepatotoxicity. The effect of Mo and/or Cd on sheep hepatocytes was investigated by determining the relationship between the mitochondria-associated endoplasmic reticulum membrane (MAM) and NLRP3 inflammasome. Sheep hepatocytes were divided into four groups: the control group, Mo group (600 μM Mo), Cd group (4 μM Cd) and Mo + Cd group (600 μM Mo+4 μM Cd). The results showed that Mo and/or Cd exposure increased the levels of lactate dehydrogenase (LDH) and nitric oxide (NO) in the cell culture supernatant, elevated the levels of intracellular Ca2+ and mitochondrial Ca2+, downregulated the expression of MAM-related factors (IP3R, GRP75, VDAC1, PERK, ERO1-α, Mfn1, Mfn2, ERP44), shortened the length of the MAM and reduced the formation of the MAM structure, eventually causing MAM dysfunction. Moreover, the expression levels of NLRP3 inflammasome-related factors (NLRP3, Caspase1, IL-1β, IL-6, TNF-α) were also dramatically increased after Mo and Cd exposure, triggering NLRP3 inflammasome production. However, an IP3R inhibitor, 2-APB treatment significantly alleviated these changes. Overall, the data indicate that Mo and Cd coexposure leads to structural disruption and dysfunction of MAM, disrupts cellular Ca2+ homeostasis, and increases NLRP3 inflammasome production in sheep hepatocytes. However, the inhibition of IP3R alleviates NLRP3 inflammasome production induced by Mo and Cd.
Collapse
Affiliation(s)
- Huifeng Chang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, PR China
| | - Fan Yang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, PR China
| | - He Bai
- Medical Research Center, Mudanjiang Medical University, No. 3 Tongxiang street, Aimin District, Mudanjiang, 157011, Heilongjiang, PR China
| | - Zengting Lu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, PR China
| | - Chenghong Xing
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, PR China
| | - Xueyan Dai
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, PR China
| | - Wengen Wan
- Jiangxi Agricultural Technology Extension Center, Nanchang, 330096, Jiangxi, PR China
| | - Shuxian Liao
- Fengxin County Modern Agricultural Technology Service Center, Fengxin, PR China
| | - Huabin Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, PR China.
| |
Collapse
|
45
|
Wang X, Wang T, Lam E, Alvarez D, Sun Y. Ocular Vascular Diseases: From Retinal Immune Privilege to Inflammation. Int J Mol Sci 2023; 24:12090. [PMID: 37569464 PMCID: PMC10418793 DOI: 10.3390/ijms241512090] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The eye is an immune privileged tissue that insulates the visual system from local and systemic immune provocation to preserve homeostatic functions of highly specialized retinal neural cells. If immune privilege is breached, immune stimuli will invade the eye and subsequently trigger acute inflammatory responses. Local resident microglia become active and release numerous immunological factors to protect the integrity of retinal neural cells. Although acute inflammatory responses are necessary to control and eradicate insults to the eye, chronic inflammation can cause retinal tissue damage and cell dysfunction, leading to ocular disease and vision loss. In this review, we summarized features of immune privilege in the retina and the key inflammatory responses, factors, and intracellular pathways activated when retinal immune privilege fails, as well as a highlight of the recent clinical and research advances in ocular immunity and ocular vascular diseases including retinopathy of prematurity, age-related macular degeneration, and diabetic retinopathy.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - Tianxi Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - Enton Lam
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - David Alvarez
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| |
Collapse
|
46
|
Prasad K, Kulkarni A, K N, Gowda V, Shaikh MA. Serum Cystatin C Levels as a Predictor of Severity and Mortality Among Patients With COVID-19 Infection. Cureus 2023; 15:e42003. [PMID: 37593314 PMCID: PMC10428180 DOI: 10.7759/cureus.42003] [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] [Accepted: 07/16/2023] [Indexed: 08/19/2023] Open
Abstract
INTRODUCTION The pandemic caused by SARS Corona Virus-2 (COVID-19) has caused widespread mortality globally. The hallmark of the disease is the "cytokine storm," which is caused due to dysregulated immune system activation. Numerous inflammatory markers are used to predict the severity and mortality of the infection. Serum Cystatin C levels are associated with immune responses to exogenous and endogenous antigens. Our study was done to assess serum cystatin C as a marker of severity and mortality among patients admitted with COVID-19 infection. METHODOLOGY This cross-sectional study was conducted in a tertiary care center in South India. Sixty-nine patients with mild and severe COVID-19 infection admitted to the hospital were included in the study. Serum Cystatin C levels were estimated at admission. The levels were correlated with disease severity and mortality. Receiver operating characteristic curves (ROCs) was constructed for Cystatin C to predict severity and mortality. The computation of sensitivity, specificity, and positive and negative predictive values was done using optimal cut-off points. SPSS 18 was used for the statistical analysis. Version 18.0 of PASW Statistics for Windows. SPSS Inc., Chicago. RESULTS Out of 69 patients, 28 (40.5%) had a mild illness, and 41 patients (59.4%) had severe COVID-19 illness. Mean serum Cystatin C levels measured at the time of admission among patients with mild illness was 1.83 (SD-1.53), and among patients with severe illness was 3.84 (SD- 2.59) (p<0.001). The area under receiver operating characteristic curves (ROC) for serum cystatin C for predicting COVID-19 severity and mortality was 0.904 and 0.768, respectively (p<0.001). CONCLUSION Patients with severe COVID-19 disease had considerably higher serum levels of Cystatin C than those with mild COVID-19 illness. Cystatin C levels can be useful for predicting mortality and severity among patients admitted with COVID-19 infection.
Collapse
Affiliation(s)
- Kavya Prasad
- Internal Medicine, Ramaiah Medical College, Bengaluru, IND
| | | | - Navikala K
- Biochemistry, Ramaiah Medical College, Bengaluru, IND
| | - Vanitha Gowda
- Biochemistry, Ramaiah Medical College, Bengaluru, IND
| | | |
Collapse
|
47
|
Zhao X, Yue Y, Wang X, Zhang Q. Bioinformatics analysis of PLA2G7 as an immune-related biomarker in COPD by promoting expansion and suppressive functions of MDSCs. Int Immunopharmacol 2023; 120:110399. [PMID: 37270927 DOI: 10.1016/j.intimp.2023.110399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND Immune mechanism is involved in the pathogenesis of chronic obstructive pulmonary disease (COPD). However, the exact immune pathogenesis still remains unclear. This study aimed to identify the immune-related biomarkers in COPD through bioinformatics analysis and its potential molecular mechanism. METHODS GSE76925 was downloaded from Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were screened, and enrichment analysis was performed. Single sample gene enrichment analysis (ssGSEA) was conducted to score the infiltration levels of immune cells. Weighted gene co-expression network analysis (WGCNA) was applied to identify trait-related modules and to further determine the key module-related DEGs. Moreover, the correlations between the key genes and clinical parameters and infiltration levels of immune cells were analyzed. Furthermore, expression of the selected one key gene, PLA2G7, the frequency of MDSCs, and the expression of MDSCs-related immunosuppressive mediators were determined among healthy, smokers and COPD patients. Finally, effects of PLA2G7 abnormal expression on the frequency of MDSCs and the expression of MDSCs-related immunosuppressive mediators were examined. RESULTS A total of 352 DEGs were observed. These DEGs were mainly related to RNA metabolism and positive regulation of organelle organization. In addition, the black module was the most correlated with COPD. Six key genes (ADAMDEC1, CCL19, CHIT1, MMP9, PLA2G7, and TM4SF19) were identified between the black module and DEGs. Serum Lp-PLA2 and mRNA levels of PLA2G7, MDSCs, and MDSCs-related immunosuppressive mediators were found to be upregulated in COPD patients compared to the controls. The expression of PLA2G7 represented positive impact on the frequency of MDSCs and the expression of MDSCs-related immunosuppressive mediators. CONCLUSION PLA2G7 may serve as a potential immune-related biomarker contributing to the progression of COPD by promoting expansion and suppressive functions of MDSCs.
Collapse
Affiliation(s)
- Xiaoyu Zhao
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, China
| | - Yuanyi Yue
- Department of Gastroenterology, Shengjing Hospital of China Medical University, China
| | - Xueqing Wang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, China.
| | - Qiang Zhang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
48
|
Zhang L, Chen L, Qi M, Yu F, Ni X, Hong H, Xu H, Xu S. Glyphosate induces autophagy in hepatic L8824 cell line through NO-mediated activation of RAS/RAF/MEK/ERK signaling pathway and energy metabolism disorders. FISH & SHELLFISH IMMUNOLOGY 2023; 137:108772. [PMID: 37100311 DOI: 10.1016/j.fsi.2023.108772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/22/2023]
Abstract
Glyphosate is an herbicide commonly used worldwide, and its substantial use causes widespread pollution with runoff. However, research on glyphosate toxicity has mostly remained at the embryonic level and existing studies are limited. In the present study, we investigated whether glyphosate can induce autophagy in hepatic L8824 cells by regulating energy metabolism and rat sarcoma (RAS)/rapidly accelerated fibrosarcoma (RAF)/mitogen-activated extracellular signal-regulated kinase (MEK)/extracellular regulated protein kinases (ERK) signaling by activating nitric oxide (NO). First, we selected 0, 50, 200, and 500 μg/mL as the challenge doses, according to the inhibitory concentration of 50% (IC50) of glyphosate. The results showed that glyphosate exposure increased the enzyme activity of inducible nitric oxide synthase (iNOS), which in turn increased the NO content. The activity and expression of enzymes related to energy metabolism, such as hexokinase (HK)1, HK2, phosphofructokinase (PFK), phosphokinase (PK), succinate dehydrogenase (SDH), and nicotinamide adenine dinucleotide with hydrogen (NADH), were inhibited, and the RAS/RAF/MEK/ERK signaling pathway was activated. This led to the negative expression of mammalian target of rapamycin (mTOR) and P62 in hepatic L8824 cells and the activation of the autophagy marker genes microtubule-associated proteins light chain 3 (LC3) and Beclin1 to induce autophagy. The above results were dependent on glyphosate concentration. To verify whether autophagy can be excited by the RAS/RAF/MEK/ERK signaling pathway, we treated L8824 cells with the ERK inhibitor U0126 and found that the autophagy gene LC3 was reduced due to the inhibition of ERK, thus demonstrating the reliability of the results. In conclusion, our results demonstrate that glyphosate can induce autophagy in hepatic L8824 cells by activating NO, thus regulating energy metabolism and the RAS/RAF/MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Linlin Zhang
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Lu Chen
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Meng Qi
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Fuchang Yu
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Xiaotong Ni
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Haozheng Hong
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Haotian Xu
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China.
| | - Shiwen Xu
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China; Key Laboratory of Tarim Animal Husbandry Technology Corps, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China.
| |
Collapse
|
49
|
Wei QH, Cao XX, Xu DF, Wang ST, Zhang JS, Zhang H. Anti-inflammatory labdane diterpenoids from the aerial parts of Leonurus japonicus. PHYTOCHEMISTRY 2023; 210:113646. [PMID: 36958706 DOI: 10.1016/j.phytochem.2023.113646] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/16/2023] [Accepted: 03/19/2023] [Indexed: 06/18/2023]
Abstract
Twenty-two labdane-type diterpenoids, including ten pairs of 15-epimers and a pair of 13,15-epimers, were obtained from the aerial parts of a well-known medicinal plant Leonurus japonicus Houtt. While these epimers were separated by chiral HPLC, their structures were established mainly via spectroscopic methods especially NMR, X-ray crystallography and ECD techniques. Among them, seventeen compounds, encompassing three pairs of solvolysis artefacts likely due to the use of ethanol as extracting solvent, were reported for the first time in the current work. Our preliminary anti-inflammatory screening demonstrated that seven diterpenoids displayed noteworthy inhibitory effect on the NO production in LPS-induced RAW264.7 cells. In addition, the release of pro-inflammatory factors TNF-α, IL-1β and IL-6, as well as the expression of iNOS and COX-2 proteins, was also suppressed by the unreported 15,16-epoxy-6β-hydroxy-15α-methoxy-7,16-dioxolabd-8,13-diene. Further investigation into the preliminary anti-inflammatory mechanism of this compound indicated that it could block the activation of NF-κB signaling pathway.
Collapse
Affiliation(s)
- Quan-Hao Wei
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, China; School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Xin-Xin Cao
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - De-Feng Xu
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Shu-Ting Wang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Jun-Sheng Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Hua Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China.
| |
Collapse
|
50
|
Peng SJ, Feng Y, Li X, Wang XX, Wang Y, Zhou BT, Liu Y, Liu T, Wu YC. Thymopentin (TP-5) prevents lipopolysaccharide-induced neuroinflammation and dopaminergic neuron injury by inhibiting the NF-κB/NLRP3 signaling pathway. Int Immunopharmacol 2023; 119:110109. [PMID: 37121113 DOI: 10.1016/j.intimp.2023.110109] [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: 09/14/2022] [Revised: 03/03/2023] [Accepted: 03/24/2023] [Indexed: 05/02/2023]
Abstract
Neuroinflammation plays a pivotal role in neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis and stroke, and is accompanied by excessive release of inflammatory cytokines and mediators by activated microglia. Microglial inflammatory response inhibition may be an effective strategy for preventing inflammatory disorders. However, the reciprocal connections between the central nervous system (CNS) and immune system have not been elucidated. Thus far, these links have been proven to mainly involve immuno- and neuropeptides. The pentapeptide thymopentin (TP-5) exerts a significant immunomodulatory effect; however, its antineuroinflammatory effects and underlying mechanism are still unclear. In this study, lipopolysaccharide (LPS) was used to establish an inflammation model, and the therapeutic effect of TP-5 was evaluated. Behavioral tests showed that TP-5 treatment could improve the performance of LPS-treated mice in the open field and pole test, but not hanging wire test. TP-5 also attenuated neuronal lesions in the brains of LPS-treated mice. TP-5 reduced cytotoxicity and morphological changes in activated microglia. Label-free quantitative analysis indicated that the expression of multiple proteins and the activation of associated signaling pathways were altered by TP-5. Moreover, TP-5 could inhibit LPS-induced neuroinflammation in the brain and BV2 microglia and the expression of major genes in the NF-κB/NLRP3 signaling pathway. Additionally, tyrosine hydroxylase (TH) expression downregulation was rescued in the LPS + TP-5 group compared with the LPS group. We conclude that TP-5 exerts neuroprotection by alleviating LPS-induced inflammatory damage and dopaminergic neurodegeneration. The protective effect of TP-5 may involve the NF-κB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Si-Jia Peng
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Ya Feng
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Xuan Li
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Xi-Xi Wang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Yu Wang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Bu-Tian Zhou
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Ye Liu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China.
| | - Yun-Cheng Wu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China.
| |
Collapse
|