1
|
Rațiu S, Mariș MI, Furdui-Lința AV, Stanciu-Lelcu T, Borza C, Olariu S, Bratu T, Sturza A, Muntean DM. Vitamin D alleviates oxidative stress in varicose veins: a pilot study in obese and non-obese patients. Mol Cell Biochem 2025:10.1007/s11010-025-05292-1. [PMID: 40261445 DOI: 10.1007/s11010-025-05292-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025]
Abstract
Chronic venous disease and varicose veins of the lower extremities represent a widespread pathology, particularly in individuals with obesity. A high prevalence of varicose vein disease has been observed in obese patients in association with lower plasma levels of vitamin D. The present pilot study aimed to investigate the acute effects of 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], the biologically active form of vitamin D, on oxidative stress in varicose veins obtained from both obese and non-obese patients undergoing cryostripping surgery for varicose vein ablation. Varicose venous samples treated or not with 1,25(OH)2D3 (100 nM, 12-h incubation) were analysed for reactive oxygen species (ROS) generation using the ferrous xylenol orange oxidation (FOX) assay and immunofluorescence technique. Additionally, the gene expression of endothelial nitric oxide synthase (eNOS), neuronal nitric oxide synthase (nNOS), and inducible nitric oxide synthase (iNOS) was assessed via qPCR. We report a significant reduction in circulating 25-hydroxyvitamin D3 [25(OH)D3] levels in obese as compared to non-obese patients. Ex vivo incubation of the venous samples with 1,25(OH)2D3 resulted in: (i) significant reduction in ROS level, (ii) upregulation of eNOS and nNOS expression, and (iii) downregulation of iNOS expression in both groups of patients. Vitamin D did not exhibit a ROS scavenger effect, and the antioxidant effect is presumably mediated via its receptor whose presence was confirmed in the varicose venous samples. In conclusion, vitamin D exerts protective effects in venous pathology, which may be beneficial in acute administration prior to the surgical intervention. Large clinical trials are required to assess the optimal dosage and time/duration of administration in patients with chronic venous disease with surgical indication.
Collapse
Affiliation(s)
- Sonia Rațiu
- Doctoral School Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Mihaela I Mariș
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Adina V Furdui-Lința
- Doctoral School Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Theia Stanciu-Lelcu
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Claudia Borza
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Sorin Olariu
- Department X - First University Clinic of Surgery, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Tiberiu Bratu
- Doctoral School Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Adrian Sturza
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania.
- Department III - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania.
- Department of Functional Sciences - Chair of Pathophysiology, Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timişoara, 2, Eftimie Murgu Sq., 300041, Timişoara, Romania.
| | - Danina M Muntean
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III - Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| |
Collapse
|
2
|
Alami M, Morvaridzadeh M, El Khayari A, Boumezough K, El Fatimy R, Khalil A, Fulop T, Berrougui H. Reducing Alzheimer's disease risk with SGLT2 inhibitors: From glycemic control to neuroprotection. Ageing Res Rev 2025; 108:102751. [PMID: 40204129 DOI: 10.1016/j.arr.2025.102751] [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: 02/11/2025] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Recent research has established a strong link between metabolic abnormalities and an increased risk of dementia. In parallel, there is growing epidemiological evidence supporting the neuroprotective effects of antidiabetic medications against cognitive impairments. Among these, sodium-glucose co-transporter (SGLT2) inhibitors have emerged as pharmacological candidates with promising potential in alleviating the burden of age-related diseases, particularly neurodegenerative diseases (NDD). SGLT2 inhibitor therapies are FDA-approved medications routinely prescribed to manage diabetes. This novel class was initially developed to address cardiovascular disorders and to reduce the risk of hypoglycemia associated with insulin-secretagogue agents. It subsequently attracted growing interest for its beneficial effects on central nervous system (CNS) disorders. However, the molecular mechanisms through which these glucose-lowering therapies mitigate cognitive decline and limit the progression of certain brain degenerative diseases remain largely unexplored. Consequently, the neuroscientific community needs further studies that gather, analyze, and critically discuss the available mechanistic evidence regarding the neuroprotective effects of SGLT2 inhibitors. This review aims to critically examine the most relevant published findings, both in vitro and in vivo, as well as human studies evaluating the impact of SGLT2 inhibitors exposure on Alzheimer's disease (AD). It seeks to integrate the current understanding of their beneficial effects at the molecular level and their role in addressing the pathophysiology and neuropathology of AD. These insights will help extend our knowledge of how SGLT2 inhibitor therapies are associated with reduced risk of dementia and thus shed light on the link between diabetes and AD.
Collapse
Affiliation(s)
- Mehdi Alami
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Mojgan Morvaridzadeh
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Abdellatif El Khayari
- Faculty of Medical Sciences, UM6P Hospitals, Mohammed VI Polytechnic University, Ben-Guerir 43150, Morocco; Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kaoutar Boumezough
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Rachid El Fatimy
- Faculty of Medical Sciences, UM6P Hospitals, Mohammed VI Polytechnic University, Ben-Guerir 43150, Morocco
| | - Abdelouahed Khalil
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Tamas Fulop
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Hicham Berrougui
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada.
| |
Collapse
|
3
|
Marzookian K, Aliakbari F, Hourfar H, Sabouni F, Otzen DE, Morshedi D. The neuroprotective effect of human umbilical cord MSCs-derived secretome against α-synuclein aggregates on the blood-brain barrier. Int J Biol Macromol 2025; 304:140387. [PMID: 39880228 DOI: 10.1016/j.ijbiomac.2025.140387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
The blood-brain barrier (BBB) is a specialized network that maintains central nervous system homeostasis. Disruption of the BBB can lead to neuronal damage and contribute to neurodegenerative diseases like Parkinson's disease (PD), characterized by alpha-synuclein (αSN) aggregation, which forms intracellular inclusions. Mesenchymal stem cells (MSCs) have shown promise in alleviating the severity of neurological diseases through their paracrine secretions. However, the impact of MSCs secretome on the BBB remains largely unclear. In this study, we investigated the effect of human umbilical cord-derived MSCs (hUC-MSCs) secretome on the BBB in the presence of toxic αSN-aggregates (αSN-AGs). Using in vitro BBB models established through mono- and co-culture systems of hCMEC/D3 cells, we assessed the influence of the secretome on the cytotoxicity and inflammatory responses induced by αSN-AGs. Our results demonstrate that the hUC-MSCs secretome exerts protective effects by mitigating the toxic effects of αSN-AGs on the BBB. Specifically, this study shows a notable reduction in cytotoxicity and inflammation. Our findings highlight the potential of hUC-MSCs secretome as a promising candidate for innovative, cell-free therapies in PD treatment. Furthermore, we propose an optimized method for isolating MSCs from umbilical cord tissue, aimed at facilitating future research on the therapeutic applications of these cells.
Collapse
Affiliation(s)
- Kimia Marzookian
- Department of Bioprocess Engineering, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Farhang Aliakbari
- Department of Bioprocess Engineering, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran; Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Hamdam Hourfar
- Department of Bioprocess Engineering, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Farzaneh Sabouni
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Centre (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, Aarhus C DK-8000, Denmark
| | - Dina Morshedi
- Department of Bioprocess Engineering, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.
| |
Collapse
|
4
|
Balabandian M, Manavi MA, Lesani A, Mohammad Jafari R, Shafaroodi H, Heidari N, Mirnajafi‐Zadeh J, Foroumadi A, Afrooghe A, Dehpour AR. Psilocin, A Psychedelic Drug, Exerts Anticonvulsant Effects Against PTZ- and MES-Induced Seizures in Mice via 5-HT1A and CB1 Receptors: Involvement of Nitrergic, Opioidergic, and Kynurenine Pathways. Pharmacol Res Perspect 2025; 13:e70079. [PMID: 39996441 PMCID: PMC11851161 DOI: 10.1002/prp2.70079] [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: 07/20/2024] [Revised: 02/01/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Epilepsy, a chronic neurological disorder affecting around 65 million people globally, is characterized by recurrent, unprovoked epileptic seizures. Psilocin, the active metabolite of psilocybin, a well-known psychedelic compound, has recently gained attention for its potential antidepressant and anxiolytic properties. This study aims to investigate the anticonvulsant effects of psilocin. The study utilizes behavioral seizure models and electrophysiological recordings in mice to assess the anticonvulsant efficacy of psilocin. The pentylenetetrazole (PTZ) test for clonic seizures and the maximal electroshock (MES) test for generalized tonic-clonic seizures are employed. Cortical electrical activity is monitored to provide insights into the compound's effects on neuronal activity. The involvement of kynurenine pathway, opioidergic and nitrergic systems, as well as cannabinoid receptors using agonist/antagonist paradigms. Western blotting was employed to evaluate the expression levels of key receptors and enzymes implicated in psilocin's anticonvulsant effects. The findings indicate a possible modulation of seizure activity by psilocin, with modest doses (3 mg/kg, i.p.) demonstrating potential anticonvulsant effects. Remarkably, the administration of 1-MT, L-NAME, naltrexone, sildenafil, and AM-251 led to a diminishment of the anticonvulsant effects of psilocin, underscoring the involvement of the kynurenine pathway, nitrergic and opioidergic systems, cGMP, and the CB1 receptor in mediating the anticonvulsant effects of psilocin, respectively. Based on western blotting analysis, the upregulation of 5-HT1A but not 5-HT2A and the downregulation of IDO and CB1 expression following psilocin administration were observed. Acute administration of psilocin exerts anticonvulsant effects that might be mediated at least in part through the kynurenine pathway, opioidergic, serotonergic, and nitrergic systems.
Collapse
Affiliation(s)
- Mohammad Balabandian
- Experimental Medicine Research CenterTehran University of Medical SciencesTehranIran
- School of MedicineIran University of Medical SciencesTehranIran
| | - Mohammad Amin Manavi
- Experimental Medicine Research CenterTehran University of Medical SciencesTehranIran
- Faculty of Pharmacy, Department of Medicinal ChemistryTehran University of Medical SciencesTehranIran
| | - Ali Lesani
- Experimental Medicine Research CenterTehran University of Medical SciencesTehranIran
- Department of Pharmacology, School of MedicineTehran University of Medical SciencesTehranIran
| | - Razieh Mohammad Jafari
- Experimental Medicine Research CenterTehran University of Medical SciencesTehranIran
- Department of Pharmacology, School of MedicineTehran University of Medical SciencesTehranIran
| | - Hamed Shafaroodi
- Experimental Medicine Research CenterTehran University of Medical SciencesTehranIran
- Department of Pharmacology, School of MedicineTehran University of Medical SciencesTehranIran
| | - Navid Heidari
- Department of Physiology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Javad Mirnajafi‐Zadeh
- Department of Physiology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Alireza Foroumadi
- Faculty of Pharmacy, Department of Medicinal ChemistryTehran University of Medical SciencesTehranIran
| | - Arya Afrooghe
- Experimental Medicine Research CenterTehran University of Medical SciencesTehranIran
- School of MedicineIran University of Medical SciencesTehranIran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research CenterTehran University of Medical SciencesTehranIran
- Department of Pharmacology, School of MedicineTehran University of Medical SciencesTehranIran
| |
Collapse
|
5
|
Yang H, Gao J, Wang HY, Ma XM, Liu BY, Song QZ, Cheng H, Li S, Long ZY, Lu XM, Wang YT. The effects and possible mechanisms of whole-body vibration on cognitive function: A narrative review. Brain Res 2025; 1850:149392. [PMID: 39662790 DOI: 10.1016/j.brainres.2024.149392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Whole-body vibration (WBV) is a physical stimulation method that transmits mechanical oscillations to the entire body through a vibration platform or device. Biokinetic and epidemiologic studies have shown that prolonged exposure to high-intensity WBV increases health risks, primarily to the lumbar spine and the nervous system connected to it. There is currently insufficient evidence to demonstrate a quantitative relationship between vibration exposure and risk of health effects. The positive effects of WBV on increasing muscle strength and improving balance and flexibility are well known, but its effects on cognitive function are more complex, with mixed findings, largely related to vibration conditions, including frequency, amplitude, and duration. Studies have shown that short-term low-frequency WBV may have a positive impact on cognitive function, demonstrates potential rehabilitation benefits in enhancing learning and memory, possibly by promoting neuromuscular coordination and enhancing neural plasticity. However, long term exposure to vibration may lead to chronic stress in nerve tissue, affecting nerve conduction efficiency and potentially interfering with neuroprotective mechanisms, thereby having a negative impact on cognitive ability, even causes symptoms such as cognitive decline, mental fatigue, decreased attention, and drowsiness. This literature review aimed to explore the effects of WBV on cognitive function and further to analyze the possible mechanisms. Based on the analysis of literatures, we came to the conclusion that the impact of WBV on cognitive function depends mainly on the frequency and duration of vibration, short-term low-frequency WBV may have a positive impact on cognitive function, while long term exposure to WBV may lead to cognitive decline, and the mechanisms may be involved in neuroinflammation, oxidative stress, synaptic plasticity, and neurotransmitter changes. This review may provide some theoretical foundations and guidance for the prevention and treatment of WBV induced cognitive impairment.
Collapse
Affiliation(s)
- Huan Yang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jie Gao
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Hai-Yan Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xin-Mei Ma
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Bing-Yao Liu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Qian-Zhong Song
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Hui Cheng
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Sen Li
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zai-Yun Long
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xiu-Min Lu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| | - Yong-Tang Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China.
| |
Collapse
|
6
|
Yamamoto T, Mitsunaga F, Kotani A, Tajima K, Wasaki K, Nakamura S. Safety Assessment of a Sublingual Vaccine Formulated with Poly(I:C) Adjuvant and Influenza HA Antigen in Mice and Macaque Monkeys: Comparison with Intranasal Vaccine. Vaccines (Basel) 2025; 13:261. [PMID: 40266106 PMCID: PMC11945353 DOI: 10.3390/vaccines13030261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 04/24/2025] Open
Abstract
A sublingual vaccine comprising the Poly(I:C) adjuvant and influenza HA antigen was evaluated for safety in both mice and macaque monkeys relative to its intranasal counterpart. Safety was assessed in terms of harmful effects corresponding to the upregulation of the inflammation-associated genes Saa3, Tnf, IL6, IL1b, Ccl2, Timp1, C2, Ifi47, Aif1, Omp, Nos2, and/or Gzmb in mice and SAA2, TNF, IL6, IL1B, CCL2, TIMP, C2, AIF1, and GZMB in macaques. Quantitative gene expression analyses were performed using RT-qPCR with RNA samples from four tissue types, the olfactory bulb, pons, lung, tongue, and lymph node, from both mice and macaques. In mice, the intranasally delivered vaccine markedly upregulated the inflammation-related genes in the olfactory bulb 1 day and 7 days after vaccination. The adverse effects of intranasal vaccination were also observed in macaques, albeit to a lesser extent than in mice. The intranasal vaccination also upregulated these genes in the pons of both mice and macaques. In contrast, the sublingual vaccine did not adversely affect the olfactory bulb or pons in either mice or macaques. The intranasally administered vaccine significantly upregulated these genes in the lungs only 1 day after vaccination, but not 7 days later, in both mice and macaques. We conclude that intranasal vaccination results in unfavorable side effects corresponding to upregulated inflammatory genes in the brain (olfactory bulb and pons). Sublingual vaccination, however, did not induce these side effects in either mice or macaques and was hence evaluated as safe.
Collapse
Affiliation(s)
- Tetsuro Yamamoto
- Innovation Research Center, EPS Holdings, Inc., 2-1 Tsukudohachimancho, Shinjuku-ku, Tokyo 162-0815, Japan; (T.Y.); (A.K.); (K.T.); (K.W.)
- EP Mediate Co., Ltd., 1-8 Tsukudocho, Shinjuku-ku, Tokyo 162-0821, Japan
- Research Center, EPS Innovative Medicine Co., Ltd., 1-8 Tsukudocho, Shinjuku-ku, Tokyo 162-0821, Japan
| | - Fusako Mitsunaga
- Intelligence & Technology Lab, Inc., 52-1 Fukue, Kaizu-cho, Kaizu 503-0628, Japan;
- Biomedical Institute, NPO Primate Agora, 52-2 Fukue, Kaizu-cho, Kaizu 503-0628, Japan
| | - Atsushi Kotani
- Innovation Research Center, EPS Holdings, Inc., 2-1 Tsukudohachimancho, Shinjuku-ku, Tokyo 162-0815, Japan; (T.Y.); (A.K.); (K.T.); (K.W.)
- Research Center, EPS Innovative Medicine Co., Ltd., 1-8 Tsukudocho, Shinjuku-ku, Tokyo 162-0821, Japan
| | - Kazuki Tajima
- Innovation Research Center, EPS Holdings, Inc., 2-1 Tsukudohachimancho, Shinjuku-ku, Tokyo 162-0815, Japan; (T.Y.); (A.K.); (K.T.); (K.W.)
- Research Center, EPS Innovative Medicine Co., Ltd., 1-8 Tsukudocho, Shinjuku-ku, Tokyo 162-0821, Japan
| | - Kunihiko Wasaki
- Innovation Research Center, EPS Holdings, Inc., 2-1 Tsukudohachimancho, Shinjuku-ku, Tokyo 162-0815, Japan; (T.Y.); (A.K.); (K.T.); (K.W.)
- EP Mediate Co., Ltd., 1-8 Tsukudocho, Shinjuku-ku, Tokyo 162-0821, Japan
| | - Shin Nakamura
- Intelligence & Technology Lab, Inc., 52-1 Fukue, Kaizu-cho, Kaizu 503-0628, Japan;
- Biomedical Institute, NPO Primate Agora, 52-2 Fukue, Kaizu-cho, Kaizu 503-0628, Japan
| |
Collapse
|
7
|
Bhuia MS, Chowdhury R, Hasan R, Hasan MSA, Ansari SA, Ansari IA, Mubarak MS, Coutinho HDM, Domiciano CB, Islam MT. trans-Ferulic Acid Antagonizes the Anti-Inflammatory Activity of Etoricoxib: Possible Interaction of COX-1 and NOS. Biotechnol Appl Biochem 2025. [PMID: 39985155 DOI: 10.1002/bab.2739] [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: 05/13/2024] [Accepted: 02/06/2025] [Indexed: 02/24/2025]
Abstract
This study emphasizes to investigate the modulatory activity of trans-ferulic acid (TFA) on anti-inflammatory activity of etoricoxib (ETO) and underlying mechanisms via formalin-induced licking and paw edema model and in silico study. Inflammation was induced by injecting formalin (50 µL) into the right hind paw of mice. The animals were treated with different doses of TFA (25, 50, and 75 mg/kg, p.o.). The vehicle and ETO (35 mg/kg, p.o.) were provided as positive and negative control, respectively. ETO also served combined with TFA to evaluate the modulatory activity. The licking behavior was counted for the early and late phases, whereas the paw edema diameter was measured by using a slide caliper. All treatment was continued for 7 days until the edema was totally minimized to determine the inflammation's recovery capability for a specific group. Different computed and web tools were used to estimate molecular binding affinity, binding interactions, and pharmacokinetics. The findings demonstrated that TFA significantly (p < 0.05) enhanced the onset of licking and reduced the number of licks compared to vehicle group. TFA also showed a significant (p < 0.05) diminished in paw edema and complete recovered of the edema after 5 days of treatment indicating the anti-inflammatory effects. However, TFA with ETO notably diminished the anti-inflammatory effects of ETO by enhancing paw edema diameter and licking number. TFA also expressed elevated binding affinity of -7.5 and -6.5 kcal/mol toward nitric oxide (NO) synthase and COX-1, respectively. In conclusion, TFA exerted anti-inflammatory effects and reduces anti-inflammatory capability of ETO.
Collapse
Affiliation(s)
- Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- BioLuster Research Center Ltd., Gopalganj, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- BioLuster Research Center Ltd., Gopalganj, Bangladesh
| | - Rubel Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- BioLuster Research Center Ltd., Gopalganj, Bangladesh
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Mohammad S Mubarak
- Department of Chemistry, The University of Jordan, Amman, Jordan
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA
| | | | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- BioLuster Research Center Ltd., Gopalganj, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, Bangladesh
| |
Collapse
|
8
|
Shi YF, Zhu YT, Zhang ZH, Chen MS, Gao S, Zhang Q, Li CH. Structurally diverse chromane meroterpenoids from Rhododendron capitatum with multifunctional neuroprotective effects. Eur J Med Chem 2025; 283:117188. [PMID: 39709792 DOI: 10.1016/j.ejmech.2024.117188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/05/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Eleven new chromane meroterpenoids (1-11), along with 24 known ones (12-35) were isolated from Rhododendron capitatum, a Tibetan medicine. Their structures were determined via extensive spectroscopic methods. The absolute configurations of 1 and 2 were determined by comparison of the experimental and theoretically calculated ECD data. For compounds 3-9, the absolute configurations at the C-2 were assigned according to the empirical chromane helicity rule. The stereochemistry of the chiral alcohols at C-13 in 3 and C-15 in 4 were determined using the Rh2(OCOCF3)4-induced ECD spectra based on the bulkiness rule. Additionally, the absolute configurations of secondary alcohols at C-13 in 8 and 9 were unambiguously established by Mosher's method. Neuroprotection evaluations in vitro and in vivo revealed that compounds 1, 18, and 21 can significantly inhibit the inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2) protein expressions. Compound 21 also down-regulated MAPK signal pathway in BV-2 cells. The PC-12 cell damage induced by H2O2 and 6-hydroxydopamine (6-OHDA) was attenuated by compounds 1, 21, and 22, especially for 22. Moreover, compounds 3, 6, 22, 23, and 28 significantly enhanced NGF-induced neurite growth in PC-12 cells. Notably, compound 6 demonstrated the most potent neurite growth promotion with a rate of 22.93 ± 2.24 % at 10 μM, which was approximately 3-fold higher than that induced by nerve growth factor (NGF). In AD Caenorhabditis elegans CL4176 model, compounds 1 and 21 delayed Aβ-induced paralysis and reduced ROS expression levels. These studies provide new potential neuroprotective agents for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ye-Fan Shi
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, Shaanxi Engineering Center of Bioresource Chemistry & Sustainable Utilization, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China
| | - Yue-Tong Zhu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, Shaanxi Engineering Center of Bioresource Chemistry & Sustainable Utilization, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China
| | - Zi-Han Zhang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, Shaanxi Engineering Center of Bioresource Chemistry & Sustainable Utilization, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China
| | - Meng-Song Chen
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, Shaanxi Engineering Center of Bioresource Chemistry & Sustainable Utilization, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China
| | - Song Gao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, Shaanxi Engineering Center of Bioresource Chemistry & Sustainable Utilization, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China
| | - Qiang Zhang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, Shaanxi Engineering Center of Bioresource Chemistry & Sustainable Utilization, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China.
| | - Chun-Huan Li
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, Shaanxi Engineering Center of Bioresource Chemistry & Sustainable Utilization, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China.
| |
Collapse
|
9
|
Zhang E, Dai F, Tao L, Chen Y, Chen T, Shen X. Immune cells: Mediators in the metabolites and Alzheimer's disease. J Alzheimers Dis 2025; 103:1277-1288. [PMID: 39876754 DOI: 10.1177/13872877241313140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder that predominantly affects elderly individuals across the globe. While genetic, environmental, and lifestyle factors are known to influence the onset of AD, the underlying mechanisms remain unclear. OBJECTIVE To elucidate the intricate interplay between metabolites and immune cell activation in the ethology of AD, and to determine their collective impact on AD risk. METHODS We conducted a comprehensive analysis of genome-wide association studies data to examine the relationships between metabolites, immune cell phenotypes, and the risk of AD. Our study encompassed a comprehensive examination involving 731 distinct immune cell types, 1400 metabolites, and a large cohort comprising10,520 AD cases with 401,661 controls. We employed univariate Mendelian randomization to assess bidirectional relationships between metabolites and AD, metabolites and immune cells, as well as immune cells and AD. Subsequently, multivariate Mendelian randomization was then applied to evaluate the potential mediating role of immune cells on the relationship between metabolites and AD. RESULTS Specific metabolites, the histidine/pyruvate ratio and homoarginine, were positively associated with the risk of AD, mediated by immune cells. Conversely, 4-hydroxycoumarin and glycolithocholate sulfate showed protective associations against AD. Immune cell markers, CD64 on monocytes and HLA DR on CD14+ CD16- monocytes were linked to higher AD risk, while CD33dim HLA DR+ CD11b- myeloid cells and HLA DR on CD8+ T cells were protective. CONCLUSIONS This study highlights the critical role of immune cells in the pathogenesis of AD, demonstrating how their interaction with specific metabolites influences disease risk.
Collapse
Affiliation(s)
- Erdong Zhang
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Fengqiu Dai
- Department of Anatomy, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Ling Tao
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yanqin Chen
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tingting Chen
- The Pharmacy Department, Guiyang Maternal and Child Health-Care Hospital, Guiyang, Guizhou, China
| | - Xiangchun Shen
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- The Pharmacy Department, Guiyang Maternal and Child Health-Care Hospital, Guiyang, Guizhou, China
| |
Collapse
|
10
|
Kantor S, Drzał A, Setkowicz Z, Elas M, Janeczko K. Dynamics of nitrergic system activation in the rat brain provoked by experimentally induced seizures. Neuroscience 2025; 564:290-298. [PMID: 39424265 DOI: 10.1016/j.neuroscience.2024.10.032] [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: 07/08/2024] [Revised: 10/05/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Epilepsy is a pathophysiological condition displaying a highly diverse phenotype. Consequently, comprehending the mechanisms underlying seizures necessitates moving beyond a simplistic model focused on the imbalance between the classical excitatory and inhibitory neurotransmitter systems. Nitric oxide (NO), a nonclassical and multifunctional gaseous neurotransmitter, has the potential to exert a profound influence on epileptic reactivity. Unfortunately, numerous studies have not provided clear answers about its involvement in the pathophysiology of epilepsy. The objective of our study was to delineate the temporal dynamics of alterations in nitrergic system activation after experimentally induced seizures. Seizures were induced in 2-month-old male Wistar rats (n = 55) by an administration of pilocarpine. Over a 6-hour observation period, seizure behaviour intensity was continuously evaluated using a modified Racine scale. At intervals of 6, 12, 24, 48, or 96 h post-chemoconvulsant administration, NO spin trapping was conducted with ferrous-diethyldithiocarbamate complexes (Fe(DETC)2). Electron paramagnetic resonance (EPR) spectroscopy was employed to quantify mononitrosyl iron complexes (NO-Fe(DETC)2) in the brain. The temporal kinetic of NO release after seizures revealed a rise in NO synthesis during the initial 12 h. Subsequently, a sharp decline occurred, returning to baseline 96 h after pilocarpine injection. Notably, our research suggests that the level of NO synthesis does not interfere with the severity of the epileptic seizures that occur. In light of this, we propose that the nitrergic system is quickly activated in the epileptic brain as a compensatory mechanism of the central nervous system. However, under usual conditions, this activation is insufficient to effectively attenuate seizures.
Collapse
Affiliation(s)
- Szymon Kantor
- Jagiellonian University, Institute of Zoology and Biomedical Research, Laboratory of Experimental Neuropathology, Gronostajowa 9, 30-387 Kraków, Poland; Jagiellonian University, Doctoral School of Exact and Natural Sciences, Łojasiewicza 11, 30-348 Kraków, Poland.
| | - Agnieszka Drzał
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Gronostajowa 7, 30-387 Kraków, Poland
| | - Zuzanna Setkowicz
- Jagiellonian University, Institute of Zoology and Biomedical Research, Laboratory of Experimental Neuropathology, Gronostajowa 9, 30-387 Kraków, Poland
| | - Martyna Elas
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Gronostajowa 7, 30-387 Kraków, Poland
| | - Krzysztof Janeczko
- Jagiellonian University, Institute of Zoology and Biomedical Research, Laboratory of Experimental Neuropathology, Gronostajowa 9, 30-387 Kraków, Poland
| |
Collapse
|
11
|
Kim DU, Kweon B, Oh J, Lim Y, Noh G, Yu J, Kang HR, Kwon T, Lee KY, Bae GS. A Network Pharmacology Study and Experimental Validation to Identify the Potential Mechanism of Heparan Sulfate on Alzheimer's Disease-Related Neuroinflammation. Biomedicines 2025; 13:103. [PMID: 39857687 PMCID: PMC11761859 DOI: 10.3390/biomedicines13010103] [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: 12/03/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Heparan sulfate (HS) is a polysaccharide that is found on the surface of cells and has various biological functions in the body. METHODS The purpose of this study was to predict the pharmacological effects and molecular mechanisms of HS on Alzheimer's disease (AD) and neuroinflammation (NI) through a network pharmacology analysis and to experimentally verify them. RESULTS We performed functional enrichment analysis of common genes between HS target genes and AD-NI gene sets and obtained items such as the "Cytokine-Mediated Signaling Pathway", "Positive Regulation Of MAPK Cascade", and "MAPK signaling pathway". To confirm the predicted results, the anti-inflammatory effect of HS was investigated using lipopolysaccharide (LPS)-stimulated BV2 microglia cells. HS inhibited the production of nittic oxide, interleukin (IL)-6, and tumor necrosis factor-α in LPS-stimulated BV2 cells, but not IL-1β. In addition, HS inactivated P38 in the MAPK signaling pathway. CONCLUSIONS These findings suggest the potential for HS to become a new treatment for AD and NI.
Collapse
Affiliation(s)
- Dong-Uk Kim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Bitna Kweon
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Jinyoung Oh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Yebin Lim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
| | - Gyeongran Noh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
| | - Jihyun Yu
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
| | - Hyang-Rin Kang
- Woori B&B Life Science Laboratory, Jeonju 54853, Republic of Korea; (H.-R.K.)
| | - Tackmin Kwon
- Woori B&B Life Science Laboratory, Jeonju 54853, Republic of Korea; (H.-R.K.)
| | - Kwang youll Lee
- Woori B&B Life Science Laboratory, Jeonju 54853, Republic of Korea; (H.-R.K.)
| | - Gi-Sang Bae
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
- Research Center of Traditional Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| |
Collapse
|
12
|
Olasehinde TA, Olaniran AO. Assessment of Neurotoxic Mechanisms of Individual and Binary Mixtures of Cobalt, Nickel and Lead in Hippocampal Neuronal Cells. ENVIRONMENTAL TOXICOLOGY 2025; 40:128-139. [PMID: 39365032 PMCID: PMC11628647 DOI: 10.1002/tox.24418] [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: 06/17/2023] [Revised: 06/14/2024] [Accepted: 08/20/2024] [Indexed: 10/05/2024]
Abstract
Many studies have focused on the neurotoxic effects of single metals, while investigation on the exposure to metal mixtures, which mainly occur in real-life situations, is scarce. This study sought to assess the neurotoxic effect of Ni, Co, and Pb binary mixtures and their individual effects in hippocampal neuronal cells (HT-22). Cells were exposed to Ni, Co, and Pb separately for 48 h at 37°C and 5% CO2, and cell viability was assessed. Morphological assessment of the cells exposed to binary mixtures of Co, Ni, and Pb and single metals was assessed using a microscope. Furthermore, acetylcholinesterase (AChE) activity, oxidative stress biomarkers (glutathione [GSH] and malondialdehyde [MDA] levels, catalase [CAT], and glutathione-S transferase [GST] activities) and nitric oxide [NO] levels were evaluated after treatment with the binary mixtures and single metals. Binary mixtures of the metals reduced cell viability, exerting an additivity action. The combinations also exerted synergistic action, as revealed by the combination index. Furthermore, a significant reduction in AChE activity, GSH levels, CAT and GST activities, and high MDA and NO levels were observed in neuronal cells. The additive interactions and synergistic actions of the binary mixtures might contribute to the significant reduction of AChE activity, GSH levels, GST, and CAT activities, and an increase in MDA and NO levels. The findings from this study revealed significant evidence that binary mixtures of Co, Pb, and Ni may induce impaired neuronal function and, ultimately, neurodegeneration.
Collapse
Affiliation(s)
- Tosin A. Olasehinde
- Nutrition and Toxicology Division, Food Technology DepartmentFederal Institute of Industrial ResearchLagosNigeria
- Discipline of Microbiology, School of Life SciencesUniversity of Kwazulu‐NatalDurbanSouth Africa
| | - Ademola O. Olaniran
- Discipline of Microbiology, School of Life SciencesUniversity of Kwazulu‐NatalDurbanSouth Africa
| |
Collapse
|
13
|
Lechkova B, Shishmanova-Doseva M, Benbassat N, Gevrenova R, Atanassova P, Penkova N, Peychev L, Hrischev P, Peychev Z, Ivanova S. Evaluation of the Influence of Tanacetum vulgare Extract on Cognitive Functions and Hippocampal BDNF Expression. Molecules 2024; 29:5723. [PMID: 39683883 DOI: 10.3390/molecules29235723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/26/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Tanacetum vulgare L. has been traditionally applied as a remedy for headaches, rheumatism, digestion, respiratory and neurological problems, and other medical conditions. However, the literature data on its effects on cognitive function are scarce. The aim of the present work was to evaluate the effects of two doses of T. vulgare alcohol extract on cognitive functions, hippocampal brain-derived neurotrophic factor (BDNF) expression, and organ toxicity in rats. Rats were treated with T. vulgare 200 mg/kg (TV 200) and 1000 mg/kg (TV 1000) for 28 days. After one week of pre-treatment, the animals were subjected to a series of tests. We found that in the active avoidance test, only TV 200 improved learning and memory, while in the passive avoidance test, both doses facilitated these processes. In addition, the two doses enhanced spatial memory. In the elevated plus maze test, only the higher dose of TV 1000 resulted in anxiolytic-like behavior. Both doses of the extract significantly increased the hippocampal expression of BDNF. We suggest that increased neurotrophic factor expression could be one of the important mechanisms underlying the cognition-enhancing effects of T. vulgare extract.
Collapse
Affiliation(s)
- Borislava Lechkova
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Michaela Shishmanova-Doseva
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Niko Benbassat
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Reneta Gevrenova
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University-Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria
| | - Pepa Atanassova
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Nadya Penkova
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Lyudmil Peychev
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Petar Hrischev
- Department of Physiology, Faculty of Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Zhivko Peychev
- Department of Medical Informatics, Biostatistics and E-Learning, Faculty of Public Health, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Stanislava Ivanova
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| |
Collapse
|
14
|
Kweon B, Oh J, Lim Y, Noh G, Yu J, Kim D, Jang M, Kim D, Bae G. Anti-Inflammatory Effects of Honeysuckle Leaf Against Lipopolysaccharide-Induced Neuroinflammation on BV2 Microglia. Nutrients 2024; 16:3954. [PMID: 39599739 PMCID: PMC11597670 DOI: 10.3390/nu16223954] [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: 09/07/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Neurodegenerative disorders have emerged as a major global public health concern, and the burden is predicted to increase over time. Modulating neuroinflammation and microglial activity is considered a promising target for improving neurodegenerative disorders. The leaf of honeysuckle (LH), which has anti-inflammatory properties, has long been collected, regardless of the season, and used for medicinal purposes. However, research on its effects on neuroinflammation is scarce. In this study, we determined the neuroprotective effects of LH water extract by inhibiting microglial activation induced by lipopolysaccharide (LPS). METHODS The production or secretion of pro-inflammatory mediators was examined in LPS-exposed BV2 cells to ascertain the efficacy of LH water extract in improving neuroinflammation. In addition, the phosphorylation of mitogen-activated protein kinases (MAPKs) and the degradation of inhibitory κBα (IκBα) were analyzed to elucidate the regulatory mechanisms of LH water extract. Ultra-performance liquid chromatography (UPLC) analysis was conducted to identify the active component of the LH. RESULTS LH water extract suppressed the formation of inducible nitric oxide synthase (iNOS), nitric oxide (NO), and pro-inflammatory cytokines, including interleukin (IL)-1β and tumor necrosis factor (TNF)-α, in LPS-activated BV2 cells. LH impeded the activation of c-Jun N-terminal kinase (JNK). Moreover, chlorogenic acid was found in LH. CONCLUSIONS The above findings suggest that LH water extract could improve neuroinflammation.
Collapse
Affiliation(s)
- Bitna Kweon
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Jinyoung Oh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Yebin Lim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Gyeongran Noh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Jihyun Yu
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Donggu Kim
- Department of Herbology, College of Korean Medicine, Dong-Eui University, 176 Eomgwang-ro, Busan 47340, Republic of Korea
| | - Mikyung Jang
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Donguk Kim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| | - Gisang Bae
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
- Research Center of Traditional Korean Medicine, Wonkwang University, 460 Iksan-daero, Iksan 54538, Republic of Korea
| |
Collapse
|
15
|
Luo B. Insights into the advances in therapeutic drugs for neuroinflammation-related diseases. Int J Neurosci 2024; 134:1256-1281. [PMID: 37722706 DOI: 10.1080/00207454.2023.2260088] [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: 08/11/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
Studies have shown that neurodegenerative diseases such as AD and PD are related to neuroinflammation. Neuroinflammation is a common inflammatory condition that can lead to a variety of dysfunction in the body. At present, it is no medications specifically approved to prevent or cure neuroinflammation, so even though many drugs can temporarily control the neurological symptoms of neuroinflammation, but no one can reverse the progress of neuroinflammation, let al.one completely cure neuroinflammation. Therefore, it is urgent to develop new drug development for neuroinflammation treatment. In this review, we highlight the therapeutic advancement in the field of neurodegenerative disorders, by focusing on the impact of neuroinflammation treatment has on these conditions, and the effective drugs for the treatment of neuroinflammation and neurodegenerative diseases and their latest research progress are reviewed according to the related signaling pathway, as well as the prospect of their clinical application is also discussed. The purpose of this review is to enable specialists to better understand the mechanisms underlying neuroinflammation and anti-inflammatory drugs, promote the development of therapeutic drugs for neuroinflammation and neurodegenerative diseases, and further provide therapeutic references for clinical neurologists.
Collapse
Affiliation(s)
- Bozhi Luo
- School of Basic Medicine, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
16
|
Adewoyin M, Hamarsha A, Akinsola R, Teoh SL, Azmai MNA, Abu Bakar N, Nasruddin NS. Intraperitoneal Injection of the Porphyromonas gingivalis Outer Membrane Vesicle (OMV) Stimulated Expressions of Neuroinflammatory Markers and Histopathological Changes in the Brains of Adult Zebrafish. Int J Mol Sci 2024; 25:11025. [PMID: 39456807 PMCID: PMC11506875 DOI: 10.3390/ijms252011025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Porphyromonas gingivalis is the major pathogenic bacteria found in the subgingival plaque of patients with periodontitis, which leads to neuroinflammation. The bacteria destroy periodontal tissue through virulence factors, which are retained in the bacteria's outer membrane vesicles (OMV). This study aimed to determine the real-time effect of an intraperitoneal injection of P. gingivalis OMV on the production and expression of inflammatory markers and histopathological changes in adult zebrafishes' central nervous systems (CNS). Following the LD50 (21 µg of OMV), the zebrafish were injected intraperitoneally with 18 µg of OMVs, and the control group were injected with normal saline at seven different time points. Brains of experimental zebrafish were dissected at desired time points for colorimetric assays, ELISA, and histology. This study discovered that nitric oxide and PGE2 were significantly increased at 45 min, while IL-1β and IL-6 were expressed at subsequent 12 h and 24 h time points, respectively. Histopathological changes such as blood coagulation, astrocytosis, edema, spongiosis, and necrosis were observed between the 6hour and 24 h time points. The two apoptotic enzymes, caspases 3 and 9, were not expressed at any point. In summary, the OMV-induced neuroinflammatory responses and histopathological changes in adult zebrafish were time-point dependent. This study will enrich our understanding of the mechanism of P. gingivalis OMVs in neuroinflammation in a zebrafish model, most especially the timing of the expression of inflammatory mediators in relation to observable changes in brain tissues.
Collapse
Affiliation(s)
- Malik Adewoyin
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (M.A.); (A.H.)
| | - Ahmed Hamarsha
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (M.A.); (A.H.)
| | - Rasaq Akinsola
- Department of Medicine, Division of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Mohammad Noor Amal Azmai
- Aquatic Animal Health and Therapeutics Laboratory, Institute of Bioscience, Universiti Putra Malaysia, UPM, Serdang 43400, Selangor, Malaysia;
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Selangor, Malaysia;
| | - Noraini Abu Bakar
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Selangor, Malaysia;
| | - Nurrul Shaqinah Nasruddin
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (M.A.); (A.H.)
| |
Collapse
|
17
|
Simões JLB, de Carvalho Braga G, Eichler SW, da Silva GB, Bagatini MD. Implications of COVID-19 in Parkinson's disease: the purinergic system in a therapeutic-target perspective to diminish neurodegeneration. Purinergic Signal 2024; 20:487-507. [PMID: 38460075 PMCID: PMC11377384 DOI: 10.1007/s11302-024-09998-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 02/21/2024] [Indexed: 03/11/2024] Open
Abstract
The pathophysiology of Parkinson's disease (PD) is marked by degeneration of dopaminergic neurons in the substantia nigra. With advent of COVID-19, which is closely associated with generalized inflammation and multiple organ dysfunctions, the PD patients may develop severe conditions of disease leading to exacerbated degeneration. This condition is caused by the excessive release of pro-inflammatory markers, called cytokine storm, that is capable of triggering neurodegenerative conditions by affecting the blood-brain barrier (BBB). A possible SARS-CoV-2 infection, in serious cases, may compromise the immune system by triggering a hyperstimulation of the neuroimmune response, similar to the pathological processes found in PD. From this perspective, the inflammatory scenario triggers oxidative stress and, consequently, cellular dysfunction in the nervous tissue. The P2X7R seems to be the key mediator of the neuroinflammatory process, as it acts by increasing the concentration of ATP, allowing the influx of Ca2+ and the occurrence of mutations in the α-synuclein protein, causing activation of this receptor. Thus, modulation of the purinergic system may have therapeutic potential on the effects of PD, as well as on the damage caused by inflammation of the BBB, which may be able to mitigate the neurodegeneration caused by diseases. Considering all the processes of neuroinflammation, oxidative stress, and mitochondrial dysfunction that PD propose, we can conclude that the P2X7 antagonist acts in the prevention of viral diseases, and it also controls purinergic receptors formed by multi-target compounds directed to self-amplification circuits and, therefore, may be a viable strategy to obtain the desired disease-modifying effect. Thus, purinergic system receptor modulations have a high therapeutic potential for neurodegenerative diseases such as PD.
Collapse
Affiliation(s)
| | | | | | - Gilnei Bruno da Silva
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Margarete Dulce Bagatini
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil.
| |
Collapse
|
18
|
Turiaco F, Iannuzzo F, Bruno A, Drago A. Genetics of suicide ideation. A role for inflammation and neuroplasticity? Eur Arch Psychiatry Clin Neurosci 2024; 274:1527-1541. [PMID: 38878077 PMCID: PMC11422468 DOI: 10.1007/s00406-024-01836-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 05/27/2024] [Indexed: 09/25/2024]
Abstract
Suicide is a leading cause of death worldwide. Suicide ideation (SI) is a known risk factor for suicide behaviour (SB). The current psychobiology and genetic predisposition to SI and SB are poorly defined. Despite convincing relevance of a genetic background for SI, there is no current implementable knowledge about the genetic makeup that identifies subjects at risk for it. One of the possible reasons for the absence of a clear-cut evidence is the polygenetic nature of SI along with the very large sample sizes that are needed to observe significant genetic association result. The CATIE sample was instrumental to the analysis. SI was retrieved as measured by the Calgary test. Clinical possible covariates were identified by a nested regression model. A principal component analysis helped in defining the possible genetic stratification factors. A GWAS analysis, polygenic risk score associated with a random forest analysis and a molecular pathway analysis were undertaken to identify the genetic contribution to SI. As a result, 741 Schizophrenic individuals from the CATIE were available for the genetic analysis, including 166,325 SNPs after quality control and pruning. No GWAS significant result was found. The random forest analysis conducted by combining the polygenic risk score and several clinical variables resulted in a possibly overfitting model (OOB error rate < 1%). The molecular pathway analysis revealed several molecular pathways possibly involved in SI, of which those involved in microglia functioning were of particular interest. A medium-small sample of SKZ individuals was analyzed to shed a light on the genetic of SI. As an expected result from the underpowered sample, no GWAS positive result was retrieved, but the molecular pathway analysis indicated a possible role of microglia and neurodevelopment in SI.
Collapse
Affiliation(s)
- Fabrizio Turiaco
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98125, Contesse, Messina, Italy
| | - Fiammetta Iannuzzo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98125, Contesse, Messina, Italy
| | - Antonio Bruno
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98125, Contesse, Messina, Italy
| | - Antonio Drago
- Unit for Psychiatric Research, Psychiatry, Aalborg University Hospital, 9100, Aalborg, Denmark.
| |
Collapse
|
19
|
Hassan ZR, El-Sayed S, Zekry KM, Ahmed SG, Hassan Abd Elhamid A, Salama DEA, Taha AK, Mahmoud NA, Mohammed SF, Amin MM, Mohamed RE, Eraque AMS, Mohamed SA, Abdelgalil RM, Atta SA, Fahmy NT, Badr MS. Evaluation of muscular apoptotic changes and myogenin gene expression in experimental trichinosis after stem cells and atorvastatin added to ivermectin treatment. Exp Parasitol 2024; 265:108823. [PMID: 39187057 DOI: 10.1016/j.exppara.2024.108823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 04/20/2024] [Accepted: 08/08/2024] [Indexed: 08/28/2024]
Abstract
Trichinosis is a common parasitic disease that affects the striated skeletal muscles, causing apoptotic and degenerative changes associated with myogenin expression in the affected myocytes. Hence, this study aimed to assess the ameliorative effects of stem cells and atorvastatin added to ivermectin on the infected myocytes during the muscular phase of murine trichinosis. 120 laboratory Swiss albino male mice were divided into 10 groups, and each group was subdivided into intestinal and muscular phases (each n = 6); uninfected control; untreated infected control; infected received ivermectin monotherapy; infected received atorvastatin monotherapy; infected received stem cells monotherapy; infected received ivermectin and atorvastatin dual therapy; infected received ivermectin and stem cells dual therapy; infected received atorvastatin and stem cells dual therapy; infected received ivermectin 0.2, atorvastatin 40, and stem cells triple therapy; and infected received ivermectin 0.1, atorvastatin 20, and stem cells triple therapy. Intestinal phase mice were sacrificed on the 5th day post-infection, while those of the muscular phase were sacrificed on the 35th day post-infection. Parasitological, histopathological, ultrastructural, histochemical, biochemical, and myogenin gene expression assessments were performed. The results revealed that mice that received ivermectin, atorvastatin, and stem cell triple therapies showed the maximum reduction in the adult worm and larvae burden, marked improvement in the underlying muscular degenerative changes (as was noticed by histopathological, ultrastructural, and histochemical Feulgen stain assessment), lower biochemical levels of serum NK-κB and tissue NO, and lower myogenin expression. Accordingly, the combination of stem cells, atorvastatin, and ivermectin affords a potential synergistic activity against trichinosis with considerable healing of the underlying degenerative sequel.
Collapse
Affiliation(s)
- Zeinab R Hassan
- Departments of Parasitology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt; Departments of Parasitology, Benha National University (BNU), Qalyubia, Egypt.
| | - Samar El-Sayed
- Departments of Parasitology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Kareman M Zekry
- Departments of Parasitology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Samah G Ahmed
- Histology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | | | - Doaa E A Salama
- Pathology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt; Departments of Pathology, School of Medicine, Badr University in Cairo (BUC), Cairo, Egypt
| | - Azza Kamal Taha
- Pathology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Nihal A Mahmoud
- Physiology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | | | - Mona M Amin
- Pharmacology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | | | - Ayat M S Eraque
- Biochemestry, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Shimaa A Mohamed
- Biochemestry, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Ranya M Abdelgalil
- Anatomy, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | | | - Nermeen Talaat Fahmy
- Molecular Biology and Genomics, Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Mohamed S Badr
- Molecular Biology and Genetic-Bioinformatics Nano-Robot Diagnostics, Medical Research Centre, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
20
|
Tsuji S, Kuramoto Y, Rajbhandari S, Takeda Y, Yamahara K, Yoshimura S. Intravenous administration of human amnion-derived mesenchymal stem cells improves gait and sensory function in mouse models of spinal cord injury. Front Cell Dev Biol 2024; 12:1464727. [PMID: 39324071 PMCID: PMC11422150 DOI: 10.3389/fcell.2024.1464727] [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: 07/15/2024] [Accepted: 08/21/2024] [Indexed: 09/27/2024] Open
Abstract
Introduction Spinal cord injury (SCI) leads to severe disabilities and remains a significant social and economic challenge. Despite advances in medical research, there are still no effective treatments for SCI. Human amnion-derived mesenchymal stem cells (hAMSCs) have shown potential due to their anti-inflammatory and neuroprotective effects. This study evaluates the therapeutic potential of intravenously administered hAMSCs in SCI models. Methods Three days after induction of SCI with forceps calibrated with a 0.2 mm gap, hAMSCs or vehicle were administered intravenously. Up to 4 weeks of SCI induction, motor function was assessed by scores on the Basso Mouse Locomotor Scale (BMS) and the Basso-Beattie-Bresnahan Scale (BBB), and sensory function by hindlimb withdrawal reflex using von Frey filaments. Six weeks after SCI induction, gait function was assessed using three-dimensional motion analysis. Immunohistochemistry, polymerase chain reaction (PCR), flow cytometry, and ELISA assay were performed to clarify the mechanisms of functional improvement. Results The hAMSC treatment significantly improved sensory response and gait function. In the SCI site, immunohistochemistry showed a reduction in Iba1-positive cells and PCR revealed decreased TNFα and increased BDNF levels in the hAMSC-treated group. In assessing the systemic inflammatory response, hAMSC treatment reduced monocytic bone marrow-derived suppressor cells (M-MDSCs) and Ly6C-positive inflammatory macrophages in the bone marrow by flow cytometry and serum NO levels by ELISA assay. Discussion This study demonstrates the therapeutic potential of the hAMSC in SCI, with improvements in gait and sensory functions and reduced inflammation both locally and systemically. The findings support further investigation of the hAMSC as a potential treatment for SCI, focusing on their ability to modulate inflammation and promote neuroprotection.
Collapse
Affiliation(s)
- Shoichiro Tsuji
- Department of Neurosurgery, Hyogo Medical University, Hyogo, Japan
| | - Yoji Kuramoto
- Department of Neurosurgery, Hyogo Medical University, Hyogo, Japan
| | | | - Yuki Takeda
- Department of Neurosurgery, Hyogo Medical University, Hyogo, Japan
| | - Kenichi Yamahara
- Laboratory of Molecular and Cellular Therapy, Institute for Advanced Medical Sciences, Hyogo Medical University, Hyogo, Japan
| | | |
Collapse
|
21
|
Abdallah SM, Muhammed RE, Mohamed RE, Khalil WKB, Taha DA, Shalaby MB, Elgohary I, Abdallah AA, Habib HM, El-Yazbi AF. Integrated Biomarker Response Emphasizing Neuronal Oxidative Stress and Genotoxicity Induced by Oxamyl in Sprague Dawley Rats: Ameliorative Effect of Ginseng as a Neuroprotective Agent. TOXICS 2024; 12:655. [PMID: 39330583 PMCID: PMC11435561 DOI: 10.3390/toxics12090655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024]
Abstract
Climate change has led to increased and varying pest infestation patterns, triggering a rise in pesticide usage and exposure. The effects of oxamyl, a widely used nematicide in Egypt, encompasses typical signs of carbamate intoxication; nevertheless, long-term effects of oxamyl exposure, particularly on the nervous system, require further elucidation. This study systematically investigated the mechanism and manifestations of repeated subacute exposure to sublethal doses of oxamyl in male SD rats. Data showed a dose-dependent genotoxic effect, manifested as increased bone marrow micronuclei and decreased brain expression of key genes involved in neurogenesis and neuronal development. Coincidently, brain histopathology showed dose-dependent neurodegeneration in various regions, associated with a significant increase in GFAP immunoreactivity, indicative of neuroinflammation. Biochemical examination revealed a typical pattern of cholinesterase inhibition by carbamates in serum and brain tissue, as well as increased oxidative stress markers in the brain such as SOD activity reduction, alongside an increase in NO and MDA. The ability of Ginseng at a 100 mg/Kg dose to ameliorate the effects of oxamyl exposure was investigated. Ginseng use, either as a protective or therapeutic regimen, attenuated the observed genotoxic, neuroinflammatory, and biochemical alterations. Our results indicate that repeated exposure to oxamyl triggers an integrative neurotoxic response, driven by genotoxicity, oxidative stress, and neuroinflammation, that could trigger an increase in neurological and cognitive disorders. These findings emphasize the urgent need for confirmatory translational studies in human subjects to assess these changes and inform policy decisions regarding safe levels of usage and appropriate agricultural and public health practices.
Collapse
Affiliation(s)
- Salwa M. Abdallah
- Center of Excellence for Toxicological Testing, Department of Mammalian and Aquatic Toxicology, Central Agricultural Pesticides Lab (CAPL), Agricultural Research Center (ARC), Dokki, Giza 12618, Egypt; (S.M.A.); (R.E.M.); (R.E.M.)
| | - Reham E. Muhammed
- Center of Excellence for Toxicological Testing, Department of Mammalian and Aquatic Toxicology, Central Agricultural Pesticides Lab (CAPL), Agricultural Research Center (ARC), Dokki, Giza 12618, Egypt; (S.M.A.); (R.E.M.); (R.E.M.)
| | - Reda E. Mohamed
- Center of Excellence for Toxicological Testing, Department of Mammalian and Aquatic Toxicology, Central Agricultural Pesticides Lab (CAPL), Agricultural Research Center (ARC), Dokki, Giza 12618, Egypt; (S.M.A.); (R.E.M.); (R.E.M.)
| | - Wagdy K. B. Khalil
- Department of Cell Biology, National Research Centre, El-Bohouth, Cairo 12262, Egypt; (W.K.B.K.); (D.A.T.)
| | - Dalia A. Taha
- Department of Cell Biology, National Research Centre, El-Bohouth, Cairo 12262, Egypt; (W.K.B.K.); (D.A.T.)
| | - Mohamed B. Shalaby
- Department of Toxicology Research, Research Institute of Medical Entomology (RIME), General Organization of Teaching Hospitals and Institutes (GOTHI), Ministry of Health and Population (MoHP), Dokki, Cairo 12311, Egypt;
| | - Islam Elgohary
- Department of Pathology, Animal Health Research Institute (AHRI), Agricultural Research Center (ARC), Dokki, Giza 12618, Egypt;
| | - Amr A. Abdallah
- Center of Excellence for Toxicological Testing, Department of Mammalian and Aquatic Toxicology, Central Agricultural Pesticides Lab (CAPL), Agricultural Research Center (ARC), Dokki, Giza 12618, Egypt; (S.M.A.); (R.E.M.); (R.E.M.)
| | - Hosam M. Habib
- Research & Innovation Hub, Alamein International University, Alamein 51718, Egypt
| | - Ahmed F. El-Yazbi
- Research & Innovation Hub, Alamein International University, Alamein 51718, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
22
|
Xiao J, Lin X, Yang Y, Yu Y, Li Y, Xu M, Liu Y. Metabolomic Profiling and Biological Investigation of the Marine Sponge-Derived Fungus Aspergillus sp. SYPUF29 in Response to NO Condition. J Fungi (Basel) 2024; 10:636. [PMID: 39330395 PMCID: PMC11433098 DOI: 10.3390/jof10090636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024] Open
Abstract
Marine-derived fungi are assuming an increasingly central role in the search for natural leading compounds with unique chemical structures and diverse pharmacological properties. However, some gene clusters are not expressed under laboratory conditions. In this study, we have found that a marine-derived fungus Aspergillus sp. SYPUF29 would survive well by adding an exogenous nitric oxide donor (sodium nitroprusside, SNP) and nitric oxide synthetase inhibitor (L-NG-nitroarginine methyl ester, L-NAME) in culture conditions. Moreover, using the LC-MS/MS, we initially assessed and characterized the difference in metabolites of Aspergillus sp. SYPUF29 with or without an additional source of nitrogen. We have found that the metabolic pathway of Arginine and proline metabolism pathways was highly enriched, which was conducive to the accumulation of alkaloids and nitrogen-containing compounds after adding an additional source of nitrogen in the cultivated condition. Additionally, the in vitro anti-neuroinflammatory study showed that the extracts after SNP and L-NAME were administrated can potently inhibit LPS-induced NO-releasing of BV2 cells with lower IC50 value than without nitric oxide. Further Western blotting assays have demonstrated that the mechanism of these extracts was associated with the TLR4 signaling pathway. Additionally, the chemical investigation was conducted and led to nine compounds (SF1-SF9) from AS1; and six of them belonged to alkaloids and nitrogen-containing compounds (SF1-SF6), of which SF1, SF2, and SF8 exhibited stronger activities than the positive control, and showed potential to develop the inhibitors of neuroinflammation.
Collapse
Affiliation(s)
- Jiao Xiao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiuping Lin
- Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Yanqiu Yang
- College of Information Science and Engineering, Northeastern University, Shenyang 110819, China
| | - Yingshu Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yinyin Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mengjie Xu
- Department of Biological Sciences, Xinzhou Normal University, Xinzhou 034000, China
| | - Yonghong Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
23
|
Guo S, Zhang J, Zhang Q, Xu S, Liu Y, Ma S, Hu X, Liu Y, Zhang X, Jiang R, Zhang Z, Zhang Z, Zhou Z, Wen L. Polygala tenuifolia willd. Extract alleviates LPS-induced acute lung injury in rats via TLR4/NF-κB pathway and NLRP3 inflammasome suppression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155859. [PMID: 38972239 DOI: 10.1016/j.phymed.2024.155859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 06/03/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND Acute lung injury (ALI) has received considerable attention in the field of critical care as it can lead to high mortality rates. Polygala tenuifolia, a traditional Chinese medicine with strong expectorant properties, can be used to treat pneumonia. Owing to the complexity of its composition, the main active ingredient is not yet known. Thus, there is a need to identify its constituent compounds and mechanism of action in the treatment of ALI using advanced technological means. PURPOSE We investigated the anti-inflammatory mechanism and constituent compounds with regard to the effect of P. tenuifolia Willd. extract (EPT) in lipopolysaccharide (LPS)-induced ALI in vivo and in vitro. METHODS The UHPLC-Q-Exactive Orbitrap MS technology was used to investigate the chemical profile of EPT. Network pharmacology was used to predict the targets and pathways of action of EPT in ALI, and molecular docking was used to validate the binding of polygalacic acid to Toll-like receptor (TLR) 4. The main compounds were determined using LC-MS. A rat model of LPS-induced ALI was established, and THP-1 cells were stimulated with LPS and adenosine triphosphate (ATP) to construct an in vitro model. Pathological changes were observed using hematoxylin and eosin staining, Wright-Giemsa staining, and immunohistochemistry. The expression of inflammatory factors (NE, MPO, Ly-6 G, TNF-α, IL-1β, IL-6, and iNOS) was determined using enzyme-linked immunosorbent assay, real-time fluorescence quantitative polymerase chain reaction, and western blotting. The LPS + ATP-induced inflammation model in THP-1 cells was used to verify the in vivo experimental results. RESULTS Ninety-nine compounds were identified or tentatively deduced from EPT. Using network pharmacology, we found that TLR4/NF-κB may be a relevant pathway for the prevention and treatment of ALI by EPT. Polygalacic acid in EPT may be a potential active ingredient. EPT could alleviate LPS-induced histopathological lung damage and reduce the wet/dry lung weight ratio in the rat model of ALI. Moreover, EPT decreased the white blood cell and neutrophil counts in the bronchoalveolar lavage fluid and decreased the expression of genes and proteins of relevant inflammatory factors (NE, MPO, Ly-6 G, TNF-α, IL-1β, IL-6, and iNOS) in lung tissues. It also increased the expression of endothelial-type nitric oxide synthase expression. Western blotting confirmed that EPT may affect TLR4/NF-κB and NLRP3 signaling pathways in vivo. Similar results were obtained in THP-1 cells. CONCLUSION EPT reduced the release of inflammatory factors by affecting TLR4/NF-κB and NLRP3 signaling pathways, thereby attenuating the inflammatory response of ALI. Polygalacic acid is the likely compounds responsible for these effects.
Collapse
Affiliation(s)
- Shuyun Guo
- School of Basic Medicine, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Jianguang Zhang
- Qinghai-Tibetan Plateau Ethnic Medicinal Resources Protection and Utilization Key Laboratory of National Ethnic Affairs Commission of the People's Republic of China, Southwest Minzu University, Chengdu 610225, China; Qinzhou Provincial Health School, Qinzhou 535000, China
| | - Qian Zhang
- School of Basic Medicine, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Shuang Xu
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Yuezhen Liu
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Shangzhi Ma
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Xiaodi Hu
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Yanju Liu
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Xiuqiao Zhang
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Ruixue Jiang
- School of Basic Medicine, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Zhifeng Zhang
- Qinghai-Tibetan Plateau Ethnic Medicinal Resources Protection and Utilization Key Laboratory of National Ethnic Affairs Commission of the People's Republic of China, Southwest Minzu University, Chengdu 610225, China
| | - Zhihua Zhang
- School of Basic Medicine, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China.
| | - Zhongshi Zhou
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China.
| | - Li Wen
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China.
| |
Collapse
|
24
|
Abudouaini H, Zhang X, Dai Y, Meng Y, Lu Q, Ren Q, Sun H, Ma Y, He B, Wang S. Activating the iNOS regulatory pathway by arginine deprivation targets energy metabolism to induce autophagy-dependent apoptosis against spinal echinococcosis. Biochem Pharmacol 2024; 227:116453. [PMID: 39059773 DOI: 10.1016/j.bcp.2024.116453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Spinal echinococcosis is one of the most overlooked zoonotic parasitic diseases worldwide. There is currently no safe and effective treatment to eradicate it, and research based on the physiological-metabolic signature of the disease is lacking. Herein, we repurposed agrimol B as a potent anti-hydatid compound and validated its pharmacological mechanism based on arginine uptake as a target through multi-omics sequencing. This herbal component suppressed energy metabolism and activated ROS aggregation by inducing mitochondrial membrane potential depolarization, which subsequently triggered autophagy-dependent apoptosis leading to parasite death. Moreover, we discovered that arginine deprivation induced metabolic changes led to a shift from ornithine to nitrogen oxide synthesis, thus boosting the iNOS enzyme-regulated dominant metabolic pathway. The excess NO targeted the mitochondrial respiratory chain complex IV to disrupt energy metabolic homeostasis and induced a downstream pathological waterfall effect to kill the hydatid. A novel metabolic regulatory mechanism targeting mitochondrial damage for arginine starvation therapy was discovered. Finally, arginine depletion was found to be superior to the anti-spinal echinococcosis effect of albendazole and accompanied by the potential for disc protection. This study unveils the role of arginine in the physiological metabolism of Echinococcus granulosus and reveals the value of targeting arginine metabolism as a potential therapy. In addition, agrimol B is proposed as a promising therapeutic strategy for spinal echinococcosis to block arginine uptake and break this parasite's metabolic balance.
Collapse
Affiliation(s)
- Haimiti Abudouaini
- Department of Spine Surgery, Xi'an Jiaotong University Affiliated HongHui Hospital, Beilin District, Xi'an, Shanxi Province, 710000, China
| | - Xuefang Zhang
- Department of Spine Surgery, Xi'an Jiaotong University Affiliated HongHui Hospital, Beilin District, Xi'an, Shanxi Province, 710000, China
| | - Yi Dai
- The First Affiliated Hospital of Shihezi University, Xinjiang Uygur Autonomous Region, Shihezi City, 832000, China
| | - Yibin Meng
- Department of Spine Surgery, Xi'an Jiaotong University Affiliated HongHui Hospital, Beilin District, Xi'an, Shanxi Province, 710000, China
| | - Qing Lu
- Department of Spine Surgery, Xi'an Jiaotong University Affiliated HongHui Hospital, Beilin District, Xi'an, Shanxi Province, 710000, China
| | - Qian Ren
- The First Affiliated Hospital of Shihezi University, Xinjiang Uygur Autonomous Region, Shihezi City, 832000, China
| | - Haohao Sun
- The First Affiliated Hospital of Shihezi University, Xinjiang Uygur Autonomous Region, Shihezi City, 832000, China
| | - Yibo Ma
- The First Affiliated Hospital of Shihezi University, Xinjiang Uygur Autonomous Region, Shihezi City, 832000, China
| | - Baorong He
- Department of Spine Surgery, Xi'an Jiaotong University Affiliated HongHui Hospital, Beilin District, Xi'an, Shanxi Province, 710000, China.
| | - Sibo Wang
- Department of Spine Surgery, Xi'an Jiaotong University Affiliated HongHui Hospital, Beilin District, Xi'an, Shanxi Province, 710000, China.
| |
Collapse
|
25
|
Behairi N, Samer A, Sahraoui L, Mataam DH, Trari R, Flissi B, Belguendouz H, Amir ZC, Touil-Boukoffa C. Neuroinflammation, neurodegeneration and alteration of spatial memory in BALB/c mice through ampicillin-induced gut dysbiosis; NOS2 and NFL involvement in a microbiota-gut-brain axis model. J Neuroimmunol 2024; 392:578374. [PMID: 38797060 DOI: 10.1016/j.jneuroim.2024.578374] [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: 02/19/2024] [Revised: 04/05/2024] [Accepted: 05/18/2024] [Indexed: 05/29/2024]
Abstract
We aimed to investigate ampicillin (AMP) mechanisms in microbiota-gut-brain axis. We evaluated its effect on two gut and brain regions and behavioral performances. We administred AMP (1 g/l) to BALB/c mice for 21 days. Then, we analyzed body weigth change, stool consistency scoring, gut length, intestinal microbiota composition, nitric oxide synthase 2 (NOS2) expression and tissue integrity. We subsequently evaluated NOS2, GFAP, CD68 and NFL cerebral expression and spatial memory.Interestingly, our data showed gut microbiota disruption, NOS2 upregulation and tissue damage, associated to cerebral NOS2, GFAP, CD68 and NFL over-expression and behavioral alteration. Antiobiotic therapy should be prescribed with great caution.
Collapse
Affiliation(s)
- Nassima Behairi
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Arezki Samer
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Lynda Sahraoui
- Laboratory of Animal Health and Production, Higher National Veterinary School of Issad-Abbes Oued-Smar, Algiers, Algeria
| | - Djehane Houria Mataam
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Ryad Trari
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Billel Flissi
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Houda Belguendouz
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Zine-Charaf Amir
- Department of Anatomy and Pathological Cytology, University Hospital Center Mustapha Pacha, 1945 Pl. May 1st, Sidi M'Hamed, 16000 Algiers, Algeria
| | - Chafia Touil-Boukoffa
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria.
| |
Collapse
|
26
|
Kessas K, Lounis W, Chouari Z, Vejux A, Lizard G, Kharoubi O. Benefits of rutin on mitochondrial function and inflammation in an aluminum-induced neurotoxicity rat model: Potential interest for the prevention of neurodegeneration. Biochimie 2024; 222:1-8. [PMID: 38408719 DOI: 10.1016/j.biochi.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 02/28/2024]
Abstract
Rutin, a phenolic compound, exhibits a diverse range of biological properties, including antioxidant, anti-inflammatory, and antimicrobial effects. In this study, we aimed to investigate the potential of rutin, a naturally occurring plant bioactive molecule, to mitigate the neurotoxic effects induced by aluminum chloride (AlCl3). Over a period of 6 weeks, rats were intraperitoneally injected with AlCl3 at a weekly dose of 60 mg/kg, while rutin treatment was administered orally via gavage at a daily dose of 30 mg/kg. AlCl3 exposure resulted in a significant increase lipid peroxidation (LPO) by 316.24%, nitrate levels by 504.14%, and tumor necrosis factor-alpha (TNF-α) levels by 93.82% in brain mitochondria. Additionally, AlCl3 exposure led to a reduction in glutathione levels and the activity of antioxidant enzymes, including superoxide dismutase (SOD) by 19.74%, glutathione peroxidase (GPx) by 44.76%, and catalase by 50.50%. There was also a significant decline in the activity of mitochondrial complex enzymes. In contrast, rutin treatment significantly enhanced the activity of antioxidant enzymes while concurrently reducing lipid peroxidation levels in rats. Specifically, rutin administration exerted a modulatory effect on the inflammatory response triggered by aluminum exposure, effectively suppressing the excessive production of nitrate and TNF-α. These findings highlight the potential of rutin as an effective therapeutic strategy in mitigating and combating neuro-inflammation and oxidative stress associated with aluminum-induced toxicity, thereby effectively restoring mitochondrial function.
Collapse
Affiliation(s)
- Khadidja Kessas
- Laboratory of Biotoxicology Experimentale, Biodepollution and Phytoremediation, Faculty of Life and Natural Sciences, University Oran1 ABB, Oran, 31100, Algeria.
| | - Wafaa Lounis
- Laboratory of Biotoxicology Experimentale, Biodepollution and Phytoremediation, Faculty of Life and Natural Sciences, University Oran1 ABB, Oran, 31100, Algeria
| | - Zehor Chouari
- Laboratory of Biotoxicology Experimentale, Biodepollution and Phytoremediation, Faculty of Life and Natural Sciences, University Oran1 ABB, Oran, 31100, Algeria
| | - Anne Vejux
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, University Bourgogne Franche-Comté, 21000, Dijon, France
| | - Gérard Lizard
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, University Bourgogne Franche-Comté, 21000, Dijon, France
| | - Omar Kharoubi
- Laboratory of Biotoxicology Experimentale, Biodepollution and Phytoremediation, Faculty of Life and Natural Sciences, University Oran1 ABB, Oran, 31100, Algeria
| |
Collapse
|
27
|
He WC, Hou SL, Wang KB, Xu N, Li K, Xiong T, Luo J. Treadmill running on neuropathic pain: via modulation of neuroinflammation. Front Mol Neurosci 2024; 17:1345864. [PMID: 38989156 PMCID: PMC11233809 DOI: 10.3389/fnmol.2024.1345864] [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: 11/28/2023] [Accepted: 05/24/2024] [Indexed: 07/12/2024] Open
Abstract
Neuropathic pain is a type of chronic pain caused by an injury or somatosensory nervous system disease. Drugs and exercise could effectively relieve neuropathic pain, but no treatment can completely stop neuropathic pain. The integration of exercise into neuropathic pain management has attracted considerable interest in recent years, and treadmill training is the most used among exercise therapies. Neuropathic pain can be effectively treated if its mechanism is clarified. In recent years, the association between neuroinflammation and neuropathic pain has been explored. Neuroinflammation can trigger proinflammatory cytokines, activate microglia, inhibit descending pain modulatory systems, and promote the overexpression of brain-derived neurotrophic factor, which lead to the generation of neuropathic pain and hypersensitivity. Treadmill exercise can alleviate neuropathic pain mainly by regulating neuroinflammation, including inhibiting the activity of pro-inflammatory factors and over activation of microglia in the dorsal horn, regulating the expression of mu opioid receptor expression in the rostral ventromedial medulla and levels of γ-aminobutyric acid to activate the descending pain modulatory system and the overexpression of brain-derived neurotrophic factor. This article reviews and summarizes research on the effect of treadmill exercise on neuropathic pain and its role in the regulation of neuroinflammation to explore its benefits for neuropathic pain treatment.
Collapse
Affiliation(s)
- Wei-Chun He
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
| | - Shuang-Long Hou
- Department of Sport Rehabilitation, Xi’an Physical Education University, Xi’an, China
| | - Kai-Bin Wang
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
| | - Ning Xu
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
| | - Ke Li
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
| | - Ting Xiong
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
| | - Jing Luo
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
- Department of Sport Rehabilitation, Xi’an Physical Education University, Xi’an, China
| |
Collapse
|
28
|
Eghbali F, Dehkordi HT, Amini-Khoei H, Lorigooini Z, Rahimi-Madiseh M. The potential role of nitric oxide in the anticonvulsant effects of betulin in pentylenetetrazole (PTZ)-induced seizures in mice. IBRO Neurosci Rep 2024; 16:527-534. [PMID: 38706971 PMCID: PMC11068554 DOI: 10.1016/j.ibneur.2024.04.003] [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: 02/03/2024] [Revised: 04/03/2024] [Accepted: 04/13/2024] [Indexed: 05/07/2024] Open
Abstract
Epilepsy poses a significant challenge, especially for drug-resistant cases, necessitating novel treatment avenues. This study explores the potential interplay between nitric oxide (NO) and the anticonvulsant effects of betulin, a triterpene with promising neuroprotective properties. While betulin exhibits anticonvulsant effects, the specific involvement of NO remains inadequately understood, constituting a pivotal gap in current knowledge. One hundred NMRI mice were randomly assigned to diverse treatment groups, with seizures induced by pentylenetetrazol (PTZ). Parameters such as seizure threshold, nitrite levels, total antioxidant capacity (TAC), malondialdehyde (MDA) levels, and iNOS/nNOS gene expressions were assessed. Betulin significantly increased seizure thresholds and mitigated PTZ-induced NO levels. These findings suggest a potential modulation of NO-related pathways, emphasizing betulin's anti-inflammatory and antioxidant attributes. The study sheds light on betulin's multifaceted impact on oxidative stress, NO regulation, and iNOS/nNOS gene expressions. The ability of betulin to suppress iNOS/nNOS gene expressions, leading to reduce NO production, underscores its potential as an anticonvulsant.
Collapse
Affiliation(s)
- Fatemeh Eghbali
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hossein Tahmasebi Dehkordi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Rahimi-Madiseh
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
29
|
Galindo AN, Frey Rubio DA, Hettiaratchi MH. Biomaterial strategies for regulating the neuroinflammatory response. MATERIALS ADVANCES 2024; 5:4025-4054. [PMID: 38774837 PMCID: PMC11103561 DOI: 10.1039/d3ma00736g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/07/2024] [Indexed: 05/24/2024]
Abstract
Injury and disease in the central nervous system (CNS) can result in a dysregulated inflammatory environment that inhibits the repair of functional tissue. Biomaterials present a promising approach to tackle this complex inhibitory environment and modulate the mechanisms involved in neuroinflammation to halt the progression of secondary injury and promote the repair of functional tissue. In this review, we will cover recent advances in biomaterial strategies, including nanoparticles, hydrogels, implantable scaffolds, and neural probe coatings, that have been used to modulate the innate immune response to injury and disease within the CNS. The stages of inflammation following CNS injury and the main inflammatory contributors involved in common neurodegenerative diseases will be discussed, as understanding the inflammatory response to injury and disease is critical for identifying therapeutic targets and designing effective biomaterial-based treatment strategies. Biomaterials and novel composites will then be discussed with an emphasis on strategies that deliver immunomodulatory agents or utilize cell-material interactions to modulate inflammation and promote functional tissue repair. We will explore the application of these biomaterial-based strategies in the context of nanoparticle- and hydrogel-mediated delivery of small molecule drugs and therapeutic proteins to inflamed nervous tissue, implantation of hydrogels and scaffolds to modulate immune cell behavior and guide axon elongation, and neural probe coatings to mitigate glial scarring and enhance signaling at the tissue-device interface. Finally, we will present a future outlook on the growing role of biomaterial-based strategies for immunomodulation in regenerative medicine and neuroengineering applications in the CNS.
Collapse
Affiliation(s)
- Alycia N Galindo
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon Eugene OR USA
| | - David A Frey Rubio
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon Eugene OR USA
| | - Marian H Hettiaratchi
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon Eugene OR USA
- Department of Chemistry and Biochemistry, University of Oregon Eugene OR USA
| |
Collapse
|
30
|
AlRuwaili R, Al-Kuraishy HM, Alruwaili M, Khalifa AK, Alexiou A, Papadakis M, Saad HM, Batiha GES. The potential therapeutic effect of phosphodiesterase 5 inhibitors in the acute ischemic stroke (AIS). Mol Cell Biochem 2024; 479:1267-1278. [PMID: 37395897 PMCID: PMC11116240 DOI: 10.1007/s11010-023-04793-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/16/2023] [Indexed: 07/04/2023]
Abstract
Acute ischemic stroke (AIS) is a focal neurological disorder that accounts for 85% of all stroke types, due to occlusion of cerebral arteries by thrombosis and emboli. AIS is also developed due to cerebral hemodynamic abnormality. AIS is associated with the development of neuroinflammation which increases the severity of AIS. Phosphodiesterase enzyme (PDEs) inhibitors have neuro-restorative and neuroprotective effects against the development of AIS through modulation of the cerebral cyclic adenosine monophosphate (cAMP)/cyclic guanosine monophosphate (cGMP)/nitric oxide (NO) pathway. PDE5 inhibitors through mitigation of neuroinflammation may decrease the risk of long-term AIS-induced complications. PDE5 inhibitors may affect the hemodynamic properties and coagulation pathway which are associated with thrombotic complications in AIS. PDE5 inhibitors reduce activation of the pro-coagulant pathway and improve the microcirculatory level in patients with hemodynamic disturbances in AIS. PDE5 inhibitors mainly tadalafil and sildenafil improve clinical outcomes in AIS patients through the regulation of cerebral perfusion and cerebral blood flow (CBF). PDE5 inhibitors reduced thrombomodulin, P-selectin, and tissue plasminogen activator. Herein, PDE5 inhibitors may reduce activation of the pro-coagulant pathway and improve the microcirculatory level in patients with hemodynamic disturbances in AIS. In conclusion, PDE5 inhibitors may have potential roles in the management of AIS through modulation of CBF, cAMP/cGMP/NO pathway, neuroinflammation, and inflammatory signaling pathways. Preclinical and clinical studies are recommended in this regard.
Collapse
Affiliation(s)
- Raed AlRuwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Amira Karam Khalifa
- Department of Medical Pharmacology, Kasr El-Ainy School of Medicine, Cairo University, El Manial, Cairo, 11562, Egypt
- Lecturer of Medical Pharmacology, Nahda Faculty of Medicine, Beni Suef, Egypt
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matrouh, 51744, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
31
|
Belenichev I, Popazova O, Bukhtiyarova N, Savchenko D, Oksenych V, Kamyshnyi O. Modulating Nitric Oxide: Implications for Cytotoxicity and Cytoprotection. Antioxidants (Basel) 2024; 13:504. [PMID: 38790609 PMCID: PMC11118938 DOI: 10.3390/antiox13050504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/26/2024] Open
Abstract
Despite the significant progress in the fields of biology, physiology, molecular medicine, and pharmacology; the designation of the properties of nitrogen monoxide in the regulation of life-supporting functions of the organism; and numerous works devoted to this molecule, there are still many open questions in this field. It is widely accepted that nitric oxide (•NO) is a unique molecule that, despite its extremely simple structure, has a wide range of functions in the body, including the cardiovascular system, the central nervous system (CNS), reproduction, the endocrine system, respiration, digestion, etc. Here, we systematize the properties of •NO, contributing in conditions of physiological norms, as well as in various pathological processes, to the mechanisms of cytoprotection and cytodestruction. Current experimental and clinical studies are contradictory in describing the role of •NO in the pathogenesis of many diseases of the cardiovascular system and CNS. We describe the mechanisms of cytoprotective action of •NO associated with the regulation of the expression of antiapoptotic and chaperone proteins and the regulation of mitochondrial function. The most prominent mechanisms of cytodestruction-the initiation of nitrosative and oxidative stresses, the production of reactive oxygen and nitrogen species, and participation in apoptosis and mitosis. The role of •NO in the formation of endothelial and mitochondrial dysfunction is also considered. Moreover, we focus on the various ways of pharmacological modulation in the nitroxidergic system that allow for a decrease in the cytodestructive mechanisms of •NO and increase cytoprotective ones.
Collapse
Affiliation(s)
- Igor Belenichev
- Department of Pharmacology and Medical Formulation with Course of Normal Physiology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Olena Popazova
- Department of Histology, Cytology and Embryology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Nina Bukhtiyarova
- Department of Clinical Laboratory Diagnostics, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Dmytro Savchenko
- Department of Pharmacy and Industrial Drug Technology, Bogomolets National Medical University, 01601 Kyiv, 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
|
32
|
Silva BD, Viero FT, Rodrigues P, Trevisan G. Nitric oxide involvement in the disability and active disease of multiple sclerosis: Systematic review and meta-analysis. Nitric Oxide 2024; 145:8-20. [PMID: 38331311 DOI: 10.1016/j.niox.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/27/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic and immune-mediated disease of unknown etiology and leading to a physical and cognitive disability. Different studies suggest that nitrosative stress may play a pivotal role in the pathogenesis and disability in MS. Besides, reports evaluated NO and their metabolites, expressed by nitrite and nitrate (NOx) levels of MS patients compared with other pathologies, but did not evaluate disability and relapse/remission phases. OBJECTIVE Thus, this study aimed to conduct a systematic review and meta-analysis of NOx levels in MS patients in relapse/remission phases and its involvement in patient disability. METHODS The protocol was registered in PROSPERO (CRD42022327161). We used GRADE to estimate the articles' quality and evaluated the publication bias using Egger's and Begg's tests. RESULTS Here, through a search in the Pubmed, Scopus, and EMBASE databases, 5.276 studies were found, and after the selection process, 20 studies were included in this systematic review and meta-analysis. The studies included data from 1.474 MS patients and 1.717 healthy controls, 1.010 RRMS and 221 primary progressive MS (PPMS). CONCLUSION NOx levels are increased in relapsing-remitting MS (RRMS) patients in the relapse phase. Also, NOx levels were increased in MS patients with higher disability. However, further studies are still needed to control lifestyle habits, pain, and MS treatment effects in biased NOx levels.
Collapse
Affiliation(s)
- Brenda da Silva
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), 97105-900, Santa Maria, RS, Brazil.
| | - Fernanda Tibolla Viero
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), 97105-900, Santa Maria, RS, Brazil.
| | - Patrícia Rodrigues
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), 97105-900, Santa Maria, RS, Brazil.
| | - Gabriela Trevisan
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
33
|
Lee JY, Kim JH, Choi JM, Noh BW, Kim HY, Cho EJ. Anti-Inflammatory Effects of Artemisia argyi H. Fermented by Lactobacillus plantarum in the LPS-Induced RAW 264.7 Cells and DSS-Induced Colitis Model. Foods 2024; 13:998. [PMID: 38611304 PMCID: PMC11011819 DOI: 10.3390/foods13070998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Ulcerative colitis is a chronic inflammatory disease caused by abnormal immune responses in the intestinal mucosa and gut microorganisms. Unlike other mugworts, Artemisia argyi H. (A. argyi H.) enhances antioxidant, anti-inflammatory, and anticancer effects, but the improvement effects against gut inflammation have not yet been reported. Therefore, this study aimed to confirm the alleviation of the inflammatory state in the gut by A. argyi H. fermented with Lactobacillus plantarum (FAA), using lipopolysaccharide (LPS)-induced RAW 264.7 cells and dextran sulfate sodium (DSS)-induced colitis models. In vitro, FAA (10, 50, 100, and 200 μg/mL) was pretreated into RAW 264.7 cells, followed with LPS (100 ng/mL), which induced the cell damage. Meanwhile, in vivo, FAA (100, 200 mg/kg/day) was orally administered into 6-week-old C57BL/6N mice for 3 weeks. During the last week of FAA administration, 2.5% DSS was used to induce colitis. The results showed that FAA reduced the production of nitric oxide (p < 0.0001), tumor necrosis factor (TNF)-α, interleukin (IL)-6 (p < 0.0001), and IL-1β (p < 0.0001) in the LPS-induced RAW 264.7 cells. Moreover, in the DSS-induced colitis model, FAA alleviated clinical symptoms (p < 0.001), inhibited the inflammatory state by reducing the production of TNF-α (p < 0.0001) and interferon-γ in intestinal immune cells (p < 0.0001), and strengthened the intestinal barrier by increasing the number of goblet cells (p < 0.0001). Furthermore, the anti-inflammatory effects were confirmed by the alleviation of histological damage (p < 0.001) and down-regulation of the expression of inflammatory proteins (TLR4, p < 0.0001; MyD88, p < 0.0001; Cox-2, p < 0.0001). These results suggest the potential of FAA as a dietary ingredient for preventing inflammation in the gut.
Collapse
Affiliation(s)
- Ji Yun Lee
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Republic of Korea; (J.Y.L.); (J.-H.K.); (B.W.N.)
| | - Ji-Hyun Kim
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Republic of Korea; (J.Y.L.); (J.-H.K.); (B.W.N.)
| | - Ji Myung Choi
- Department of Food and Nutrition, Kyungsung University, Busan 48434, Republic of Korea;
- Department of Food Science and Nutrition, Gyeongsang National University, Jinju 52725, Republic of Korea
| | - Byeong Wook Noh
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Republic of Korea; (J.Y.L.); (J.-H.K.); (B.W.N.)
| | - Hyun Young Kim
- Department of Food Science and Nutrition, Gyeongsang National University, Jinju 52725, Republic of Korea
| | - Eun Ju Cho
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Republic of Korea; (J.Y.L.); (J.-H.K.); (B.W.N.)
| |
Collapse
|
34
|
Xia J, Dong S, Yang L, Wang F, Xing S, Du J, Li Z. Design, synthesis, and biological evaluation of novel tryptanthrin derivatives as selective acetylcholinesterase inhibitors for the treatment of Alzheimer's disease. Bioorg Chem 2024; 143:106980. [PMID: 38006789 DOI: 10.1016/j.bioorg.2023.106980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 11/27/2023]
Abstract
Two novel series of tryptanthrin (TRYP) derivatives were designed and synthesized as multifunctional agents for the treatment of Alzheimer's disease (AD). Inhibition assay against cholinesterase (ChE) indicated that these derivatives can act as acetylcholinesterase (AChE) inhibitors with selectivity over butyrylcholinesterase (BuChE). Among them, n1 exhibited the most excellent ChE inhibitory potency (AChE, IC50 = 12.17 ± 1.50 nM; BuChE, IC50 = 6.29 ± 0.48 μΜ; selectivity index = 517). Molecular docking studies indicated that compound n1 can interact with amino acid residues in the catalytic active site and peripheral anionic site of AChE and the molecular dynamics (MD) simulation studies demonstrated that the AChE-n1 complex had good stability. N1 also exhibited anti-amyloid-β (Aβ) aggregation (63.48 % ± 1.02 %, 100 μΜ) and anti-neuroinflammation activity (NO, IL-1β, TNF-α; IC50 = 2.13 ± 0.54 μΜ, 2.21 ± 0.37 μΜ, 2.47 ± 0.07 μΜ, respectively), and n1 had neuroprotective and metal-chelating properties. Further studies indicated n1 had proper blood-brain barrier permeability in the Parallel artificial membrane permeation assay. In vivo studies found that n1 effectively improved learning and memory impairment in scopolamine-induced AD mouse models. Nissl staining ofmice hippocampaltissue sections revealed that n1 restored neuronal cells in the hippocampus CA3 and CA1 regions. These findings suggested that n1 can be a promising compound for further development of multifunctional agents for AD treatment.
Collapse
Affiliation(s)
- Jucheng Xia
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, PR China
| | - Shuanghong Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, PR China
| | - Lili Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, PR China
| | - Fang Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, PR China
| | - Siqi Xing
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, PR China
| | - Jiyu Du
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, PR China
| | - Zeng Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, PR China.
| |
Collapse
|
35
|
Bartra C, Yuan Y, Vuraić K, Valdés-Quiroz H, Garcia-Baucells P, Slevin M, Pastorello Y, Suñol C, Sanfeliu C. Resveratrol Activates Antioxidant Protective Mechanisms in Cellular Models of Alzheimer's Disease Inflammation. Antioxidants (Basel) 2024; 13:177. [PMID: 38397775 PMCID: PMC10886200 DOI: 10.3390/antiox13020177] [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: 12/14/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Resveratrol is a natural phenolic compound with known benefits against neurodegeneration. We analyzed in vitro the protective mechanisms of resveratrol against the proinflammatory monomeric C-reactive protein (mCRP). mCRP increases the risk of AD after stroke and we previously demonstrated that intracerebral mCRP induces AD-like dementia in mice. Here, we used BV2 microglia treated with mCRP for 24 h in the presence or absence of resveratrol. Cells and conditioned media were collected for analysis. Lipopolysaccharide (LPS) has also been implicated in AD progression and so LPS was used as a resveratrol-sensitive reference agent. mCRP at the concentration of 50 µg/mL activated the nitric oxide pathway and the NLRP3 inflammasome pathway. Furthermore, mCRP induced cyclooxygenase-2 and the release of proinflammatory cytokines. Resveratrol effectively inhibited these changes and increased the expression of the antioxidant enzyme genes Cat and Sod2. As central mechanisms of defense, resveratrol activated the hub genes Sirt1 and Nfe2l2 and inhibited the nuclear translocation of the signal transducer NF-ĸB. Proinflammatory changes induced by mCRP in primary mixed glial cultures were also protected by resveratrol. This work provides a mechanistic insight into the protective benefits of resveratrol in preventing the risk of AD induced by proinflammatory agents.
Collapse
Affiliation(s)
- Clara Bartra
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (DIBAPS), 08036 Barcelona, Spain
- PhD Program in Biotechnology, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08034 Barcelona, Spain
| | - Yi Yuan
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
| | - Kristijan Vuraić
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
| | - Haydeé Valdés-Quiroz
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
| | - Pau Garcia-Baucells
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
| | - Mark Slevin
- School of Life Sciences, John Dalton Building, Manchester Metropolitan University, Manchester M15 6BH, UK;
- Centru Avansat de Cercetari Medicale si Farmaceutice (CCAMF), Universitatea de Medicina, Farmacie, Stiinte si Tehnologie “George Emil Palade” din Targu Mures, 540142 Targu Mures, Romania
| | - Ylenia Pastorello
- Department of Anatomy and Embryology, Universitatea de Medicina, Farmacie, Stiinte si Tehnologie “George Emil Palade” din Targu Mures, 540142 Targu Mures, Romania;
| | - Cristina Suñol
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (DIBAPS), 08036 Barcelona, Spain
| | - Coral Sanfeliu
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (DIBAPS), 08036 Barcelona, Spain
| |
Collapse
|
36
|
Wang R, Zeng J, Chen L, Sun L, Wang Y, Xu J, He X. Diterpenoid WT-29 isolated from Wedelia exerted anti-inflammatory and anti-allergic activities. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117265. [PMID: 37783409 DOI: 10.1016/j.jep.2023.117265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/23/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wedelia (Sphagneticola trilobata) is a traditional anti-inflammatory herb native to tropical America. It is commonly used to treat some inflammatory related diseases clinically, such as pertussis, pharyngitis, etc. However, its specific anti-inflammatory mechanism is still unclear. AIM OF THE STUDY WT-29 (3α-angeloyloxy-9β-hydroxyent-kaura-16-en-19-oic acid) is a main bioactive diterpenoid isolated and purified from Wedelia. This study aims to explore the potential anti-inflammatory and anti-allergic properties of WT-29 on RAW264.7 cells stimulated with LPS and P815 cells induced by C48/80, as well as investigating their underlying molecular mechanisms. METHODS The anti-inflammatory mechanism of WT-29 was analyzed and predicted using network pharmacology, and then verified through experiments. The Griess reagent assay was employed to evaluate the impact of WT-29 on the generation of nitric oxide (NO) in RAW264.7 cells induced by LPS, the expression of various inflammatory cytokines and the release of histamine in cells were measured through qRT-PCR and ELISA techniques. The impact of WT-29 on the translocation of the NF-κB p65 protein to the nucleus was assessed through immunofluorescence staining. Western blot technique was utilized to investigate protein expression in inflammation, allergy, and autophagy pathways. RESULTS The study found that WT-29 can reduce the secretion of inflammatory factors (NO, iNOS, COX-2, IL-6, IL-1β and TNF-α), inhibit NF-κB activation and MAPK family phosphorylation, and induce autophagy in RAW264.7 cells stimulated with LPS. In addition, it demonstrated that WT-29 could inhibit histamine release and degranulation, as well as inhibit the MAPK family in C48/80-induced P815 cells. CONCLUSION WT-29 isolated from Wedelia exerts anti-inflammatory and anti-allergic effects mainly through NF-κB, Nrf2/Keap-1, MAPK pathways and regulating of autophagy, suggesting that it might be a potential anti-inflammatory and anti-allergic agent and could be used as medicine or health benefit product.
Collapse
Affiliation(s)
- Ru Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou, 510006, China.
| | - Jia Zeng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Lu Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou, 510006, China.
| | - Lianlian Sun
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou, 510006, China.
| | - Yihai Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou, 510006, China.
| | - Jingwen Xu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou, 510006, China.
| | - Xiangjiu He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou, 510006, China.
| |
Collapse
|
37
|
Räuber S, Förster M, Schüller J, Willison A, Golombeck KS, Schroeter CB, Oeztuerk M, Jansen R, Huntemann N, Nelke C, Korsen M, Fischer K, Kerkhoff R, Leven Y, Kirschner P, Kölsche T, Nikolov P, Mehsin M, Marae G, Kokott A, Pul D, Schulten J, Vogel N, Ingwersen J, Ruck T, Pawlitzki M, Meuth SG, Melzer N, Kremer D. The Use of Nitrosative Stress Molecules as Potential Diagnostic Biomarkers in Multiple Sclerosis. Int J Mol Sci 2024; 25:787. [PMID: 38255863 PMCID: PMC10815836 DOI: 10.3390/ijms25020787] [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: 12/02/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) of still unclear etiology. In recent years, the search for biomarkers facilitating its diagnosis, prognosis, therapy response, and other parameters has gained increasing attention. In this regard, in a previous meta-analysis comprising 22 studies, we found that MS is associated with higher nitrite/nitrate (NOx) levels in the cerebrospinal fluid (CSF) compared to patients with non-inflammatory other neurological diseases (NIOND). However, many of the included studies did not distinguish between the different clinical subtypes of MS, included pre-treated patients, and inclusion criteria varied. As a follow-up to our meta-analysis, we therefore aimed to analyze the serum and CSF NOx levels in clinically well-defined cohorts of treatment-naïve MS patients compared to patients with somatic symptom disorder. To this end, we analyzed the serum and CSF levels of NOx in 117 patients (71 relapsing-remitting (RR) MS, 16 primary progressive (PP) MS, and 30 somatic symptom disorder). We found that RRMS and PPMS patients had higher serum NOx levels compared to somatic symptom disorder patients. This difference remained significant in the subgroup of MRZ-negative RRMS patients. In conclusion, the measurement of NOx in the serum might indeed be a valuable tool in supporting MS diagnosis.
Collapse
Affiliation(s)
- Saskia Räuber
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Moritz Förster
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
- Department of Neurology, Kliniken Maria Hilf GmbH, Academic Teaching Hospital of the RWTH Aachen University Hospital, 41063 Moenchengladbach, Germany
| | - Julia Schüller
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Alice Willison
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Kristin S. Golombeck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Christina B. Schroeter
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Menekse Oeztuerk
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Robin Jansen
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Niklas Huntemann
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Christopher Nelke
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Melanie Korsen
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Katinka Fischer
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Ruth Kerkhoff
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
- Department of Neurology, Kliniken Maria Hilf GmbH, Academic Teaching Hospital of the RWTH Aachen University Hospital, 41063 Moenchengladbach, Germany
| | - Yana Leven
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Patricia Kirschner
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Tristan Kölsche
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Petyo Nikolov
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Mohammed Mehsin
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Gelenar Marae
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Alma Kokott
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Duygu Pul
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Julius Schulten
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Niklas Vogel
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Jens Ingwersen
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Tobias Ruck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Sven G. Meuth
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Nico Melzer
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - David Kremer
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
- Department of Neurology and Neurorehabilitation, Hospital Zum Heiligen Geist, Academic Teaching Hospital of the Heinrich Heine University Düsseldorf, 47906 Kempen, Germany
| |
Collapse
|
38
|
Serrano-Marín J, Marin S, Bernal-Casas D, Lillo A, González-Subías M, Navarro G, Cascante M, Sánchez-Navés J, Franco R. A metabolomics study in aqueous humor discloses altered arginine metabolism in Parkinson's disease. Fluids Barriers CNS 2023; 20:90. [PMID: 38049870 PMCID: PMC10696737 DOI: 10.1186/s12987-023-00494-5] [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: 07/27/2023] [Accepted: 11/23/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND The lack of accessible and informative biomarkers results in a delayed diagnosis of Parkinson's disease (PD), whose symptoms appear when a significant number of dopaminergic neurons have already disappeared. The retina, a historically overlooked part of the central nervous system (CNS), has gained recent attention. It has been discovered that the composition of cerebrospinal fluid influences the aqueous humor composition through microfluidic circulation. In addition, alterations found in the brain of patients with PD have a correlate in the retina. This new paradigm highlights the potential of the aqueous humor as a sample for identifying differentially concentrated metabolites that could, eventually, become biomarkers if also found altered in blood or CSF of patients. In this research we aim at analyzing the composition of the aqueous humor from healthy controls and PD patients. METHODS A targeted metabolomics approach with concentration determination by mass spectrometry was used. Statistical methods including principal component analysis and linear discriminants were used to select differentially concentrated metabolites that allow distinguishing patients from controls. RESULTS In this first metabolomics study in the aqueous humor of PD patients, elevated levels of 16 compounds were found; molecules differentially concentrated grouped into biogenic amines, amino acids, and acylcarnitines. A biogenic amine, putrescine, alone could be a metabolite capable of differentiating between PD and control samples. The altered levels of the metabolites were correlated, suggesting that the elevations stem from a common mechanism involving arginine metabolism. CONCLUSIONS A combination of three metabolites, putrescine, tyrosine, and carnitine was able to correctly classify healthy participants from PD patients. Altered metabolite levels suggest altered arginine metabolism. The pattern of metabolomic disturbances was not due to the levodopa-based dopamine replacement medication because one of the patients was not yet taking levodopa but a dopamine receptor agonist.
Collapse
Affiliation(s)
- Joan Serrano-Marín
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine of University of Barcelona (IBUB), University of Barcelona (UB), Barcelona, 08028, Spain
- CIBEREHD. Network Center for Hepatic and Digestive Diseases, National Spanish Health Institute Carlos III (ISCIII), Madrid, 28029, Spain
| | - David Bernal-Casas
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, 08028, Spain
| | - Alejandro Lillo
- Department of Biochemistry and Physiology, Universitat de Barcelona, Barcelona, Spain
| | - Marc González-Subías
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Navarro
- Department of Biochemistry and Physiology, Universitat de Barcelona, Barcelona, Spain
- CiberNed. Network Center for Biomedical Research in Neurodegenerative Diseases., Spanish National Health Institute Carlos iii, Av. Monforte de Lemos, 3-5, Madrid, 28029, Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine of University of Barcelona (IBUB), University of Barcelona (UB), Barcelona, 08028, Spain
- CIBEREHD. Network Center for Hepatic and Digestive Diseases, National Spanish Health Institute Carlos III (ISCIII), Madrid, 28029, Spain
| | - Juan Sánchez-Navés
- Department of Ophthalmology, Ophthalmedic and I.P.O. Institute of Ophthalmology, Palma de Mallorca, Spain
| | - Rafael Franco
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.
- CiberNed. Network Center for Biomedical Research in Neurodegenerative Diseases., Spanish National Health Institute Carlos iii, Av. Monforte de Lemos, 3-5, Madrid, 28029, Spain.
- School of Chemistry, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
39
|
Han YH, Liu XD, Jin MH, Sun HN, Kwon T. Role of NLRP3 inflammasome-mediated neuronal pyroptosis and neuroinflammation in neurodegenerative diseases. Inflamm Res 2023; 72:1839-1859. [PMID: 37725102 DOI: 10.1007/s00011-023-01790-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Neurodegenerative diseases are a common group of neurological disorders characterized by progressive loss of neuronal structure and function leading to cognitive impairment. Recent studies have shown that neuronal pyroptosis mediated by the NLRP3 inflammasome plays a crucial role in the pathogenesis of neurodegenerative diseases. OBJECTIVE AND METHOD The NLRP3 inflammasome is a multiprotein complex that, when activated within cells, triggers an inflammatory response, ultimately leading to pyroptotic cell death of neurons. Pyroptosis is a typical pro-inflammatory programmed cell death process occurring downstream of NLRP3 inflammasome activation, characterized by the formation of pores on the cell membrane by the GSDMD protein, leading to cell lysis and the release of inflammatory factors. It has been found that NLRP3 inflammasome-mediated neuronal pyroptosis is closely associated with the development of various neurodegenerative diseases, such as Alzheimer's disease, traumatic brain injury, and Parkinson's disease. Therefore, inhibiting NLRP3 inflammasome activation and attenuating neuronal pyroptosis could potentially serve as novel strategies for the treatment of neurodegenerative diseases. RESULTS The aim of this review is to explore the role of NLRP3 activation-mediated neuronal pyroptosis and neuroinflammation in neurodegenerative diseases. Firstly, we extensively discuss the relationship between NLRP3 inflammasome-mediated neuronal pyroptosis and neuroinflammation in various neurodegenerative diseases. Subsequently, we further explore the mechanisms driving NLRP3 activation and assembly, as well as the post-translational modifications regulating NLRP3 inflammasome activation. CONCLUSION Understanding these mechanisms will contribute to a deeper understanding of the link between neuronal pyroptosis and neurodegenerative diseases, and hold significant implications for the treatment and prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ying-Hao Han
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
| | - Xiao-Dong Liu
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Mei-Hua Jin
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Hu-Nan Sun
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup-si, Jeonbuk, 56216, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
40
|
Wang L, Dan Q, Xu B, Chen Y, Zheng T. Research progress on gas signal molecular therapy for Parkinson's disease. Open Life Sci 2023; 18:20220658. [PMID: 37588999 PMCID: PMC10426759 DOI: 10.1515/biol-2022-0658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/22/2023] [Accepted: 06/14/2023] [Indexed: 08/18/2023] Open
Abstract
The pathogenesis of Parkinson's disease (PD) remains unclear. Among the pathological manifestations is the progressive degeneration of the nigrostriatal dopaminergic pathway, leading to massive loss of neurons in the substantia nigra pars compacta and dopamine (DA) depletion. Therefore, the current drug treatment is primarily based on DA supplementation and delaying the progression of the disease. However, as patients' symptoms continue to worsen, the drug effect will gradually decrease or even disappear, thereby further aggravating clinical symptoms. Gas signaling molecules, such as hydrogen sulfide (H2S), nitric oxide (NO), carbon monoxide (CO), and hydrogen (H2), exhibit pleiotropic biological functions and play crucial roles in physiological and pathological effects. In common neurodegenerative diseases including Alzheimer's disease and PD, gas signal molecules can prevent or delay disease occurrence via the primary mechanisms of antioxidation, anti-inflammatory response, and antiapoptosis. This article reviews the therapeutic progress of gas signaling molecules in PD models and discusses the possibility of their clinical applications.
Collapse
Affiliation(s)
- Linlin Wang
- Department of Hubei University of Medicine, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen518036, P. R. China
| | - Qing Dan
- Department of Hubei University of Medicine, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen518036, P. R. China
| | - Bingxuan Xu
- Department of Hubei University of Medicine, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen518036, P. R. China
| | - Yun Chen
- Department of Hubei University of Medicine, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen518036, P. R. China
| | - Tingting Zheng
- Department of Hubei University of Medicine, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen518036, P. R. China
| |
Collapse
|
41
|
Vasanthi SS, Rao NS, Samidurai M, Massey N, Meyer C, Gage M, Kharate M, Almanza A, Wachter L, Mafuta C, Trevino L, Carlo AM, Bryant E, Corson BE, Wohlgemuth M, Ostrander M, Showman L, Wang C, Thippeswamy T. Disease-modifying effects of a glial-targeted inducible nitric oxide synthase inhibitor (1400W) in mixed-sex cohorts of a rat soman (GD) model of epilepsy. J Neuroinflammation 2023; 20:163. [PMID: 37438764 PMCID: PMC10337207 DOI: 10.1186/s12974-023-02847-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Acute exposure to seizurogenic organophosphate (OP) nerve agents (OPNA) such as diisopropylfluorophosphate (DFP) or soman (GD), at high concentrations, induce immediate status epilepticus (SE), reactive gliosis, neurodegeneration, and epileptogenesis as a consequence. Medical countermeasures (MCMs-atropine, oximes, benzodiazepines), if administered in < 20 min of OPNA exposure, can control acute symptoms and mortality. However, MCMs alone are inadequate to prevent OPNA-induced brain injury and behavioral dysfunction in survivors. We have previously shown that OPNA exposure-induced SE increases the production of inducible nitric oxide synthase (iNOS) in glial cells in both short- and long- terms. Treating with a water soluble and highly selective iNOS inhibitor, 1400W, for 3 days significantly reduced OPNA-induced brain changes in those animals that had mild-moderate SE in the rat DFP model. However, such mitigating effects and the mechanisms of 1400W are unknown in a highly volatile nerve agent GD exposure. METHODS Mixed-sex cohort of adult Sprague Dawley rats were exposed to GD (132 μg/kg, s.c.) and immediately treated with atropine (2 mg/kg, i.m) and HI-6 (125 mg/kg, i.m.). Severity of seizures were quantified for an hour and treated with midazolam (3 mg/kg, i.m.). An hour post-midazolam, 1400W (20 mg/kg, i.m.) or vehicle was administered daily for 2 weeks. After behavioral testing and EEG acquisition, animals were euthanized at 3.5 months post-GD. Brains were processed for neuroinflammatory and neurodegeneration markers. Serum and CSF were used for nitrooxidative and proinflammatory cytokines assays. RESULTS We demonstrate a significant long-term (3.5 months post-soman) disease-modifying effect of 1400W in animals that had severe SE for > 20 min of continuous convulsive seizures. 1400W significantly reduced GD-induced motor and cognitive dysfunction; nitrooxidative stress (nitrite, ROS; increased GSH: GSSG); proinflammatory cytokines in the serum and some in the cerebrospinal fluid (CSF); epileptiform spikes and spontaneously recurring seizures (SRS) in males; reactive gliosis (GFAP + C3 and IBA1 + CD68-positive glia) as a measure of neuroinflammation, and neurodegeneration (especially parvalbumin-positive neurons) in some brain regions. CONCLUSION These findings demonstrate the long-term disease-modifying effects of a glial-targeted iNOS inhibitor, 1400W, in a rat GD model by modulating reactive gliosis, neurodegeneration (parvalbumin-positive neurons), and neuronal hyperexcitability.
Collapse
Affiliation(s)
- Suraj S. Vasanthi
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Nikhil S. Rao
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Manikandan Samidurai
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Nyzil Massey
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Christina Meyer
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Meghan Gage
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Mihir Kharate
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Aida Almanza
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Logan Wachter
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Candide Mafuta
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Lily Trevino
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Adriana M. Carlo
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Elijah Bryant
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Brooke E. Corson
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Morgan Wohlgemuth
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Morgan Ostrander
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Lucas Showman
- W.M. Keck Metabolomics Research Laboratory, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Chong Wang
- Department of Veterinary Diagnostic and Production Animal Medicine and Statistics, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Thimmasettappa Thippeswamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| |
Collapse
|
42
|
Zhang W, Xiao D, Mao Q, Xia H. Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther 2023; 8:267. [PMID: 37433768 PMCID: PMC10336149 DOI: 10.1038/s41392-023-01486-5] [Citation(s) in RCA: 401] [Impact Index Per Article: 200.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 07/13/2023] Open
Abstract
Studies in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and Amyotrophic lateral sclerosis, Huntington's disease, and so on, have suggested that inflammation is not only a result of neurodegeneration but also a crucial player in this process. Protein aggregates which are very common pathological phenomenon in neurodegeneration can induce neuroinflammation which further aggravates protein aggregation and neurodegeneration. Actually, inflammation even happens earlier than protein aggregation. Neuroinflammation induced by genetic variations in CNS cells or by peripheral immune cells may induce protein deposition in some susceptible population. Numerous signaling pathways and a range of CNS cells have been suggested to be involved in the pathogenesis of neurodegeneration, although they are still far from being completely understood. Due to the limited success of traditional treatment methods, blocking or enhancing inflammatory signaling pathways involved in neurodegeneration are considered to be promising strategies for the therapy of neurodegenerative diseases, and many of them have got exciting results in animal models or clinical trials. Some of them, although very few, have been approved by FDA for clinical usage. Here we comprehensively review the factors affecting neuroinflammation and the major inflammatory signaling pathways involved in the pathogenicity of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic lateral sclerosis. We also summarize the current strategies, both in animal models and in the clinic, for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Weifeng Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China
| | - Dan Xiao
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, P.R. China
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Qinwen Mao
- Department of Pathology, University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China.
| |
Collapse
|
43
|
Mutti V, Carini G, Filippini A, Castrezzati S, Giugno L, Gennarelli M, Russo I. LRRK2 Kinase Inhibition Attenuates Neuroinflammation and Cytotoxicity in Animal Models of Alzheimer's and Parkinson's Disease-Related Neuroinflammation. Cells 2023; 12:1799. [PMID: 37443833 PMCID: PMC10340668 DOI: 10.3390/cells12131799] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/22/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Chronic neuroinflammation plays a crucial role in the progression of several neurodegenerative diseases (NDDs), including Parkinson's disease (PD) and Alzheimer's disease (AD). Intriguingly, in the last decade, leucine-rich repeat kinase-2 (LRRK2), a gene mutated in familial and sporadic PD, was revealed as a key mediator of neuroinflammation. Therefore, the anti-inflammatory properties of LRRK2 inhibitors have started to be considered as a disease-modifying treatment for PD; however, to date, there is little evidence on the beneficial effects of targeting LRRK2-related neuroinflammation in preclinical models. In this study, we further validated LRRK2 kinase modulation as a pharmacological intervention in preclinical models of AD- and PD-related neuroinflammation. Specifically, we reported that LRRK2 kinase inhibition with MLi2 and PF-06447475 (PF) molecules attenuated neuroinflammation, gliosis and cytotoxicity in mice with intracerebral injection of Aβ1-42 fibrils or α-syn preformed fibrils (pffs). Moreover, for the first time in vivo, we showed that LRRK2 kinase activity participates in AD-related neuroinflammation and therefore might contribute to AD pathogenesis. Overall, our findings added evidence on the anti-inflammatory effects of LRRK2 kinase inhibition in preclinical models and indicate that targeting LRRK2 activity could be a disease-modifying treatment for NDDs with an inflammatory component.
Collapse
Affiliation(s)
- Veronica Mutti
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (V.M.)
| | - Giulia Carini
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (V.M.)
- Biology and Genetics Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Alice Filippini
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (V.M.)
- Biology and Genetics Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Stefania Castrezzati
- Human Anatomy Unit, Department of Biomedical Sciences and Biotechnologies, University of Brescia, 25123 Brescia, Italy
| | - Lorena Giugno
- Human Anatomy Unit, Department of Biomedical Sciences and Biotechnologies, University of Brescia, 25123 Brescia, Italy
| | - Massimo Gennarelli
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (V.M.)
- Biology and Genetics Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Isabella Russo
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (V.M.)
- Biology and Genetics Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
44
|
Popova NA, Soodaeva SK, Klimanov IA, Misharin VM, Temnov AA. Autoregulation and Autoinhibition of the Main NO Synthase Isoforms (Brief Review). Sovrem Tekhnologii Med 2023; 15:53-59. [PMID: 38435476 PMCID: PMC10904358 DOI: 10.17691/stm2023.15.3.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Indexed: 03/05/2024] Open
Abstract
Nitric oxide (II) (NO) is the most important mediator of a wide range of physiological and pathophysiological processes. It is synthesized by NO synthases (NOSs), which have three main isoforms differing from each other in terms of activation and inhibition features, levels of NO production, subcellular localization, etc. At the same time, all isoforms are structurally very similar, and these differences are determined by NOS autoregulatory elements. The article presents an analysis of the autoregulatory and autoinhibitory mechanisms of the NOS reductase domain that determine differences in the productivity of isoforms, as well as their dependence on the concentration of Ca2+ ions. The main regulatory elements in NOS that modulate the electron transfer from flavin to heme include calmodulin (CaM), an autoinhibitory insert (AI), and the C-terminal tail (C-tail). Hydrophobic interactions of CaM with the surface of the NOS oxidase domain are assumed to facilitate electron transfer from flavin mononucleotide (FMN). CaM binding causes a change in the inter-domain distances, a shift of AI and the C-tail, and, as a result, a decrease in their inhibitory effect. CaM also shifts the conformational equilibrium of the reductase domain towards more open conformations, reduces the lifetime of conformations, their stereometric distribution, and accelerates the flow of electrons through the reductase domain. The AI element, apparently, induces a conformational change that hinders electron transfer within the reductase domain, similar to the hinge domain in cytochrome P450. Together with CaM, the C-tail regulates the electron flow between flavins, the distance and relative orientation of isoalloxane rings, and also modulates the electron flow from FMN to the terminal acceptor. Together with the C-tail, AI also predetermines the dependence of neuronal and endothelial forms of NOS on the concentration of Ca2+ ions, and the C-tail length affects differences in the productivity of NO synthesis. The inhibitory effect of the C-tail is likely to be reduced by CaM binding due to the C-tail shift due to the electrostatic repulsive forces of the negatively charged phosphate and aspartate residues. The autoregulatory elements of NOS require further study, since the mechanisms of their interaction are complex and multidirectional, and hence provide a wide range of characteristics of the observed isoforms.
Collapse
Affiliation(s)
- N A Popova
- Researcher, Laboratory of Clinical and Experimental Biophysics; Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 28 Orekhovy Boulevard, Moscow, 115682, Russia; Researcher, Laboratory of Chemical and Biotechnological Synthesis; Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy Per., Dolgoprudny, Moscow Region, 141701, Russia
| | - S K Soodaeva
- Head of Laboratory of Clinical and Experimental Biophysics; Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 28 Orekhovy Boulevard, Moscow, 115682, Russia; Leading Researcher, Laboratory of Chemical and Biotechnological Synthesis; Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy Per., Dolgoprudny, Moscow Region, 141701, Russia
| | - I A Klimanov
- Senior Researcher, Laboratory of Clinical and Experimental Biophysics; Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 28 Orekhovy Boulevard, Moscow, 115682, Russia
| | - V M Misharin
- Acting Director of the Institute; Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 28 Orekhovy Boulevard, Moscow, 115682, Russia
| | - A A Temnov
- Head of Laboratory of Chemical and Biotechnological Synthesis; Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy Per., Dolgoprudny, Moscow Region, 141701, Russia
| |
Collapse
|
45
|
Liu Y, Chang D, Zhou X. Development of Novel Herbal Compound Formulations Targeting Neuroinflammation: Network Pharmacology, Molecular Docking, and Experimental Verification. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:2558415. [PMID: 37266321 PMCID: PMC10232107 DOI: 10.1155/2023/2558415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/05/2023] [Accepted: 04/20/2023] [Indexed: 06/03/2023]
Abstract
Neuroinflammation plays an important role in the onset and progression of neurodegenerative diseases. The multicomponent and multitarget approach may provide a practical strategy to address the complex pathological mechanisms of neuroinflammation. This study aimed to develop synergistic herbal compound formulas to attenuate neuroinflammation using integrated network pharmacology, molecular docking, and experimental bioassays. Eight phytochemicals with anti-neuroinflammatory potential were selected in the present study. A compound-gene target-signaling pathway network was constructed to illustrate the mechanisms of action of each phytochemical and the interactions among them at the molecular level. Molecular docking was performed to verify the binding affinity of each phytochemical and its key gene targets. An experimental study was conducted to identify synergistic interactions among the eight phytochemicals, and the associated molecular mechanisms were examined by immunoblotting based on the findings from the network pharmacology analysis. Two paired combinations, andrographolide and 6-shogaol (AN-SG) (IC50 = 2.85 μg/mL), and baicalein-6-shogaol (BA-SG) (IC50 = 3.28 μg/mL), were found to synergistically (combination index <1) inhibit the lipopolysaccharides (LPS)-induced nitric oxide production in microglia N11 cells. Network pharmacology analysis suggested that MAPK14, MAPK8, and NOS3 were the top three relevant gene targets for the three phytochemicals, and molecular docking demonstrated strong binding affinities of the phytochemicals to their coded proteins. Immunoblotting suggested that the AN-SG and BA-SG both showed prominent effects in inhibiting inducible nitric oxide synthase (iNOS) (p < 0.01 and p < 0.05, respectively) and MAPKp-p38 (both p < 0.05) compared with those induced by the LPS stimulation only. The AN-SG combination exhibited greater inhibitions of the protein expressions of iNOS (p < 0.05 vs. individual components), which may partly explain the mechanisms of the synergy observed. This study established a practical approach to developing novel herbal-compound formulations using integrated network pharmacology analysis, molecular docking, and experimental bioassays. The study provides a scientific basis and new insight into the two synergistic combinations against neuroinflammation.
Collapse
Affiliation(s)
- Yang Liu
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia
| |
Collapse
|
46
|
Vasanthi SS, Rao NS, Samidurai M, Massey N, Meyer C, Gage M, Kharate M, Almanza A, Wachter L, Mafuta C, Trevino L, Carlo AM, Bryant E, Corson BE, Wohlgemuth M, Ostrander M, Wang C, Thippeswamy T. Disease-Modifying Effects of a Glial-targeted Inducible Nitric Oxide Synthase Inhibitor (1400W) in Mixed-sex Cohorts of a Rat Soman (GD) Model of Epilepsy. RESEARCH SQUARE 2023:rs.3.rs-2883247. [PMID: 37214912 PMCID: PMC10197763 DOI: 10.21203/rs.3.rs-2883247/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Background Acute exposure to seizurogenic organophosphate (OP) nerve agents (OPNA) such as diisopropylfluorophosphate (DFP) or soman (GD), at high concentrations, induce immediate status epilepticus (SE), reactive gliosis, neurodegeneration, and epileptogenesis as a consequence. Medical countermeasures (MCMs- atropine, oximes, benzodiazepines), if administered in < 20 minutes of OPNA exposure, can control acute symptoms and mortality. However, MCMs alone are inadequate to prevent OPNA-induced brain injury and behavioral dysfunction in survivors. We have previously shown that OPNA exposure-induced SE increases the production of inducible nitric oxide synthase (iNOS) in glial cells in both short- and long- terms. Treating with a water soluble and highly selective iNOS inhibitor, 1400W, for three days significantly reduced OPNA-induced brain changes in those animals that had mild-moderate SE in the rat DFP model. However, such mitigating effects and the mechanisms of 1400W are unknown in a highly volatile nerve agent GD exposure. Methods Mixed-sex cohort of adult Sprague Dawley rats were exposed to GD (132µg/kg, s.c.) and immediately treated with atropine (2mg/kg, i.m) and HI-6 (125mg/kg, i.m.). Severity of seizures were quantified for an hour and treated with midazolam (3mg/kg, i.m.). An hour post-midazolam, 1400W (20mg/kg, i.m.) or vehicle was administered daily for two weeks. After behavioral testing and EEG acquisition, animals were euthanized at 3.5 months post-GD. Brains were processed for neuroinflammatory and neurodegeneration markers. Serum and CSF were used for nitrooxidative and proinflammatory cytokines assays. Results We demonstrate a significant long-term (3.5 months post-soman) disease-modifying effect of 1400W in animals that had severe SE for > 20min of continuous convulsive seizures. 1400W significantly reduced GD-induced motor and cognitive dysfunction; nitrooxidative stress (nitrite, ROS; increased GSH: GSSG); proinflammatory cytokines in the serum and some in the cerebrospinal fluid (CSF); epileptiform spikes and spontaneously recurring seizures (SRS) in males; reactive gliosis (GFAP + C3 and IBA1 + CD68 positive glia) as a measure of neuroinflammation, and neurodegeneration (including parvalbumin positive neurons) in some brain regions. Conclusion These findings demonstrate the long-term disease-modifying effects of a glial-targeted iNOS inhibitor, 1400W, in a rat GD model by modulating reactive gliosis, neurodegeneration, and neuronal hyperexcitability.
Collapse
|
47
|
Wong TS, Li G, Li S, Gao W, Chen G, Gan S, Zhang M, Li H, Wu S, Du Y. G protein-coupled receptors in neurodegenerative diseases and psychiatric disorders. Signal Transduct Target Ther 2023; 8:177. [PMID: 37137892 PMCID: PMC10154768 DOI: 10.1038/s41392-023-01427-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Neuropsychiatric disorders are multifactorial disorders with diverse aetiological factors. Identifying treatment targets is challenging because the diseases are resulting from heterogeneous biological, genetic, and environmental factors. Nevertheless, the increasing understanding of G protein-coupled receptor (GPCR) opens a new possibility in drug discovery. Harnessing our knowledge of molecular mechanisms and structural information of GPCRs will be advantageous for developing effective drugs. This review provides an overview of the role of GPCRs in various neurodegenerative and psychiatric diseases. Besides, we highlight the emerging opportunities of novel GPCR targets and address recent progress in GPCR drug development.
Collapse
Affiliation(s)
- Thian-Sze Wong
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Wei Gao
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Shiyi Gan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Manzhan Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China.
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China.
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China.
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, 518116, Shenzhen, Guangdong, China.
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China.
| |
Collapse
|
48
|
de Sousa JAC, Azul FVCS, de Araújo AB, Tomé RC, Silva FRM, de Vasconcelos SMM, Rios FJ, Leal LKAM. Epiisopiloturine, an Alkaloid from Pilocarpus microphyllus, Attenuates LPS-Induced Neuroinflammation by Interfering in the TLR4/NF- κB-MAPK Signaling Pathway in Microglial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:4752502. [PMID: 37151606 PMCID: PMC10162877 DOI: 10.1155/2023/4752502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 03/18/2023] [Indexed: 05/09/2023]
Abstract
Neuroinflammation is present in the pathophysiological mechanisms of several diseases that affect the central nervous system (CNS). Microglia have a prominent role in initiating and sustaining the inflammatory process. Epiisopiloturine (EPI) is an imidazole alkaloid obtained as a by-product of pilocarpine extracted from Pilocarpus microphyllus (jaborandi) and has shown promising anti-inflammatory and antinociceptive properties. In the present study, we investigated the effects of EPI on the inflammatory response in microglial cells (BV-2 cells) induced by lipopolysaccharide (LPS) and explored putative underlying molecular mechanisms. Cell viability was not affected by EPI (1-100 μg/mL) as assessed by both LDH activity and the MTT test. Pretreatment with EPI (25, 50, and 100 μg/mL) significantly reduced the proinflammatory response induced by LPS, as observed by a decrease in nitrite oxide production and iNOS protein expression. EPI (25 μg/mL) reduced IL-6 and TNF-α production, by 40% and 34%, respectively. However, no changes were observed in the anti-inflammatory IL-10 production. Mechanistically, EPI inhibited the TLR4 expression and phosphorylation of NF-κB p65 and MAPKs (JNK and ERK1/2) induced by LPS, but no changes were observed in TREM2 receptor expression in LPS-stimulated cells. In conclusion, our data demonstrated the potent anti-inflammatory properties of EPI in microglial cells. These effects are associated with the reduction of TLR4 expression and inhibition of intracellular signaling cascades, including NF-κB and MAPKs (JNK and ERK1/2).
Collapse
Affiliation(s)
- João Antônio Costa de Sousa
- Center of Cosmetics and Pharmaceutical Studies, CEFAC, Faculty of Pharmacy, Odontology, and Nursing, Department of Pharmacy, Federal University of Ceará, CEFAC, Fortaleza, CE, Brazil
| | - Francisco Vinícius Clemente Serra Azul
- Center of Cosmetics and Pharmaceutical Studies, CEFAC, Faculty of Pharmacy, Odontology, and Nursing, Department of Pharmacy, Federal University of Ceará, CEFAC, Fortaleza, CE, Brazil
| | - Ana Bruna de Araújo
- Center of Cosmetics and Pharmaceutical Studies, CEFAC, Faculty of Pharmacy, Odontology, and Nursing, Department of Pharmacy, Federal University of Ceará, CEFAC, Fortaleza, CE, Brazil
| | - Rebeca Colares Tomé
- Center of Cosmetics and Pharmaceutical Studies, CEFAC, Faculty of Pharmacy, Odontology, and Nursing, Department of Pharmacy, Federal University of Ceará, CEFAC, Fortaleza, CE, Brazil
| | - Francisca Raysse Mesquita Silva
- Center of Cosmetics and Pharmaceutical Studies, CEFAC, Faculty of Pharmacy, Odontology, and Nursing, Department of Pharmacy, Federal University of Ceará, CEFAC, Fortaleza, CE, Brazil
| | | | - Francisco José Rios
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Luzia Kalyne Almeida Moreira Leal
- Center of Cosmetics and Pharmaceutical Studies, CEFAC, Faculty of Pharmacy, Odontology, and Nursing, Department of Pharmacy, Federal University of Ceará, CEFAC, Fortaleza, CE, Brazil
| |
Collapse
|
49
|
Iova OM, Marin GE, Lazar I, Stanescu I, Dogaru G, Nicula CA, Bulboacă AE. Nitric Oxide/Nitric Oxide Synthase System in the Pathogenesis of Neurodegenerative Disorders-An Overview. Antioxidants (Basel) 2023; 12:antiox12030753. [PMID: 36979000 PMCID: PMC10045816 DOI: 10.3390/antiox12030753] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/24/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Nitric oxide, a ubiquitous molecule found throughout the natural world, is a key molecule implicated in many central and benefic molecular pathways and has a well-established role in the function of the central nervous system, as numerous studies have previously shown. Dysregulation of its metabolism, mainly the upregulation of nitric oxide production, has been proposed as a trigger and/or aggravator for many neurological affections. Increasing evidence supports the implication of this molecule in prevalent neurodegenerative diseases, such as Parkinson's disease, Alzheimer's disease, or amyotrophic lateral sclerosis. The mechanisms proposed for its neurotoxicity mainly center around the increased quantities of nitric oxide that are produced in the brain, their cause, and, most importantly, the pathological metabolic cascades created. These cascades lead to the formation of neuronal toxic substances that impair the neurons' function and structure on multiple levels. The purpose of this review is to present the main causes of increased pathological production, as well as the most important pathophysiological mechanisms triggered by nitric oxide, mechanisms that could help explain a part of the complex picture of neurodegenerative diseases and help develop targeted therapies.
Collapse
Affiliation(s)
- Olga-Maria Iova
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Gheorghe-Eduard Marin
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Izabella Lazar
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Stanescu
- Department of Neurology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Gabriela Dogaru
- Department of Physical Medicine and Rehabilitation, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Viilor Street, No. 46-50, 400347 Cluj-Napoca, Romania
| | - Cristina Ariadna Nicula
- Department of Ophthalmology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Adriana Elena Bulboacă
- Department of Pathophysiology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
50
|
Liu Y, Chang D, Liu T, Zhou X. Natural product-based bioactive agents in combination attenuate neuroinflammation in a tri-culture model. Front Pharmacol 2023; 14:1135934. [PMID: 36873986 PMCID: PMC9979791 DOI: 10.3389/fphar.2023.1135934] [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: 01/02/2023] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
Introduction: Neuroinflammation is an important pathological event contributing to the onset and progression of neurodegenerative diseases. The hyperactivation of microglia triggers the release of excessive proinflammatory mediators that lead to the leaky blood-brain barrier and impaired neuronal survival. Andrographolide (AN), baicalein (BA) and 6-shogaol (6-SG) possess anti-neuroinflammatory properties through diverse mechanisms of action. The present study aims to investigate the effects of the pair-combinations of these bioactive compounds in attenuating neuroinflammation. Methods: A tri-culture model with microglial N11 cells, microvascular endothelial MVEC(B3) cells, and neuroblastoma N2A cells was established in a transwell system. AN, BA and 6-SG used alone (25 µM) or in pair-wised combinations (12.5 + 12.5 µM) were subjected to the tri-culture system. Upon the stimulation of lipopolysaccharides (LPS) at 1 μg/mL, tumor necrosis factor-alpha (TNF-α) and interleukin 6 (IL-6) levels were determined by ELISA assays. Immunofluorescence staining was applied to investigate the nuclear translocation of nuclear factor kappa B p65 (NF-κB p65) on N11 cells, expressions of protein zonula occludens-1 (ZO-1) on MVEC cells and phosphorylated tau (p-tau) on N2A cells, respectively. The endothelial barrier permeability of MVEC cells was assessed by the Evans blue dye, and the resistance from the endothelial barrier was measured by transepithelial/endothelial electrical resistance (TEER) value. Neuronal survival of N2A cells was determined by Alamar blue and MTT assays. Results: Combinations of AN-SG and BA-SG synergistically lowered the TNF and IL-6 levels in LPS-induced N11 cells. Remarkably, the combined anti-neuroinflammatory effects of AN-SG and BA-SG remained significantly greater compared to their individual components at the same concentration level. The molecular mechanism of the attenuated neuroinflammation was likely to be mediated by downregulation of NF-κB p65 translocation (p < 0.0001 vs. LPS stimulation) in N11 cells. In the MVEC cells, both AN-SG and BA-SG restored TEER values, ZO-1 expression and reduced permeability. Furthermore, AN-SG and BA-SG significantly improved neuronal survival and reduced expressions of p-tau on N2A cells. Discussion: The AN-SG and BA-SG combinations showed greater anti-neuroinflammatory potential than those used alone in mono- and tri-cultured N11 cells, thereby further protecting endothelial tight junction and neuronal survival. Taken together, AN-SG and BA-SG may provide improved anti-neuroinflammatory and neuroprotective activities.
Collapse
Affiliation(s)
- Yang Liu
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia.,School of Science, Western Sydney University, Penrith, NSW, Australia
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| |
Collapse
|