1
|
Abo-Saif MA, Ragab AE, Talaat IM, Saber-Ayad M, Ibrahim AO, Selim HM. Cranberry Extract Ameliorates Diabetic Cognitive Impairment in Rats Via LncRNA GAS-5 Downregulation and Pyroptosis Pathway Inhibition. J Neuroimmune Pharmacol 2025; 20:44. [PMID: 40257540 PMCID: PMC12011949 DOI: 10.1007/s11481-025-10199-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/22/2025] [Indexed: 04/22/2025]
Abstract
The pathophysiology of diabetes-induced brain injury involves pyroptosis, an inflammatory programmed cell death. This study aimed to investigate the potential protective effect of cranberry extract (CE) against diabetes-induced brain injury. Type 1 diabetes was induced by intraperitoneal injection of streptozotocin in rats. Brain tissue samples were investigated for biochemical determination of the reduced glutathione (GSH), superoxide dismutase (SOD), and malondialdehyde (MDA), and the quantitative RT-PCR for the gene expression of glial cell-derived neurotrophic factor (GDNF), lncRNA GAS-5, and pyroptosis markers. ELISA was used to determine the caspase-1 level and immunohistochemical staining for assessing IL-1β. Prophylactic dosing of the CE in diabetic rats improved cognitive behavior and significantly suppressed MDA concentration, pyroptosis genes expression (gasdermin D and caspase 1), and lncRNA GAS-5. In addition, CE significantly elevated GSH concentration, SOD activity, and gene expression of GDNF and markedly reduced IL-1β positive stained cells score in the brain. Phytochemical characterization of the CE by FT-IR and UPLC-PDA-MS/MS revealed cyanidin arabinoside, procyanidins, quercetin, and isorhamnetin as key components. CE protects against diabetes-induced cognitive dysfunction in rats by targeting redox-related signaling pathways and inducing an anti-inflammatory effect. LncRNA GAS-5 downregulation and pyroptosis pathway inhibition may contribute to its beneficial effects, suggesting its therapeutic potential.
Collapse
Affiliation(s)
- Mariam Ali Abo-Saif
- Biochemistry Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Amany E Ragab
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Iman M Talaat
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates.
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates.
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, 21131, Egypt.
| | - Maha Saber-Ayad
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Pharmacology, College of Medicine, Cairo University, Giza, 11956, Egypt
| | - Amera O Ibrahim
- Biochemistry Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Hend Mostafa Selim
- Biochemistry Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
2
|
Goel F, Kumar D, Sharma A. Impact of corticoid receptors on Alzheimer's disease: a neuroendocrine perspective. Inflammopharmacology 2025:10.1007/s10787-025-01734-w. [PMID: 40249479 DOI: 10.1007/s10787-025-01734-w] [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: 02/19/2025] [Accepted: 03/27/2025] [Indexed: 04/19/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that has been strongly associated with changes in corticoid receptor function and HPA axis dysregulation. This review gives an overview of the complex role of GC and MC receptors in AD, especially how chronic exposure to elevated cortisol contributes to hippocampal degeneration, oxidative stress, and cognitive decline. Specific emphasis lies with cortisol, brought to the attention of neurotoxicity, and relates it to Cushing syndrome with chronic hyper-cortisolism simulating cognitive and structural impairments seen in AD. The impact of HPA axis over-activity in AD pathology is presented, demonstrating its contribution to neuro-inflammation and possible utilization as a biomarker for disease progression. This review further includes pharmacological strategies that modulate corticoid receptors for the reduction of GC-induced neurotoxicity and includes selective GR antagonists and MR agonists. Lifestyle modifications, which modulate HPA activity, are the other non-pharmacological approach to managing AD. Finally, novel drugs and interventions targeting the regulation of GC, anti-inflammatory pathways, as well as attenuation of oxidative stress are emerging strategies. Such a strategy implies that it is possible that receptor activity balance can delay or arrest AD progression.
Collapse
Affiliation(s)
- Falguni Goel
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology (MIET), Meerut, India.
| | - Daksh Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology (MIET), Meerut, India
| | - Anushka Sharma
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology (MIET), Meerut, India
| |
Collapse
|
3
|
Xu G, Sun X, An J, Sun F, Zhang C, Williams JP. Ozone protects from myocardial ischemia-reperfusion injury via inhibition of the NLRP3 inflammasome. Eur J Pharmacol 2025; 997:177631. [PMID: 40246138 DOI: 10.1016/j.ejphar.2025.177631] [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: 01/07/2025] [Revised: 04/01/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Ischemic heart disease (IHD) is a leading cause of morbidity and mortality worldwide. Myocardial ischemia/reperfusion injury (MIRI) is the primary cause of myocardial injury triggered by post-myocardial infarction reperfusion therapy. Its pathogenesis involves Ca2+ overload, the production of large amounts of oxygen-free radicals, inflammation, and cell necrosis. Growing evidence suggests that the NLRP3 inflammasome significantly contributes to the sterile inflammatory response and pyroptosis in MIRI, linking damage sensing to the initiation and amplification of the inflammatory response. Reportedly, ozone exerts anti-inflammatory and anti-infection effects by activating the antioxidant system. Additional evidence suggests that ozone inhibits NLRP3 inflammasome expression to relieve ischemic injury. In this study, we aimed to explore whether pretreating the myocardium with ozone protects it from MIRI by inhibiting the NLRP3 inflammasome. Rats were subjected to rectal infusion of ozone for 5 consecutive days, followed by ligation of the left anterior descending coronary artery for 30 min and reperfusion for 120 min to induce MIRI. Experimental results were obtained using echocardiography, triphenyltetrazolium chloride and hematoxylin and eosin staining, western blotting, and enzyme-linked immunosorbent assay. The results showed that ozone significantly improved the diastolic function of the heart, reduced the area of myocardial infarction, and decreased the expression levels of NLRP3, pro-caspase-1, ASC, and the secretion of caspase-1, interleukin (IL)-1β, and IL-18. In summary, these findings reveal that ozone pretreatment can alleviate the damage that occurs during MIRI by inhibiting the NLRP3 Inflammasome.
Collapse
Affiliation(s)
- Guohao Xu
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China; Institute for Lnnovation Diagnosis & Treatment in Anesthesiology, Shandong Second Medical University, Weifang, Shandong Province, China; Pain and Sleep Medicine Center, Rapid Anti-depression Center, The Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, China
| | - Xiaotong Sun
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China
| | - Jianxiong An
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China; Institute for Lnnovation Diagnosis & Treatment in Anesthesiology, Shandong Second Medical University, Weifang, Shandong Province, China; Pain and Sleep Medicine Center, Rapid Anti-depression Center, The Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, China; Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Brain Disease Institute & Department of Anesthesiology for the Integrated Traditional Chinese and Western Medicine Hospital, Southern Medical University, Guangzhou, China.
| | - Fan Sun
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China
| | - Chengming Zhang
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China.
| | - John P Williams
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Xu Z, Liu K, Zhang G, Yang F, He Y, Nan W, Li Y, Lin J. Transcriptome analysis reveals that the injection of mesenchymal stem cells remodels extracellular matrix and complement components of the brain through PI3K/AKT/FOXO1 signaling pathway in a neuroinflammation mouse model. Genomics 2025; 117:111033. [PMID: 40122474 DOI: 10.1016/j.ygeno.2025.111033] [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: 10/28/2024] [Revised: 02/23/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Neurological disorders are often accompanied by neuroinflammatory responses. Our previous research indicated that mesenchymal stem cells (MSCs) suppressed neuroinflammation in the brain. The mechanism of action remains not fully understood. In this study, we analyzed the impact of injected MSCs on the transcriptome in the brains of neuroinflammatory mouse model (NIM) with bioinformatical methods and conducted experimental validation with qPCR and Western blot. The results showed that the expression of extracellular matrix components changed, and the complement cascade was activated in the NIM brains. Injection of MSCs reversed the expression of ECM components and inhibited complement activation. MSCs may promote the improvement of neuronal synaptic function and alter the infiltration of immune cells into the brain. MSCs regulated the PI3K/AKT/Foxo1 signaling pathway. These findings will be very helpful for the development of MSCs-based therapy and the treatment of neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Zhihao Xu
- Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China; Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Keqin Liu
- Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Guoqing Zhang
- Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Fen Yang
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Ya''nan He
- Zhongyuan Stem Cell Research Institute, Xinxiang 453003, China
| | - Wenbin Nan
- Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Yonghai Li
- Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China; Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Juntang Lin
- Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China; Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
5
|
Manna PR, Yang S, Manna C, Waters H, Islam MA, Reddy AP, Rawat P, Reddy PH. Steroidogenic acute regulatory protein mediated variations of gender-specific sex neurosteroids in Alzheimer's disease: Relevance to hormonal and neuronal imbalance. Neurosci Biobehav Rev 2025; 169:105969. [PMID: 39631487 DOI: 10.1016/j.neubiorev.2024.105969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/24/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
The steroidogenic acute regulatory (StAR) protein mediates the rate-liming step in neuro/steroid biosynthesis. Multifaceted and delicate changes during aging, disrupting hormonal and neuronal homeostasis, constitute human senescence, an inevitable phenomenon that attributes to increased morbidity and mortality. Aging, along with progressive decreases in bioactive neurosteroids, is the primary risk factor for Alzheimer's disease (AD), which preferentially impacts two-thirds of women and one-third of men. AD is neuropathologically characterized by the accumulation of extracellular amyloid-β and intracellular phosphorylated Tau containing neurofibrillary tangles, resulting in dementia. Postmortem brains pertaining to gender-specific AD patients exhibit varied suppression of StAR and sex neurosteroid levels compared with age-matched cognitively healthy subjects, in which the attenuation of StAR is inversely correlated with the AD pathological markers. Interestingly, retinoid signaling upregulates StAR-motivated neurosteroid biosynthesis and reinstates various neurodegenerative vulnerabilities that promote AD pathogenesis. This review summarizes current understanding of StAR-driven alterations of sex neurosteroids in gender-specific AD risks and provides biochemical and molecular insights into therapeutic interventions for preventing and/or alleviating dementia for healthy aging.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Shengping Yang
- Department of Biostatistics, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Chayan Manna
- Baylor College of Medicine, Ben Taub Research Center, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Hope Waters
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Priyanka Rawat
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
6
|
Liu Z, Hu B, Tang J, Liu X, Cheng B, Jia C, Zhang L. Frontiers and hotspots evolution between air pollution and Alzheimer's disease: A bibliometric analysis from 2013 to 2023. J Alzheimers Dis 2024; 102:257-274. [PMID: 39573870 DOI: 10.1177/13872877241289381] [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: 11/26/2024]
Abstract
In recent years, the study of air pollution has received increasing attention from researchers, but a summary of Alzheimer's disease (AD) and air pollution is missed. Through combing the documents in the core dataset of Web of Science, this study analyzes current research based on specific keywords. CiteSpace and VOSviewer perform statistical analysis of measurement metrics to visualize a network of relevant content elements. The research devotes discussion to the relationship between air pollution and AD. Keyword hotspots include AD, children, oxidative stress, and system inflammation. Overall, 304 documents on air pollution and AD from 2013 to 2023 were retrieved from Web of Science. One hundred twenty-two journals published relevant articles, and the number of articles has increased gradually since the past decade. Research and development in AD and air pollution are progressing rapidly, but there is still a need for more connections with multidisciplinary technologies to explore cutting-edge hotspots.
Collapse
Affiliation(s)
- Zhirong Liu
- Department of General Surgery, The Affiliated Hospital of Chengdu Medical College, Chengdu Second People's Hospital, Chengdu, China
| | - BingShuang Hu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Ju Tang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - XinLian Liu
- Development and Regeneration Key Laboratory of Sichuan Province, Institute of Neuroscience, Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China
| | - BaoJing Cheng
- President Office, Chengdu Medical College, Chengdu, China
| | - Cui Jia
- Development and Regeneration Key Laboratory of Sichuan Province, Institute of Neuroscience, Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China
| | - LuShun Zhang
- Development and Regeneration Key Laboratory of Sichuan Province, Institute of Neuroscience, Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
7
|
Warren A. The relationship between gender differences in dietary habits, neuroinflammation, and Alzheimer's disease. Front Aging Neurosci 2024; 16:1395825. [PMID: 38694261 PMCID: PMC11061392 DOI: 10.3389/fnagi.2024.1395825] [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: 03/04/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
Neurocognitive decline is one of the foremost dire issues in medicine today. The mechanisms by which dementia pathogenesis ensues are complicated and multifactorial, particularly in the case of Alzheimer's disease (AD). One irrefutable, yet unexplained factor is the gender disparity in AD, in which women are disproportionately affected by AD, both in the rate and severity of the disease. Examining the multifaceted contributing causes along with unique gender dynamics in modifiable risk factors, such as diet, may lend some insight into why this disparity exists and potential paths forward. The aim of this brief narrative review is to summarize the current literature of gender differences in dietary habits and how they may relate to neuroinflammatory states that contribute to AD pathogenesis. As such, the interplay between diet, hormones, and inflammation will be discussed, along with potential interventions to inform care practices.
Collapse
Affiliation(s)
- Alison Warren
- The Department of Clinical Research and Leadership, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| |
Collapse
|
8
|
Liu S, Zhong H, Zhu J, Wu L. Identification of blood metabolites associated with risk of Alzheimer's disease by integrating genomics and metabolomics data. Mol Psychiatry 2024; 29:1153-1162. [PMID: 38216726 PMCID: PMC11176029 DOI: 10.1038/s41380-023-02400-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 12/17/2023] [Accepted: 12/22/2023] [Indexed: 01/14/2024]
Abstract
Specific metabolites have been reported to be potentially associated with Alzheimer's disease (AD) risk. However, the comprehensive understanding of roles of metabolite biomarkers in AD etiology remains elusive. We performed a large AD metabolome-wide association study (MWAS) by developing blood metabolite genetic prediction models. We evaluated associations between genetically predicted levels of metabolites and AD risk in 39,106 clinically diagnosed AD cases, 46,828 proxy AD and related dementia (proxy-ADD) cases, and 401,577 controls. We further conducted analyses to determine microbiome features associated with the detected metabolites and characterize associations between predicted microbiome feature levels and AD risk. We identified fourteen metabolites showing an association with AD risk. Five microbiome features were further identified to be potentially related to associations of five of the metabolites. Our study provides new insights into the etiology of AD that involves blood metabolites and gut microbiome, which warrants further investigation.
Collapse
Affiliation(s)
- Shuai Liu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Hua Zhong
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Jingjing Zhu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA.
| |
Collapse
|
9
|
Intestinal Flora Affect Alzheimer's Disease by Regulating Endogenous Hormones. Neurochem Res 2022; 47:3565-3582. [DOI: 10.1007/s11064-022-03784-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/13/2022] [Accepted: 10/01/2022] [Indexed: 11/25/2022]
|
10
|
The Mito-Hormetic Mechanisms of Ozone in the Clearance of SARS-CoV2 and in the COVID-19 Therapy. Biomedicines 2022; 10:biomedicines10092258. [PMID: 36140358 PMCID: PMC9496465 DOI: 10.3390/biomedicines10092258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
An increasing body of evidence in the literature is reporting the feasibility of using medical ozone as a possible alternative and adjuvant treatment for COVID-19 patients, significantly reducing hospitalization time, pro-inflammatory indicators, and coagulation markers and improving blood oxygenation parameters. In addition to the well-described ability of medical ozone in counteracting oxidative stress through the upregulation of the main anti-oxidant and scavenging enzymes, oxygen–ozone (O2–O3) therapy has also proved effective in reducing chronic inflammation and the occurrence of immune thrombosis, two key players involved in COVID-19 exacerbation and severity. As chronic inflammation and oxidative stress are also reported to be among the main drivers of the long sequelae of SARS-CoV2 infection, a rising number of studies is investigating the potential of O2–O3 therapy to reduce and/or prevent the wide range of post-COVID (or PASC)-related disorders. This narrative review aims to describe the molecular mechanisms through which medical ozone acts, to summarize the clinical evidence on the use of O2–O3 therapy as an alternative and adjuvant COVID-19 treatment, and to discuss the emerging potential of this approach in the context of PASC symptoms, thus offering new insights into effective and safe nonantiviral therapies for the fighting of this devastating pandemic.
Collapse
|
11
|
Proaño B, Casani-Cubel J, Benlloch M, Rodriguez-Mateos A, Navarro-Illana E, Lajara-Romance JM, de la Rubia Ortí JE. Is Dutasteride a Therapeutic Alternative for Amyotrophic Lateral Sclerosis? Biomedicines 2022; 10:biomedicines10092084. [PMID: 36140184 PMCID: PMC9495995 DOI: 10.3390/biomedicines10092084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that is characterized by the loss of upper and lower motor neurons (MNs) in the cerebral cortex, brainstem and spinal cord, with consequent weakness, atrophy and the progressive paralysis of all muscles. There is currently no medical cure, and riluzole and edaravone are the only two known approved drugs for treating this condition. However, they have limited efficacy, and hence there is a need to find new molecules. Dutasteride, a dual inhibitor of type 1 and type 2 5α-reductase (5AR) enzymes, the therapeutic purposes of which, to date, are the treatment of benign prostatic hyperplasia and androgenic alopecia, shows great anti-ALS properties by the molecular-topology methodology. Based on this evidence, this review aims to assess the effects of dutasteride on testosterone (T), progesterone (PROG) and 17β-estradiol (17BE) as a therapeutic alternative for the clinical improvement of ALS, based on the hormonal, metabolic and molecular pathways related to the pathogenesis of the disease. According to the evidence found, dutasteride shows great neuroprotective, antioxidant and anti-inflammatory effects. It also appears effective against glutamate toxicity, and it is capable of restoring altered dopamine activity (DA). These effects are achieved both directly and through steroid hormones. Therefore, dutasteride seems to be a promising molecule for the treatment of ALS, although clinical studies are required for confirmation.
Collapse
Affiliation(s)
- Belén Proaño
- Doctoral Degree School, Health Sciences, Catholic University of Valencia San Vicente Mártir, 46001 Valencia, Spain
| | - Julia Casani-Cubel
- School of Medicine and Health Sciences, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - María Benlloch
- Department Nursing, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - Ana Rodriguez-Mateos
- Department of Nutritional Sciences, King’s College London, Franklin Wilkins Building, London SE1 9NH, UK
| | | | | | | |
Collapse
|
12
|
Bette M, Cors E, Kresse C, Schütz B. Therapeutic Treatment of Superoxide Dismutase 1 (G93A) Amyotrophic Lateral Sclerosis Model Mice with Medical Ozone Decelerates Trigeminal Motor Neuron Degeneration, Attenuates Microglial Proliferation, and Preserves Monocyte Levels in Mesenteric Lymph Nodes. Int J Mol Sci 2022; 23:ijms23063403. [PMID: 35328829 PMCID: PMC8950555 DOI: 10.3390/ijms23063403] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 12/15/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable and lethal neurodegenerative disease in which progressive motor neuron loss and associated inflammation represent major pathology hallmarks. Both the prevention of neuronal loss and neuro-destructive inflammation are still unmet challenges. Medical ozone, an ozonized oxygen mixture (O3/O2), has been shown to elicit profound immunomodulatory effects in peripheral organs, and beneficial effects in the aging brain. We investigated, in a preclinical drug testing approach, the therapeutic potential of a five-day O3/O2i.p. treatment regime at the beginning of the symptomatic disease phase in the superoxide dismutase (SOD1G93A) ALS mouse model. Clinical assessment of SOD1G93A mice revealed no benefit of medical ozone treatment over sham with respect to gross body weight, motor performance, disease duration, or survival. In the brainstem of end stage SOD1G93A mice, however, neurodegeneration was found decelerated, and SOD1-related vacuolization was reduced in the motor trigeminal nucleus in the O3/O2 treatment group when compared to sham-treated mice. In addition, microglia proliferation was less pronounced in the brainstem, while the hypertrophy of astroglia remained largely unaffected. Finally, monocyte numbers were reduced in the blood, spleen, and mesenteric lymph nodes at postnatal day 60 in SOD1G93A mice. A further decrease in monocyte numbers seen in mesenteric lymph nodes from sham-treated SOD1G93A mice at an advanced disease stage, however, was prevented by medical ozone treatment. Collectively, our study revealed a select neuroprotective and possibly anti-inflammatory capacity for medical ozone when applied as a therapeutic agent in SOD1G93A ALS mice.
Collapse
Affiliation(s)
- Michael Bette
- Institute of Anatomy and Cell Biology, Philipps-University, 35037 Marburg, Germany; (E.C.); (C.K.)
- Correspondence: (M.B.); (B.S.); Tel.: +49-6421-286-6780 (M.B.); +49-6421-286-4040 (B.S.)
| | - Eileen Cors
- Institute of Anatomy and Cell Biology, Philipps-University, 35037 Marburg, Germany; (E.C.); (C.K.)
- Department of Mitochondrial Proteostasis, Max-Planck-Institute for Biology of Ageing, 50931 Cologne, Germany
| | - Carolin Kresse
- Institute of Anatomy and Cell Biology, Philipps-University, 35037 Marburg, Germany; (E.C.); (C.K.)
| | - Burkhard Schütz
- Institute of Anatomy and Cell Biology, Philipps-University, 35037 Marburg, Germany; (E.C.); (C.K.)
- Correspondence: (M.B.); (B.S.); Tel.: +49-6421-286-6780 (M.B.); +49-6421-286-4040 (B.S.)
| |
Collapse
|
13
|
Liu L, Wang N, Kalionis B, Xia S, He Q. HMGB1 plays an important role in pyroptosis induced blood brain barrier breakdown in diabetes-associated cognitive decline. J Neuroimmunol 2022; 362:577763. [PMID: 34844084 DOI: 10.1016/j.jneuroim.2021.577763] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus increases the risk of dementia, and evidence suggests hyperglycemia is a key contributor to neurodegeneration. However, our understanding of diabetes-associated cognitive decline, an important complication of diabetes mellitus, is lacking and the underlying mechanism is unclear. Blood brain barrier (BBB) breakdown is a possible cause of dementia in diabetes mellitus and Alzheimer's disease. Accumulating evidence shows BBB dysfunction caused by hyperglycemia contributes to cognitive decline. A specific type of inflammatory programmed cell death, called pyroptosis, has potential as a therapeutic target for BBB-associated diseases. Potential inducers of pyroptosis include inflammasomes such as NLRP3, whose activation relies on damage-associated molecular patterns. High mobility group box 1 (HMGB1) is a highly conserved, ubiquitous protein found in most cell types, and acts as a damage-associated molecular pattern when released from the nucleus. We propose that HMGB1 influences vascular inflammation by activating the NLRP3 inflammasome and thereby initiating pyroptosis in vascular cells. Moreover, HMGB1 plays a pivotal role in the pathogenesis of diabetes mellitus and diabetic complications. Here, we review the role of HMGB1 in BBB dysfunction induced by hyperglycemia and propose that HMGB1 is a promising therapeutic target for countering diabetes-associated cognitive decline.
Collapse
Affiliation(s)
- Lumei Liu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, PR China
| | - Neng Wang
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, PR China
| | - Bill Kalionis
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Australia; University of Melbourne, Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Australia
| | - Shijin Xia
- Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai, PR China.
| | - Qinghu He
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, PR China; Hunan University of Medicine, Huaihua, PR China.
| |
Collapse
|
14
|
Ding J, Huang J, Yin D, Liu T, Ren Z, Hu S, Ye Y, Le C, Zhao N, Zhou H, Li Z, Qi X, Huang J. Trilobatin Alleviates Cognitive Deficits and Pathologies in an Alzheimer's Disease Mouse Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3298400. [PMID: 34777683 PMCID: PMC8589506 DOI: 10.1155/2021/3298400] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease nowadays that causes memory impairments. It is characterized by extracellular aggregates of amyloid-beta (Aβ), intracellular aggregates of hyperphosphorylated Tau (p-Tau), and other pathological features. Trilobatin (TLB), a natural flavonoid compound isolated from Lithocarpuspolystachyus Rehd., has emerged as a neuroprotective agent. However, the effects and mechanisms of TLB on Alzheimer's disease (AD) remain unclear. In this research, different doses of TLB were orally introduced to 3×FAD AD model mice. The pathology, memory performance, and Toll-like receptor 4- (TLR4-) dependent inflammatory pathway protein level were assessed. Here, we show that TLB oral treatment protected 3×FAD AD model mice against the Aβ burden, neuroinflammation, Tau hyperphosphorylation, synaptic degeneration, hippocampal neuronal loss, and memory impairment. The TLR4, a pattern recognition immune receptor, has been implicated in neurodegenerative disease-related neuroinflammation. We found that TLB suppressed glial activation by inhibiting the TLR4-MYD88-NFκB pathway, which leads to the inflammatory factor TNF-α, IL-1β, and IL-6 reduction. Our study shows that TLR4 might be a key target of TLB in AD treatment and suggests a multifaceted target of TLB in halting AD. Taken together, our findings suggest a potential therapeutic effect of TLB in AD treatment.
Collapse
Affiliation(s)
- Jiuyang Ding
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Jian Huang
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Dan Yin
- Laboratory of Electron Microscopy, School of Basic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550004, China
| | - Zheng Ren
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Shanshan Hu
- Good Clinical Practice Center, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Yuanliang Ye
- Department of Neurosurgery, Liuzhou People's Hospital, Liuzhou, China
| | - Cuiyun Le
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Na Zhao
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Hongmei Zhou
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Zhu Li
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
| | - Jiang Huang
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| |
Collapse
|
15
|
Blood-Based Biomarkers of Neuroinflammation in Alzheimer's Disease: A Central Role for Periphery? Diagnostics (Basel) 2021; 11:diagnostics11091525. [PMID: 34573867 PMCID: PMC8464786 DOI: 10.3390/diagnostics11091525] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation represents a central feature in the development of Alzheimer’s disease (AD). The resident innate immune cells of the brain are the principal players in neuroinflammation, and their activation leads to a defensive response aimed at promoting β-amyloid (Aβ) clearance. However, it is now widely accepted that the peripheral immune system—by virtue of a dysfunctional blood–brain barrier (BBB)—is involved in the pathogenesis and progression of AD; microglial and astrocytic activation leads to the release of chemokines able to recruit peripheral immune cells into the central nervous system (CNS); at the same time, cytokines released by peripheral cells are able to cross the BBB and act upon glial cells, modifying their phenotype. To successfully fight this neurodegenerative disorder, accurate and sensitive biomarkers are required to be used for implementing an early diagnosis, monitoring the disease progression and treatment effectiveness. Interestingly, as a result of the bidirectional communication between the brain and the periphery, the blood compartment ends up reflecting several pathological changes occurring in the AD brain and can represent an accessible source for such biomarkers. In this review, we provide an overview on some of the most promising peripheral biomarkers of neuroinflammation, discussing their pathogenic role in AD.
Collapse
|