Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 5 de 5
1.
Food Chem Toxicol ; 187: 114608, 2024 May.
Article En | MEDLINE | ID: mdl-38522498

Metal-organic frameworks (MOFs) are known as potential pharmaceutical carriers because of their structure. Here, we evaluated the sub-acute administrations of MOF-5 on behavioral parameters, oxidative stress, and inflammation levels in rats. Thirty-two male Wistar rats received four injections of saline or MOF-5 at different doses which were 1, 10, and 50 mg/kg via caudal vein. Y-Maze and Morris-Water Maze (MWM) tests were used to explore working memory and spatial learning and memory, respectively. The antioxidant capacity and oxidative stress level of brain samples were assessed by ferric reducing antioxidant power (FRAP) and thiobarbituric acid-reacting substance (TBARS) assay, respectively. The expression levels of GFAP, IL-1ß, and TNF-α were also measured by quantitative real-time reverse-transcription PCR (qRT-PCR). Sub-acute administration of MOF-5 reduced the spatial learning and memory as well as working memory, dose-dependently. The levels of FRAP were significantly reduced in rats treated with MOF-5 at higher doses. The Malondialdehyde (MDA) levels increased at the dose of 50 mg/kg. Additionally, the expression levels of IL-1ß and TNF-α were significantly elevated in the rats' brains that were treated with MOF-5. Our findings indicate that sub-acute administration of MOF-5 induces cognitive impairment dose-dependently which might be partly mediated by increasing oxidative stress and inflammation.


Antioxidants , Metal-Organic Frameworks , Rats , Animals , Male , Rats, Wistar , Antioxidants/metabolism , Memory Disorders/drug therapy , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Hippocampus/metabolism , Brain/metabolism , Oxidative Stress , Inflammation/chemically induced , Inflammation/metabolism , Maze Learning
2.
J Cell Physiol ; 239(5): e31230, 2024 May.
Article En | MEDLINE | ID: mdl-38403972

Multiple sclerosis (MS) is a chronic central nervous system (CNS) disorder characterized by demyelination, neuronal damage, and oligodendrocyte depletion. Reliable biomarkers are essential for early diagnosis and disease management. Emerging research highlights the role of mitochondrial dysfunction and oxidative stress in CNS disorders, including MS, in which mitochondria are central to the degenerative process. Adenosine monophosphate-activated protein kinase (AMPK) regulates the mitochondrial energy balance and initiates responses in neurodegenerative conditions. This systematic review, following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, aimed to comprehensively assess the literature on AMPK pathways, mitochondrial dysfunction, and in vivo studies using MS animal models. The search strategy involved the use of AMPK syntaxes, MS syntaxes, and animal model syntaxes. The PubMed, Scopus, Web of Science, and Google Scholar databases were systematically searched on August 26, 2023 without publication year restrictions. The review identified and analyzed relevant papers to provide a comprehensive overview of the current state of related research. Eight studies utilizing various interventions and methodological approaches were included. Risk of bias assessment revealed some areas of low risk but lacked explicit reporting in others. These studies collectively revealed a complex relationship between AMPK, mitochondrial dysfunction, and MS pathogenesis, with both cuprizone and experimental autoimmune encephalomyelitis models demonstrating associations between AMPK and mitochondrial disorders, including oxidative stress and impaired expression of mitochondrial genes. These studies illuminate the multifaceted role of AMPK in MS animal models, involving energy metabolism, inflammatory processes, oxidative stress, and gene regulation leading to mitochondrial dysfunction. However, unanswered questions about its mechanisms and clinical applications underscore the need for further research to fully harness its potential in addressing MS-related mitochondrial dysfunction.


AMP-Activated Protein Kinases , Encephalomyelitis, Autoimmune, Experimental , Mitochondria , Multiple Sclerosis , Animals , Humans , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Mitochondria/pathology , Mitochondria/genetics , Mitochondria/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Multiple Sclerosis/enzymology , Oxidative Stress
3.
Int J Clin Pract ; 2023: 5734675, 2023.
Article En | MEDLINE | ID: mdl-37547100

Background: Curcumin is a polyphenol derivative of the Curcuma longa rhizome, with potential antioxidant, anticancer, antidepressant, antiviral, and anti-inflammatory effects. This compound can be prepared as biodegradable polymer nanoparticles, called nanocurcumin, to improve its solubility, stability, half-life, and bioavailability. Aim: We explored nanocurcumin's effect on the clinical manifestations of patients hospitalized with mild-to-moderate COVID-19. Methods: This double-blind, randomized clinical trial involved 76 COVID-19 patients admitted to Ali-Asghar Hospital from December 2021 to March 2022. All patients received standard coronavirus treatment as per national guidelines. In addition, four times a day for two weeks, the curcumin group received 40 mg of nanocurcumin, while the control group received a placebo. Clinical manifestations were examined and recorded by the associate doctors working in the department. Statistical analysis was done using SPSS v. 21. Results: Thirty-nine people from the control group and 29 from the curcumin group completed the study. At baseline, the groups were comparable in age, gender, body mass index, hospitalization duration, and background diseases. The mean age of patients in the control and treatment groups was 53.9 ± 11.9 and 54.6 ± 13.4, respectively. Compared with the placebo, nanocurcumin minimized coughs (P=0.036), fatigue (P=0.0001), myalgia (P=0.027), oxygen demand (P=0.036), oxygen usage (P=0.05), and respiratory rate (P < 0.0001). By discharge, the curcumin group had a significantly greater increase in SPO2 than the control group (P=0.006). Conclusions: This preliminary study suggests that nanocurcumin has a potentiating anti-inflammatory effect when combined with standard COVID-19 treatment, helping the recovery from the acute inflammatory phase of the disease in hospitalized patients with mild-to-moderate disease severity. This trial is registered with Iranian Registry of Clinical Trials: IRCT20211126053183N1 (registered while recruiting on 13/12/2021).


COVID-19 , Curcumin , Humans , Curcumin/therapeutic use , Iran , COVID-19 Drug Treatment , Treatment Outcome , Oxygen , Anti-Inflammatory Agents/therapeutic use , Double-Blind Method
4.
Cytokine ; 169: 156248, 2023 09.
Article En | MEDLINE | ID: mdl-37307689

BACKGROUND: One of the regulators in severe acute respiratory syndrome coronavirus2 (SARS-CoV2) infection is miRNAs. In COVID-19 patients, immunological responses to SARS-CoV2 infection may be impacted by miR-155, a miRNA associated to inflammation. MATERIALS AND METHODS: Peripheral blood mononuclear cells (PBMCs) of 50 confirmed COVID-19 patients /Healthy Controls (HCs) was isolated by Ficoll. The frequency of T helper 17 and regulatory T cells was analyzed by flowcytometry. The RNA was extracted from each sample and after synthesis of c-DNA, the relative expression of miR-155, suppressor of cytokine signaling (SOCS-1), Signal transducer and activator of transcription 3(STAT3), and Fork Head Box Protein 3 (FoxP3) was evaluated by real-time PCR. The protein level of STAT3, FoxP3 and RORγT in the isolated PBMCs measured by western blotting. The serum level of IL-10, TGF-ß, IL-17 and IL21 was assessed by ELISA method. RESULTS: The population of Th17 cells showed a significant rise, whereas Treg cells reduced in COVID-19 cases. The master transcription factor of Treg (FoxP3) and Th17 (RORγT) relative expression showed the same pattern as flowcytometry. STAT3 level of expression at RNA and protein level increased in COVID-19 cases. FOXP3 and SOCS-1 proteins were down-regulated. The relative expression of miR-155, up-regulated in PBMC of COVID-19 patients and revealed a negative correlation with SOCS-1. The serum cytokine profile showed a reduction in TGF-ß, on the other hand an increase was seen in IL-17, IL-21 and IL-10 in COVID-19 cases toward control group. CONCLUSION: Based on the studies conducted in this field, it can be suggested that Th17/Treg in covid-19 patients can be affected by miR-155 and it can be considered a valuable diagnostic and prognostic factor in this disease.


COVID-19 , MicroRNAs , Suppressor of Cytokine Signaling 1 Protein , T-Lymphocytes, Regulatory , Th17 Cells , Humans , COVID-19/immunology , COVID-19/metabolism , COVID-19/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Interleukin-10/metabolism , Interleukin-17/metabolism , Leukocytes, Mononuclear/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , RNA, Viral , SARS-CoV-2/metabolism , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/metabolism
5.
Brain Sci ; 13(3)2023 Feb 27.
Article En | MEDLINE | ID: mdl-36979225

Understanding the transmission pathways of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) will aid in developing effective therapies directed at the virus's life cycle or its side effects. While severe respiratory distress is the most common symptom of a coronavirus 2019 (COVID-19) infection, the virus is also known to cause damage to almost every major organ and system in the body. However, it is not obvious whether pathological changes in extra-respiratory organs are caused by direct infection, indirect, or combination of these effects. In this narrative review, we first elaborate on the characteristics of SARS-CoV-2, followed by the mechanisms of this virus on various organs such as brain, eye, and olfactory nerve and different systems such as the endocrine and gastrointestinal systems.

...