Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
J Biochem Mol Toxicol ; 38(1): e23635, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38229313

ABSTRACT

Arsenic is a toxic metalloid found in the environment in different organic and inorganic forms. Molecular mechanisms implicated in arsenic hepatotoxicity are complex but include oxidative stress, apoptosis, and autophagy. The current study focused on the potential protective capacity of melatonin against arsenic-induced hepatotoxicity. Thirty-six male Wistar rats were allocated into control, arsenic (15 mg/kg; orally), arsenic (15 mg/kg) plus melatonin (10, 20, and 30 mg/kg; intraperitoneally), and melatonin alone (30 mg/kg) groups for 28 days. After the treatment period, the serum sample was separated to measure liver enzymes (AST and ALT). The liver tissue was removed and then histological alterations, oxidative stress markers, antioxidant capacity, the levels of Nrf2 and HO-1, apoptosis (Bcl-2, survivin, Mcl1, Bax, and caspase-3), and autophagy (Sirt1, Beclin-1, and LC3 II/I ratio) proteins, as well as the expression level of miR-34a, were evaluated on this tissue. Arsenic exposure resulted in the enhancement of serum AST, ALT, and substantial histological damage in the liver. Increased levels of malondialdehyde, a lipid peroxidation marker, and decreased levels of physiological antioxidants including glutathione, superoxide dismutase, and catalase were indicators of arsenic-induced oxidative damage. The levels of Nrf2, HO-1, and antiapoptotic proteins diminished, while proapoptotic and autophagy proteins were elevated in the arsenic group concomitant with a low level of hepatic miR-34a. The co-treatment of melatonin and arsenic reversed the changes caused by arsenic. These findings showed that melatonin reduced the hepatic damage induced by arsenic due to its antioxidant and antiapoptotic properties as well as its regulatory effect on the miR-34a/Sirt1/autophagy pathway.


Subject(s)
Arsenic , Chemical and Drug Induced Liver Injury , Melatonin , MicroRNAs , Rats , Male , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Melatonin/pharmacology , Arsenic/toxicity , NF-E2-Related Factor 2/metabolism , Sirtuin 1/metabolism , Rats, Wistar , Liver/metabolism , Oxidative Stress , Apoptosis , MicroRNAs/genetics , MicroRNAs/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Autophagy
3.
Biol Trace Elem Res ; 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37853305

ABSTRACT

Arsenic (As) exposure is known to cause several neurological disorders through various molecular mechanisms such as oxidative stress, apoptosis, and autophagy. In the current study, we assessed the effect of melatonin (Mel) on As-induced neurotoxicity. Thirty male Wistar rat were treated daily for 28 consecutive days. As (15 mg/kg, gavage) and Mel (10 and 20 mg/kg, i.p.) were administered to rats. Morris water maze test was done to evaluate learning and memory impairment in training days and probe trial. Oxidative stress markers including MDA and GSH levels, SOD activity, and HO-1 levels were measured. Besides, the levels of apoptosis (caspase 3, Bax/Bcl2 ratio) and autophagy markers (Sirt1, Beclin-1, and LC3 II/I ratio) as well as the expression of miR-144 and miR-34a in cortex tissue were determined. As exposure disturbed learning and memory in animals and Mel alleviated these effects. Also, Mel recovered cortex pathological damages and oxidative stress induced by As. Furthermore, As increased the levels of apoptosis and autophagy proteins in cortex, while Mel (20 mg/kg) decreased apoptosis and autophagy. Also, Mel increased the expression of miR-144 and miR-34a which inhibited by As. In conclusion, Mel administration attenuated As-induced neurotoxicity through anti-oxidative, anti-apoptotic, and anti-autophagy mechanisms, which may be recommended as a therapeutic target for neurological disorders.

4.
Iran J Basic Med Sci ; 26(8): 953-959, 2023.
Article in English | MEDLINE | ID: mdl-37427324

ABSTRACT

Objectives: Natural coumarin called osthole is regarded as a medicinal herb with widespread applications in Traditional Chinese Medicine. It has various pharmacological properties, including antioxidant, anti-inflammatory, and anti-apoptotic effects. In some neurodegenerative diseases, osthole also shows neuroprotective properties. In this study, we explored how osthole protects human neuroblastoma SH-SY5Y cells from the cytotoxicity of 6-hydroxydopamine (6-OHDA). Materials and Methods: Using the MTT assay and DCFH-DA methods, respectively, the viability of the cells and the quantity of intracellular reactive oxygen species (ROS) were evaluated. Signal Transducers and Activators of Transcription (STAT), Janus Kinase (JAK), extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), and caspase-3 activation levels were examined using western blotting. Results: In SH-SY5Y cells, the results showed that a 24-hour exposure to 6-OHDA (200 µM) lowered cell viability but markedly elevated ROS, p-JAK/JAK, p-STAT/STAT, p-ERK/ERK, p-JNK/JNK ratio, and caspase-3 levels. Interestingly, osthole (100 µM) pretreatment of cells for 24 hr prevented 6-OHDA-induced cytotoxicity by undoing all effects of 6-OHDA. Conclusion: In summary, our data showed that osthole protects SH-SY5Y cells against 6-OHDA-induced cytotoxicity by inhibiting ROS generation and reducing the activity of the JAK/STAT, MAPK, and apoptotic pathways.

5.
Toxicol Ind Health ; 39(5): 248-257, 2023 May.
Article in English | MEDLINE | ID: mdl-37022282

ABSTRACT

Benzo(a)pyrene is a ubiquitous environmental contaminant, which could induce renal injury. It is reported that melatonin has a protective effect against multiple organ injuries by regulating oxidative stress, apoptosis, and autophagy. The aim of this study was to estimate the melatonin effects on benzo(a)pyrene renal toxicity in mice and the possible molecular mechanisms involved in this model. Thirty male mice were allocated to five groups and treated with benzo(a)pyrene (75 mg/kg, oral gavage) and/or melatonin (10 and 20 mg/kg, intraperitoneally). The oxidative stress factors were evaluated in renal tissue. The levels of apoptotic (the Bax/Bcl-2 ratio and caspase-3) and autophagic (the LC3 II/I, Beclin-1, and Sirt1) proteins were examined using Western blot. Following the administration of benzo(a)pyrene, malondialdehyde, caspase-3 and the Bax/Bcl-2 ratio increased in renal tissue, while Sirt1, Beclin-1, and the LC3 II/I ratio diminished. Interestingly, the co-administration of 20 mg/kg melatonin along with benzo(a)pyrene reduced the oxidative stress markers, apoptotic and autophagic proteins. Collectively, melatonin exhibited a protective effect against benzo(a)pyrene-induced renal injury through the suppression of oxidative stress and apoptosis and the inhibition of Sirt1/autophagy pathway.


Subject(s)
Melatonin , Mice , Male , Animals , Melatonin/pharmacology , Benzo(a)pyrene , Sirtuin 1/metabolism , Sirtuin 1/pharmacology , Caspase 3 , Beclin-1/metabolism , Beclin-1/pharmacology , bcl-2-Associated X Protein/metabolism , Oxidative Stress , Apoptosis , Proto-Oncogene Proteins c-bcl-2/metabolism
6.
Food Chem Toxicol ; 175: 113691, 2023 May.
Article in English | MEDLINE | ID: mdl-36871878

ABSTRACT

Arsenic is a notorious metalloid that exists in the earth's crust and is considered toxic for humans and the environment. Both cancerous and non-cancerous complications are possible after arsenic exposure. Target organs include the liver, lungs, kidney, heart, and brain. Arsenic-induced neurotoxicity, the main focus of our study, can occur in central and peripheral nervous systems. Symptoms can develop in a few hours, weeks, or years depending on the quantity of arsenic and the duration of exposure. In this review, we aimed to gather all the compounds, natural and chemical, that have been studied as protective agents in cellular, animal, and human reports. Oxidative stress, apoptosis, and inflammation are frequently described as destructive mechanisms in heavy metal toxicity. Moreover, reduced activity of acetylcholinesterase, the altered release of monoamine neurotransmitters, down-regulation of N-methyl-D-aspartate receptors, and decreased brain-derived neurotrophic factor are important underlying mechanisms of arsenic-induced neurotoxicity. As for neuroprotection, though some compounds have yet limited data, there are others, such as curcumin, resveratrol, taurine, or melatonin which have been studied more deeply and might be closer to a reliable protective agent. We collected the available information on all protective agents and the mechanisms by which they fight against arsenic-induced neurotoxicity.


Subject(s)
Arsenic , Melatonin , Neurotoxicity Syndromes , Animals , Humans , Arsenic/toxicity , Acetylcholinesterase , Oxidative Stress , Melatonin/pharmacology , Brain , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control
7.
Biofactors ; 49(3): 620-635, 2023.
Article in English | MEDLINE | ID: mdl-36609811

ABSTRACT

Chronic arsenic (As) exposure, mainly as a result of drinking contaminated water, is associated with cardiovascular diseases. Mitochondrial dysfunction, oxidative stress, inflammation, apoptosis, and autophagy have been suggested as the molecular etiology of As cardiotoxicity. Melatonin (Mel) is a powerful antioxidant. Mel improves diabetic cardiomyopathy, cardiac remodeling, and heart failure. Following pre-treatment with Mel (10, 20, or 30 mg/kg/day i.p.), rats were orally gavaged with As (15 mg/kg/day) for 28 days. Electrocardiographic findings showed that Mel decreased the As-mediated QT interval prolongation. The effects of As on cardiac levels of glutathione (GSH) and malondialdehyde (MDA) were reversed by Mel pretreatment. Mel also modulated the Sirt1 and Nrf2 expressions promoted by As. Mel down-regulated autophagy markers such as Beclin-1 expression and the LC3-II/I ratio. Moreover, the cardiac expression of cleaved-caspase-3 and Bax/Bcl-2 ratio was decreased by Mel pretreatment. Reduced expression of miR-34a and miR-144 by As were reversed by Mel. The histopathological changes of cardiac injury associated with As exposure was moderated by Mel. Mel may improve As-induced cardiac dysfunction through anti-oxidative, anti-apoptotic, and anti-autophagic mechanisms.


Subject(s)
Arsenic , Melatonin , MicroRNAs , Rats , Animals , Melatonin/pharmacology , Arsenic/toxicity , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Cardiotoxicity/drug therapy , Cardiotoxicity/genetics , Sirtuin 1/genetics , Sirtuin 1/metabolism , Oxidative Stress , Glutathione/metabolism , Apoptosis , MicroRNAs/genetics , MicroRNAs/metabolism
8.
Cell Cycle ; 22(9): 1062-1073, 2023 05.
Article in English | MEDLINE | ID: mdl-36703306

ABSTRACT

In recent years, researchers have begun to realize the importance of the role of non-coding RNAs in the treatment of cancer and cardiovascular and neurological diseases. LncRNAs and miRNAs are important non-coding RNAs, which regulate gene expression and activate mRNA translation through binding to diverse target sites. Their involvement in the regulation of protein function and the modulation of physiological and pathological conditions continues to be investigated. Sirtuins, especially Sirt1, have a critical function in regulating a variety of physiological processes such as oxidative stress, inflammation, apoptosis, and autophagy. The lncRNAs/miRNAs/Sirt1 axis may be a novel regulatory mechanism, which is involved in the progression and/or prevention of numerous diseases. This review focuses on recent findings on the crosstalk between non-coding RNAs and Sirt1 in myocardial and cerebral injuries and may provide some insight into the development of novel approaches in the treatment of these disorders.Abbreviation: BMECs, brain microvascular endothelial cells; C2dat1, calcium/calmodulin-dependent protein kinase type II subunit delta (CAMK2D)-associated transcript 1; EPCs, endothelial progenitor cells; FOXOs, forkhead transcription factors; GAS5, growth arrest-specific 5; HAECs, human aortic endothelial cells; HAND2-AS1, HAND2 Antisense RNA 1; HIF-1α, hypoxia-inducible factor-1α; ILF3-AS1, interleukin enhancer-binding factor 3-antisense RNA 1; KLF3-AS1, KLF3 antisense RNA 1; LncRNA, long noncoding RNA; LUADT1, Lung Adenocarcinoma Associated Transcript 1; MALAT1, Metastasis-associated lung adenocarcinoma transcript 1; miRNA, microRNA; NEAT1, nuclear enriched abundant transcript 1; NF-κB, nuclear factor kappa B; OIP5-AS1, Opa-interacting protein 5-antisense transcript 1; Sirt1-AS, Sirt1 Antisense RNA; SNHG7, small nucleolar RNA host gene 7; SNHG8, small nucleolar RNA host gene 8; SNHG12, small nucleolar RNA host gene 12; SNHG15, small nucleolar RNA host gene 15; STAT3, signal transducers and activators of transcription 3; TUG1, taurine up-regulated gene 1; VSMCs, vascular smooth muscle cells; XIST, X inactive specific transcript; ZFAS1, ZNFX1 Antisense RNA 1.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Humans , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Sirtuin 1/metabolism , Endothelial Cells/metabolism , RNA, Small Nucleolar , NF-kappa B , Kruppel-Like Transcription Factors
9.
Avicenna J Phytomed ; 13(1): 85-96, 2023.
Article in English | MEDLINE | ID: mdl-36698739

ABSTRACT

Objective: Portulaca oleracea, commonly known as Purslane, is traditionally used as a sour, diuretic, and cooling herb with hemostatic properties. The present study evaluates the antianemic effect of methanolic and aqueous extracts of P. oleracea in a phenylhydrazine model of anemia. Materials and Methods: Phenylhydrazine (60 mg/kg/day, i.p., two consecutive days) was used to induce anemia in rats. The aqueous and methanolic extracts of P. oleracea were prepared, and three methods of treatment were defined with two doses (500 and 750 mg/kg, i.p.). The hematological parameters and blood cell morphology, total and direct bilirubin, and morphology, and pathology of bone marrow were evaluated. Results: The results showed that the methanolic extract has better effects than aqueous extract in improving phenylhydrazine-induced anemia. Our results showed that administration of 500 and 750 mg/kg of P. oleracea methanolic extracts for 4 days could protect against the development of anemia caused by phenylhydrazine. Conclusion: In summary, the methanolic extracts of P. oleracea might be effective in phenylhydrazine-induced anemia.

10.
Environ Sci Pollut Res Int ; 30(12): 33113-33123, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36474038

ABSTRACT

Benzo(a)pyrene (BaP) is a polycyclic aromatic hydrocarbon and a serious environmental pollutant. BaP is formed by the incomplete combustion of organic matter at high temperatures. In addition, tobacco smoke and many foods, especially charbroiled food and grilled meats, contain BaP and can cause it to enter human body. Melatonin, a pineal gland hormone, has antioxidant, anti-apoptosis, and autophagy regulatory properties. The possible protective impact of melatonin on cardiopulmonary toxicity induced by BaP was investigated by examining the antioxidant effects and the apoptosis and autophagy properties of melatonin. Thirty male mice were divided into 5 groups and treated for 28 days as follows: (I) control (BaP and melatonin solvent), (II) BaP (75 mg/kg, oral gavage), (III and IV) BaP (75 mg/kg) + melatonin (10 and 20 mg/kg, intraperitoneally), (V) melatonin (20 mg/kg). The oxidative stress factors (MDA and GSH content) were assessed in the heart and lung tissues. The levels of apoptotic (Caspase-3 and the Bax/Bcl-2 ratio) and autophagic (the LC3 ӀӀ/Ӏ, Beclin-1, and Sirt1) proteins were examined by using western blot analysis. Following the administration of BaP, MDA, the Bax/Bcl-2 ratio, and the Caspase-3 proteins increased in the heart and lung tissues, while GSH, Sirt1, Beclin-1, and the LC3 II/I ratio diminished. The coadministration of melatonin along with BaP, MDA, and apoptotic proteins returned to the control values, while GSH and the autophagy proteins were enhanced in both the heart and lungs. Melatonin exhibited a protective effect against BaP-induced heart and lung injury through the suppression of oxidative stress and apoptosis and the induction of the Sirt1/autophagy pathway.


Subject(s)
Melatonin , Mice , Male , Humans , Animals , Melatonin/pharmacology , Benzo(a)pyrene/toxicity , Sirtuin 1 , Caspase 3 , bcl-2-Associated X Protein , Beclin-1/pharmacology , Antioxidants/metabolism , Proto-Oncogene Proteins c-bcl-2 , Autophagy
11.
Biomed Pharmacother ; 151: 113135, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35598369

ABSTRACT

Arsenic (As), a metalloid chemical element, is classified as heavy metal. Previous studies proposed that As induces vascular toxicity by inducing autophagy, apoptosis, and oxidative stress. It has been shown that melatonin (Mel) can decrease oxidative stress and apoptosis, and modulate autophagy in different pathological situations. Hence, this study aimed to investigate the Mel effect on As-induced vascular toxicity through apoptosis and autophagy regulation. Forty male rats were treated with As (15 mg/kg; oral gavage) and Mel (10 and 20 mg/kg, intraperitoneally; i.p.) for 28 days. The systolic blood pressure (SBP) changes, oxidative stress markers, the aorta histopathological injuries, contractile and relaxant responses, the level of apoptosis (Bnip3 and caspase-3) and autophagy (Sirt1, Beclin-1 and LC3 II/I ratio) proteins were determined in rats aorta. The As exposure significantly increased SBP and enhanced MDA level while reduced GSH content. The exposure to As caused substantial histological damage in aorta tissue and changed vasoconstriction and vasorelaxation responses to KCl, PE, and Ach in isolated rat aorta. The levels of HO-1 and Nrf-2, apoptosis markers, Sirt1, and autophagy proteins also enhanced in As group. Interestingly, Mel could reduce changes in oxidative stress, blood pressure, apoptosis, and autophagy induced by As. On the other hand, Mel led to more increased the levels of Nrf-2 and HO-1 proteins compared with the As group. In conclusion, our findings showed that Mel could have a protective effect against As-induced vascular toxicity by inhibiting apoptosis and the Sirt1/autophagy pathway.


Subject(s)
Arsenic , Hypertension , Melatonin , Animals , Apoptosis , Arsenic/toxicity , Autophagy/drug effects , Hypertension/chemically induced , Hypertension/drug therapy , Male , Melatonin/pharmacology , Oxidative Stress , Rats , Sirtuin 1/metabolism
12.
Naunyn Schmiedebergs Arch Pharmacol ; 393(12): 2241-2251, 2020 12.
Article in English | MEDLINE | ID: mdl-32632567

ABSTRACT

Benzo(a)pyrene (BaP), a toxic polycyclic aromatic hydrocarbon, is spread in different ways as an environmental pollutant. It has been proposed that BaP can induce toxicity through oxidative stress and apoptosis in vital organs. The present study evaluated the protective effect of melatonin, a circadian hormone of the pineal gland, on BaP-induced neurotoxicity focused on oxidative stress, autophagy, and apoptosis pathways. Thirty male mice in 5 groups were treated daily for 28 consecutive days: (I) control group (BaP and melatonin solvent), (II) BaP (75 mg/kg, orally), (III) and (IV) BaP + melatonin (10 and 20 mg/kg, i.p.), (V) melatonin (20 mg/kg). The oxidative stress markers were determined in the brain. Western blot was conducted for the level of LC3 II/I and Beclin1, as autophagy markers, caspase3 and Bcl2, as apoptosis proteins, and Sirt1 in the brain. The exposure of mice to BaP caused a marked increase in the malondialdehyde (MDA) level and decrease of glutathione (GSH) content in the brain. Furthermore, the Sirt1 level upregulated as well as LC3 II/I, Beclin1, and cleaved caspase3 proteins, while the level of Bcl2 did not change. Melatonin at 20 mg/kg concurrently with BaP restored the BaP alteration in the brain compared with the BaP group. In conclusion, BaP induced brain toxicity via the induction of oxidative stress, apoptosis, and autophagy, whereas melatonin afforded neuroprotection against BaP due to inhibition of these mechanisms.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Benzo(a)pyrene/toxicity , Brain Injuries/chemically induced , Brain Injuries/prevention & control , Melatonin/administration & dosage , Animals , Antioxidants/administration & dosage , Apoptosis/physiology , Autophagy/physiology , Brain Injuries/metabolism , Dose-Response Relationship, Drug , Male , Mice , Neuroprotective Agents/administration & dosage , Oxidative Stress/drug effects , Oxidative Stress/physiology
13.
Ecotoxicol Environ Saf ; 196: 110556, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32247962

ABSTRACT

Benzo(a)pyrene (BaP), an important environmental pollutant, is produced as the result of incomplete combustion of organic materials in many industries and food cooking process. It has been purposed that BaP induces hepatotoxicity through oxidative stress and apoptosis. Several studies have shown that melatonin can protect against chemical-induced apoptosis through autophagy pathway. In this study, we assessed the modulating effect of melatonin, a well-known antioxidant, on BaP-induced hepatotoxicity through induction of autophagy. Thirty male mice were treated daily for 28 consecutive days. BaP (75 mg/kg; oral gavage) and melatonin (10 and 20 mg/kg, i.p.) were administered to mice. The liver histopathology and the levels of apoptosis and autophagy proteins as well as the expression of miR-34a were determined. The BaP exposure induced severe liver histological injury and markedly enhanced AST, ALT and MDA level. Also, apoptosis proteins and hepatic miR-34a expression increased. However, the level of Sirt1 and autophagy markers such as LC3 II/I ratio and Beclin-1 reduced. The co-administration of melatonin reversed all changes caused by BaP. In summary, melatonin appears to be effective in BaP-induced hepatotoxicity maybe through the miR-34a/Sirt1/autophagy molecular pathway.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Benzo(a)pyrene/toxicity , Liver/drug effects , Melatonin/pharmacology , MicroRNAs/metabolism , Sirtuin 1/metabolism , Animals , Antioxidants/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Liver/metabolism , Liver/pathology , Male , Mice , Protective Agents/pharmacology
14.
Foot Ankle Int ; 41(1): 63-68, 2020 01.
Article in English | MEDLINE | ID: mdl-31587569

ABSTRACT

BACKGROUND: The objective of this study was to determine whether the injection of botulinum toxin A (BTA) in the medial head of the gastrocnemius muscle could yield improvements in function and disability in patients with chronic plantar fasciitis with follow-up 12 months after treatment. METHODS: Thirty-two patients with chronic plantar fasciitis were included in the study and randomly allocated to the BTA and placebo groups. The visual analog scale (VAS) and American Orthopaedic Foot & Ankle Society (AOFAS) scores were used to evaluate pain levels pre- and postinjection as well as function of the foot, respectively. Patients were also asked to rate their treatment satisfaction 1 year after injection. The range of dorsiflexion was measured before and 12 months after the injection. RESULTS: At the 12-month follow-up, the mean VAS decreased from 7.8 to 4 in the placebo group and from 8 to 0.33 in the BTA group. Furthermore, the mean AOFAS scores increased from 48.4 to 65.3 in the placebo group and from 45.5 to 90.6 in the BTA group. The postinjection scores in the BTA group were significantly higher than those in the placebo group (P < .001). Patient satisfaction in the BTA group was higher than that in the placebo group at the 12-month follow-up. CONCLUSION: In patients with chronic plantar fasciitis, the use of BTA had a positive effect on improvement in pain and foot function 1 year after treatment. LEVEL OF EVIDENCE: Level I, prospective randomized controlled trial.


Subject(s)
Botulinum Toxins, Type A/therapeutic use , Fasciitis, Plantar/therapy , Muscle, Skeletal/drug effects , Adult , Fasciitis, Plantar/physiopathology , Female , Humans , Injections, Intramuscular , Male , Middle Aged , Muscle, Skeletal/physiopathology , Pain Measurement , Patient Satisfaction , Prospective Studies , Surveys and Questionnaires , Ultrasonography, Interventional
15.
Pharmacol Res ; 142: 22-29, 2019 04.
Article in English | MEDLINE | ID: mdl-30742900

ABSTRACT

Long non-coding RNA (lncRNA) is a class of non-coding RNA with ≥200 nucleotides in length which are involved as critical regulators in various cellular processes. LncRNAs contribute to the development and progression of many human diseases. Autophagy is a key catabolic process which helps to maintain the cellular homeostasis through the decay of damaged or unwanted proteins and dysfunctional cytoplasmic organelles. The impairment of the autophagy process has been described in numerous diseases. The autophagy possess can have either a protective or a detrimental role in cells depending on its activation status and other cellular conditions. LncRNAs have been shown to have an important function in the regulation of important biological processes such as autophagy. The relationship between lncRNAs and autophagy has been shown to be involved in the progression and possibly in the prevention of many diseases. In this review, recent findings on the regulatory roles of lncRNAs in the cell autophagy pathway, as well as their relevance to different diseases such as cardiovascular disease, cerebral ischemic stroke and cancer are highlighted.


Subject(s)
Autophagy , RNA, Long Noncoding , Animals , Disease , Humans
16.
Crit Rev Food Sci Nutr ; 58(7): 1230-1241, 2018 May 03.
Article in English | MEDLINE | ID: mdl-28925721

ABSTRACT

Atrial fibrillation (AF) is the most common cardiac arrhythmia that occurs because of several different risk factors, e.g., valvular heart disease, coronary artery disease, age ≥75 years, hypertension and diabetes mellitus. One key risk factor that results in AF, is oxidative stress. Evidence suggests that there is a correlation between oxidative processes and the genesis of AF. Oxidative stress occurs when the generation of reactive oxygen species (ROS) increase due to excessive activity of enzymes including NADPH oxidase (NOX) and xanthine oxidase; or its degradation decrease by dysfunctional antioxidant enzyme systems, such as superoxide dismutase (SOD), catalase and glutathione peroxidase (GPx). Afterwards, elevated ROS may shift ion channel activity to increase AF susceptibility. The outbreak of AF continues to grow. Unfortunately, current treatment strategies may have limited efficacy or adverse effects. On the other hand, the inhibition of ROS formation and alteration of ion channel activity could be important therapeutic targets for prevention or treatments of AF. Additionally, many studies have been shown that several natural compounds have the ability to inhibit NADPH oxidases directly. This review focuses on natural compounds which specially inhibit NOX isoforms and have direct effects on ion channels, suggesting these compounds can be helpful in AF treatment.


Subject(s)
Atrial Fibrillation/drug therapy , Biological Products/pharmacology , Ion Channels/antagonists & inhibitors , NADPH Oxidases/antagonists & inhibitors , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...