جستجوی مقالات مرتبط با کلیدواژه "hmgb1" در نشریات گروه "پزشکی"
-
Objective (s)
The expression of pro-inflammatory substances is closely related to various diseases. Genistein (GEN), a soy isoflavone, has been proven to inhibit the production of pro-inflammatory substances in macrophages. This study aimed to determine whether GEN exerts its inhibitory effect on the expression of pro-inflammatory substances by suppressing the release of these substances via attenuating pyroptotic cell lysis.
Materials and MethodsMice were treated with lipopolysaccharide (LPS) and GEN. J774A.1 cells were treated with LPS, adenosine triphosphate (ATP), and GEN. The expression of pro-inflammatory cytokines and high mobility group box 1 (HMGB1) was measured by qRT-PCR and ELISA. The activation of caspase-1 (CASP1) and cleavage of gasdermin D (GSDMD) were determined by Western blot assay. Lactic dehydrogenase (LDH) assay and CCK8 assay were performed to determine the integrity of the cell membrane and cell viability. The concentration of intracellular potassium (K+) and the production of reactive oxygen species (ROS) were determined by the colorimetric method and flow cytometry, respectively.
ResultsGEN inhibited the production of IL-1β and HMGB1 in LPS-challenged mice and LPS+ATP-treated mouse macrophages by inhibiting GSDMD-mediated pyroptotic cell lysis. Mechanistically, GEN could prevent the loss of intracellular K+ and the production of ROS caused by LPS+ATP treatment, thereby inhibiting the activation of CASP1. The pathological significance of the release of HMGB1 could be partially attributed to its ability to induce cell apoptosis.
ConclusionGEN inhibits CASP1/GSDMD-mediated pyroptotic cell lysis and the following release of pro-inflammatory substances by suppressing K+ loss and ROS production of macrophages.
Keywords: Gasdermin D, Genistein, HMGB1, Inhibition, Pyroptosis, Release -
Objective (s)
To investigate whether 3-methyladenine (3-MA) can protect the kidney of streptozotocin (STZ) - induced diabetes mice, and explore its possible mechanism.
Materials and MethodsSTZ was used to induce diabetes in C57BL/6J mice. The mice were divided into normal control group (NC), diabetes group (DM), and diabetes+3-MA intervention group (DM+3-MA). Blood glucose, water consumption, and body weight were recorded weekly. At the end of the 6th week of drug treatment, 24-hour urine was collected. Blood and kidneys were collected for PAS staining to evaluate the degree of renal injury. Sirius red staining was used to assess collagen deposition. Blood urea nitrogen (BUN), serum creatinine, and 24-hour urine albumin were used to evaluate renal function. Western blot was used to detect fibrosis-related protein, inflammatory mediators, high mobility group box 1 (HMGB1)/NF-κB signal pathway molecule, vascular endothelial growth factor (VEGF), and podocin, and immunohistochemistry (IHC) was used to detect the expression and localization of autophagy-related protein and fibronectin.
ResultsCompared with the kidney of normal control mice, the kidney of diabetes control mice was more pale and hypertrophic. Hyperglycemia induces renal autophagy and activates the HMGB1/NF-κB signal pathway, leading to the increase of inflammatory mediators, extracellular matrix (ECM) deposition, and proteinuria in the kidney. In diabetic mice treated with 3-MA, blood glucose decreased, autophagy and HMGB1/NF-κB signaling pathways in the kidneys were inhibited, and proteinuria, renal hypertrophy, inflammation, and fibrosis were improved.
Conclusion3-MA can attenuate renal injury in STZ-induced diabetic mice through inhibition of autophagy and HMGB1/NF-κB signaling pathway.
Keywords: 3-Methyladenine, Autophagy, Diabetes, Diabetic nephropathy, HMGB1, NF-κB signaling - pathway -
An immune system response known as inflammation can be carried on by a variety of things, such as infections, damaged cells, and noxious substances. These factors may cause acute or chronic inflammatory responses in the heart, pancreas, liver, kidney, lungs, brain, colon, and reproductive system, which may cause disease or tissue damage. Inflammatory cells and signaling pathways are activated by both pathogenic and non-pathogenic agents, cell injury, and infectious agents. The most ubiquitous types of these include tumor necrosis factor-alpha (TNF-α), nuclear factor kappa B (NF-κB), High mobility group box 1 protein (HMGB1), mitogen-activated protein kinase (MAPK), monocyte chemoattractant protein (MCP1), interleukin 1 beta (IL1β), and Janus kinase-signal transducer and activator of transcription (JAK-STAT). Severe inflammation has the potential to cause systemic inflammatory response syndrome. The most severe forms of this condition are characterized by hyperinflammation and can cause organ damage, shock, and even death. We concentrate on the origin of inflammation, all conceivable inflammatory mechanisms, and organ-specific inflammatory responses in this study on inflammatory reactions inside organs.
Keywords: HMGB1, Inflammation, Immune System, IL1β, NF-кB, TNF-α -
Objective
The growth and migration of airway smooth muscle cells (ASMCs) are dysregulated in asthma. MicroRNAs (miRNAs) are associated with the pathogenesis of many diseases including asthma. Instead, the function of miR-140- 3p in ASMCs’ dysregulation in asthma remains inconclusive. This study aimed to explore the role and mechanism of miR-140-3p in ASMCs’ dysregulation.
Materials and MethodsIn this experimental study, ASMCs were stimulated with platelet-derived growth factor (PDGF)- BB to construct an asthma cell model in vitro. MiR-140-3p expression level in the plasma of 50 asthmatic patients and 50 healthy volunteers was measured with quantitative real-time polymerase chain reaction (qRT-PCR). Besides, the enzyme-linked immunosorbent assay (ELISA) was applied to detect the contents of interleukin (IL) -1β, IL-6, and tumor necrosis factor-α (TNF-α) in the cell culture supernatant of ASMCs. Additionally, CCK-8 and transwell assays were adopted to probe the multiplication and migration of ASMCs. In addition, the western blot was employed to examine HMGB1, JAK2, and STAT3 protein expressions in ASMCs after miR-140-3p and HMGB1 were selectively regulated.
ResultsmiR-140-3p expression was declined in asthmatic patients' plasma and ASMCs stimulated by PDGF-BB. Upregulating miR-140-3p suppressed the viability and migration of the cells and alleviated the inflammatory response while inhibiting miR-140-3p showed opposite effects. Additionally, HMGB1 was testified as the target of miR-140-3p. HMGB1 overexpression could reverse the impact of miR-140-3p upregulation on the inflammatory response of ASMCs stimulated by PDGF-BB. MiR-140-3p could repress the activation of JAK2/STAT3 via suppressing HMGB1.
ConclusionIn ASMCs, miR-140-3p can inhibit the JAK2/STAT3 signaling pathway by targeting HMGB1, thus ameliorating airway inflammation and remodeling in the pathogenesis of asthma.
Keywords: Asthma, HMGB1, JAK2, STAT3, miR-140-3p -
Cardiovascular diseases (CVD) affect millions of people and spend a lot of medical costs around the world each year. Taxifolin is a natural anti-oxidative reagent obtained from multiple plants and exhibits a wide range of pharmacological effects. High mobility group box protein 1 (HMGB1) is expressed in multiple types of cells in the extracellular environment, regulating the pro-inflammatory process. Here, we detected the viability of cells using MTT assay, and the expression of each target protein was detected using western blotting analysis. The expression of each target mRNA was detected using the qPCR method, and the concentration of each cytokine in serum samples was detected using the ELISA method. In this study, we found that taxifolin could decrease the expression of hypoxia-inducible factor-1α (HIF-1α) while increasing the expression of endothelial nitric oxide synthase (eNOS), presented a protective role. Besides, taxifolin could also increase the expression of vascular endothelial growth factor-α (VEGF-α), transforming growth factor-β (TGF-β) and fibroblast growth factor21 (FGF21), resulting in viability rate increasing. And these effects were mediated by phosphatidylinositol 3-hydroxy kinase (PI3K)/AKT/mTOR signaling pathway; a similar trend was also observed in HMGB1 knockdown mice. We also found that inhibition of HMGB1 could enhance the cardioprotective effect of taxifolin and might be a new therapeutic strategy for cardiovascular disease.Keywords: HMGB1, Taxifolin, Cardiomyocytes, PI3K, AKT signaling pathway
-
Background
Radiation not only kills tumor cells, but also damages other sites. The mechanisms of damage caused by the bystander effect of irradiation in animal models are unclear and the time node is single. In this study, we aimed to investigate the inflammatory response of thymus tissue injury in non-irradiated areas at different times after irradiating rat skin.
Materials and MethodsRats were irradiated with an X-ray dose of 38 Gy, and at 15 d after irradiation, when the skin wound was most severe, the pro-inflammatory drug high mobility group box1 (HMGB1) and the anti-inflammatory drug glycyrrhizic acid (GA) were injected intraperitoneally into rats. After irradiation, skin tissues were collected for histology, and thymus tissues were collected for gene and protein testing.
ResultsAnimal model of skin damage was successfully established. The expression of macrophage (F4/80) increased after irradiation, and F4/80 produced cytokines. Through the flow which was activated by inflammatory factors in the blood, DNA damage and the expression of inflammatory-related cytokines in non-irradiated area of the thymus peaked at 15 d after irradiation. Moreover, HMGB1 treatment increased the expression at 1 d after intraperitoneal injection, and GA solution decreased the expression of inflammatory-related cytokines.
ConclusionWhen radiation damages the skin, it can cause damage to other organs through the circulation, and an anti-inflammatory GA solution reduced inflammatory responses, which are required to modify radiation-induced systemic effects with anti-inflammatory drugs or agents that affect pathways that cause bystander instability.
Keywords: Radiation induced bystander effects, thymus, γH2AX, NLRP3, HMGB1 -
Analysis of Single Nucleotide Polymorphisms in HLA-DRA, IL2RA, and HMGB1 Genes in Multiple SclerosisBackground
Multiple sclerosis (MS) is a common demyelinating neurodegenerative disorder with significant heritability. Previous studies have associated genetic variants in human leukocyte antigen (HLA) complex, IL2RA, and HMGB1 genes with the pathophysiology of MS.
MethodsIn order to investigate the gene association in the Iranian population, we performed a genotyping study of 36 variants in the mentioned genes using Sanger sequencing in 102 MS patients and 113 healthy controls.
ResultsOur results identified significant associations as well as significant allele frequency differences in some of the studied single-nucleotide polymorphisms including rs4935356, rs3177928, and rs7197 from HLA-DRA gene, and rs12722489 and rs12722490 variants from IL2RA gene (p< 0.05). Moreover, the strong linkage disequilibrium of two common haplotypes was estimated from the HLA-DRA gene.
ConclusionsThis association study may suggest the role of these polymorphisms in the genetic susceptibility of MS in the Iranian population and would facilitate the recognition of causative variants in this disease.
Keywords: HMGB1, HLA-DRA, IL2RA, Multiple sclerosis, Polymorphism -
مقدمه و هدف
التهاب عصبی نقش محوری در توسعه درد نوروپاتی دیابت بازی می کنند. هدف از پژوهش حاضر بررسی تاثیر تمرین هوازی بر سطوح پروتئین HMGB1 و برخی شاخص های استرس اکسیداتیو در موش های صحرایی با درد نوروپاتی دیابت است.
مواد و روش هادر این بررسی، 40 سر موش صحرایی نر ویستار 8 هفته ای (محدوده وزنی 2/10±220 گرم) به طور تصادفی در چهار گروه نوروپاتی دیابت، نوروپاتی دیابت تمرین، سالم تمرین و کنترل سالم قرار گرفتند. دیابت با تزریق STZ(mg/kg 50) القاء شد. پس از تائید ایجاد نوروپاتی دیابت توسط تست های رفتاری، گروه های تمرین، شش هفته تمرین هوازی تداومی با شدت متوسط 15 متر در دقیقه برای 30 دقیقه روی تردمیل اجرا کردند. سطح سرمی پروتئین HMGB1 با تکنیک الایزا و غلظت مالون دی آلدهید (MAD) و فعالیت آنزیم های سوپراکسید دیسموتاز (SOD) و کاتالاز (CAT) در بافت نخاع توسط روش های بیوشیمیایی اندازه گیری شد. آزمون آنالیز واریانس دو راهه و آزمون تعقیبی توکی برای تحلیل آماری استفاده گردید.
نتایجتمرین هوازی باعث کاهش معنی دار سطح سرمی پروتئین HMGB1 و غلظت MAD و افزایش فعالیت آنزیم های SOD و CAT نسبت به گروه نوروپاتی دیابت شد (P<0.05). همچنین سطوح پروتئین HMGB1 و غلظت MAD افزایش و فعالیت آنزیم های SOD و CAT در گروه نوروپاتی دیابت کاهش داشت P<0.05)).
نتیجه گیریبه نظر می رسد تمرین هوازی سطوح پروتئین HMGB1 و استرس اکسیداتیو را تعدیل و حساسیت نوسیسپتورها به عوامل درد زا را بهبود می بخشد. پیشنهاد می شود که تمرین هوازی به عنوان یک مداخله درمانی غیردارویی برای بیماران دیابتی به منظور کاهش درد نوروپاتیک استفاده شود.
کلید واژگان: تمرین هوازی, HMGB1, استرس اکسیداتیو, نوروپاتی محیطی دیابتBackground and ObjectiveNeuroinflammation plays a pivotal role in the development of diabetic neuropathic pain. The purpose of this study was to investigate the effect of aerobic exercise on HMGB1 protein levels and some oxidative stress biomarkers in a rat model of diabetic neuropathic pain.
Materials and Methods40 male Wistar rats (weighing 220 ± 10.2 g) were randomly divided into four groups: diabetic neuropathy, diabetic neuropathy + exercise, healthy exercise, and healthy control.Diabetes was induced by STZ injection (50 mg/kg).After confirming the development of diabetes neuropathy by behavioral tests, exercise groups received 6 weeks of continuous aerobic exercise with an average intensity of 15 m/min for 30 minutes on a treadmill.Serum levels of HMGB1 were measured by ELISA and malondialdehyde (MAD) concentrations and the activity of superoxide dismutase (SOD) and catalase (CAT) enzymes in spinal cord were determined by biochemical methods.Two-way ANOVA and Tukey's post hoc tests were used for statistical analysis.
ResultsAerobic exercise significantly reduced the serum level of HMGB1 protein and MDA concentration and increased the activity of SOD and CAT enzymes compared to diabetic neuropathy group (p <0.05).Also, HMGB1 levels and MDA increased and the activity of SOD and CAT enzymes decreased in the diabetic neuropathy group (p <0.05).
ConclusionAerobic exercise seems to modify the HMGB1 protein levels and oxidative stress and improve the sensitivity of the nociceptors to painful agents.It is suggested that aerobic exercise be used as a non-prescriptive therapeutic intervention for diabetic patients to reduce neuropathic pain.
Keywords: Aerobic exercise, HMGB1, Oxidative stress, Diabetic peripheral neuropathy -
Objective(s)Empagliflozin, a sodium-glucose cotransporter-2 (SGLT-2) inhibitor, possesses verified anti-inflammatory and anti-oxidative stress effects against diabetic nephropathy. The present investigation aims to examine empagliflozin effects on the renal levels of high mobility group box-1 (HMGB1), a potent inflammatory cytokine, and its respective receptor toll-like receptor-4 (TLR-4) in STZ-induced diabetic rats.Materials and MethodsEmpagliflozin at 10 mg/kg per os (p.o.) was administered for 4 weeks, starting 8 weeks after the induction of diabetes. Renal function, kidney inflammation, oxidative stress, and apoptosis markers as well as renal HMGB1, receptor for advanced glycation end products (RAGE), and TLR-4 levels were assessed.ResultsIn addition to down-regulating NF-κB activity in renal cortices, empagliflozin reduced renal levels of HMGB1, RAGE, and TLR-4. It alleviated renal inflammation as indicated by diminished renal expressions of inflammatory cytokines and chemokines like tumor necrosis factor-alpha (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) and also decreased urinary levels of interleukin-6 (IL-6) and alpha-1 acid glycoprotein (AGP). Moreover, empagliflozin ameliorated renal oxidative stress as demonstrated by decreased renal malondialdehyde (MDA) and elevated renal activities of superoxide dismutase (SOD) and glutathione peroxidase (GPX). It also suppressed renal caspase-3, the marker of apoptosis; and furthermore, enhanced renal function noticed by the declined levels of serum urea and creatinine.ConclusionThese findings underline that empagliflozin is able to attenuate diabetes-related elevations in renal HMGB1 levels, an influential inflammatory cytokine released from the necrotic and activated cells, and its correspondent receptors, i.e., RAGE and TLR-4.Keywords: Diabetic nephropathy, Empagliflozin, HMGB1, Inflammation, TLR-4
-
مقدمهالتهاب نقش مهمی در بیماری زایی کاردیومیوپاتی دیابتی دارد. بیان ژن HMGB1 که یک سایتوکاین پیش التهابی است و ژن های دخیل در مسیر پیام رسان پایین دست آن در دیابت افزایش می یابد. هدف از مطالعه حاضر بررسی تاثیر هشت هفته تمرین هوازی بر میزان بیان ژن های مسیر HMGB1-RAGE/TLR4-NF-kB در بافت قلبی موش های صحرایی نر دارای هایپرگلیسمی بود.مواد و روش هاسی و شش سر موش صحرایی نر نژاد ویستار با میانگین وزنی 25±231 گرم به طور تصادفی به سه گروه 12 تایی (شاهد سالم، هایپرگلیسمی شاهد و هایپرگلیسمی آزمون) تقسیم شدند. هایپرگلیسمی با تزریق درون صفاقی استرپتوزوتوسین و نیکوتین آمید القاء شد. چهل و هشت ساعت پس از اتمام برنامه تمرینی (هشت هفته تمرین هوازی) ، بافت قلبی تحت شرایط استریل جدا گردید. میزان بیان ژن های HMGB1، RAGE، TLR4و NF-kB به وسیله روش Real-Time PCR اندازه گیری شد. تجزیه و تحلیل داده ها با استفاده از آزمون تحلیل واریانس یک طرفه و آزمون تعقیبی توکی انجام شد و سطح معنی داری 0/05P< در نظر گرفته شد.یافته هامیزان بیان ژن های HMGB1، RAGE، TLR4و NF-kB در بافت قلبی گروه هایپرگلیسمی شاهد نسبت به گروه شاهد سالم به طور معنی داری (0/001P<) افزایش یافت. هشت هفته تمرین هوازی باعث کاهش بیان ژن های مورد مطالعه شد (0/001P<).نتیجه گیریبه نظر می رسد تمرین هوازی می تواند از طریق کاهش بیان ژن های HMGB1، RAGE، TLR4و NF-kB در بافت قلبی موش های صحرایی داری هایپرگلیسمی از اثرات منفی هایپرگلیسمی جلوگیری کند که این موضوع می تواند یک سازوکار مهم برای عملکرد قلبی و پیش گیری از کاردیومیوپاتی دیابتی باشد.کلید واژگان: تمرین هوازی, HMGB1, RAGE, TLR4, NF-kB, کاردیومیوپاتی دیابتی, هایپرگلیسمیIntroductionInflammation plays a critical role in the pathogenesis of diabetic cardiomyopathy. The expression of HMGB1, a proinflammatory cytokine, and its downstream signaling pathway is upregulated in diabetes. The aim of this study was to investigate the effect of 8 weeks aerobic training on the expression levels of the HMGB1-RAGE/TLR4-NF-kB pathway in cardiac tissue of male rats with hyperglycemia.Materials and MethodsThirty-six male Wistar rats (mean weight, 231±25g) were randomly divided into three groups (n=12 each): Healthy control, control hyperglycemia and trained hyperglycemia. Hyperglycemia was induced by intraperitoneal injection of strept ozotocin and nicotinamide. Forty-eight hours after completion of the training program (eight weeks aerobic training), cardiac tissue was removed under sterile conditions. Gene expression of HMGB1, RAGE, TLR4 and NF-kB were investigated, using Real-time PCR. For data analysis, one-way ANOVA and post-hoc Tukey’s test were used, with P<0.05 considered statistically significant.ResultsGene expression levels of HMGB1, RAGE, TLR4 and NF-kB were significantly (P=0.001) increased in the cardiac tissue of the hyperglycemic control group, compared with healthy controls. Eight weeks of aerobic training decreased the expression levels of the studied genes (P=0.001).ConclusionIt seems that aerobic training can prevent the negative effects of hyperglycemia via by attenuating gene expression levels of HMGB1, RAGE, TLR4 and NF-kB in the cardiac tissue of rats with hyperglycemia, and could hence be an important mechanism for cardiac function and preventing diabetic cardiomyopathy.Keywords: Aerobic Training, HMGB1, RAGE, TLR4, NF-kB, Diabetic Cardiomyopathy, Hyperglycemia
-
Objective(s)High Mobility Group Box1 (HMGB1) is a nonhistone, DNA-binding protein that serves a crucial role in regulating gene transcription and is involved in a variety of proinflammatory, extracellular activities. The aim of this study was to explore whether HMGB1 stimulation can up-regulate the expression of Toll-like Receptor 2 (TLR2) and Toll-like Receptor 4 (TLR4) on macrophages from pulpitis and to clarify the subsequent events involving Th17 cells and Th17 cell-associated cytokine changes.Materials And MethodsHaving prepared dental pulp tissues of pulpitis and healthy controls, macrophage were isolated and cultured. Macrophages were thereafter stimulated by HMGB1 time course. RT-QPCR, flowcytometer, immunofluorescence, Western blotting, and ELISA techniques were used in the present research.ResultsOur results showed that the expression of TLR2 and TLR4 on macrophages stimulated with HMGB1 increased in pulpitis compared with controls (macrophages without HMGB1 stimulation) with a statistical significance (PConclusionThe evidence from the present study suggests that the enhanced TLR2 and TLR4 pathways and Th17 cell polarization may be due to HMGB1 stimulation in pulpitis.Keywords: HMGB1, Pulpitis, TLR2, TLR4
-
One of the inflammatory mediators which is secreted by inflammatory cells is high-mobility group protein B1 (HMGB1). Interaction of HMGB1 and toll-like receptors (TLRs) leads to increased production of inflammatory cytokines. On the other hand, it was shown that triggering receptor expressed on myeloid cells (TREM-1) also can be activated by TLRs, and its soluble form (sTREM-1) can be formed by cleaving of membrane-bound form of TREM-1 proteinases. Since there is not enough knowledge about the precise role of HMGB1 and sTREM-1 in periodontal diseases, the aim of this study was to evaluate the concentration of HMGB1 and sTREM-1 in gingival crevicular fluid (GCF) samples of patients with chronic periodontitis. Gingival crevicular fluid (GCF) samples were obtained from a total of 24 individuals with clinically healthy gingiva and 24 patients with moderate to severe chronic periodontitis. For collecting GCF samples, periopapers were placed at the entrance of the crevice and left in position for 30 seconds. Then, they were stored at -80°C. Enzyme-linked immunosorbent assay (ELISA) was used for measuring the concentration of HMGB1 and sTREM-1 in GCF samples. The concentration of HMGB1 (pKeywords: Alarmins, Chronic periodontitis, DAMP, High, mobility group protein, HMGB1, Inflammation, sTREM1
-
BackgroundHepatocellular carcinoma (HCC) is one of the most important health problems in China..ObjectivesThis study analyzed expression of high-mobility group protein B1 (HMGB1) and inhibitor of apoptosis protein-2 (c-IAP2) proteins in HCC compared to paired para-tumor tissue samples to assess the association with HCC pathogenesis and progression..Materials And MethodsSixty-eight HCC and para-tumor tissue samples were collected for Western blot, qRT-PCR and immunohistochemical analyses of HMGB1 and c-IAP2..ResultsHMGB1 and c-IAP2 proteins were highly expressed in HCC tissue samples [85.3% (58/68) and 82.4% (56/68), respectively] compared to para-tumor tissue samples [32.3% and 27.9%, respectively]. Furthermore, expression of HMGB1 was significantly associated with enhanced c-IAP2 expression in HCC tissue samples (r = 0.878, P < 0.01). Expression of HMGB1 was associated with tumor multiplicity and size, alpha-fetoprotein (AFP) level and advanced TNM stage, while expression of c-IAP2 was associated with tumor size, AFP level and advanced TNM stage..ConclusionsExpression of HMGB1 and c-IAP2 proteins was associated with HCC development and progression, and the expression of HMGB1 and c-IAP2 proteins in HCC were significantly associated with each other. Additionally, these proteins may show promise as biomarkers to predict HCC progression..Keywords: Hepatocellular Carcinoma, HMGB1, c, IAP2 Protein, Biomarkers
-
ObjectiveNutrients and antioxidants in the medium of immature oocyte have a profound effect on maturation، fertilization and development of resulting embryos. In this study the effects of melatonin as an antioxidant agent on maturation، glutathione level and expression of high mobility group box-1 (HMGB1) gene were evaluated in immature oocytes of mice stained with brilliant cresyl blue (BCB).Materials And MethodsIn this experimental study، immature oocytes were harvested from ovaries of Naval Medical Research Institute (NMRI) mice. Oocytes were stained with 26 μM BCB for 90 minutes and transferred to in vitro maturation medium containing varying doses of melatonin (10-12، 10-9، 10-6، 10-3 M) and without melatonin، for 22-24 hours. Maturation was monitored using an inverted microscope. Glutathione was assessed by monochlorobimane (MCB) staining and HMGB1 expression in mature oocyte was analyzed using real-time polymerase chain reaction (PCR).ResultsMelatonin in the concentration of 10-6 M had the most effect on maturation and HMGB1 expression of BCB+ oocytes (p<0. 05). Meanwhile melatonin had no effects on glutathione levels. Additionally in immature BCB- oocytes، compared to the control group، melatonin did not affect cytoplasm maturation (p>0. 05).ConclusionIn vitro treatment with melatonin increases the maturation and HMGB1 expression in BCB+ immature oocytes and has no significant effect on glutathione levels.Keywords: Melatonin, Glutathione, Oocyte, Brilliant Cresyl Blue Staining, HMGB1
- نتایج بر اساس تاریخ انتشار مرتب شدهاند.
- کلیدواژه مورد نظر شما تنها در فیلد کلیدواژگان مقالات جستجو شدهاست. به منظور حذف نتایج غیر مرتبط، جستجو تنها در مقالات مجلاتی انجام شده که با مجله ماخذ هم موضوع هستند.
- در صورتی که میخواهید جستجو را در همه موضوعات و با شرایط دیگر تکرار کنید به صفحه جستجوی پیشرفته مجلات مراجعه کنید.