به جمع مشترکان مگیران بپیوندید!

تنها با پرداخت 70 هزارتومان حق اشتراک سالانه به متن مقالات دسترسی داشته باشید و 100 مقاله را بدون هزینه دیگری دریافت کنید.

برای پرداخت حق اشتراک اگر عضو هستید وارد شوید در غیر این صورت حساب کاربری جدید ایجاد کنید

عضویت

جستجوی مقالات مرتبط با کلیدواژه « blood brain barrier » در نشریات گروه « پزشکی »

  • Yang Qiao, Qing Yuan *, Zhen Liu
    Objective (s)

    Chronic alcohol abuse causes cognitive deficits. Huangqi Gegen Decoction (HGD), a traditional Chinese herbal formula comprising Huangqi and Gegen, has been documented for its therapeutic efficacy in the treatment of alcoholic liver injury. However, its potential neuroprotective effects against alcohol-induced brain injury remain unexplored. This study aims to evaluate the neuroprotection of HGD on alcohol-induced cognitive dysfunction and the associated mechanism.

    Materials and Methods

    Wistar rats were orally administered 50% ethanol for 10 weeks, followed by treatment with HGD at doses of 16, 32, or 64 mg/kg/day for an additional 6 weeks. The spatial learning and memory abilities of rats were assessed through the Morris Water Maze experiment. The pathological condition in the hippocampus was assessed using H&E and Nissl staining. Tight junction proteins, oxidative stress, and inflammation cytokines were measured by IF, ELISA, PCR, and western blot. The mRNA and protein expression of Keap1, Nrf-2, HO-1, and NQO-1 were tested by PCR and western blot.

    Results

    Results showed that HGD effectively mitigated cognitive dysfunction and pathological changes in alcohol-induced rats while enhancing the expression of ZO-1, Occludin, and Claudin-5. Furthermore, HGD effectively mitigated oxidative stress by reducing levels of ROS and MDA, while elevating levels of SOD, CAT, and GSH-PX in brain tissue. Moreover, HGD significantly suppressed microglial activation and down-regulated expressions of IL-1β, IL-6, and TNF-α. Mechanistically, HGD remarkably up-regulated the expression of Nrf-2, HO-1, and NQO-1 while down-regulating Keap1 expression.

    Conclusion

    These findings suggest that HGD may be a promising therapeutic agent for alleviating alcohol-induced cognitive dysfunction.

    Keywords: Alcohols, Blood-Brain Barrier, Cognitive Dysfunction Huangqi, Oxidative Stress, Pueraria}
  • سجاد جمشیدی، محمدهادی باقرصاد*
    زمینه و هدف

    ترکیبات آلی فسفر یا ارگانوفسفره شامل عوامل عصبی، آفت کش ها، معادل های عوامل شیمیایی و نوویکوک یک مخاطره جدی برای سلامتی انسان هستند که استفاده از آن ها بر ضد انسان ها طی کنوانسیون منع سلاح های شیمیایی به وسیله 192 کشور در سال 1993، ممنوع شده است. اثرات سمی این ترکیبات شامل فسفریله شدن و در نتیجه مهار آنزیم استیل کولین استراز است که باعث تجمع استیل کولین در سیناپس های سیستم اعصاب مرکزی و محیطی و بروز علائم مسمومیت نوروتوکسیک می شود. داروهای مورد استفاده برای درمان این مسمومیت ترکیباتی هستند که حداکثر 10 درصد می توانند از سد خونی مغزی عبور کنند و بنابراین قابلیت سم زدایی در سیستم اعصاب مرکزی را به طور موثر ندارند. هدف از این تحقیق طراحی و سنتز یک نانو فعال کننده با خاصیت پارامغناطیسی به منظور فعال سازی آنزیم مهار شده در سیستم اعصاب مرکزی است.

    روش ها

    در این پژوهش نانوذرات اکسید آهن کانجوگه شده با 2-پیریدینیوم آلدوکسیم طراحی و طی چهار مرحله سنتز شد. مشخصات نانوذرات توسط روش های طیف سنجی مادون قرمز، طیف سنجی تفرق انرژی پرتو ایکس (EDS)، آنالیز عنصری، پراکندگی نور دینامیکی (DLS) و میکروسکوپ الکترونی (SEM) تعیین شد و اثر فعال سازی آن ها در مقایسه با پرالیدوکسیم به عنوان داروی در دسترس تجاری، با استفاده از روش المن به صورت برون تنی مورد بررسی قرار گرفت.

    یافته ها

    نتایج حاصل از روش المن نشان داد که درصد فعال سازی آنزیم مهار شده با پاراکسون به عنوان یک عامل ارگانوفسفره، توسط پرالیدوکسیم به عنوان آنتی دوت تجاری در دسترس و نانوذرات سنتز شده درغلظت 5 میلی مولار و طی 30 دقیقه، به ترتیب 88/6 و 39/3 درصد است.

    نتیجه گیری

    این مطالعه نشان داد که نانوذرات اکسید آهن کانجوگه شده با 2-پیریدینیوم آلدوکسیم طراحی شده به خوبی قابل سنتز و دارای قابلیت فعال سازی آنزیم استیل کولین استراز مهار شده با پاراکسون هستند. بنابراین در مطالعات آینده باید اثر گروه های دیگر مانند 4-پیریدینیوم آلدوکسیم، 2 و 4-پیریدینیوم الدوکسیم و... در قدرت فعال سازی آنزیم و همچنین قابلیت عبور این نانوذرات از سد خونی- مغزی مورد بررسی قرار گیرد.

    کلید واژگان: آنتی دوت ها, عوامل عصبی, سد خونی- مغزی, فعال کننده های کولین استراز, مسمومیت با ارگانوفسفره, نانوذرات اکسید آهن ابرپارامغناطیسی}
    Sajjad Jamshidi, Mohammadhadi Baghersad *
    Background and Aim

    Organophosphate compounds, including nerve agents, pesticides, chemical agent analogs, and Novichok, pose a serious health hazard to humans. Their use against humans was banned by 192 countries in 1993 under the Chemical Weapons Convention. The toxic effects of these compounds involve the phosphorylation and subsequent inhibition of the enzyme acetylcholinesterase, leading to the accumulation of acetylcholine in the synapses of the central and peripheral nervous systems, resulting in neurotoxic poisoning symptoms. The drugs used to treat this poisoning are compounds that can cross the blood-brain barrier by a maximum of 10% and therefore do not effectively detoxify the central nervous system. This research aims to design and synthesize a superparamagnetic nano-reactivator to activate the inhibited enzyme in the central nervous system.

    Methods

    In this study, iron oxide nanoparticles conjugated with 2-pyridinium aldoxime were designed and synthesized in four steps. The characteristics of the nanoparticles were determined by infrared spectroscopy, energy-dispersive X-ray spectroscopy (EDS), elemental analysis, dynamic light scattering (DLS), and electron microscopy (SEM). Their activation effect was compared with pralidoxime, a commercially available drug, using the Ellman method in vitro.

    Results

    The results of the Ellman method showed that the percentage of activation of the paraoxon-inhibited enzyme by pralidoxime as a commercially available antidote and the synthesized nanoparticles at a concentration of 5 mM over 30 minutes was 88.6% and 39.3%, respectively.

    Conclusion

    This study demonstrated that the designed iron oxide nanoparticles conjugated with 2-pyridinium aldoxime can be synthesized and activate acetylcholinesterase inhibited by paraoxon. Therefore, future studies should investigate the effect of other groups such as 4-pyridinium aldoxime, 2,4-pyridinium aldoxime, etc., on the enzyme activation performance and the ability of these nanoparticles to cross the blood-brain barrier.

    Keywords: Antidotes, Nerve Agents, Blood-Brain Barrier, Cholinesterase Reactivators, Organophosphate Poisoning, Super Paramagnetic Iron Oxide Nanoparticles}
  • حمیدرضا مرادی، سحر عبدالهی نژاد، سهراب حیدریان*
    مقدمه

    بیماری های پارکینسون و آلزایمر دو بیماری شایع تحلیل برنده عصبی هستند که علت آن ها تا حد زیادی ناشناخته مانده است. این دو بیماری ویژگی های بیماری زایی مشابهی مانند از دست دادن پیشرونده نورون های خاص، و حضور پروتئین های انباشته شده دارند. اگزوزوم ها با ساختار دو لایه لیپیدی مشابه غشاء سلولی از اکثر سلول های مختلف بدن مشتق می شوند و به راحتی می توانند از انواع غشاهای بیولوژیک مانند سد خونی- مغزی عبور کنند. اگزوزوم ها برای انتقال واسطه ها و اطلاعات بین سلول ها مهم هستند. بنابراین، آن ها می توانند نقش حیاتی در شرایط طبیعی و پاتولوژی مغز، از جمله اختلالات عصبی مانند بیماری پارکینسون و بیماری آلزایمر داشته باشند. این مقاله نقش و کاربرد اگزوزوم ها در بیماری زایی و درمان بیماری های پارکینسون و آلزایمر را بررسی می کند.

    نتیجه گیری

    ساختار و بیوژنز اگزوزوم ها ممکن است نقش های مهمی در تشخیص و پیشرفت بیماری های تحلیل برنده عصبی داشته باشد. علاوه بر این، درک مکانیسم های پیچیده حاکم بر تشکیل و ترکیب اگزوزوم در شرایط پاتولوژی می تواند بینش ارزشمندی در مورد پاتوفیزیولوژی زمینه ای بیماری های پارکینسون و آلزایمر ارائه دهد.

    کلید واژگان: بیماری های تحلیل برنده عصبی, سد خونی- مغزی, سیستم اعصاب مرکزی, غشاء سلولی}
    Hamireza Moradi, Sahar Abdollahinezhad, Sohrab Heydarian*
    Introduction

    Parkinson's and Alzheimer's diseases are two common neurodegenerative diseases whose etiology remains largely unknown. These two diseases share similar pathogenesis features, including the progressive loss of specific neurons and the accumulation of deposited proteins. Exosomes, characterized by a lipid bilayer structure akin to that of the cell membrane, originate from various cells throughout the body and can readily traverse different biological membranes, such as the blood-brain barrier. Exosomes are important for the transfer of mediators and information between cells. Therefore, they can play a vital role in the normal and pathological conditions of the brain, including neurodegenerative disorders such as Parkinson’s disease and Alzheimer's disease. This article reviews the role and application of exosomes in the pathogenesis and treatment of Parkinson's and Alzheimer's diseases.

    Conclusion

    The structure and biogenesis of exosomes may play crucial roles in both the diagnosis and progression of neurodegenerative diseases. Moreover, understanding the complex mechanisms governing exosome formation and composition in pathological conditions could offer valuable insights into the underlying pathophysiology of Parkinson's and Alzheimer's diseases.

    Keywords: Neurodegenerative Diseases, Blood-Brain Barrier, Central Nervous System, Cell Membrane}
  • Morteza Fathi, Majid Hassanpourezatti*
    Background

    Vascular endothelial growth factor (VEGF) signaling pathway plays an important role in the pathogenesis of seizure. The oxidant/antioxidant factors and miRNA expression in the brain are differentially regulated in seizure.

    Objectives

    We aim to investigate the potential mechanism of action for the recombinant human VEGF (rhVEGF) in mice with maximal electroshock (MES)-induced seizure.

    Materials & Methods

    A total of 40 male mice (weight: 20-25 g) were treated intraperitoneally with normal saline, or rhVEGF (50, 100, and 150 µg/kg, daily for 4 consecutive days). One hour after the last injection, seizures were induced in each animal by MES. The latency for the onset of the first clonus and the duration of hind limb extension (HLE) were recorded. The levels of nitric oxide (NO), total antioxidant capacity (TAC), and micoRNA-142-5p expression were determined in the hippocampus of mice. Blood-brain barrier (BBB) permeability was also estimated by Evans blue dye extravasation method.

    Results

    The administration of rhVEGF at all doses significantly reduced the HLE duration. However, latency for the seizure onset increased after administration of 50 and 150 μg/kg rhVEGF and decreased after administration of 100 μg/kg rhVEGF. In the brain, the NO level decreased, while TAC level and microRNA-142-5p expression increased by rhVEGF treatment in mice with MES-induced seizure. Pretreatment with rhVEGF at doses of 100 and 150 μg/kg reversed the increase in BBB leakage induced by MES-induced seizures.

    Conclusion

    The rhVEGF administration can prevent MES-induced seizures by regulating NO, TAC, and miR-142-5 expression levels in the hippocampus and reducing BBB leakage in mice.

    Keywords: Vascular endothelial growth factor, Generalized tonic-clonic seizure, Blood-brain Barrier, microRNAs, mouse, Oxidative stress}
  • Seyyed mohammadtaghi Razavi-Toosi, Yasin Asadi, Nahid Aboutaleb, Masoumeh Faezi*
    Introduction

    Stem cells isolated from the amniotic membrane can produce and release substances that can regenerate damaged tissues and contain proteins and other factors that via numerous major and minor mechanisms lead to increasing angiogenesis and tissue survival. This research was conducted to prove the defensive characteristics of the secretome in the face of temporary focal cerebral ischemia in mouse stroke models.

    Methods

    Cerebral ischemia protocol in a specific area was implemented in rats with middle cerebral artery occlusion for 60 minutes and then reperfusion was given for 6, 20, and 30 minutes. Within 30 minutes after the start of reperfusion, conditioned medium derived from the human amniotic membrane (AMSC-CM) was poured into the right ventricle (ICV) at a dose of 0.5 µL. Finally, the volume of the injury, cerebral tissue water, sensorimotor activity, and the strength of the blood-brain barrier integrity were evaluated 24 hours after drug injection.

    Results

    ICV injection of conditioned medium at the start of reperfusion phase considerably decreased the volume of the injury 6, 20, and 30 hours after reperfusion compared to the MCAO-operated group (P<0.01). Cerebral tissue water in the treatment group decreased considerably after the intervention in comparison with the MCAO group in the core and penumbral area not in the subcortical area (P<0.05). Also, the amount of Evans blue infiltration at all times in the core and half-foot area in the AMSC-CM group was significantly reduced in parallel with the MCAO group (P<0.05). 

    Conclusion

    Treatment with AMSC-CM during 6-30 h after ischemia-reperfusion insult exerts some beneficial effects against ischemia-reperfusion injury. These findings provide an important vision for more complementary research and treatment of stroke.

    Keywords: Conditioned medium, Mesenchymal Stem Cells, Blood-brain barrier, Stroke, Rats}
  • آرش عبدالملکی*، مهدی تمجید، فرشته اشرفیان نانسا، فاطمه قلیوند، زهرا چاوشی لاهرود، نیلوفر بهاری، شوکر واسمن اسماعیل، ژیکال عمر خوندهور
    مقدمه

    مغز پیچیده ترین و تکامل یافته ترین اندام انسان است، بنابراین حفاظت از عملکرد آن امری حیاتی می باشد. سد خونی- مغزی که توسط سیستم میکروواسکولار مغز تشکیل شده است، یک نوار غشایی است که خون را از بخش خارج سلولی مغز در سیستم عصبی مرکزی اکثر مهره داران جدا می کند. سد خونی- مغزی یک لایه از سلول های اندوتلیال است که از پنج قسمت تشکیل شده است: پری سیت ها، آستروسیت ها، نورون ها، غشای پایه و بافت های همبند. سد خونی- مغزی مانع اصلی انتقال دارو به مغز است. برای رهاسازی موثر داروها در مغز، روش های مختلفی توسعه یافته است. در این میان، تحویل دارو با نانوذرات دارای مزایای بسیاری از جمله غیر تهاجمی بودن، مقرون به صرفه بودن، زیست تخریب پذیری بهتر و پایداری طولانی مدت است.

    نتیجه گیری

    بررسی ساختار و عملکرد سد خونی- مغزی و همچنین ارزیابی سیستم های مختلف موثر بر این ساختار به ویژه استفاده از نانوذرات می تواند نقش مهمی در کمک به درمان بیماری های سیستم عصبی مرکزی داشته باشد.

    کلید واژگان: نانوذرات, سد خونی مغزی, سیستم عصبی مرکزی}
    Arash Abdolmaleki*, Mehdi Tamjid, Fereshteh Ashrafiyan Nansa, Fatemeh Golivand, Zahra Chavoshi Lahrod, Niloufar Bahari, Shukur Wasman Smail, Zhikal Omar Khudhur
    Introduction

    The brain is the most complex and evolved human organ, so protecting its function is a vital issue. The blood-brain barrier formed by the microvascular system of the brain is a membrane strip that separates the blood from the extracellular compartment of the brain in the central nervous system of most vertebrates. The blood-brain barrier is a single layer of endothelial cells that consists of five parts: pericytes, astrocytes, neurons, basement membrane, and connective tissues. The blood-brain barrier is a major barrier to drug delivery to the brain. To effectively release drugs into the brain, various methods have been developed. Among them, drug delivery with nanoparticles has many advantages, including non-invasiveness, cost-effectiveness, better biodegradability, and long-term stability.

    Conclusion

    Investigating the structure and function of the blood-brain barrier, as well as the evaluation of various systems affecting this structure, especially the use of nanoparticles, can play an important role in helping to treat central nervous system diseases.

    Keywords: Nanoparticles, Blood-Brain Barrier, Central Nervous System}
  • Marziyeh Soltani, Yousef Mirzaei, Ali Hussein Mer, Mina Mohammad-Rezaei, Zahra Shafaghat, Soheila Fattahi, Fatemeh Azadegan-Dehkordi, Meghdad Abdollahpour-Alitappeh, Nader Bagheri

    Schizophrenia is one of the most severely debilitating mental disorders that affects 1.1% of the world's population. The exact cause of the disease is not known, but genetics, environmental factors (such as infectious agents, season and region of birth, exposure to viruses, low birth weight, advanced paternal age, and tobacco), and immune system dysfunction can all contribute to the development of schizophrenia. Recently, the role of the immune system in schizophrenia has received much attention. Both acquired and innate immune systems are involved in the pathogenesis of schizophrenia and facilitate the disease's progression. Almost all cells of the immune system including microglia, B cells, and T cells play an important role in the blood-brain barrier damage, inflammation, and in the progression of this disease. In schizophrenia, the integrity of the blood-brain barrier is reduced and then the immune cells are recruited into the endothelium following an increase in the expression of cell adhesion molecules. The entry of immune cells and cytokines leads to inflammation and antibody production in the brain. Accordingly, the results of this study strengthen the hypothesis that the innate and acquired immune systems are involved in the pathogenesis of schizophrenia.

    Keywords: Blood-brain barrier, Immune system, Inflammation, Cytokines, Psychoneuroimmunology, Schizophrenia, Therapeutics}
  • زهرا جاوید، مهناز طاهریان فرد*، جواد ساجدیان فرد، حمیدرضا مرادی
    زمینه و هدف

    بیماری های دمیلینه کننده از اختلالات اتوایمیون سیستم عصبی مرکزی هستند. هدف از مطالعه حاضر تاثیر عصاره آبی-الکلی زعفران بر تخریب سد خونی-مغزی، توزیع پروتئین اسید فیبریلاری گلیال و فاکتور رونویسی الیگودندروسیت 2 در سلولهای الیگودندروسیت و آستروسیت و تعداد سلول های مذکور است.

    روش ها

    از 40 موش صحرایی ماده بالغ نژاد اسپراگ داولی در چهار گروه شم، کنترل درمان، درمان زعفران، درمان اینفلکسیمب استفاده شد. دمیلیناسیون با تزریق اتیدیوم بروماید 0/01 درصد در شکنج دندانه دار القا شد. توزیع پروتئین اسید فیبریلاری گلیال و فاکتور رونویسی الیگودندروسیت 2 سلول های اولیگودندروسیت و آستروسیت توسط ایمونوهیستوشیمی شناسایی و توسط نرم افزار آنالیزور تصویری بررسی شد. وسعت دمیلیناسیون توسط رنگ آمیزی لوکسول فست بلو مشخص و با برنامه تصویر-j مورد ارزیابی قرار گرفت. وسعت تخریب سد خونی-مغزی توسط تکنیک الایزا و اندازه گیری میزان خروج اوانس بلو از رگ مورد بررسی قرار گرفت.

    یافته ها

    تخریب در سد خونی-مغزی به طور معنی داری (0/05<p ) در گروه کنترل درمان در مقایسه با سایر گروه ها بیشتر بود. توزیع پروتئین اسید فیبریلاری گلیال و فاکتور رونویسی الیگودندروسیت 2 سلول های اولیگودندروسیت و آستروسیت و تعداد این سلول ها به طور قابل توجهی (0/05<p ) در گروه کنترل درمان نسبت به سایر گروه ها کمتر بود. 

    نتیجه گیری

    عصاره آبی-الکلی زعفران باعث بهبود سد خونی-مغزی، افزایش توزیع پروتئین اسید فیبریلاری گلیال و فاکتور رونویسی الیگودندروسیت 2 بر روی سلول های آستروسیت و الیگودندروسیت و تعداد این سلول ها در گروه درمان زعفران شده است.

    کلید واژگان: پروتئین اسید فیبریلاری گلیال, دمیلینه شدن, سد خونی مغزی, فاکتور رونویسی الیگودندروسیت 2}
    Zahra Javid, Mahnaz Taherianfard*, Javad Sajedianfard, Hamid Reza Moradi
    Background and aim

     Demyelinating diseases are autoimmune disorders of the nervous system. The present study aims to effect of the hydro-alcoholic extract of saffron on blood brain-barrier destruction, Glial fibrillary acidic protein, and Oligodendrocyte transcription factor 2 distribution in astrocyte and oligodendroglia cells and the number of mentioned cells. 

    Methods

     Forty female Sprague Dawley adult rats in four groups, Sham, treatment control, saffron treatment, and infliximab treatment were used. Demyelination was induced by 3 µl of 0.01% ethidium bromide injection in the dentate gyrus. Immunohistochemistry was used, for the distribution of Glial fibrillary acidic protein and Oligodendrocyte transcription factor 2 determination. The Image Analyzer software have evaluated the distribution of Glial fibrillary acidic protein and Oligodendrocyte transcription factor 2. Demyelination was determined by Luxol fast blue staining and image-j software. The ELISA method and Evans blue extravasation measurement have investigated the extent of blood brain-barrier disruption.

    Results

     Blood brain-barrier disruption was meaningfully (p < 0.05) more in the treatment control compared to the other groups. The distribution of Glial fibrillary acidic protein and Oligodendrocyte transcription factor 2 and the number of both cells were considerably (p < 0.001) lower in the treatment control relative to the other groups. Glial fibrillary acidic protein and Oligodendrocyte transcription factor 2 remarkably (p < 0.05) were lesser in the treatment control compared to the other groups. 

    Conclusion

     The hydro-alcoholic extract of saffron ameliorates blood brain-barrier destruction, improved Glial fibrillary acidic protein, and Oligodendrocyte transcription factor 2 distribution and number of astrocyte and oligodendroglia cells.

    Keywords: ‎Glial fibrillary acidic protein, Demyelination, Blood Brain Barrier, Oligodendrocyte transcription factor}
  • سهراب حیدریان*
    مقدمه

    گلیوبلاستوما مولتی فرم (GBM) یکی از شایع ترین تومورها با مرگ و میر بالا و ماهیت بسیار تهاجمی است. سد خونی- مغزی (BBB) ورود داروهای شیمی درمانی به بافت تومور را محدود می کند. مطالعات اخیر نشان داده است که کاربرد انواع نانوحامل های دارویی به منظور درمان GBM می تواند راهبردهای جدید و موثر را ارایه دهد. این نانوحامل ها برای تحویل هدفمند دارو موفق عمل کرده اند و عمدتا شامل نانولوله های کربنی، نانوذرات فلزی، نانوامولسیون ها، میکروکپسول ها، میسل های پلی مری دندریمرها، نیوزوم ها، لیپوزوم ها و فیتوزوم می باشند. قابلیت تحویل دارو به صورت دقیق و مستقیم به سلول های سرطانی GBM توسط نانوحامل ها توانسته است موجب افزایش اثربخشی و کاهش عوارض جانبی داروهای شیمی درمانی تجویزی شود.  

    نتیجه گیری

    در این مقاله مروری، استفاده از انواع مختلف نانوحامل های دارویی در درمان گلیوبلاستوما مولتی فرم مرور شده است و دیدگاه های درمانی آینده این روش برای غلبه بر محدودیت های درمانی GBM مورد بحث قرار گرفته است.

    کلید واژگان: گلیوبلاستوما, سد خونی-مغزی, نانوتکنولوژی}
    Sohrab Heydarian*
    Introduction

    Glioblastoma multiforme (GBM) is one of the most common tumors with high mortality and a very aggressive nature. Blood-brain barrier (BBB) limits the penetration of chemotherapy drugs into the tumor tissue. Recent studies have shown that the use of various drug nanocarriers to treat GBM can provide new and effective strategies. These nanocarriers have been successful in targeted drug delivery and mainly include carbon nanotubes, metal nanoparticles, nanoemulsions, microcapsules, polymeric micelles, dendrimers, niosomes, liposomes, and pyrosomes. The ability to accurately and directly deliver drugs to GBM cells via nanocarriers has been able to increase the effectiveness and reduce the side effects of chemotherapy drugs.

    Conclusion

    In this review article, the use of different types of drug nanocarriers is reviewed in the treatment of glioblastoma multiforme, and the future therapeutic perspectives of this method are discussed to overcome the limitations of GBM therapy.

    Keywords: Glioblastoma, Blood-Brain Barrier, Nanotechnology}
  • Soheil Mehrdadi *

    Brain, predisposed to local and metastasized tumors, has always been the focus of oncological studies. Glioblastoma multiforme (GBM), the most common invasive primary tumor of the brain, is responsible for 4% of all cancer-related deaths worldwide. Despite novel technologies, the average survival rate is 2 years. Physiological barriers such as blood-brain barrier (BBB) prevent drug molecules penetration into brain. Most of the pharmaceuticals present in the market cannot infiltrate BBB to have their maximum efficacy and this in turn imposes a major challenge. This mini review discusses GBM and physiological and biological barriers for anticancer drug delivery, challenges for drug delivery across BBB, drug delivery strategies focusing on SLNs and NLCs and their medical applications in on-going clinical trials. Numerous nanomedicines with various characteristics have been introduced in the last decades to overcome the delivery challenge. Solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) were introduced as oral drug delivery nanomedicines which can be encapsulated by both hydrophilic and lipophilic pharmaceutical compounds. Their biocompatibility, biodegradability, lower toxicity and side effects, enhanced bioavailability, solubility and permeability, prolonged half-life and stability and finally tissue-targeted drug delivery makes them unique among all.

    Keywords: Brain tumors, Glioblastomamultiforme, Blood-brain barrier, Solid lipid nanoparticles, Nanostructured lipid carriers, Brain drug delivery}
  • Hariom Kumar, G.T. Kulkarni, Vishal Diwan, Bhupesh Sharma*
    Introduction

    A neurodevelopmental disorder, autism is typically identified with three primary behavioral consequences, such as social impairment, communication problems, and limited or stereotypical behavior. Because of its co-morbidity and lack of therapeutic options, autism is a global economic burden. A short chain of fatty acid, propionic acid is formed biologically by the gut microbiome. Propionic acid levels that are too high can cause leaky intestines, which can lead to autism-like symptoms.

    Methods

    To induce autism, male Albino Wistar rats were given propionic acid (250 mg/kg/po on the 21st, 22nd, and 23rd postnatal days). Rats also received a ryanodine receptor antagonist (Ruthenium red: 3 mg/kg/po; postnatal 21st to 50th day) to see what influence it had on propionic acid-induced autism. Anxiety, social behavior, and repeated behaviors were all assessed, as well as oxidative stress, inflammatory indicators, neuro signaling proteins, and blood-brain barrier permeability.

    Results

    Ruthenium red was found to counter the propionic acid-induced increases in anxiety, repetitive behavior prefrontal cortex levels of IL-6, TNF-α, TBARS, Evans blue leakage, and water content along with decreases in social behavior, IL-10, and GSH followed by hippocampus CREB and BDNF levels. 

    Conclusion

    Ryanodine receptor antagonists presented a neuroprotective effect in propionic acid-induced conditions like autism by modulatory effects on social and repetitive behavior, oxidative stress, neuroinflammation, and neuroprotein changes. Ryanodine receptors can be further explored in depth to manage autism as a condition.

    Keywords: BDNF, Ryanodine, Interleukins-6, Mitochondria, Blood brain barrier, ASST}
  • Tahmineh Mokhtari, Maryam Shayan, Amirmohammad Rezaei Rashnudi, Gholamreza Hassanzadeh *, Kobra Mehran Nia
    Objective(s)
    Intracerebroventricular (ICV) injections of mesenchymal stem cells (MSCs) may improve the function and structure of blood-brain barrier (BBB), possibly by preserving the BBB integrity. This study examined the impact of Wharton’s jelly (WJ)-MSCs on cognitive dysfunction and BBB disruption following a protracted hypoxic state.
    Materials and Methods
    Twenty-four male Wistar rats were randomly studied in four groups: Control (Co): Healthy animals, Sham (Sh): Rats were placed in the cage without hypoxia induction and with ICV injection of vehicle, Hypoxic (Hx)+vehicle: Hypoxic rats with ICV injection of vehicle (5 μl of PBS), and Hx+MSCs: Hypoxic rats with ICV injection of MSCs. Spatial learning and memory were evaluated one week after WJ-MSCs injection, and then animals were sacrificed for molecular research.
    Results
    Hypoxia increased latency and lowered the time and distance required reaching the target quarter, according to the findings. Furthermore, hypoxic rats had lower gene expression and protein levels of hippocampus vascular endothelial (VE)-cadherin, claudin 5, and tricellulin gene expression than Co and Sh animals (P<0.05). Finally, administering WJ-MSCs after long-term hypoxia effectively reversed the cognitive deficits and prevented the BBB breakdown via the upregulation of VE-cadherin, claudin 5, and tricellulin genes (P<0.05).
    Conclusion
    These findings suggest that prolonged hypoxia induces spatial learning and memory dysfunction and increases BBB disruption, the potential mechanism of which might be via reducing VE-cadherin, claudin 5, and tricellulin genes. Hence, appropriate treatment with WJ-MSCs could reverse ischemia adverse effects and protect the BBB integrity following prolonged hypoxia.
    Keywords: Blood-brain barrier, Hippocampus, Hypoxia, Intraventricular, Mesenchymal stem cells, Wharton jelly}
  • محمود رمدان، محمدرضا بیگدلی*، حسین شاهسوارانی*، سلما احمدلو
    زمینه و هدف

    سکته مغزی دومین عامل مرگ و میر و ناتوانی جسمی در جهان می باشد. سلول درمانی به دلیل توانایی در ترمیم و جایگزینی نورون های آسیب دیده می تواند به عنوان گزینه مناسبی در نظر گرفته شود. سلول های بنیادی جنینی موش (mESCs) به دلیل آزادسازی فاکتورهای رشد و نروتروفیک به عنوان کاندید  بالقوه برای تحقیق حاضر در نظر گرفته شد.

    روش ها :

    موش ها به سه گروه اصلی شاهد، کنترل و گیرنده پیوند سلول های بنیادی جنینی موش (mESCs) تقسیم بندی شدند. درگروه mESCs، القای ایسکمی مغزی به روش انسداد شریان مغزی میانی (MCAO) ، چهار روز پس از تزریق نیم میلیون سلول به استریاتوم مغز موش به روش جراحی استریوتاکسی صورت گرفت. موش ها به زیرگروه هایی جهت بررسی نواقص نورولوژیک، حجم انفارکتوس، میزان استحکام سد خونی-مغزی، ادم، بررسی فعالیت آنزیم سوپر اکسید دسموتاز (SOD) و بیان ژن های گیرنده یونوتروپیک گلوتامات GRIN1 و نیتریک اکسید سنتاز NOS1 تقسیم بندی شدند.

    یافته ها: 

    در گروه mESCs، کاهش معناداری در نواقص نورولوژیک، حجم انفارکتوس، آسیب سد خونی-مغزی و ادم نسبت به گروه کنترل مشاهده گردید. همچنین بیان ژن هایGRIN1 و NOS1 کاهش معناداری را در گروه mESCs نسبت به گروه کنترل نشان داد. همچنین افزایش معنادار بیان آنزیم SOD در گروه mESCs نسبت به گروه کنترل مشاهده گردید.

    نتیجه گیری :

    نتایج مطالعه حاضر نشان داده است که پیوندmESCs  موجب کاهش آسیب های ایسکمی گردید که احتمالا بخشی از این اثربه واسطه کاهش بیان ژن های مرتبط با التهاب و تحریک پذیری نورونی و افزایش آنزیم آنتی اکسیدانی است.

    کلید واژگان: ادم, انفارکتوس, سد خونی-مغزی, سلول های بنیادی جنینی, نقایص نورولوژیک}
    Mahmoud Ramdan, MohammadReza Bigdeli*, Hosein Shahsavarani, Salma Ahmadloo
    Background and Aim

    Stroke is the second cause of death and disability in the world. Cell therapy can be considered as a suitable option due to its ability to restore and replace damaged neurons. Mouse embryonic stem cells (mESCs) were considered as potential candidates for the present research due to the release of growth and neurotrophic factors.

    Methods

    Rats were divided into three main groups: Intact, control, and mouse embryonic stem cell (mESCs) transplant recipients. In mESCs group, ischemia was induced by middle cerebral artery occlusion (MCAO) method four days after injection of half a million cells into striatum of rat brain by stereotaxic method. Rats were divided into subgroups to investigate neurological deficits, infarct volume, blood-brain barrier integrity, edema, superoxide dismutase (SOD) enzyme activity, and the expression of glutamate ionotropic receptor (GRIN1) and nitric oxide synthase (NOS1) genes.

    Results

    A significant reduction in neurological deficits, infarct volume, blood-brain barrier damage and edema was observed in the mESCs group compared to the control group. Also, the expression of GRIN1 and NOS1 showed a significant decrease in the mESCs group compared to the control group. In addition, a significant increase in SOD enzyme expression was observed in the mESCs group compared to the control group.

    Conclusion

    The present results showed that transplantation of mESCs reduced ischemic damages. This effectiveness is probably due to reduction of inflammatory and excitotoxicity related-genes and increase of antioxidant enzyme.

    Keywords: Edema, Infarct volume, Blood-brain barrier, Embryonic stem cells, Neurological defects}
  • علیرضا تقی زاده، فروزان صادقی محلی، علی سیاه پشت خاچکی*
    سابقه و هدف

    آسیب مغزی ناشی از تروما (TBI) یکی از پیچیده ترین بیماری های شناخته شده درسیستم عصبی مرکزی می باشد. نسفاتین یک پلیپپتید 82-آمینو اسید است که بر سیستم عصبی مرکزی موثر است. در این پژوهش نقش محافظت نورونی نسفاتین در فرایند ضربه مغزی منتشر در موش صحرایی و هم چنین تاثیر آن روی میزان ماتریکس متالوپروتییناز-9 (MMP-9) بررسی شد.

    مواد و روش ها: 

    در این مطالعه تجربی، 56 موش صحرایی نر نژاد ویستار به روش مارمارو تحت TBI قرار گرفتند و 30 دقیقه بعد نسفاتین با دوزهای مختلف به صورت داخل صفاقی تزریق و در روز دوم و سوم تکرار شد. در زمان های قبل از ضربه، بلافاصله پس از به هوش آمدن پس از القا ضربه، 24، 48 و 72 ساعت بعد از ضربه، Veterinary Coma Scale و تست های حرکتی و تعادلی Beam Walk و Beam Balance در رت ها ثبت شد. پس از 72 ساعت،CSF از سیسترنا مگنا جمع آوری و جهت انجام آزمایش الایزا برای بررسی ماتریکس متالوپروتییناز-9 استفاده شد.

    یافته ها:

     تجویز داخل صفاقی نسفاتین در دوزهای0 5 و µg/kg 10 می تواند باعث کاهش ادم مغزی، تخریب سد خونی مغزی و نمرات عصبی و تعادلی حرکتی (0/0001<p) در مقایسه با گروه کنترل (شم و سالم) شود (0/001<p). لازم به ذکر است که نسفاتین در دوز µg/kg 20 موثر نبود (0/05>P).

    استنتاج

    نسفاتین توانسته است بر پیامدهای تروما تاثیر بگذارد و ادم مغزی را کاهش دهد، بهبود سد خونی مغزی و نمرات عصبی و تعادلی را تسریع بخشد. لازم به ذکر است احتمالا بخشی از این اثرات محافظتی عصبی نسفاتین از طریق کاهش MMP-9 می باشد.

    کلید واژگان: نسفاتین, نوروپروتکتیو, ترومای مغزی, ادم مغزی, سد خونی- مغزی, MMP-9}
    Alireza Taghizadeh, Forouzan Sadeghimahalli, Ali Siahposht –Khachaki*
    Background and purpose

    Traumatic brain injury (TBI) is one of the most complex diseases of the central nervous system (CNS). Nesfatin is an 82-amino acid effective polypeptide in CNS. In this study, we investigated the role of nesfatin in neuron protection in the process of diffuse concussion in rats and also its effect on the level of matrix metalloproteinase-9.

    Materials and methods

    In this experimental study, 56 Wistar rats were subjected to TBI by the Marmaru method. After 30 minutes, nesfatin was injected intraperitoneally at different doses, and repeated on second and third days. The results of Veterinary Coma Scale, beam walk and beam balance tests were recorded at Pre-TBI, immediately after TBI, 24, 48, and 72 hours after TBI. After 72 hours, CSF was collected from the cisterna magna and used for ELISA test to measure the matrix metalloproteinase-9.

    Results

    Intraperitoneal administration of nesfatin at 50 µg/kg and 10 µg/kg can reduce cerebral edema, destruction of the blood-brain barrier, and neurological and balance-motor scores (P<0.0001) compared with the control group (Sham and Intact) (P<0.0001). Nesfatin was not found effective at 20µg/kg (P>0.05).

    Conclusion

    Nesfatin has been able to affect the consequences of trauma and reduce brain edema, accelerate the improvement of blood-brain barrier and neurological and balance scores. Probably part of these neuroprotective effects is through reduction of MMP-9.

    Keywords: nesfatin, neuroprotective, brain trauma, cerebral edema, blood-brain barrier, MMP-9}
  • Nazanin Kianinejad, Young Min Kwon*

    The delivery of chemotherapies to brain tumors faces the difficult task of crossing the blood-brain barrier (BBB).1-4 The brain capillary endothelial cells (BCECs) along with other cell lines, such as astrocytes and pericytes, form the BBB. This highly selective semipermeable barrier separates the blood from the brain parenchyma. The BBB controls the movement of drug molecules in a selective manner5 and maintains central nervous system (CNS) homeostasis. Depending on the properties of drugs such as their hydrophilic-lipophilic balance (HLB), some can cross the BBB through passive diffusion.6 However, this approach alone has not led to successful drug developments due to low net diffusion rates and systemic toxicity. Although the use of nanomedicine has been proposed to overcome these drawbacks, many recent studies still rely on the so-called ‘enhanced permeability and retention (EPR)’ effect though there is a realization in the field of drug delivery that EPR effect may not be sufficient for successful drug delivery to brain tumors. Since, compared to many other solid tumors, brain tumors pose additional challenges such as more restrictive blood-tumor barrier as well as the well-developed lymphatic drainage, the selection of functional moieties on the nanocarriers under consideration must be carried out with care to propose better solutions to this challenge.

    Keywords: Blood-brain-barrier, Targeted delivery, EPR effect, Lymphatic drainage, Brain tumors, Nanomedicine}
  • سید رضا سیدین، مسلم محمدی، علی سیاه پشت خاچکی*
    سابقه و هدف

    آلیسین دارای طیف گسترده‎ای از عملکردهای دارویی است که همه آن‎ها را می‎توان در فعالیت‎های ضد التهابی، آنتی‎اکسیدانی، ضدقارچی و ضدتومور نشان داد، لذا در این مطالعه نقش محافظت نورونی آلیسین در فرایند ضربه مغزی منتشر در موش صحرایی و هم چنین تاثیر آن بر روی میزان اینترلوکین‎ها و تغییرات هیستولوژیکی مورد بررسی قرار گرفت.

    مواد و روش ها: 

    در این مطالعه مداخله ای تجربی، 56 سر موش صحرایی نر نژاد ویستار پس از القای بیهوشی و کانول‎گذاری در نای، تحت ضربه مغزی کنترل شده منتشر به روش مار مارو قرار گرفتند و 30 دقیقه بعد به رت‎‎ها داروی آلیسین با دوزهای مختلف به صورت داخل صفاقی تزریق شد. در زمان های قبل از ضربه، بلافاصله پس از به هوش آمدن پس از القای ضربه، 24، 48 و 72 ساعت بعد از ضربه Veterinary Coma Scale و تست های حرکتی و تعادلی Beam Walk و Beam Balance از رت ها گرفته و ثبت شد.

    یافته ها:

     آلیسین در دوزهای 25 و 50 mg/kg می تواند اختلاف موارد ذکرشده  را نسبت به گروه کنترل (Sham و Intact) کم تر کند (0/001>P). لازم به ذکر است آلیسین در دوز 25 mg/kg موثرتر بوده است (0/0001>P).

    استنتاج

    براساس این مطالعه می توان دریافت که آلیسین دارای اثرات محافظت نورونی در مغز بود و توانسته بر روی پیامدهای ناشی از ضربه مغزی اثر بگذارد و هم چنین می تواند موجب کاهش ادم مغزی، تسریع در بهبود وضعیت سد خونی- مغزی و نمرات نورولوژیکی و تعادلی شود.

    کلید واژگان: آلیسین, نوروپروتکتیو, ترومای مغزی, ادم مغزی, سد خونی- مغزی, اینترلوکین ها}
    Seyed Reza Seyedin, Moslem Mohammadi, Ali Siahposht-Khachaki*
    Background and purpose

    Allicin has a wide range of pharmacological functions, all of which can be demonstrated in anti-inflammatory, antioxidant, antifungal and anti-tumor activities. In this research, we investigated the neuroprotective role of allicin in the process of diffuse traumatic brain injury and its effect on interleukin levels and histological changes in rats.

    Materials and methods

    In this experimental study, Wistar rats (n=56) underwent diffuse controlled brain injury by Marmarou method, and 30 minutes later, the drug was injected intraperitoneally at different doses. Veterinary Coma Scale was used and Beam Walk and Beam Balance movement and balance tests were taken at pre-traumatic times, immediately after recovery from the trauma, and 24, 48, and 72 hours after the trauma.

    Results

    Findings showed that allicin at 25 mg/kg and 50 mg/kg can reduce these differences compared to the control group (Sham and Intact) (P<0.001). Allicin was found to be more effective at 25 mg/kg (P<0.0001).

    Conclusion

    Allicin showed neuroprotective effects in the brain and can affect the consequences of trauma, reduce cerebral edema, accelerate improvement of blood-brain barrier, and improve neurological scores and the function of vestibular system.

    Keywords: allicin, neuroprotective, brain trauma, brain edema, blood-brain barrier, interleukins}
  • Ahmad Alinaghi Langari, Nazanin Sabet, Hamideh Bashiri, Sobhan Mohammadi Jorjafki, Reyhaneh Rezaie, Nafise Esmaeilpour, Sedigheh Amiresmaili, Mohammad Khaksari, Shahryar Dabiri, Mahmoud Amiri, Zahra Soltani *
    Background
    In our previous studies, the effect of sex hormones on brain edema reduction after traumatic brain injury (TBI) was demonstrated. In the current study, alone and combined effects of 17-β estradiol (E2) and vitamin D (Vit D) on TBI in male rats were investigated.
    Methods
    Male rats were divided into six groups, including sham, TBI, vehicle, E2, Vit D, and E2+Vit D. In all groups except sham, moderate-intensity diffuse TBI was induced by the Marmarou’s method. Vehicle, E2, Vit D and their combination were intramusculary injected one and 12 hours after the TBI. The brain water content, permeability of blood brain barrier (BBB) and histopathological outcome were assessed 24h after TBI. The neurological outcome score was determined using the veterinary coma scale (VCS).
    Results
    Significant reductions in brain water content (P<0.001, P<0.05 and P<0.01, respectively) and BBB permeability (P<0.001) appeared in the treated groups with E2, Vit D, and E2+Vit D compared to the vehicle group. Twenty-four hours after the injury, the neurological scores in the E2, Vit D, and E2+Vit D groups increased significantly compared to the vehicle group (P<0.05). Dramatic improvement in histopathological outcome was also observed in the treated groups compared to the vehicle group.
    Conclusion
    Alone and combined consumption of estrogen and vitamin D may similarly decrease the development of brain edema and improve the neurological and histopathological consequences of TBI. Therefore, consumption of vitamin D did not enhance the neuroprotective effect of estrogen in TBI.
    Keywords: Traumatic brain injury, Vitamin D, estrogen, Brain edema, Blood brain barrier}
  • Avinash Ramrao Tekade*, Pradeep Sadanand Mittha, Charushila Sopan Pisal

    Neurological disorders such as Alzheimer’s disease, Parkinson’s disease, dementia, epilepsy,depression, migraine etc. are affecting more and more elderly people’s day by day. Conventionalroute of administration to treat these diseases has to face a major hindrance that is bloodbrain and blood-cerebrospinal fluid (CSF) barrier to achieve desired concentration of drug atthe site of action for therapeutic effect. Hence, intranasal route of delivery is considered aspromising and alternative route to achieve desired goals. In last four decades, brain targetingstrategies are widely studied and considered having great potential by researchers; especiallyintranasal delivery owing to its benefits. Various nano formulations such as nanoemulsions,nanosuspensions, hydrogels, in situ gels, dendrimers and lipidic formulations are studiedwidely. Lipid nano formulations especially second generation nanostructured lipid carriersoffer greater advantages in terms of stability, fabrication techniques, scalability, drug loadingand drug targeting. Nanostructured lipid carrier (NLCs) constitute of two major componentsviz solid lipid and liquid lipid in a specific ratio. In this review, authors have discussed aboutthe possible synergistic actions of oils/liquid lipids with synthetic drugs resulting into greattherapeutic benefits.

    Keywords: CNS targeting, Blood brain barrier, Intranasal route, NLC, Liquid lipid, Synergistic action}
  • Rajalakshmi Rajendran, Krishnakumar Neelakandha Menon*, Sreeja Chandrasekharan Nair*

    Schizophrenia is a neuropsychiatric disorder mainly affecting the central nervous system (CNS), presented with auditory and visual hallucinations, delusion and withdrawal from society. Abnormal dopamine levels mainly characterise the disease; various theories of neurotransmitters explain the pathophysiology of the disease. The current therapeutic approach deals with the systemic administration of drugs other than the enteral route, altering the neurotransmitter levels within the brain and providing symptomatic relief. Fluid biomarkers help in the early detection of the disease, which would improve the therapeutic efficacy. However, the major challenge faced in CNS drug delivery is the blood-brain barrier (BBB). Nanotherapeutic approaches may overcome these limitations, which will improve safety, efficacy, and targeted drug delivery. This review article addresses the main challenges faced in CNS drug delivery and the significance of current therapeutic strategies and nanotherapeutic approaches for a better understanding and enhanced drug delivery to the brain, which improve the quality of life of schizophrenia patients.

    Keywords: Schizophrenia, Blood-brain barrier, Biomarkers, Drug delivery systems, Nanotechnology}
  • Mahsa Eskandari, Ali Awsat Mellati*
    Purpose

    blood-brain barrier (BBB) is made of specialized cells that are responsible for the selective passage of substances directed to the brain. The integrated BBB is essential for precise controlling of the different substances passage as well as protecting the brain from various damages. In this article, we attempted to explain the role of liver X receptor (LXR) in maintaining BBB integrity as a possible drug target.

    Methods

    In this study, various databases, including PubMed, Google Scholar, and Scopus were searched using the following keywords: blood-brain barrier, BBB, liver X receptor, and LXR until July, 2020. Additionally, contents close to the subject of our study were surveyed.

    Results

    LXR is a receptor the roles of which in various diseases have been investigated. LXR can affect maintaining BBB by affecting various ways such as ATP-binding cassette transporter A1 (ABCA1), matrix metalloproteinase-9 (MMP9), insulin-like growth factor 1 (IGF1), nuclear factor-kappa B (NF-κB) signaling, mitogen-activated protein kinase (MAPK), tight junction molecules, both signal transducer and activator of transcription 1 (STAT1), Wnt/β-catenin Signaling, transforming growth factor beta (TGF-β) signaling, and expressions of Smad 2/3 and Snail.

    Conclusion

    LXR could possibly be used either as a target for drug delivery to brain tissue or as a target for maintaining the BBB integrity in different diseases; thereby the drug will be conducted to tissues, other than the brain. If it is verified that only LXRα is necessary for protecting BBB, some specific LXRα ligands must be found and then used in medication.

    Keywords: Blood-brain barrier, Liver X receptor, Drug delivery, LXR, BBB}
نکته
  • 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.
درخواست پشتیبانی - گزارش اشکال