Adeno-associated viral vector

腺相关病毒载体
  • 文章类型: Journal Article
    近年来,基因治疗取得了长足的进步。4000多个蛋白质编码基因与6000多种遗传疾病有关;下一代测序极大地彻底改变了遗传疾病的诊断。大多数遗传疾病被认为是非常罕见的或超罕见的,这里定义为少于1:100,000的案例,但在12种已批准的基因疗法(不包括RNA疗法)中,只有一种针对的是超微病变.本文探讨了三种适用于多种罕见遗传病的基因补充治疗方法:慢病毒载体修饰的自体CD34+造血干细胞移植,将腺相关病毒(AAV)载体全身递送至肝脏,和局部AAV递送到脑脊液和大脑。连同RNA疗法,我们为这些基因疗法提出了一个潜在的商业模式。
    Gene therapy has made considerable strides in recent years. More than 4000 protein-coding genes have been implicated in more than 6000 genetic diseases; next-generation sequencing has dramatically revolutionized the diagnosis of genetic diseases. Most genetic diseases are considered very rare or ultrarare, defined here as having fewer than 1:100,000 cases, but only one of the 12 approved gene therapies (excluding RNA therapies) targets an ultrarare disease. This article explores three gene supplementation therapy approaches suitable for various rare genetic diseases: lentiviral vector-modified autologous CD34+ hematopoietic stem cell transplantation, systemic delivery of adeno-associated virus (AAV) vectors to the liver, and local AAV delivery to the cerebrospinal fluid and brain. Together with RNA therapies, we propose a potential business model for these gene therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    脊髓性肌萎缩症(SMA)是最常见的遗传性疾病之一,直到最近,婴儿死亡的主要遗传原因。三种疾病修饰治疗方法极大地改变了受严重影响的婴儿(SMA1型)的疾病轨迹和结果。特别是在症状前阶段开始。这些治疗方法之一是基于腺相关病毒载体9(AAV9)的基因疗法,它是全身性递送的,并已被欧洲医学机构批准用于具有多达三个SMN2基因拷贝或具有SMA1型临床表现的SMA患者。虽然这种广泛的适应症为患者选择提供了灵活性,对于支持治疗的证据有限或没有证据的患者,这也引起了人们对风险-获益比的担忧.2020年,我们召集了一个欧洲神经肌肉专家工作组,以支持合理使用onasemnogeneabeparvovec,采用改进的德尔菲法。三年后,我们已经召集了一个类似但更大的欧洲专家小组,他们评估了新出现的证据,即asemnogeneabeparvovec在治疗老年和体重较重的SMA患者中的作用,整合来自最近临床试验和现实世界证据的见解。这项努力导致了12项协商一致声明,在9个方面达成了强烈共识,在其余3个方面达成了共识,反映了onasemnogeneabeparvovec在治疗SMA中不断发展的作用。
    Spinal muscular atrophy (SMA) is one of the most common genetic diseases and was, until recently, a leading genetic cause of infant mortality. Three disease-modifying treatments have dramatically changed the disease trajectories and outcome for severely affected infants (SMA type 1), especially when initiated in the presymptomatic phase. One of these treatments is the adeno-associated viral vector 9 (AAV9) based gene therapy onasemnogene abeparvovec (Zolgensma®), which is delivered systemically and has been approved by the European Medicine Agency for SMA patients with up to three copies of the SMN2 gene or with the clinical presentation of SMA type 1. While this broad indication provides flexibility in patient selection, it also raises concerns about the risk-benefit ratio for patients with limited or no evidence supporting treatment. In 2020, we convened a European neuromuscular expert working group to support the rational use of onasemnogene abeparvovec, employing a modified Delphi methodology. After three years, we have assembled a similar yet larger group of European experts who assessed the emerging evidence of onasemnogene abeparvovec\'s role in treating older and heavier SMA patients, integrating insights from recent clinical trials and real-world evidence. This effort resulted in 12 consensus statements, with strong consensus achieved on 9 and consensus on the remaining 3, reflecting the evolving role of onasemnogene abeparvovec in treating SMA.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    We previously demonstrated that cross-talk between α5β1 integrin and the angiopoietin-1 (Ang1) / Tie2 receptor plays an important role in regulating brain endothelial angiogenic responses in the ischemic penumbra following cerebral ischemic stroke (CIS). However, a recent study suggested that stimulation of the α5β1 integrin also has the potential of increasing blood-brain barrier (BBB) permeability after CIS, raising doubt about whether α5β1 integrin stimulation by itself will protect against ischemic injury. In light of these conflicting roles, the goal of this study was to evaluate the impact of co-overexpression of α5 integrin and Ang1 on vascular remodeling and repair under cerebral ischemic conditions both in vivo following 90 min of ischemia by temporary occlusion of the middle cerebral artery, and in vitro. Our results demonstrate that as compared to mock-transfected controls, overexpression of α5 integrin alone didn\'t improve the outcomes in neurological score and size of infarct and caused worse BBB breakdown in the ischemic hemisphere, offsetting its beneficial angiogenic effects during the early stages of CIS. However, co-overexpression of α5 integrin with Ang1 led to smaller infarcts and improved neurological deficits, which at the molecular level was underpinned by reduced BBB breakdown and increased expression of endothelial tight junction proteins in the ischemic penumbra during the early stages of CIS. Furthermore, co-overexpression of α5 integrin and Ang1 synergistically promoted BEC proliferation during the early stage of CIS, resulting in increased blood vessel density at later stages. Positive effects of α5 integrin and Ang1 co-overexpression on endothelial proliferation and tight junction protein expression were also confirmed in vitro. Collectively, these data indicate that co-overexpression of Ang-1 and α5 integrin in combination confers synergistic vascular protection against cerebral ischemic injury without the negative side effects on BBB permeability, suggesting a novel combinatorial approach for the treatment of CIS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Ang-(1-7) inhibits lung cancer cell growth both in vitro and in vivo. However, the molecular mechanism of action is unclear and also the rapid degradation of Ang-(1-7) in vivo limits its clinical application. Here, we have demonstrated that Ang- (1-7) inhibits lung cancer cell growth by interrupting pre-replicative complex assembly and restrains epithelial-mesenchymal transition via Cdc6 inhibition. Furthermore, we constructed a mutant adeno-associated viral vector AAV8 (Y733F) that produced stable and high efficient Ang-(1-7) expression in a xenograft tumor model. The results show that AAV8-mediated Ang-(1-7) over-expression can remarkably suppress tumor growth in vivo by down-regulating Cdc6 and anti-angiogenesis. Ang-(1-7) over-expression via the AAV8 method may be a promising strategy for lung cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    腺相关病毒(AAV)载体已广泛用于基因治疗和基因功能研究,因为强转基因表达是积极结果的先决条件。AAV8被报道为用于肝脏转导的最有效的AAV血清型,与其他血清型相比,大脑和肌肉。然而,与小鼠肝细胞相比,AAV8介导的人肝细胞转导相当差,效率低约20倍。因此,我们应用土拨鼠肝炎病毒转录后调控元件(WPRE),以增强由CMV-IE增强子和鸡β-肌动蛋白启动子联合启动子(CAG启动子)驱动的AAV8介导的转基因表达,从而获得更有效的病毒载体.在体外和体内评估了具有或不具有WPRE的带有红色荧光蛋白(RFP)报告基因的重组AAV8(rAAV8)载体的转基因表达。结果表明,WPRE改善了AAV8介导的RFP在不同细胞系中的表达,肝脏中的转基因表达明显增加,动物的大脑或肌肉。这项研究的发现将有助于大大减少必须注射的病毒颗粒的数量,以达到基因治疗中转基因表达的治疗水平。因此,这种剂量减少可能会降低与高剂量病毒载体相关的潜在风险.
    Adeno-associated virus (AAV) vectors have been utilized extensively in gene therapy and gene function studies, as strong transgene expression is a prerequisite for positive outcomes. AAV8 was reported as the most efficient AAV serotype for transduction of the liver, brain and muscle compared with other serotypes. However, AAV8-mediated transduction of human hepatocytes is rather poor with approximately 20-fold lower efficiency compared with that of mouse hepatocytes. Therefore, we applied the woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) to enhance AAV8-mediated transgene expression driven by a combination promoter (CAG promoter) with a CMV-IE enhancer and chicken beta-actin promoter for a more efficient viral vector. Transgene expression from recombinant AAV8 (rAAV8) vectors harboring a red fluorescent protein (RFP) reporter gene with or without WPRE were evaluated in vitro and in vivo. The results demonstrated that WPRE improved AAV8-mediated RFP expression in different cell lines with clear increases of transgene expression in the liver, brain or muscle of animals. The findings of this study will help to substantially reduce the quantity of viral particles that must be injected in order to reach a therapeutic level of transgene expression in gene therapy. Consequently, such dose reductions may lessen the potential risks associated with high doses of viral vectors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    BACKGROUND: Rupture of the repair and adhesion around a tendon are two major problems after tendon surgery. Novel biological therapies which enhance healing and reduce adhesions are goals of many investigations. Gene therapy offers a new and promising approach to tackle these difficult problems. In the past decade, we sought to develop methods to augment tendon healing and reduce tendon adhesion through gene therapy.
    METHODS: This review discusses the methods and results of adeno-associated viral (AAV) type 2 vector gene therapy to increase tendon healing strength and reduce adhesions in a chicken model. Micro-RNA related gene therapy is also discussed. We also developed a controlled release system, which incorporates nanoparticles to deliver micro-RNAs to regulate tendon healing.
    CONCLUSIONS: We obtained promising results of enhancement of tendon healing strength in a chicken model using AAV2-mediated gene transfer. AAV2-mediated micro-RNA transfer also limited adhesions around the tendon. Controlled release systems incorporating nanoparticles have ideally delivered genes to the healing tendons and resulted in a moderate (but incomplete) reduction of adhesions. It remains to be determined what the best doses are and what other factors are in play in adhesion formation. These are two targets in our future investigations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Viral vectors have been utilized extensively to introduce genetic material into the central nervous system. In order to investigate gene functions in cardiovascular control regions of rat brain, we applied WPRE (woodchuck hepatitis virus post-transcriptional regulatory element) enhanced-adenoviral (Ad) and adeno-assoicated virus (AAV) type 2 vectors to mediate neuronal gene delivery to the paraventricular nucleus of the hypothalamus, the nucleus tractus solitarius and the rostral ventrolateral medulla, three important cardiovascular control regions known to express renin-angiotensin system (RAS) genes. Ad or AAV2 harboring an enhanced green fluorescent protein (EGFP) reporter gene or the angiotensin type 2 receptor gene were microinjected into these brain regions in adult rats. Our results demonstrated that both AAV2 and Ad vectors elicited long-term neuronal transduction in these regions. Interestingly, we found that the WPRE caused expression of GFP driven by the synapsin1 promoter in pure glial cultures or co-cultures of neurons and glia derived from rat hypothalamus and brainstem. However, in rat paraventricular nucleus WPRE did not cause expression of GFP in glia. This demonstrates the potential use of these vectors in studies of physiological functions of certain genes in the cardiovascular control regions of the brain.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号