lentiviral gene therapy

慢病毒基因疗法
  • 文章类型: Journal Article
    Pompe病是由酸性α-葡萄糖苷酶(GAA)缺乏引起的溶酶体贮积症,导致骨骼肌中糖原积累,病理深刻。我们最近开发了一种针对庞贝氏病的慢病毒基因疗法的优化形式,其中GAA转基因(LV-GAAco)的密码子优化版本与胰岛素样生长因子2(IGF2)肽(LV-IGF2)融合。GAAco),通过阳离子非依赖性甘露糖-6-磷酸/IGF2受体促进细胞摄取。用LV-IGF2进行慢病毒基因疗法。GAAco在心脏方面表现出优异的疗效,骨骼肌,与未标记LV-GAAco的基因治疗相比,Gaa-/-小鼠的大脑。这里,我们使用TMT标记的定量质谱来分析Gaa-/-小鼠的肌肉蛋白质组和对基因治疗的反应。我们发现Gaa-/-小鼠的肌肉显示出改变的蛋白质水平,包括那些在CLEAR信号通路中具有功能的蛋白质,自噬,细胞质糖原代谢,钙稳态,氧化还原信号,线粒体功能,脂肪酸运输,肌肉收缩,细胞骨架组织,吞噬体成熟,和炎症。LV-GAAco基因治疗导致肌肉蛋白质组的部分校正,而LV-IGF2基因治疗。GAAco导致几乎完全恢复到野生型水平,而不会引起额外的蛋白质组变化,支持Pompe病的慢病毒基因疗法的临床发展。意义:溶酶体糖原积累是庞皮病的主要原因,并导致心脏和骨骼肌以及中枢神经系统的一系列病理事件。在这项研究中,我们在小鼠模型的骨骼肌中鉴定了由Pompe病引起的蛋白质组变化。我们证明了用LV-IGF2进行慢病毒基因治疗。GAAco几乎完全纠正了疾病相关的蛋白质组变化。本研究支持LV-IGF2慢病毒基因治疗的未来临床发展。GAAco作为Pompe病的新治疗选择。
    Pompe disease is a lysosomal storage disorder caused by deficiency of acid alpha-glucosidase (GAA), resulting in glycogen accumulation with profound pathology in skeletal muscle. We recently developed an optimized form of lentiviral gene therapy for Pompe disease in which a codon-optimized version of the GAA transgene (LV-GAAco) was fused to an insulin-like growth factor 2 (IGF2) peptide (LV-IGF2.GAAco), to promote cellular uptake via the cation-independent mannose-6-phosphate/IGF2 receptor. Lentiviral gene therapy with LV-IGF2.GAAco showed superior efficacy in heart, skeletal muscle, and brain of Gaa -/- mice compared to gene therapy with untagged LV-GAAco. Here, we used quantitative mass spectrometry using TMT labeling to analyze the muscle proteome and the response to gene therapy in Gaa -/- mice. We found that muscle of Gaa -/- mice displayed altered levels of proteins including those with functions in the CLEAR signaling pathway, autophagy, cytoplasmic glycogen metabolism, calcium homeostasis, redox signaling, mitochondrial function, fatty acid transport, muscle contraction, cytoskeletal organization, phagosome maturation, and inflammation. Gene therapy with LV-GAAco resulted in partial correction of the muscle proteome, while gene therapy with LV-IGF2.GAAco resulted in a near-complete restoration to wild type levels without inducing extra proteomic changes, supporting clinical development of lentiviral gene therapy for Pompe disease. SIGNIFICANCE: Lysosomal glycogen accumulation is the primary cause of Pompe disease, and leads to a cascade of pathological events in cardiac and skeletal muscle and in the central nervous system. In this study, we identified the proteomic changes that are caused by Pompe disease in skeletal muscle of a mouse model. We showed that lentiviral gene therapy with LV-IGF2.GAAco nearly completely corrects disease-associated proteomic changes. This study supports the future clinical development of lentiviral gene therapy with LV-IGF2.GAAco as a new treatment option for Pompe disease.
    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
    艾杜糖醛酸2-硫酸酯酶(IDS)的缺乏导致II型粘多糖贮积症(MPSII),溶酶体贮积症,以糖胺聚糖(GAG)的系统性积累为特征,导致毁灭性的认知能力下降和危及生命的呼吸和心脏并发症。我们以前发现造血干细胞和祖细胞介导的慢病毒基因疗法(HSPC-LVGT)采用标记的IDS与胰岛素样生长因子2(IGF2)或ApoE2,但不是受体相关蛋白最小肽(RAP12x2),在MPSII的小鼠模型中有效地预防了脑病理学。在这项研究中,我们报道了HSPC-LVGT对外周病理学的影响,并分析了IDS的生物分布.我们发现HSPC-LVGT与所有载体完全纠正GAG积累和溶酶体病理在肝脏,脾,脾肾,气管粘膜,和心脏瓣膜。只有使用IDS才能完全校正大心脏血管的膜介质。IGF2co基因治疗,而其他向量几乎完全提供(IDS。ApoE2co)或没有(IDSco和IDS。RAP12x2co)校正。相比之下,气管,骨phy,所有测试的载体仍未纠正关节软骨。这些功效与HSPC-LVGT后的IDS蛋白水平紧密匹配。我们的研究结果证明了HSPC-LVGT在高临床相关性组织中纠正病理的能力,包括心脏和呼吸系统,而软骨病理学的矫正仍然存在挑战。
    Deficiency of iduronate 2-sulfatase (IDS) causes Mucopolysaccharidosis type II (MPS II), a lysosomal storage disorder characterized by systemic accumulation of glycosaminoglycans (GAGs), leading to a devastating cognitive decline and life-threatening respiratory and cardiac complications. We previously found that hematopoietic stem and progenitor cell-mediated lentiviral gene therapy (HSPC-LVGT) employing tagged IDS with insulin-like growth factor 2 (IGF2) or ApoE2, but not receptor-associated protein minimal peptide (RAP12x2), efficiently prevented brain pathology in a murine model of MPS II. In this study, we report on the effects of HSPC-LVGT on peripheral pathology and we analyzed IDS biodistribution. We found that HSPC-LVGT with all vectors completely corrected GAG accumulation and lysosomal pathology in liver, spleen, kidney, tracheal mucosa, and heart valves. Full correction of tunica media of the great heart vessels was achieved only with IDS.IGF2co gene therapy, while the other vectors provided near complete (IDS.ApoE2co) or no (IDSco and IDS.RAP12x2co) correction. In contrast, tracheal, epiphyseal, and articular cartilage remained largely uncorrected by all vectors tested. These efficacies were closely matched by IDS protein levels following HSPC-LVGT. Our results demonstrate the capability of HSPC-LVGT to correct pathology in tissues of high clinical relevance, including those of the heart and respiratory system, while challenges remain for the correction of cartilage pathology.
    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
    II型粘多糖贮积症(OMIM309900)是由艾杜糖醛酸2-硫酸酯酶(IDS)缺乏和糖胺聚糖积累引起的溶酶体贮积症,导致进行性神经变性.由于静脉输注酶替代疗法不能穿过血脑屏障(BBB),它不能治疗脑部病理,强调开发替代疗法的未满足的医疗需求。这里,我们使用IDS标记与普遍存在的MND启动子组合测试造血干细胞和祖细胞(HSPC)介导的慢病毒基因疗法(LVGT)的改良版本,以优化脑中的功效并研究其作用机制。我们发现用IGF2或ApoE2而不是RAP12x2标记的IDS可改善临床相关载体拷贝数的脑硫酸乙酰肝素和神经炎症的校正。在脑中移植的HSPC来源的细胞在血管周围区域显示出最高的效率,较低的脉络丛和脑膜,薄壁组织最低。重要的是,校正的疗效与脑移植细胞的数量无关.这些结果表明,IDS的标记版本可以优于HSPC-LVGT中未标记的IDS,以纠正MPSII中的脑部病理,它们暗示了细胞介导和标签介导的矫正机制,包括通过血脑屏障和增加摄取,强调他们的临床翻译潜力。
    Mucopolysaccharidosis type II (OMIM 309900) is a lysosomal storage disorder caused by iduronate 2-sulfatase (IDS) deficiency and accumulation of glycosaminoglycans, leading to progressive neurodegeneration. As intravenously infused enzyme replacement therapy cannot cross the blood-brain barrier (BBB), it fails to treat brain pathology, highlighting the unmet medical need to develop alternative therapies. Here, we test modified versions of hematopoietic stem and progenitor cell (HSPC)-mediated lentiviral gene therapy (LVGT) using IDS tagging in combination with the ubiquitous MND promoter to optimize efficacy in brain and to investigate its mechanism of action. We find that IDS tagging with IGF2 or ApoE2, but not RAP12x2, improves correction of brain heparan sulfate and neuroinflammation at clinically relevant vector copy numbers. HSPC-derived cells engrafted in brain show efficiencies highest in perivascular areas, lower in choroid plexus and meninges, and lowest in parenchyma. Importantly, the efficacy of correction was independent of the number of brain-engrafted cells. These results indicate that tagged versions of IDS can outperform untagged IDS in HSPC-LVGT for the correction of brain pathology in MPS II, and they imply both cell-mediated and tag-mediated correction mechanisms, including passage across the BBB and increased uptake, highlighting their potential for clinical translation.
    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)的特征在于由于存活运动神经元(SMN)缺乏而导致的下脊髓运动神经元的损失。运动神经元细胞自主和非细胞自主疾病机制驱动早期谷氨酸能功能障碍,运动神经元细胞丢失之前的治疗可靶向表型,仍然不清楚。使用微电极阵列分析,我们证明,正常突触调节所需的分泌组和细胞表面蛋白可能在人SMA星形胶质细胞中被破坏,并导致运动神经元活性降低.虽然健康的星形胶质细胞条件培养基不能改善SMA运动神经元活性,在直接接触培养物中,SMA运动神经元对健康的星形胶质细胞神经调节反应强劲。这表明星形胶质细胞突触相关的质膜蛋白和接触介导的细胞相互作用对于SMA中正确的运动神经元功能具有重要作用。具体来说,我们发现人SMA星形胶质细胞和SMA腰脊髓组织中谷氨酸钠依赖性兴奋性氨基酸转运蛋白EAAT1显著减少。在健康共培养物中EAAT1的选择性抑制表现出在SMA星形胶质细胞共培养物中观察到的神经活性降低。Caveolin-1,一种以前与质膜局部翻译相关的SMN相互作用蛋白,在人SMA星形胶质细胞中异常升高。尽管慢病毒SMN递送至SMA星形胶质细胞部分挽救了EAAT1表达,在SMN转导的SMA星形胶质细胞共培养物中仍观察到健康运动神经元的有限活性.一起,这些数据强调了星形胶质细胞介导的疾病机制对SMA运动神经元功能的有害影响,并且SMN递送可能不足以完全恢复突触处的星形胶质细胞功能.
    Spinal muscular atrophy (SMA) is characterized by the loss of the lower spinal motor neurons due to survival motor neuron (SMN) deficiency. The motor neuron cell autonomous and non-cell autonomous disease mechanisms driving early glutamatergic dysfunction, a therapeutically targetable phenotype prior to motor neuron cell loss, remain unclear. Using microelectrode array analysis, we demonstrate that the secretome and cell surface proteins needed for proper synaptic modulation are likely disrupted in human SMA astrocytes and lead to diminished motor neuron activity. While healthy astrocyte conditioned media did not improve SMA motor neuron activity, SMA motor neurons robustly responded to healthy astrocyte neuromodulation in direct contact cultures. This suggests an important role of astrocyte synaptic-associated plasma membrane proteins and contact-mediated cellular interactions for proper motor neuron function in SMA. Specifically, we identified a significant reduction of the glutamate Na+ dependent excitatory amino acid transporter EAAT1 within human SMA astrocytes and SMA lumbar spinal cord tissue. The selective inhibition of EAAT1 in healthy co-cultures phenocopied the diminished neural activity observed in SMA astrocyte co-cultures. Caveolin-1, an SMN-interacting protein previously associated with local translation at the plasma membrane, was abnormally elevated in human SMA astrocytes. Although lentiviral SMN delivery to SMA astrocytes partially rescued EAAT1 expression, limited activity of healthy motor neurons was still observed in SMN-transduced SMA astrocyte co-cultures. Together, these data highlight the detrimental impact of astrocyte-mediated disease mechanisms on motor neuron function in SMA and that SMN delivery may be insufficient to fully restore astrocyte function at the synapse.
    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
    肾上腺脑白质营养不良(ALD)是一种X连锁的过氧化物酶体疾病,发病率为14-17,000名男性中的1名。由ABCD1基因内的致病变异引起。到了成年,大约40%的病人发展为脑ALD,一个严重的,通常是进行性和致命的神经炎症性疾病,没有干预。
    历史上,只有异基因造血干细胞移植(HSCT)已被证明可以阻止脑ALD的进展,在疾病过程早期进行HSCT时获得了更好的结果。最近,已经研究了一种基于慢病毒的基因治疗方法,作为同种异体移植的替代方法,用于治疗脑ALD。使用术语“造血干细胞移植”进行了重点文献综述,\'\'基因治疗\'和\'肾上腺脑白质营养不良\'包括相关文献,特别是比较基因治疗和HSCT结果的经验。我们回顾了HSCT在脑ALD中的历史和经验及其局限性,以及目前与脑ALD基因治疗试验相关的信息。
    关于这种基于慢病毒的基因治疗方法及其相对风险和益处的数据仍在评估中。在使用同种异体HSCT进行脑ALD的经验的背景下,对这些信息进行了探索。
    Adrenoleukodystrophy (ALD) is an X-linked peroxisomal disorder with an incidence of 1 in 14-17,000 male births, caused by pathogenic variants within the ABCD1 gene. By adulthood, approximately 40% of the patients develop cerebral ALD, a severe, neuroinflammatory condition that is generally progressive and fatal without intervention.
    Historically, only allogeneic hematopoietic stem cell transplantation (HSCT) has been shown to halt progression of cerebral ALD, with superior outcomes obtained when HSCT is performed early in the disease process. More recently, a lentiviral-based gene therapy approach has been investigated as therapy for cerebral ALD as an alternative to allogeneic transplantation. A focused literature review was performed using the terms \'hematopoietic stem cell transplantation,\' \'gene therapy\' and \'adrenoleukodystrophy\' to include relevant literature, especially comparing the experience with gene therapy and HSCT outcomes. We review the history and experience with HSCT in cerebral ALD and its limitations, as well as the information currently available in association with the gene therapy trials for cerebral ALD.
    The data regarding this lentiviral-based gene therapy approach and its relative risks and benefits is still being evaluated. This information is explored in the context of the experience with allogeneic HSCT for cerebral ALD.
    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
    严重的联合免疫缺陷(SCID)导致T细胞严重缺乏以及B和NK细胞的可变缺陷。未治疗,这种情况在生命的头两年内是致命的。HSCT传统上是唯一的治疗方法;然而,缺乏HLA匹配供体的患者的成功率并不理想,预处理方案可导致显著的毒性.基因治疗是30多年前腺苷脱氨酶(ADA-SCID)的先驱,并产生了非常成功的结果。X-SCID的令人鼓舞的数据以及Artemis-SCID和RAG1-SCID的临床前工作为该疗法成为可行的治愈性治疗选择铺平了道路。
    Severe combined immune deficiency (SCID) causes profound deficiency in T cells and variable deficiencies in B and NK cells. Untreated, the condition is fatal within the first 2 years of life. HSCT has traditionally been the only curative approach; however, success rates are suboptimal in those lacking an HLA-matched donor and conditioning regimens can cause significant toxicity. Gene therapy was pioneered for adenosine deaminase (ADA-SCID) over 3 decades ago and has produced highly successful results. Encouraging data for X-SCID and preclinical work for Artemis-SCID and RAG1-SCID are paving the way for the therapy to become a viable curative treatment option.
    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
    酶替代疗法(ERT)是目前治疗庞贝氏症的标准疗法,由溶酶体酶酸性α-葡糖苷酶(GAA)缺乏引起的溶酶体贮积障碍。ERT已被证明可以挽救经典婴儿庞贝病患者的生命。然而,一个主要缺点是针对ERT的中和抗体的开发.造血干细胞和祖细胞介导的慢病毒基因疗法(HSPC-LVGT)提供了一种新的,潜在的终身治疗与单一干预和可能诱导免疫耐受。这里,我们研究了在Pompe病小鼠模型中,在HSPC-LVGT后,ERT是否可以安全地作为额外或替代疗法应用.我们发现亚治疗剂量的慢病毒表达足以诱导对转基因产物的耐受性,以及随后进行的ERT。在基因治疗后4-6周内建立免疫耐受。小鼠耐受ERT剂量高达100mg/kg,允许ERT消除心肌和骨骼肌中的糖原积累并使运动功能正常化。胸腺中表达GAA的HSPC衍生细胞的存在表明建立了中枢免疫耐受。这些发现表明,鼠类庞贝氏症的慢病毒基因疗法可诱导对转基因表达或作为ERT提供的GAA的强大而长期的免疫耐受。
    Enzyme replacement therapy (ERT) is the current standard treatment for Pompe disease, a lysosomal storage disorder caused by deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). ERT has shown to be lifesaving in patients with classic infantile Pompe disease. However, a major drawback is the development of neutralizing antibodies against ERT. Hematopoietic stem and progenitor cell-mediated lentiviral gene therapy (HSPC-LVGT) provides a novel, potential lifelong therapy with a single intervention and may induce immune tolerance. Here, we investigated whether ERT can be safely applied as additional or alternative therapy following HSPC-LVGT in a murine model of Pompe disease. We found that lentiviral expression at subtherapeutic dose was sufficient to induce tolerance to the transgene product, as well as to subsequently administered ERT. Immune tolerance was established within 4-6 weeks after gene therapy. The mice tolerated ERT doses up to 100 mg/kg, allowing ERT to eliminate glycogen accumulation in cardiac and skeletal muscle and normalizing locomotor function. The presence of HSPC-derived cells expressing GAA in the thymus suggested the establishment of central immune tolerance. These findings demonstrate that lentiviral gene therapy in murine Pompe disease induced robust and long-term immune tolerance to GAA either expressed by a transgene or supplied as ERT.
    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
    Chronic granulomatous disease (CGD) is an inherited blood disorder of phagocytic cells that renders patients susceptible to infections and inflammation. A recent clinical trial of lentiviral gene therapy for the most frequent form of CGD, X-linked, has demonstrated stable correction over time, with no adverse events related to the gene therapy procedure. We have recently developed a parallel lentiviral vector for p47phox-deficient CGD (p47phoxCGD), the second most common form of this disease. Using this vector, we have observed biochemical correction of CGD in a mouse model of the disease. In preparation for clinical trial approval, we have performed standardized preclinical studies following Good Laboratory Practice (GLP) principles, to assess the safety of the gene therapy procedure. We report no evidence of adverse events, including mutagenesis and tumorigenesis, in human hematopoietic stem cells transduced with the lentiviral vector. Biodistribution studies of transduced human CD34+ cells indicate that the homing properties or engraftment ability of the stem cells is not negatively affected. CD34+ cells derived from a p47phoxCGD patient were subjected to an optimized transduction protocol and transplanted into immunocompromised mice. After the procedure, patient-derived neutrophils resumed their function, suggesting that gene correction was successful. These studies pave the way to a first-in-man clinical trial of lentiviral gene therapy for the treatment of p47phoxCGD.
    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
    Genetically modified, autologous hematopoietic stem and progenitor cells (HSPCs) represent a new class of genetic medicine. Following this therapeutic paradigm, we are developing a product candidate, designated CD68-ET3-LV CD34+, for the treatment of the severe bleeding disorder, hemophilia A. The product consists of autologous CD34+ cells transduced with a human immunodeficiency virus 1-based, monocyte lineage-restricted, self-inactivating lentiviral vector (LV), termed CD68-ET3-LV, encoding a bioengineered coagulation factor VIII (fVIII) transgene, termed ET3, designed for enhanced expression. This vector was shown capable of high-titer manufacture under clinical scale and Good Manufacturing Practice. Biochemical and immunogenicity testing of recombinant ET3, as well as safety and efficacy testing of CD68-ET3-LV HSPCs, were utilized to demonstrate overall safety and efficacy in murine models. In the first model, administration of CD68-ET3-LV-transduced stem-cell antigen-1+ cells to hemophilia A mice resulted in sustained plasma fVIII production and hemostatic correction without signs of toxicity. Patient-derived, autologous mobilized peripheral blood (mPB) CD34+ cells are the clinical target cells for ex vivo transduction using CD68-ET3-LV, and the resulting genetically modified cells represent the investigational drug candidate. In the second model, CD68-ET3-LV gene transfer into mPB CD34+ cells isolated from normal human donors was utilized to obtain in vitro and in vivo pharmacology, pharmacokinetic, and toxicology assessment. CD68-ET3-LV demonstrated reproducible and efficient gene transfer into mPB CD34+ cells, with vector copy numbers in the range of 1 copy per diploid genome equivalent without affecting clonogenic potential. Differentiation of human CD34+ cells into monocytes was associated with increased fVIII production, supporting the designed function of the CD68 promoter. To assess in vivo pharmacodynamics, CD68-ET3-LV CD34+ cell product was administered to immunodeficient mice. Treated mice displayed sustained plasma fVIII levels and no signs of product related toxicity. Collectively, the findings of the current study support the preclinical safety and efficacy of CD68-ET3-LV CD34+.
    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
    胰高血糖素样肽-1(GLP-1)是一种具有促胰岛素作用的代谢激素,可促进餐后葡萄糖诱导的胰岛胰岛素分泌。在它介导的各种影响中,GLP-1诱导生长抑素的δ细胞分泌,抑制α细胞释放胰高血糖素,减少胃排空,减缓食物摄入。随着时间的推移,这些事件共同导致体重减轻。在2型糖尿病(T2DM)期间,然而,肠促胰岛素对葡萄糖的反应降低,并伴随着GLP-1分泌的适度减少。为了补偿减少的肠促胰岛素效应,产生基于人类免疫缺陷病毒的慢病毒载体以递送编码人GLP-1(LentiGLP-1)的DNA,在高脂饮食/链脲佐菌素诱导的T2DM模型中测试了LentiGLP-1的抗糖尿病功效。LentiGLP-1的治疗性给药降低肥胖糖尿病SpragueDawley大鼠的血糖水平,同时改善胰岛素敏感性和糖耐量。正常血糖与LentiGLP-1处理的大鼠血液GLP-1和胰腺β细胞再生增加相关。LentiGLP-1注射后血浆甘油三酯水平也正常化。总的来说,这些数据提示GLP-1基因转移治疗2型糖尿病的临床潜力.
    Postprandial glucose-induced insulin secretion from the islets of Langerhans is facilitated by glucagon-like peptide-1 (GLP-1)-a metabolic hormone with insulinotropic properties. Among the variety of effects it mediates, GLP-1 induces delta cell secretion of somatostatin, inhibits alpha cell release of glucagon, reduces gastric emptying, and slows food intake. These events collectively contribute to weight loss over time. During type 2 diabetes (T2DM), however, the incretin response to glucose is reduced and accompanied by a moderate reduction in GLP-1 secretion. To compensate for the reduced incretin effect, a human immunodeficiency virus-based lentiviral vector was generated to deliver DNA encoding human GLP-1 (LentiGLP-1), and the anti-diabetic efficacy of LentiGLP-1 was tested in a high-fat diet/streptozotocin-induced model of T2DM. Therapeutic administration of LentiGLP-1 reduced blood glucose levels in obese diabetic Sprague Dawley rats, along with improving insulin sensitivity and glucose tolerance. Normoglycemia was correlated with increased blood GLP-1 and pancreatic beta cell regeneration in LentiGLP-1-treated rats. Plasma triglyceride levels were also normalized after LentiGLP-1 injection. Collectively, these data suggest the clinical potential of GLP-1 gene transfer therapy for the treatment of T2DM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号