Sclerosteosis

硬化病
  • 文章类型: Journal Article
    先前已经描述了破坏硬化蛋白(由SOST编码)与其受体LRP4之间结合的致病变体会导致硬化,一种罕见的高骨量疾病.硬化蛋白-LRP4复合物抑制典型的WNT信号,这是调节成骨细胞骨形成的关键途径,也是常见骨疾病的有希望的治疗靶点,比如骨质疏松症。在目前的研究中,我们用p.Arg1170GlnLrp4敲入(Lrp4KI/KI)小鼠模型杂交Sost缺陷(Sost-/-)小鼠,以创建双突变Sost-/-;Lrp4KI/KI小鼠。我们比较了Sost-/-小鼠与Sost-/-;Lrp4KI/KI小鼠的表型,研究Lrp4中引起疾病的p.Arg1170Trp变体对Sost缺乏症的可能协同作用。有趣的是,Lrp4KI等位基因的存在部分减轻了Sost-/-表型。细胞和动态组织形态计量学未揭示对观察到的表型差异的机械见解。因此,我们通过对Lrp4KI/KI原代成骨细胞进行大量RNA测序来确定Lrp4KI等位基因的分子效应。出乎意料的是,大多数与骨吸收或重塑相关的基因(Acp5,Rankl,Mmp9)在Lrp4KI/KI原代成骨细胞中上调。在Lrp4KI/KI中验证这些标记,Sost-/-和Sost-/-;Lrp4KI/KI小鼠揭示了硬化素缺乏在Sost-/-;Lrp4KI/KI小鼠中抵消这种Lrp4KI/KI效应。因此,我们假设具有两个失活Lrp4KI等位基因的模型激活了骨重建,随着骨量的净增加,而硬化蛋白缺乏对骨形成有更强的合成代谢作用。此外,硬化蛋白和Lrp4的这些作用在雌性小鼠中更强,导致比男性更严重的表型和不同基因型之间更可检测的表型差异。
    Pathogenic variants disrupting the binding between sclerostin (encoded by SOST) and its receptor LRP4 have previously been described to cause sclerosteosis, a rare high bone mass disorder. The sclerostin-LRP4 complex inhibits canonical WNT signaling, a key pathway regulating osteoblastic bone formation and a promising therapeutic target for common bone disorders, such as osteoporosis. In the current study, we crossed mice deficient for Sost (Sost-/-) with our p.Arg1170Gln Lrp4 knock-in (Lrp4KI/KI) mouse model to create double mutant Sost-/-;Lrp4KI/KI mice. We compared the phenotype of Sost-/- mice with that of Sost-/-;Lrp4KI/KI mice, to investigate a possible synergistic effect of the disease-causing p.Arg1170Trp variant in Lrp4 on Sost deficiency. Interestingly, presence of Lrp4KI alleles partially mitigated the Sost-/- phenotype. Cellular and dynamic histomorphometry did not reveal mechanistic insights into the observed phenotypic differences. We therefore determined the molecular effect of the Lrp4KI allele by performing bulk RNA sequencing on Lrp4KI/KI primary osteoblasts. Unexpectedly, mostly genes related to bone resorption or remodeling (Acp5, Rankl, Mmp9) were upregulated in Lrp4KI/KI primary osteoblasts. Verification of these markers in Lrp4KI/KI, Sost-/- and Sost-/-;Lrp4KI/KI mice revealed that sclerostin deficiency counteracts this Lrp4KI/KI effect in Sost-/-;Lrp4KI/KI mice. We therefore hypothesize that models with two inactivating Lrp4KI alleles rather activate bone remodeling, with a net gain in bone mass, whereas sclerostin deficiency has more robust anabolic effects on bone formation. Moreover, these effects of sclerostin and Lrp4 are stronger in female mice, contributing to a more severe phenotype than in males and more detectable phenotypic differences among different genotypes.
    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
    影响骨骼系统的遗传性疾病具有广泛的症状,使诊断和治疗变得困难。全基因组关联和测序研究已经确定了与人类骨骼疾病相关的基因。斑马鱼模型的基因编辑使研究人员能够进一步检查基因型和表型之间的联系,长期目标是改善诊断和治疗。虽然目前的自动化工具能够快速和深入地对轴向骨骼进行表型分析,表征突变对颅面骨骼的影响更具挑战性。本研究的目的是评估一种半自动筛查工具,该工具可用于使用与人类骨骼疾病相关的四个基因(meox1,plod2,sost,和wnt16)作为测试用例。我们使用传统的地标来证实我们的数据集和伪地标来量化各组之间3D颅骨的变化(体细胞脆皮,种系突变体,和控制鱼)。拟议的管道确定了四个基因的脆皮或突变鱼与对照鱼之间的变异。对于所测试的四个基因中的两个,表型的变化与人类颅面症状平行。这项研究证明了我们的管道作为检查与斑马鱼颅面骨骼相关的多维表型的筛选工具的潜力和局限性。
    Genetic diseases affecting the skeletal system present with a wide range of symptoms that make diagnosis and treatment difficult. Genome-wide association and sequencing studies have identified genes linked to human skeletal diseases. Gene editing of zebrafish models allows researchers to further examine the link between genotype and phenotype, with the long-term goal of improving diagnosis and treatment. While current automated tools enable rapid and in-depth phenotyping of the axial skeleton, characterizing the effects of mutations on the craniofacial skeleton has been more challenging. The objective of this study was to evaluate a semi-automated screening tool can be used to quantify craniofacial variations in zebrafish models using four genes that have been associated with human skeletal diseases (meox1, plod2, sost, and wnt16) as test cases. We used traditional landmarks to ground truth our dataset and pseudolandmarks to quantify variation across the 3D cranial skeleton between the groups (somatic crispant, germline mutant, and control fish). The proposed pipeline identified variation between the crispant or mutant fish and control fish for four genes. Variation in phenotypes parallel human craniofacial symptoms for two of the four genes tested. This study demonstrates the potential as well as the limitations of our pipeline as a screening tool to examine multi-dimensional phenotypes associated with the zebrafish craniofacial skeleton.
    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
    背景:遗传性颅骨肥大是一种在意大利从未描述过的罕见疾病,因此,很少研究该疾病患者和携带者的神经系统表现。
    方法:我们描述了来自意大利一个患有硬化病的大型家庭的患者和基因携带者的神经和神经影像学特征。
    结果:在这个家族中,遗传测试检测到先证者中SOST基因的纯合p.Gln24X(c.70C>T)突变和9个兄弟姐妹中的杂合突变。在纯合成人中,严重的颅面骨肥厚在儿童时期表现为颅神经病,继发于颅内高压的慢性头痛,和成人阻塞性睡眠呼吸暂停综合征。在一个成年患者中,枕区有一个可压缩的皮下肿胀,由经骨颅内-颅外枕静脉引流引起,继发于颅骨肥厚的静脉引流阻塞的补偿机制。在9名杂合子受试者中,轻度颅骨增生引起频繁的头痛和打鼾。
    结论:儿童多发性颅神经病和头痛,而成人严重的慢性头痛和睡眠障碍,是第一个患有骨硬化的意大利家庭的神经系统表现。将神经和神经影像学评估扩展到基因携带者也是合理的。
    BACKGROUND: Hereditary cranial hyperostosis is a rare disease never described in Italy, so the neurological manifestations in patients and carriers of the disease have been little studied.
    METHODS: We describe the neurological and neuroimaging features of patients and carriers of the gene from a large Italian family with sclerosteosis.
    RESULTS: In this family, genetic testing detected the homozygous p.Gln24X (c.70C > T) mutation of the SOST gene in the proband and a heterozygous mutation in 9 siblings. In homozygous adults, severe craniofacial hyperostosis was manifested by cranial neuropathy in childhood, chronic headache secondary to intracranial hypertension, and an obstructive sleep apnea syndrome in adults. In one of the adult patients, there was a compressible subcutaneous swelling in the occipital region caused by transosseous intracranial-extracranial occipital venous drainage, a compensation mechanism of obstructed venous drainage secondary to cranial hyperostosis. Mild cranial hyperostosis causing frequent headache and snoring was evident in the nine heterozygous subjects.
    CONCLUSIONS: Multiple cranial neuropathies and headache in children, while severe chronic headache and sleep disturbances in adults, are the neurological manifestations of the first Italian family with osteosclerosis. It is reasonable to extend neurological and neuroimaging evaluation to gene carriers as well.
    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: Case Reports
    背景与目的:硬化蛋白是一种SOST基因产物,通过拮抗Wnt信号通路,抑制成骨细胞活性,防止骨过度形成。硬化病与SOST基因中功能突变的丧失有关。这是一种罕见的常染色体隐性遗传疾病,其特征是颅管肥大,可导致致命的小脑疝。我们的目的是描述硬化病患者的临床和放射学特征以及新的潜在SOST突变。病例:一名25岁女性,因怀疑生长激素过量而被转诊至内分泌诊所。病人抱怨头痛,渐进式视力模糊,听力障碍,增加了脚的大小,突增,下巴突出。她有正常的产前病史,但有并发症。图像显示弥漫性骨性增厚和高骨密度。生化和激素测试正常。由于进行性压迫性视神经病变,视神经开窗术与减压半颅切开术。怀疑是由于主要的颅管骨肥厚症并并指。使用外周白细胞DNA,对SOST基因进行基因组测序.这在SOST基因中发现了一个新的缺失纯合突变(c.387delG,p.Asp131ThrfsTer116)破坏硬化蛋白功能,导致硬化.结论:硬化病的分子基础的发现代表了这种致命疾病的诊断和治疗的重要进展。
    Background and Objectives: Sclerostin is an SOST gene product that inhibits osteoblast activity and prevents excessive bone formation by antagonizing the Wnt signaling pathway. Sclerosteosis has been linked to loss of function mutations in the SOST gene. It is a rare autosomal recessive disorder characterized by craniotubular hyperostosis and can lead to fatal cerebellar herniation. Our aim is to describe the clinical and radiological features and the new underlying SOST mutation in a patient with sclerosteosis. Case: A 25-year-old female who was referred to the endocrine clinic for suspected excess growth hormone. The patient complained of headaches, progressive blurred vision, hearing disturbances, increased size of feet, proptosis, and protrusion of the chin. She had normal antenatal history except for syndactyly. Images showed diffuse osseous thickening and high bone mineral density. Biochemical and hormonal tests were normal. Due to progressive compressive optic neuropathy, optic nerve fenestration with decompression hemicraniotomy was performed. Sclerosteosis was suspected due to the predominant craniotubular hyperostosis with syndactyly. Using peripheral leucocyte DNA, genomic sequencing of the SOST gene was performed. This identified a novel deletion homozygous mutation in the SOST gene (c.387delG, p.Asp131ThrfsTer116) which disrupts sclerostin function, causing sclerosteosis. Conclusions: Discovery of the molecular basis of sclerosteosis represents an important advance in the diagnosis and management of this fatal 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: Case Reports
    硬化性骨病是一种高骨量疾病,由编码硬化蛋白或LRP4的基因中的致病变异引起。两种蛋白质形成强烈抑制经典WNT信号传导活性的复合物,在骨形成中非常重要的途径。到目前为止,所有报道的致病变异都位于LRP4的第三个β-螺旋桨结构域,这对于与硬化蛋白的相互作用至关重要.这里,我们报告了两种复合杂合变体的鉴定,一个已知的p.Arg1170Gln和一个新颖的p.Arg632His变体,患有硬化性骨病表型的患者。有趣的是,新的变体位于第一个β-螺旋桨域,众所周知,这对于与agrin的相互作用是必不可少的。然而,使用荧光素酶报告基因测定,我们证明了LRP4中的p.Arg1170Gln和p.Arg632His变体均降低了硬化蛋白对经典WNT信号传导活性的抑制能力。总之,这项研究是第一个证明LRP4的第一个β-螺旋桨结构域中的致病变异体可以促进硬化的发展,这扩大了疾病的突变范围。
    Sclerosteosis is a high bone mass disorder, caused by pathogenic variants in the genes encoding sclerostin or LRP4. Both proteins form a complex that strongly inhibits canonical WNT signaling activity, a pathway of major importance in bone formation. So far, all reported disease-causing variants are located in the third β-propeller domain of LRP4, which is essential for the interaction with sclerostin. Here, we report the identification of two compound heterozygous variants, a known p.Arg1170Gln and a novel p.Arg632His variant, in a patient with a sclerosteosis phenotype. Interestingly, the novel variant is located in the first β-propeller domain, which is known to be indispensable for the interaction with agrin. However, using luciferase reporter assays, we demonstrated that both the p.Arg1170Gln and the p.Arg632His variant in LRP4 reduced the inhibitory capacity of sclerostin on canonical WNT signaling activity. In conclusion, this study is the first to demonstrate that a pathogenic variant in the first β-propeller domain of LRP4 can contribute to the development of sclerosteosis, which broadens the mutational spectrum of the disorder.
    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
    硬化,以过度骨形成为特征的严重常染色体隐性硬化性骨骼发育不良,是由于缺乏硬化蛋白引起的,结合LRP5/6Wnt共受体的骨形成的负调节物。目前的治疗仅限于由骨过度生长引起的症状的手术治疗。这项研究调查了硬化蛋白替代疗法在硬化病小鼠模型中的有效性。
    重组野生型小鼠硬化蛋白(mScl)和含有C末端人Fc(mSclhFc)的新型mScl融合蛋白,或具有聚天冬氨酸基序(mSclhFcPD)的C末端人Fc,使用哺乳动物表达和标准层析方法产生和纯化。使用三种独立的生物物理技术和体外骨结节形成测定来评估重组蛋白的体外功能和功效。在对年轻雌性野生型(WT)或SOST敲除(SOST-/-)小鼠单次施用后,在体内研究蛋白质的药代动力学性质。在为期六周的体内概念验证研究中,年轻的雌性WT或SOST-/-小鼠用10mg/kgmSclhFc或mSclhFcPD治疗(每周),或4.4mg/kgmScl(每日)。通过显微计算机断层扫描(μCT)评估重组硬骨素对股骨皮质和小梁骨参数的影响。
    重组mScl蛋白以高亲和力(nM范围)与Wnt共受体LRP6的胞外结构域结合,并在体外完全抑制基质矿化。向WT或SOST-/-小鼠施用单剂量后的药代动力学评估表明hFc的存在使蛋白质半衰期从小于5分钟增加到至少1.5天。用mSclhFcPD治疗超过6周的时间导致小梁体积骨矿物质密度(vBMD)和骨体积分数(BV/TV)的适度但显著的降低,20%和15%,分别。
    施用重组mSclhFcPD部分纠正了SOST-/-小鼠的高骨量表型,提示在SOST-/-硬化病小鼠模型中,以提高其半衰期为目标的硬化素能够负向调节骨形成。
    这些发现支持外源性硬化素能减少骨量的概念,然而,适度的疗效表明,硬骨素替代可能不是缓解硬骨病中骨过度形成的最佳策略。因此,应该探索替代方法。
    UNASSIGNED: Sclerosteosis, a severe autosomal recessive sclerosing skeletal dysplasia characterised by excessive bone formation, is caused by absence of sclerostin, a negative regulator of bone formation that binds LRP5/6 Wnt co-receptors. Current treatment is limited to surgical management of symptoms arising from bone overgrowth. This study investigated the effectiveness of sclerostin replacement therapy in a mouse model of sclerosteosis.
    UNASSIGNED: Recombinant wild type mouse sclerostin (mScl) and novel mScl fusion proteins containing a C-terminal human Fc (mScl hFc), or C-terminal human Fc with a poly-aspartate motif (mScl hFc PD), were produced and purified using mammalian expression and standard chromatography methods. In vitro functionality and efficacy of the recombinant proteins were evaluated using three independent biophysical techniques and an in vitro bone nodule formation assay. Pharmacokinetic properties of the proteins were investigated in vivo following a single administration to young female wild type (WT) or SOST knock out (SOST-/-) mice. In a six week proof-of-concept in vivo study, young female WT or SOST-/- mice were treated with 10 mg/kg mScl hFc or mScl hFc PD (weekly), or 4.4 mg/kg mScl (daily). The effect of recombinant sclerostin on femoral cortical and trabecular bone parameters were assessed by micro computed tomography (μCT).
    UNASSIGNED: Recombinant mScl proteins bound to the extracellular domain of the Wnt co-receptor LRP6 with high affinity (nM range) and completely inhibited matrix mineralisation in vitro. Pharmacokinetic assessment following a single dose administered to WT or SOST-/- mice indicated the presence of hFc increased protein half-life from less than 5 min to at least 1.5 days. Treatment with mScl hFc PD over a six week period resulted in modest but significant reductions in trabecular volumetric bone mineral density (vBMD) and bone volume fraction (BV/TV), of 20% and 15%, respectively.
    UNASSIGNED: Administration of recombinant mScl hFc PD partially corrected the high bone mass phenotype in SOST-/- mice, suggesting that bone-targeting of sclerostin engineered to improve half-life was able to negatively regulate bone formation in the SOST-/- mouse model of sclerosteosis.
    UNASSIGNED: These findings support the concept that exogenous sclerostin can reduce bone mass, however the modest efficacy suggests that sclerostin replacement may not be an optimal strategy to mitigate excessive bone formation in sclerosteosis, hence alternative approaches should be explored.
    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
    LRP4基因编码高度保守的低密度脂蛋白受体相关蛋白4(LRP4),作为硬化蛋白的共受体。硬化蛋白和LRP4负调节WNT/β-catenin信号通路,缺乏其抑制活性导致恒定的成骨细胞分化。因此,骨形成增加,在LRP4突变的情况下,导致2型硬化(SOST2)。LRP4内的改变也可能导致Cenani-Lenz综合征(CLSS),先天性肌无力或孤立性的。这里,我们报告了一个病人,在其中,我们使用全外显子组测序在LRP4中发现了一个新的纯合剪接位点变异c.1048+6T>C。患者最初被误诊为孤立的CLSS样或Malik-Percin样并指。然而,经过全面的放射学检查和分子证实的SOST2,我们终于完善了诊断。此外,我们在这里指出剪接变体是重要的致病改变,由于缺乏先进的,在分子分析中可能会被忽视,可靠的算法,内置的标准诊断管道。使用先进的剪接位点改变的计算机预测工具,包括AlamutVisual软件,我们已经证明c.1048+6T>CLRP4变体影响天然供体位点并损害SC35增强子活性。根据我们的经验,我们建议全面的放射成像,包括每种由LRP4致病变异体引起的分离的并指的颅骨X线检查。我们建议所有先前报道的携带双等位基因LRP4突变的患者,他们被诊断为孤立的并肢,实际上可能存在由于不完整的临床和放射学评估而无法识别的SOST2。
    The LRP4 gene encodes the highly conserved low-density lipoprotein receptor-related protein 4 (LRP4), which acts as a co-receptor for sclerostin. Sclerostin and LRP4 negatively regulate WNT/β-catenin signaling pathway and lack of their inhibitory activity leads to constant osteoblastic differentiation. Consequently, increased bone formation occurs, which in the case of LRP4 mutations results in sclerosteosis type 2 (SOST2). Alterations within the LRP4 may also cause Cenani-Lenz syndactyly syndrome (CLSS), congenital myasthenia or isolated syndactyly. Here, we have reported a patient, in whom we found a novel homozygous splice-site variant c.1048+6T>C in LRP4 using whole exome sequencing. The patient was initially misdiagnosed with isolated CLSS-like or Malik-Percin-like syndactyly. However, we have finally refined the diagnosis after comprehensive radiological examination and molecularly confirmed SOST2. Additionally, we have pointed here to the splicing variants as important causative alterations that may be overlooked in the molecular analysis due to the lack of advanced, reliable algorithms, built-into the standard diagnostic pipelines. Using advanced in silico prediction tools of splice-site alterations, including Alamut Visual software, we have demonstrated that the c.1048+6T>C LRP4 variant affects the native donor site and impairs an SC35 enhancer activity. Based on our experience, we recommend comprehensive radiological imaging, including X-ray of the skull in each case of isolated syndactyly resulting from pathogenic variants of LRP4. We suggest that all previously reported patients carrying biallelic LRP4 mutations, who were diagnosed with isolated syndactyly, could actually present with SOST2 that had been unrecognized due to the incomplete clinical and radiological assessment.
    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
    硬化病和范布赫姆病(VBD)是两种罕见的常染色体隐性遗传疾病,由成骨细胞多动症引起,其中进行性骨骼过度生长导致非常致密的骨骼,扭曲的脸,和脑神经的压迫。硬化是由SOST基因的功能缺失突变引起的,SOST基因编码分泌的糖蛋白,硬化蛋白.VBD是由去除了骨中SOST特异性调控元件的非编码缺失引起的。在骨头里,SOST主要由骨细胞表达,硬化蛋白通过抑制经典的Wnt信号通路抑制骨形成。在这里,我们描述了如何在硬化性和VBD患者中进行人类遗传学研究,结合转基因和基因敲除小鼠的产生,已经导致了对骨代谢中硬化蛋白的作用有了更好的理解。
    Sclerosteosis and van Buchem disease (VBD) are two rare autosomal recessive disorders that results from osteoblast hyperactivity, in which progressive bone overgrowth leads to very dense bones, distortion of the face, and entrapment of cranial nerves. Sclerosteosis is caused by loss-of-function mutations in the SOST gene which encodes a secreted glycoprotein, sclerostin. VBD is caused by a noncoding deletion that removes a SOST-specific regulatory element in bone. In bone, SOST is expressed predominantly by osteocytes and sclerostin suppresses bone formation by inhibiting the canonical Wnt signaling pathway. Here we describe how human genetics studies in sclerosteosis and VBD patients, in combination with the generation of transgenic and knockout mice, has led to a better understanding of the role of sclerostin in bone metabolism.
    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
    硬化病和vanBuchem病是两种罕见的骨硬化发育不良,由硬化素合成中的遗传缺陷引起。在这篇文章中,我们回顾了人口统计,临床,生物化学,放射学,以及硬化病和vanBuchem病患者的组织学特征,使人们更好地了解了硬化素在人类骨代谢中的作用,我们讨论了这些发现与开发治疗骨质疏松症患者的新疗法的相关性。
    Sclerosteosis and van Buchem disease are two rare bone sclerosing dysplasias caused by genetic defects in the synthesis of sclerostin. In this article we review the demographic, clinical, biochemical, radiological, and histological characteristics of patients with sclerosteosis and van Buchem disease that led to a better understanding of the role of sclerostin in bone metabolism in humans and we discuss the relevance of these findings for the development of new therapeutics for the treatment of patients with osteoporosis.
    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
    在过去的15年中,我们对调节骨重塑的细胞和分子机制的知识得到了扩展,这些机制确定了骨更新的新信号通路以及骨细胞之间以前未知的相互作用。这些发展的核心是对罕见骨骼疾病的研究。这些发现,反过来,已经导致了骨质疏松症的新治疗范例,其中一些处于临床发展的后期阶段。在这篇文章中,我们回顾了三种罕见的骨骼疾病的病例描述,肾盂畸形和颅管增生硬化病和vanBuchem病,导致组织蛋白酶K和硬化蛋白抑制剂的发展,分别,用于治疗骨质疏松症。
    During the past 15 years there has been an expansion of our knowledge of the cellular and molecular mechanisms regulating bone remodeling that identified new signaling pathways fundamental for bone renewal as well as previously unknown interactions between bone cells. Central for these developments have been studies of rare bone disorders. These findings, in turn, have led to new treatment paradigms for osteoporosis some of which are at late stages of clinical development. In this article, we review three rare skeletal disorders with case descriptions, pycnodysostosis and the craniotubular hyperostoses sclerosteosis and van Buchem disease that led to the development of cathepsin K and sclerostin inhibitors, respectively, for the treatment of osteoporosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号