primary chondrocytes

  • 文章类型: Journal Article
    安全和抗炎的基于植物的天然产物在慢性炎性疾病如骨关节炎或炎性肠病的治疗中日益受到关注。其中,藏红花,一种源自番红花污名的香料,可以具有抗炎特性,因此将是用于治疗此类病症的有希望的治疗剂。然而,藏红花在人类体内的抗炎分子机制尚不清楚。在这项研究中,结合人类血清代谢物和细胞培养物,我们评估了消耗专利藏红花提取物(Safr\'InsideTM)产生的循环代谢物对软骨细胞和结肠上皮细胞对炎症应激反应的影响。使用事后测试进行了参数或非参数方差分析。我们证明了藏红花摄入的含有代谢物的人血清减弱了软骨细胞中IL-1β刺激的PGE2和MMP-13的产生,并限制了ICAM-1,MCP-1,iNOS的增加。IL-1β和TNF-α联合炎症刺激后人上皮细胞中的MMP-3。总之,这些数据为藏红花在细胞水平和慢性炎症性疾病中对软骨细胞和肠上皮细胞有益作用的潜在机制提供了新的发现.
    Safe and anti-inflammatory plant-based natural products present an increasing focus in the treatment of chronic inflammatory diseases such as osteoarthritis or inflammatory bowel diseases. Among them, saffron, a spice derived from the stigma of Crocus sativus, could have anti-inflammatory properties and would be therefore a promising therapeutic agent for the treatment of such conditions. However, the anti-inflammatory molecular mechanisms of saffron in humans are still understudied and unclear. In this study, combining human serum metabolites and cell cultures, we evaluated the effect of circulating metabolites from the consumption of a patented saffron extract (Safr\'InsideTM) on the chondrocytes and colon epithelial cell responses to inflammatory stress. Parametric or non-parametric Analysis of Variance with post hoc tests was performed. We demonstrated that human serum containing metabolites from saffron intake attenuated IL-1β-stimulated production of PGE2 and MMP-13 in chondrocyte cells and limited the increase in ICAM-1, MCP-1, iNOS, and MMP-3 in human epithelial cells following combined IL-1β and TNF-α inflammatory stimulation. Altogether, these data provide new findings into the mechanisms underlying the beneficial effects of saffron on chondrocytes and enterocyte cells at the cellular level and in the context of chronic inflammatory disorders.
    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:基于CRISPR-Cas9的基因组工程代表了软骨组织工程和理解驱动软骨疾病的分子途径的强大治疗工具。然而,原代软骨细胞难以转染并在单层(2D)细胞培养过程中迅速去分化,使单细胞来源的编辑克隆群体的长时间扩增不可行。出于这个原因,专注于软骨和风湿性疾病的功能遗传学研究长期以来一直在细胞模型中进行,这些模型很难概括人类软骨组织的天然分子特性(例如,细胞系,诱导多能干细胞)。这里,我们着手开发一种适用于来自不同软骨来源的软骨细胞群体的非病毒CRISPR-Cas9批量基因编辑方法.
    方法:我们筛选了用于原代多指软骨细胞中核糖核蛋白(RNP)递送的电穿孔和脂质纳米粒,和优化的RNP试剂组装。我们淘汰了RELA(也称为p65),核因子κB(NF-κB)的亚基,在多指软骨细胞中,并通过RT-qPCR和Western印迹进一步表征敲除(KO)细胞。我们测试了来自不同软骨来源的软骨细胞中的RELAKO,并通过RT-qPCR表征了它们的表型。我们检查了在存在和不存在白介素1β(IL-1β)的情况下野生型(WT)和KO细胞颗粒的软骨形成潜力。
    结果:我们确立了电穿孔作为软骨细胞增强转染和编辑效率的最佳转染技术,同时保持高细胞活力。我们以前所未有的90%的效率淘汰了RELA,证实与未编辑的细胞相比,IL-1β刺激后的炎症途径激活较低。我们的方案可以很容易地转移到从骨关节炎(OA)患者收获的原代人软骨细胞,人FE002软骨祖细胞,牛软骨细胞,和人类软骨细胞系,使用相同的协议实现相当的平均RELAKO编辑水平。来自原代人软骨细胞的所有KO颗粒保留了与WT细胞相当的软骨形成能力,并在发炎条件下显示出增强的基质保留。
    结论:我们展示了我们的批量基因编辑方法的适用性,以开发有效的自体和同种异体现成的基因治疗策略,并使人类软骨细胞的功能遗传学研究能够揭示软骨疾病的分子机制。
    BACKGROUND: CRISPR-Cas9-based genome engineering represents a powerful therapeutic tool for cartilage tissue engineering and for understanding molecular pathways driving cartilage diseases. However, primary chondrocytes are difficult to transfect and rapidly dedifferentiate during monolayer (2D) cell culture, making the lengthy expansion of a single-cell-derived edited clonal population not feasible. For this reason, functional genetics studies focused on cartilage and rheumatic diseases have long been carried out in cellular models that poorly recapitulate the native molecular properties of human cartilaginous tissue (e.g., cell lines, induced pluripotent stem cells). Here, we set out to develop a non-viral CRISPR-Cas9, bulk-gene editing method suitable for chondrocyte populations from different cartilaginous sources.
    METHODS: We screened electroporation and lipid nanoparticles for ribonucleoprotein (RNP) delivery in primary polydactyly chondrocytes, and optimized RNP reagents assembly. We knocked out RELA (also known as p65), a subunit of the nuclear factor kappa B (NF-κB), in polydactyly chondrocytes and further characterized knockout (KO) cells with RT-qPCR and Western Blot. We tested RELA KO in chondrocytes from diverse cartilaginous sources and characterized their phenotype with RT-qPCR. We examined the chondrogenic potential of wild-type (WT) and KO cell pellets in presence and absence of interleukin-1β (IL-1β).
    RESULTS: We established electroporation as the optimal transfection technique for chondrocytes enhancing transfection and editing efficiency, while preserving high cell viability. We knocked out RELA with an unprecedented efficiency of ~90%, confirming lower inflammatory pathways activation upon IL-1β stimulation compared to unedited cells. Our protocol could be easily transferred to primary human chondrocytes harvested from osteoarthritis (OA) patients, human FE002 chondroprogenitor cells, bovine chondrocytes, and a human chondrocyte cell line, achieving comparable mean RELA KO editing levels using the same protocol. All KO pellets from primary human chondrocytes retained chondrogenic ability equivalent to WT cells, and additionally displayed enhanced matrix retention under inflamed conditions.
    CONCLUSIONS: We showcased the applicability of our bulk gene editing method to develop effective autologous and allogeneic off-the-shelf gene therapies strategies and to enable functional genetics studies in human chondrocytes to unravel molecular mechanisms of cartilage diseases.
    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
    骨关节炎微环境的分子生态位由天然软骨细胞组成,循环免疫细胞,和它们各自的炎症介质。虽然,M2巨噬细胞在骨关节炎(OA)期间浸润关节组织以启动软骨修复,下面的机械串扰仍然是未知的。我们的研究建立了人OA软骨细胞和M2巨噬细胞在3D球体和3D生物打印的丝明胶构建体中的共培养系统。由于Wnt/β-连环蛋白途径的上调,丝素蛋白-明胶生物墨水支持软骨形成分化已经得到充分证实。此外,抗炎M2巨噬细胞的存在显着上调了软骨形成生物标志物的表达(COL-II,ACAN),软骨细胞肥大(COL-X)的表达减弱,即使不存在白细胞介素,软骨细胞去分化(COL-I)和基质分解代谢(MMP-1和MMP-13)基因也是如此。此外,3D生物打印的共培养模型在刺激软骨再生和OA抑制方面比球体模型占上风,强调丝素蛋白明胶在促进软骨形成中的作用。此外,3D生物打印的丝明胶构建体进一步支持M2巨噬细胞的稳定抗炎表型的维持。因此,在3D环境中,原发性OAC和M2巨噬细胞之间的直接相互作用以及M2细胞释放可溶性抗炎因子显著有助于更好地理解免疫细胞介导的OA愈合的分子机制. .
    The molecular niche of an osteoarthritic microenvironment comprises the native chondrocytes, the circulatory immune cells, and their respective inflammatory mediators. Although M2 macrophages infiltrate the joint tissue during osteoarthritis (OA) to initiate cartilage repair, the mechanistic crosstalk that dwells underneath is still unknown. Our study established a co-culture system of human OA chondrocytes and M2 macrophages in 3D spheroids and 3D bioprinted silk-gelatin constructs. It is already well established that Silk fibroin-gelatin bioink supports chondrogenic differentiation due to upregulation of the Wnt/β-catenin pathway. Additionally, the presence of anti-inflammatory M2 macrophages significantly upregulated the expression of chondrogenic biomarkers (COL-II, ACAN) with an attenuated expression of the chondrocyte hypertrophy (COL-X), chondrocyte dedifferentiation (COL-I) and matrix catabolism (MMP-1 and MMP-13) genes even in the absence of the interleukins. Furthermore, the 3D bioprinted co-culture model displayed an upper hand in stimulating cartilage regeneration and OA inhibition than the spheroid model, underlining the role of silk fibroin-gelatin in encouraging chondrogenesis. Additionally, the 3D bioprinted silk-gelatin constructs further supported the maintenance of stable anti-inflammatory phenotype of M2 macrophage. Thus, the direct interaction between the primary OAC and M2 macrophages in the 3D context, along with the release of the soluble anti-inflammatory factors by the M2 cells, significantly contributed to a better understanding of the molecular mechanisms responsible for immune cell-mediated OA healing.
    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
    BACKGROUND: Osteoarthritis (OA) is an inflammatory response in chondrocytes, causing extracellular matrix (ECM) degradation and cartilage destruction, affecting millions of people worldwide. Chinese herbal formulae BuShen JianGu Fang (BSJGF) has been clinically applied for treating OA-related syndromes, but the underlying mechanism still unclear.
    METHODS: The components of BSJGF were analyzed by liquid chromatography-mass spectrometry (LC-MS). To make a traumatic OA model, the anterior cruciate ligament of 6-8-week-old male SD rats were cut and then the 0.4 mm metal was used to destroy the knee joint cartilage. OA severity was assessed by histological and Micro-CT. Mouse primary chondrocytes were utilized to investigate the mechanism of BSJGF alleviate osteoarthritis, which was examined by RNA-seq technology combined with a series of functional experiments.
    RESULTS: A total 619 components were identified by LC-MS. In vivo, BSJGF treatment result in a higher articular cartilage tissue area compared to IL-1β group. Treatment also significantly increased Tb.Th, BV/TV and BMD of subchondral bone (SCB), which implied a protective effect on maintaining the stabilization of SCB microstructure. In vitro results indicated BSJGF promoted chondrocyte proliferation, increased the expression level of cartilage-specific genes (Sox9, Col2a1, Acan) and synthesized acidic polysaccharide, while inhibiting the release of catabolic enzymes and production of reactive oxygen species (ROS) induced by IL-1β. Transcriptome analysis showed that there were 1471 and 4904 differential genes between IL-1β group and blank group, BSJGF group and IL-1β group, respectively, including matrix synthesis related genes (Col2a1, H19, Acan etc.), inflammation related genes (Comp, Pcsk6, Fgfr3 etc.) and oxidative stress related genes (Gm26917, Bcat1, Sod1 etc.). Furthermore, KEGG analysis and validation results showed that BSJGF reduces OA-mediated inflammation and cartilage damaged due to modulation of NF-κB/Sox9 signaling axis.
    CONCLUSIONS: The innovation of the present study was the elucidation of the alleviating cartilage degradation effect of BSJGF in vivo and in vitro and discovery of its mechanism through RNA-seq combined with function experiments, which provides a biological rationale for the clinical application of BSJGF for OA treatment.
    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
    背景:软骨细胞是软骨中唯一的细胞成分,再生能力差。因此,修复受损的软骨仍然是一个巨大的挑战。梅花鹿鹿角主要由软骨组织组成,具有惊人的修复和更新能力。我们先前的研究已经证明转化生长因子β(TGF-β1)被认为是参与快速生长的关键分子,在软骨层中表现最强。然而,鹿TGF-β1是否具有与小鼠等其他物种明显不同的功能,以及调节软骨生长的分子机制。
    方法:从新生小鼠肋软骨中收集原代软骨细胞。通过RNA测序(RNA-seq)技术结合定量实时聚合酶链反应(qRT-PCR)和免疫荧光测定(IFA)等验证方法,阐明了TGF-β1对原代软骨细胞活力的影响。使用DEGseq包鉴定差异表达基因。
    结果:我们的结果表明,鹿TGF-β1的过表达可能促进软骨细胞增殖和细胞外基质(ECM)的合成,同时通过调节转录因子抑制软骨细胞分化,生长因子,ECM相关基因,增殖和分化标记基因,如Comp,Fgfr3、Atf4、Stat1等。,和信号通路,如MAPK信号通路,TRP通道等的炎症介质调节。此外,通过比较鹿TGF-β1和小鼠TGF-β1的氨基酸序列和结构,我们发现鹿TGF-β1和小鼠TGF-β1蛋白在臂结构域上主要在结构上不同,这是主要的功能域。表型鉴定结果表明,鹿TGF-β1可能比小鼠TGF-β1具有更强的功能。
    结论:这些结果表明,鹿TGF-β1具有通过调节软骨细胞增殖来促进软骨形成的能力,分化和ECM合成。这项研究提供了有关鹿TGF-β1对软骨细胞活力影响的分子机制的见解。
    BACKGROUND: Chondrocytes are the only cell components in the cartilage, which has the poor regeneration ability. Thus, repairing damaged cartilage remains a huge challenge. Sika deer antlers are mainly composed of cartilaginous tissues that have an astonishing capacity for repair and renewal. Our previous study has demonstrated the transforming growth factor β (TGF-β1) is considered to be a key molecule involved in rapid growth, with the strongest expression in the cartilage layer. However, it remains to be clarified whether deer TGF-β1 has significantly different function from other species such as mouse, and what is the molecular mechanism of regulating cartilage growth.
    METHODS: Primary chondrocytes was collected from new born mouse rib cartilage. The effect of TGF-β1 on primary chondrocytes viability was elucidated by RNA sequencing (RNA-seq) technology combined with validation methods such as quantitative real-time polymerase chain reaction (qRT-PCR) and immunofluorescence assay (IFA). Differential expression genes were identified using the DEGseq package.
    RESULTS: Our results demonstrated that the overexpression of deer TGF-β1 possibly promoted chondrocyte proliferation and extracellular matrix (ECM) synthesis, while simultaneously suppressing chondrocyte differentiation through regulating transcription factors, growth factors, ECM related genes, proliferation and differentiation marker genes, such as Comp, Fgfr3, Atf4, Stat1 etc., and signaling pathways such as the MAPK signaling pathway, inflammatory mediator regulation of TRP channels etc. In addition, by comparing the amino acid sequence and structures between the deer TGF-β1 and mouse TGF-β1, we found that deer TGF-β1 and mouse TGF-β1 proteins are mainly structurally different in arm domains, which is the main functional domain. Phenotypic identification results showed that deer TGF-β1 may has stronger function than mouse TGF-β1.
    CONCLUSIONS: ​These results suggested that deer TGF-β1 has the ability to promote chondrogenesis by regulating chondrocyte proliferation, differentiation and ECM synthesis. This study provides insights into the molecular mechanisms underlying the effects of deer TGF-β1 on chondrocyte viability.
    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
    背景:骨关节炎(OA)与软骨细胞中的氧化应激损伤(OSI)和炎症反应有关。本研究旨在探讨高迁移率族蛋白A1(HMGA1)在白细胞介素-1β(IL-1β)诱导的OSI和原代软骨细胞炎症反应中的作用机制。
    方法:培养原代软骨细胞并用IL-1β处理,建立OA细胞模型。HMGA1,含有Jumonji域的3(JMJD3)的水平,通过实时定量聚合酶链反应和蛋白质印迹分析测定细胞中的ZEB1。细胞活力,肿瘤坏死因子-α的含量,IL-6和IL-10,活性氧水平,和谷胱甘肽过氧化物酶活性通过细胞计数试剂盒-8测定,酶联免疫吸附测定,和测定试剂盒。通过染色质免疫沉淀确定JMJD3启动子上HMGA1的富集水平以及ZEB1启动子区域上赖氨酸27(H3K27me3)上JMJD3和三甲基化组蛋白H3的富集水平。进行功能性拯救实验以分析ZEB1和JMJD3对IL-1β诱导的软骨细胞的影响。
    结果:IL-1β治疗诱导HMGA1上调,OSI,和软骨细胞的炎症反应。HMGA1下调降低了IL-1β诱导的软骨细胞OSI和炎症反应。机械上,HMGA1与JMJD3启动子结合,促进JMJD3转录,和JMJD3在ZEB1启动子上诱导H3K27me3去甲基化以促进ZEB1转录。JMJD3或ZEB1的过表达中和了IL-1β诱导的软骨细胞中沉默HMGA1的保护作用。
    结论:HMGA1通过促进JMJD3和ZEB1加重IL-1β诱导的软骨细胞OSI和炎症反应。
    Osteoarthritis (OA) is associated with oxidative stress injury (OSI) and inflammatory responses in chondrocytes. This study sought to explore the mechanism of high mobility group A1 (HMGA1) in interleukin-1beta (IL-1β)-induced OSI and inflammatory responses in primary chondrocytes.
    Primary chondrocytes were cultured and treated with IL-1β to establish an OA-cell model. Levels of HMGA1, Jumonji domain-containing 3 (JMJD3), and ZEB1 in cells were determined by real-time quantitative polymerase chain reaction and Western blot analysis. Cell viability, contents of tumor necrosis factor-α, IL-6, and IL-10, reactive oxygen species level, and glutathione peroxidase activity were assessed by the cell counting kit-8 assay, enzyme-linked immunosorbent assay, and assay kits. Enrichment levels of HMGA1 on the JMJD3 promoter and enrichment levels of JMJD3 and trimethylated histone H3 at lysine 27 (H3K27me3) on the ZEB1 promoter region were determined by chromatin immunoprecipitation. Functional rescue experiments were performed to analyze the impact of ZEB1 and JMJD3 on IL-1β-induced chondrocytes.
    IL-1β treatment induced HMGA1 upregulation, OSI, and inflammatory responses in chondrocytes. HMGA1 downregulation reduced IL-1β-induced OSI and inflammatory responses in chondrocytes. Mechanically, HMGA1 was bound to the JMJD3 promoter to promote JMJD3 transcription, and JMJD3 induced demethylation of H3K27me3 on the ZEB1 promoter to promote ZEB1 transcription. Overexpression of JMJD3 or ZEB1 neutralized the protective role of silencing HMGA1 in IL-1β-induced chondrocytes.
    HMGA1 aggravated IL-1β-induced OSI and inflammatory responses in chondrocytes through the promotion of JMJD3 and ZEB1.
    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
    Aging is a major risk factor of osteoarthritis, which is characterized by the degeneration of articular cartilage. CCN3, a member of the CCN family, is expressed in cartilage and has various physiological functions during chondrocyte development, differentiation, and regeneration. Here, we examine the role of CCN3 in cartilage maintenance. During aging, the expression of Ccn3 mRNA in mouse primary chondrocytes from knee cartilage increased and showed a positive correlation with p21 and p53 mRNA. Increased accumulation of CCN3 protein was confirmed. To analyze the effects of CCN3 in vitro, either primary cultured human articular chondrocytes or rat chondrosarcoma cell line (RCS) were used. Artificial senescence induced by H2O2 caused a dose-dependent increase in Ccn3 gene and CCN3 protein expression, along with enhanced expression of p21 and p53 mRNA and proteins, as well as SA-β gal activity. Overexpression of CCN3 also enhanced p21 promoter activity via p53. Accordingly, the addition of recombinant CCN3 protein to the culture increased the expression of p21 and p53 mRNAs. We have produced cartilage-specific CCN3-overexpressing transgenic mice, and found degradative changes in knee joints within two months. Inflammatory gene expression was found even in the rib chondrocytes of three-month-old transgenic mice. Similar results were observed in human knee articular chondrocytes from patients at both mRNA and protein levels. These results indicate that CCN3 is a new senescence marker of chondrocytes, and the overexpression of CCN3 in cartilage may in part promote chondrocyte senescence, leading to the degeneration of articular cartilage through the induction of p53 and p21.
    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
    OBJECTIVE: Obesity is a known risk factor for knee osteoarthritis (OA). Diabetes has been associated with progression of OA and metformin is the first-line treatment in type 2 diabetes. The effect of the body mass index (BMI) and metformin on the expression of certain matrix genes in human chondrocytes is unclear. The purpose of this study was to investigate the effect of BMI and metformin on the expression of matrix genes in primary human chondrocytes.
    METHODS: Adult female patients undergoing knee arthroplasty for end-stage OA were enrolled. Primary chondrocytes were cultivated and stimulated with metformin. Matrix gene expression was analyzed using polymerase chain reaction. Clinical data were used in multivariable regression models to assess the influence of BMI and metformin stimulation on gene expression.
    RESULTS: A total of 14 patients were analyzed. BMI was a predictor of increased expression in ADAMTS5 (β = -0.11, P = 0.03). Metformin slightly reduced expression in ADAMTS5 (β = 0.34, P = 0.04), HIF-1a (β = 0.39, P = 0.04), IL4 (β = 0.30, P = 0.02), MMP1 (β = 0.47, P < 0.01), and SOX9 (β = 0.37, P = 0.03). The hip-knee-ankle angle and proton pump inhibitors (PPIs) intake were associated with reduced SOX9 expression (β = 0.23, P < 0.01; β = 2.39, P < 0.01). Higher C-reactive protein (CRP) levels were associated with increased MMP1 expression (β = -0.16, P = 0.02).
    CONCLUSIONS: We found that BMI exerts a destructive effect via induction of ADAMTS5. Metformin reduced the expression of catabolic genes ADAMTS5 and MMP1 and might play a role in disease prevention. Limb malalignment and PPI intake was associated with a reduced expression of SOX9, and higher CRP levels correlated with increased MMP1 expression, indicating a destructive process.
    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
    There is an unmet need for a single-stage cartilage repair treatment that is cost-effective and chondrocyte-based.
    To evaluate the safety and preliminary efficacy of autologous freshly isolated primary chondrocytes and bone marrow mononucleated cells (MNCs) seeded into a PolyActive scaffold in patients with symptomatic cartilage lesions of the knee.
    Case series; Level of evidence, 4.
    A total of 40 patients with symptomatic knee cartilage lesions were treated with freshly isolated autologous chondrocytes combined with bone marrow MNCs delivered in a biodegradable load-bearing scaffold. The treatment requires only 1 surgical intervention and is potentially a cost-effective alternative to autologous chondrocyte implantation. The primary chondrocytes and bone marrow MNCs were isolated, washed, counted, mixed, and seeded into a load-bearing scaffold in the operating room. Patients were followed up at 3, 6, 12, 18, and 24 months. Primary endpoints were treatment-related adverse events up to 3 months, adverse implant effects between 3 and 24 months, and the implant success rate at 3 months as measured by lesion filling.
    Successful lesion filling (≥67% on magnetic resonance imaging) was found in 40 patients at 3 months and in 32 of the 32 patients analyzed at 24 months. Significant improvement over baseline was found for visual analog scale for pain from 3 months onward; Knee injury and Osteoarthritis Outcome Score (KOOS)-Pain and KOOS-Activities of Daily Living from 6 months onward; for KOOS-Symptoms and Stiffness, KOOS-Quality of Life and International Knee Documentation Committee from 12 months onward; and for KOOS-Sport and Recreation from 18 months onward. Hyaline-like repair tissue was found in 22 of 31 patients available for biopsy. Arthralgia and joint effusion were the most common adverse events. Scaffold delamination and adhesions led to removal of the implant in 2 patients.
    The treatment of knee cartilage lesions with autologous primary chondrocytes and bone marrow MNCs, both isolated and seeded into a load-bearing PolyActive scaffold within a single surgical intervention, is safe and clinically effective. Good lesion fill and sustained clinically important and statistically significant improvement in all patient-reported outcome scores were found throughout the 24-month study. Hyaline-like cartilage was observed on biopsy specimen in at least 22 of the 40 patients.
    NCT01041885 (ClinicalTrials.gov identifier).
    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
    目的:Hox类的Homeobox基因是骨骼元素的正确模式所必需的,并且在软骨分化中起作用。在软骨发育过程中过表达Hoxc8和Hoxd4的转基因小鼠中,作者观察到严重的缺陷,即,软骨的物理不稳定性,未成熟软骨细胞的积累,和减少成熟到肥大。为了定义这些缺陷背后的分子基础,作者使用Affymetrix微阵列平台进行了基因表达谱分析.
    结果:在妊娠18.5天从Hoxc8和Hoxd4转基因小鼠胚胎肋软骨中分离出原代软骨细胞。在这两种情况下,差异表达的基因被鉴定为在细胞增殖和细胞周期调节中起作用。两个实验组的对照之间的比较没有发现显着差异,如预期。然而,差异表达基因库在Hoxc8-和Hoxd4-转基因软骨之间没有重叠。这包括不同的Wnt基因,细胞周期,和凋亡调节剂。
    结论:在E18.5时,Hoxc8和Hoxd4转录因子的过表达改变了软骨细胞的转录谱。两种转基因条件之间改变的基因表达库的差异表明,两种转基因范式中软骨缺损的分子机制可能不同。尽管表型相似。
    OBJECTIVE: Homeobox genes of the Hox class are required for proper patterning of skeletal elements and play a role in cartilage differentiation. In transgenic mice with overexpression of Hoxc8 and Hoxd4 during cartilage development, the authors observed severe defects, namely, physical instability of cartilage, accumulation of immature chondrocytes, and decreased maturation to hypertrophy. To define the molecular basis underlying these defects, the authors performed gene expression profiling using the Affymetrix microarray platform.
    RESULTS: Primary chondrocytes were isolated from Hoxc8- and Hoxd4-transgenic mouse embryo rib cartilage at 18.5 days of gestation. In both cases, differentially expressed genes were identified that have a role in cell proliferation and cell cycle regulation. A comparison between the controls for both experimental groups did not reveal significant differences, as expected. However, the repertoires of differentially expressed genes were found not to overlap between Hoxc8- and Hoxd4-transgenic cartilage. This included different Wnt genes, cell cycle, and apoptosis regulators.
    CONCLUSIONS: Overexpression of Hoxc8 and Hoxd4 transcription factors alters transcriptional profiles in chondrocytes at E18.5. The differences in repertoires of altered gene expression between the 2 transgenic conditions suggest that the molecular mechanisms underlying the cartilage defects may be different in both transgenic paradigms, despite apparently similar phenotypes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号