skeletal stem cell

  • 文章类型: Journal Article
    临界大小的节段性骨缺损不能自发愈合,导致残疾和死亡率显著增加。然而,目前利用骨移植物的治疗面临着从供体可用性到骨整合不良的各种挑战。生长因子等药物会增加癌症风险,而且非常昂贵。这里,我们报道了一种仅通过机械生物学设计促进骨再生的多孔生物陶瓷支架。使用高精度3D打印技术创建具有高和低孔隙曲率的两种类型的支架,以制造100微米的孔隙曲率半径。虽然两者都能够支持骨形成,高曲率孔导致更高的异位骨形成和血管侵袭增加。具有高曲率孔的支架还通过激活机械敏感性途径促进了临界尺寸的节段性骨缺损的更快再生。在修复的早期阶段,高曲率孔从骨膜和骨髓中募集了骨骼干细胞和H型血管。高曲率孔增加了移植的GFP标记的骨骼干细胞(SSC)的存活率,并招募了更多的宿主SSC。一起来看,我们的生物陶瓷支架具有确定的微米级孔隙曲率,证明了骨科支架设计的机械生物学方法。本文受版权保护。保留所有权利。
    Critical-sized segmental bone defects cannot heal spontaneously, leading to disability and significant increase in mortality. However, current treatments utilizing bone grafts face a variety of challenges from donor availability to poor osseointegration. Drugs such as growth factors increase cancer risk and are very costly. Here, a porous bioceramic scaffold that promotes bone regeneration via solely mechanobiological design is reported. Two types of scaffolds with high versus low pore curvatures are created using high-precision 3D printing technology to fabricate pore curvatures radius in the 100s of micrometers. While both are able to support bone formation, the high-curvature pores induce higher ectopic bone formation and increased vessel invasion. Scaffolds with high-curvature pores also promote faster regeneration of critical-sized segmental bone defects by activating mechanosensitive pathways. High-curvature pore recruits skeletal stem cells and type H vessels from both the periosteum and the marrow during the early phase of repair. High-curvature pores have increased survival of transplanted GFP-labeled skeletal stem cells (SSCs) and recruit more host SSCs. Taken together, the bioceramic scaffolds with defined micrometer-scale pore curvatures demonstrate a mechanobiological approach for orthopedic scaffold design.
    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
    纤维发育不良(FD)是一种马赛克骨骼疾病,由编码Gαs的GNAS的体细胞激活变体引起,并导致骨髓基质细胞(BMSC)中过度的环磷酸腺苷信号传导。Gαs激活在BMSC转录组中的作用及其如何影响FD病变微环境尚不清楚。我们分析了在BMSC转录组和分泌组中由Gαs激活诱导的变化。FD患者和健康志愿者培养的BMSCs差异基因表达的RNAseq分析,从FD的诱导型小鼠模型中,被执行,并将两个模型的转录组图谱结合起来,以构建一个稳健的FDBMSC遗传签名。与Gα激活相关的途径,细胞因子信号,并鉴定了细胞外基质沉积。为了评估FD发病机制中几种关键分泌因子的调节,在培养基中测量细胞因子和其他因子.还从FD患者的血浆样本中筛选了细胞因子,几种细胞因子与其疾病负担评分呈正相关,以及彼此和骨转换标记,被发现了。这些数据支持促炎,FDBMSCs的促破骨细胞行为,并指出几种细胞因子和其他分泌因子作为FD的可能治疗靶标和/或循环生物标志物。
    Fibrous dysplasia (FD) is a mosaic skeletal disorder caused by somatic activating variants of GNAS encoding for Gαs and leading to excessive cyclic adenosine monophosphate signaling in bone-marrow stromal cells (BMSCs). The effect of Gαs activation in the BMSC transcriptome and how it influences FD lesion microenvironment are unclear. We analyzed changes induced by Gαs activation in the BMSC transcriptome and secretome. RNAseq analysis of differential gene expression of cultured BMSCs from patients with FD and healthy volunteers, and from an inducible mouse model of FD, was performed, and the transcriptomic profiles of both models were combined to build a robust FD BMSC genetic signature. Pathways related to Gαs activation, cytokine signaling, and extracellular matrix deposition were identified. To assess the modulation of several key secreted factors in FD pathogenesis, cytokines and other factors were measured in culture media. Cytokines were also screened in a collection of plasma samples from patients with FD, and positive correlations of several cytokines to their disease burden score, as well as to one another and bone turnover markers, were found. These data support the pro-inflammatory, pro-osteoclastic behavior of FD BMSCs and point to several cytokines and other secreted factors as possible therapeutic targets and/or circulating biomarkers for FD.
    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
    骨被认为是少数无纤维疤痕愈合的组织之一。骨膜的外层覆盖有纤维组织,其在骨形成中的功能是未知的。我们在此开发了一种系统,可区分纤维层骨膜细胞(FL-PC)与小鼠形成层骨膜和骨髓中的骨骼干/祖细胞(SSPC)的命运。我们发现FL-PCs不参与稳态成骨,但在骨折愈合过程中形成了纤维软骨骨痂的主体。此外,FL-PCs在骨折后侵入形成层骨膜和骨髓,形成新SSPCs,在整个成年期继续维持愈合的骨骼。与先前存在的SSPC相比,FL-PC衍生的新SSPC表达较低水平的成骨标记基因并显示较低的成骨分化活性。与此一致,愈合的骨骼比正常骨骼更薄,形成更慢。因此,纤维骨膜在骨折后成为骨骼的细胞起源,并永久改变骨骼特性。
    Bone is regarded as one of few tissues that heals without fibrous scar. The outer layer of the periosteum is covered with fibrous tissue, whose function in bone formation is unknown. We herein developed a system to distinguish the fate of fibrous-layer periosteal cells (FL-PCs) from the skeletal stem/progenitor cells (SSPCs) in the cambium-layer periosteum and bone marrow in mice. We showed that FL-PCs did not participate in steady-state osteogenesis, but formed the main body of fibrocartilaginous callus during fracture healing. Moreover, FL-PCs invaded the cambium-layer periosteum and bone marrow after fracture, forming neo-SSPCs that continued to maintain the healed bones throughout adulthood. The FL-PC-derived neo-SSPCs expressed lower levels of osteogenic signature genes and displayed lower osteogenic differentiation activity than the preexisting SSPCs. Consistent with this, healed bones were thinner and formed more slowly than normal bones. Thus, the fibrous periosteum becomes the cellular origin of bones after fracture and alters bone properties permanently.
    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
    骨形态发生蛋白2(BMP2)是一种有吸引力的成骨和软骨生长因子,可促进腱-骨愈合。最近,据报道,可溶性血管内皮生长因子(VEGF)受体1(sVEGFR1)(VEGF受体拮抗剂)可以增强BMP2诱导的骨修复和软骨再生;因此,它们的联合应用可能是改善腱-骨愈合的有希望的治疗方法。此外,BMP2可以刺激骨骼干细胞(SSC)的扩增和形成,负责受伤的腱-骨界面修复。然而,BMP2和sVEGFR1联合给药是否比单独给药BMP2能更好地增加肌腱损伤激活的SSCs还需要进一步研究.
    研究BMP2联合sVEGFR1对腱-骨愈合和损伤激活SSC谱系的影响。
    对照实验室研究。
    总共128只接受单侧冈上肌腱脱离和修复的C57BL/6小鼠被随机分为4组:(1)未经治疗的对照组;(2)水凝胶组,在损伤部位局部注射空白水凝胶;(3)BMP2组,其接受了具有BMP2的水凝胶的注射;和(4)具有sVEGFR1基团的BMP2,其接受具有BMP2和sVEGFR1的水凝胶的注射。组织学,显微计算机断层扫描,并在术后4周和8周进行生物力学测试以评估腱-骨愈合。此外,流式细胞术检测SSC及其下游分化亚型的比例,包括骨头,软骨,和基质祖细胞;骨祖细胞;术后1周,冈上肌腱内的前软骨形成祖细胞。
    BMP2与sVEGFR1组的修复界面显示出显著改善的胶原纤维连续性,纤维软骨增加,新形成的骨骼更大,力学性能较其他3组提高。有更多的SSC;骨骼,软骨,和基质祖细胞;骨祖细胞;具有sVEGFR1组的BMP2中前软骨形成祖细胞比其他组。
    我们的研究表明,BMP2和sVEGFR1的联合递送可以促进腱-骨愈合,并刺激SSC及其下游后代在受损的腱-骨界面内的扩张。
    BMP2与sVEGFR1的联合治疗可能是肌腱损伤愈合的良好临床治疗方法。
    UNASSIGNED: Bone morphogenetic protein 2 (BMP2) is an appealing osteogenic and chondrogenic growth factor for promoting tendon-bone healing. Recently, it has been reported that soluble vascular endothelial growth factor (VEGF) receptor 1 (sVEGFR1) (a VEGF receptor antagonist) could enhance BMP2-induced bone repair and cartilage regeneration; thus, their combined application may represent a promising treatment to improve tendon-bone healing. Moreover, BMP2 could stimulate skeletal stem cell (SSC) expansion and formation, which is responsible for wounded tendon-bone interface repair. However, whether the codelivery of BMP2 and sVEGFR1 increases tendon enthesis injury-activated SSCs better than does BMP2 alone needs further research.
    UNASSIGNED: To study the effect of BMP2 combined with sVEGFR1 on tendon-bone healing and injury-activated SSC lineage.
    UNASSIGNED: Controlled laboratory study.
    UNASSIGNED: A total of 128 C57BL/6 mice that underwent unilateral supraspinatus tendon detachment and repair were randomly assigned to 4 groups: (1) untreated control group; (2) hydrogel group, which received a local injection of the blank hydrogel at the injured site; (3) BMP2 group, which received an injection of hydrogel with BMP2; and (4) BMP2 with sVEGFR1 group, which received an injection of hydrogel with BMP2 and sVEGFR1. Histology, micro-computed tomography, and biomechanical tests were conducted to evaluate tendon-bone healing at 4 and 8 weeks after surgery. In addition, flow cytometry was performed to detect the proportion of SSCs and their downstream differentiated subtypes, including bone, cartilage, and stromal progenitors; osteoprogenitors; and pro-chondrogenic progenitors within supraspinatus tendon enthesis at 1 week postoperatively.
    UNASSIGNED: The repaired interface in BMP2 with sVEGFR1 group showed a significantly improved collagen fiber continuity, increased fibrocartilage, greater newly formed bone, and elevated mechanical properties compared with the other 3 groups. There were more SSCs; bone, cartilage, and stromal progenitors; osteoprogenitors; and pro-chondrogenic progenitors in the BMP2 with sVEGFR1 group than that in the other groups.
    UNASSIGNED: Our study suggests that the combined delivery of BMP2 and sVEGFR1 could promote tendon-bone healing and stimulate the expansion of SSCs and their downstream progeny within the injured tendon-bone interface.
    UNASSIGNED: Combining BMP2 with sVEGFR1 may be a good clinical treatment for wounded tendon enthesis 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: Preprint
    成骨不全症(OI)是一种低骨量和骨折风险增加的疾病,由于一系列遗传变异,主要包括编码Ⅰ型胶原蛋白的基因突变。虽然众所周知,OI反映了骨形成成骨细胞活性的缺陷,目前尚不清楚OI是否也反映了骨骼中许多其他细胞类型的缺陷,包括骨骼血管内皮或产生成骨细胞的骨骼干细胞群的缺陷,以及纠正这些更广泛的缺陷是否具有治疗作用。这里,我们发现,在Col1a2oim/oim小鼠中,骨骼干细胞(SSC)和骨骼动脉内皮细胞(AECs)的数量增加,研究良好的中度至重度OI动物模型,表明血管SSC生态位的破坏是OI发病机制的特征。此外,将Col1a2oim/oim小鼠与缺乏骨骼血管生成和骨形成负调节剂的小鼠交叉,Schnurri3(SHN3),不仅纠正了SSC和AEC表型,而且还有力地纠正了骨量和自发性骨折表型。由于这一发现表明SHN3抑制用于治疗OI的强大治疗效用,骨靶向AAV用于介导Sn3敲低,挽救Col1a2oim/oim表型,并提供靶向SHN3治疗OI的治疗概念证明。总的来说,这项工作既为抑制SHN3通路提供了概念验证,也更广泛地解决了茎/骨祖细胞生态位的缺陷,这是治疗OI的一种策略.
    Osteogenesis imperfecta (OI) is a disorder of low bone mass and increased fracture risk due to a range of genetic variants that prominently include mutations in genes encoding type collagen. While it is well known that OI reflects defects in the activity of bone-forming osteoblasts, it is currently unclear whether OI also reflects defects in the many other cell types comprising bone, including defects in skeletal vascular endothelium or the skeletal stem cell populations that give rise to osteoblasts and whether correcting these broader defects could have therapeutic utility. Here, we find that numbers of skeletal stem cells (SSCs) and skeletal arterial endothelial cells (AECs) are augmented in Col1a2oim/oim mice, a well-studied animal model of moderate to severe OI, suggesting that disruption of a vascular SSC niche is a feature of OI pathogenesis. Moreover, crossing Col1a2oim/oim mice to mice lacking a negative regulator of skeletal angiogenesis and bone formation, Schnurri 3 (SHN3), not only corrected the SSC and AEC phenotypes but moreover robustly corrected the bone mass and spontaneous fracture phenotypes. As this finding suggested a strong therapeutic utility of SHN3 inhibition for the treatment of OI, a bone-targeting AAV was used to mediate Shn3 knockdown, rescuing the Col1a2oim/oim phenotype and providing therapeutic proof-of-concept for targeting SHN3 for the treatment of OI. Overall, this work both provides proof-of-concept for inhibition of the SHN3 pathway and more broadly addressing defects in the stem/osteoprogentior niche as is a strategy to treat OI.
    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
    骨骼干/祖细胞(SSPC)是位于骨骼内的组织特异性干/祖细胞,有助于骨骼发育。稳态和再生。然而,小鼠长骨中SSPC种群的异质性及其各自的再生能力仍有待进一步阐明。在这项研究中,我们使用小鼠后肢芽的单细胞RNA测序(scRNA-seq)数据集进行整合分析,出生后的长骨,长骨骨折.我们的分析揭示了骨软骨谱系细胞的异质性,并概括了小鼠长骨生长过程中的发育轨迹。此外,我们确定了一个新的Cd168+SSPC群体,在胚胎和出生后的长骨中具有高度复制能力和骨软骨形成潜力。此外,Cd168+SSPCs可以在骨折愈合过程中促进新形成的骨骼组织。此外,多色免疫荧光的结果表明,Cd168SSPC位于关节软骨的浅表区域以及出生后小鼠长骨的生长板中。总之,我们确定了一个新的Cd168+SSPC群体,在小鼠长骨中具有再生潜力,这增加了骨骼中组织特异性干细胞的知识。
    Skeletal stem/progenitor cells (SSPCs) are tissue-specific stem/progenitor cells localized within skeletons and contribute to bone development, homeostasis, and regeneration. However, the heterogeneity of SSPC populations in mouse long bones and their respective regenerative capacity remain to be further clarified. In this study, we perform integrated analysis using single-cell RNA sequencing (scRNA-seq) datasets of mouse hindlimb buds, postnatal long bones, and fractured long bones. Our analyses reveal the heterogeneity of osteochondrogenic lineage cells and recapitulate the developmental trajectories during mouse long bone growth. In addition, we identify a novel Cd168+ SSPC population with highly replicating capacity and osteochondrogenic potential in embryonic and postnatal long bones. Moreover, the Cd168+ SSPCs can contribute to newly formed skeletal tissues during fracture healing. Furthermore, the results of multicolor immunofluorescence show that Cd168+ SSPCs reside in the superficial zone of articular cartilage as well as in growth plates of postnatal mouse long bones. In summary, we identify a novel Cd168+ SSPC population with regenerative potential in mouse long bones, which adds to the knowledge of the tissue-specific stem cells in skeletons.
    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
    骨膜干细胞和祖细胞(PSPC)是骨维持和修复的主要贡献者。破译调节其功能的分子机制对于未来疗法的成功产生和应用至关重要。这里,我们确定Hox转录因子对于骨膜干细胞功能是必需和足够的。Hox基因在骨膜干细胞中转录富集,它们在更多定型祖细胞中的过表达驱动重新编程为幼稚,自我更新的干细胞样状态。至关重要的是,单个Hox家族成员以特定位置的方式表达,只有当Hox基因与PSPC的解剖起源相匹配时,才能观察到它们的干细胞促进活性。证明了胚胎Hox代码在成人干细胞中的作用。最后,我们证明Hoxa10过表达部分恢复了与年龄相关的骨折修复下降.一起,我们的数据强调了Hox基因作为PSPC身份在骨骼稳态和修复中的关键调节因子的重要性.
    Periosteal stem and progenitor cells (PSPCs) are major contributors to bone maintenance and repair. Deciphering the molecular mechanisms that regulate their function is crucial for the successful generation and application of future therapeutics. Here, we pinpoint Hox transcription factors as necessary and sufficient for periosteal stem cell function. Hox genes are transcriptionally enriched in periosteal stem cells and their overexpression in more committed progenitors drives reprogramming to a naïve, self-renewing stem cell-like state. Crucially, individual Hox family members are expressed in a location-specific manner and their stem cell-promoting activity is only observed when the Hox gene is matched to the anatomical origin of the PSPC, demonstrating a role for the embryonic Hox code in adult stem cells. Finally, we demonstrate that Hoxa10 overexpression partially restores the age-related decline in fracture repair. Together, our data highlight the importance of Hox genes as key regulators of PSPC identity in skeletal homeostasis and repair.
    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
    Prx1的表达已被用作标记以定义在骨髓和骨膜中发现的有助于骨再生的骨骼干细胞(SSC)群体。然而,Prx1表达SSC(Prx1-SSC)不限于骨区室,但也位于肌肉内,能够促进异位骨形成。然而,鲜为人知,关于调节肌肉中Prx1-SSC的机制以及它们如何参与骨再生。这项研究比较了骨膜和肌肉来源的Prx1-SSCs的内在和外在因素,并分析了其激活的调节机制。扩散,和骨骼分化。在肌肉或骨膜中发现的Prx1-SSC存在相当大的转录组异质性,但是来自两种组织的体外细胞均显示出三谱系(脂肪,软骨和骨)分化。在稳态下,骨膜来源的Prx1细胞增殖,低水平的BMP2能够促进其分化,而肌肉来源的Prx1细胞是静止的,并且与促进骨膜细胞分化的BMP2水平相当。将Prx1-SCC从肌肉和骨膜移植到分离它们的同一部位,或它们的相对位点表明,移植到骨组织表面的骨膜细胞分化为骨骼和软骨细胞,但移植到肌肉中时无法类似的分化。来自肌肉的Prx1-SSC在任一移植部位均没有分化能力。需要骨折和十倍的BMP2剂量来促进肌肉来源的细胞快速进入细胞周期以及经历骨骼细胞分化。这项研究阐明了Prx1-SSC群体的多样性,表明不同组织部位的细胞本质上是不同的。虽然肌肉组织必须有促进Prx1-SSC保持静止的因素,骨损伤或高水平的BMP2可以激活这些细胞增殖并进行骨骼细胞分化。最后,这些研究提出了肌肉间充质干细胞是骨骼修复和骨骼疾病的潜在靶点的可能性。
    The expression of Prx1 has been used as a marker to define the skeletal stem cells (SSCs) populations found within the bone marrow and periosteum that contribute to bone regeneration. However, Prx1 expressing SSCs (Prx1-SSCs) are not restricted to the bone compartments, but are also located within the muscle and able to contribute to ectopic bone formation. Little is known however, about the mechanism(s) regulating Prx1-SSCs that reside in muscle and how they participate in bone regeneration. This study compared both the intrinsic and extrinsic factors of the periosteum and muscle derived Prx1-SSCs and analyzed their regulatory mechanisms of activation, proliferation, and skeletal differentiation. There was considerable transcriptomic heterogeneity in the Prx1-SSCs found in muscle or the periosteum however in vitro cells from both tissues showed tri-lineage (adipose, cartilage and bone) differentiation. At homeostasis, periosteal-derived Prx1 cells were proliferative and low levels of BMP2 were able to promote their differentiation, while the muscle-derived Prx1 cells were quiescent and refractory to comparable levels of BMP2 that promoted periosteal cell differentiation. The transplantation of Prx1-SCC from muscle and periosteum into either the same site from which they were isolated, or their reciprocal sites showed that periosteal cell transplanted onto the surface of bone tissues differentiated into bone and cartilage cells but was incapable of similar differentiation when transplanted into muscle. Prx1-SSCs from the muscle showed no ability to differentiate at either site of transplantation. Both fracture and ten times the BMP2 dose was needed to promote muscle-derived cells to rapidly enter the cell cycle as well as undergo skeletal cell differentiation. This study elucidates the diversity of the Prx1-SSC population showing that cells within different tissue sites are intrinsically different. While muscle tissue must have factors that promote Prx1-SSC to remain quiescent, either bone injury or high levels of BMP2 can activate these cells to both proliferate and undergo skeletal cell differentiation. Finally, these studies raise the possibility that muscle SSCs are potential target for skeletal repair and bone 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
    骨骼干细胞(SSC)为成年骨骼提供干细胞资源,以维持稳态并支持再生。这取决于SSC成骨谱系定型的精确测定。在这项研究中,使用Cre-loxP记者谱系追踪,我们鉴定并表征了骨再生中的NFATc1+SSC群体。预先存在的NFATc1+SSC参与早期骨痂形成。随后,这些NFATc1+SSC在随后的再生阶段产生骨骼系统后代。NFATc1的Ca2触发的转录活性构成了固有地协调SSC成骨的轨迹的预印迹记忆。在SSC中Ca2+/NFATc1信号传导的抑制直接损害成骨和骨再生。总之,我们的研究结果通过NFATc1+SSCs的调节提供了成人骨再生机制的理解.
    Skeletal stem cells (SSCs) fuel adult bone with stemness resources to maintain homeostasis and support regeneration, which depends on the precise determination of the osteogenic lineage commitment of SSCs. In this study, using Cre-loxP reporter lineage tracking, we identified and characterized a population of NFATc1+ SSCs in bone regeneration. Pre-existing NFATc1+ SSCs are involved in early bone callus formation. Subsequently, these NFATc1+ SSCs produce osteolineage descendants in the subsequent stages of regeneration. The Ca2+-triggered transcriptional activity of NFATc1 constitutes the pre-imprinted memory of the trajectory to intrinsically orchestrate osteogenesis of SSCs. Inhibition of Ca2+/NFATc1 signaling in SSCs directly impairs osteogenesis and bone regeneration. In summary, our findings provide a mechanistic understanding of adult bone regeneration through the regulation of NFATc1+ SSCs.
    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
    骨生物学中的一个基本问题涉及骨髓中的骨骼干/祖细胞(SSC)与骨膜对骨修复的贡献。我们发现,可以根据Leprcre和Adiponectin-cre/creER的表达来鉴定成人骨髓中的SSC,而可以根据Gli1creERT2的表达来鉴定成人骨膜中的SSC。在稳态条件下,新骨主要来自骨髓SSC。骨损伤后,两种SSC种群开始增殖,但对骨修复的贡献却大不相同.钻头损伤主要由LepR+/脂联素+骨髓SSC修复。相反,双皮质骨折主要由Gli1+骨膜SSC修复,尽管LepR/脂联素骨髓细胞在骨折部位短暂形成了小梁骨。Gli1骨膜细胞还再生了LepR骨髓基质细胞,在骨折部位表达造血生态位因子。因此,不同的SSC修复了不同的骨损伤,非稳定型骨折后,骨膜细胞再生骨骼和骨髓基质。
    A fundamental question in bone biology concerns the contributions of skeletal stem/progenitor cells (SSCs) in the bone marrow versus the periosteum to bone repair. We found that SSCs in adult bone marrow can be identified based on Leprcre and Adiponectin-cre/creER expression while SSCs in adult periosteum can be identified based on Gli1creERT2 expression. Under steady-state conditions, new bone arose primarily from bone marrow SSCs. After bone injuries, both SSC populations began proliferating but made very different contributions to bone repair. Drill injuries were primarily repaired by LepR+/Adiponectin+ bone marrow SSCs. Conversely, bicortical fractures were primarily repaired by Gli1+ periosteal SSCs, though LepR+/Adiponectin+ bone marrow cells transiently formed trabecular bone at the fracture site. Gli1+ periosteal cells also regenerated LepR+ bone marrow stromal cells that expressed hematopoietic niche factors at fracture sites. Different bone injuries are thus repaired by different SSCs, with periosteal cells regenerating bone and marrow stroma after non-stabilized fractures.
    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