osteoprogenitor

骨祖细胞
  • 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
    骨发育和重塑受磷酸肌醇-3-激酶(Pi3k)信号通路控制。我们研究了磷酸酶和张力蛋白同源物(Pten)下调的影响,Pi3k信号的负调节剂,在前成骨细胞Pten缺乏的小鼠模型中。我们旨在确定与骨转换调节有关的机制,并与骨骼疾病有关。Femora,胫骨,将从表达Osterix/Sp7的骨祖细胞中条件缺失Pten(PtencKO)的小鼠中分离的骨髓基质细胞(BMSC)与Cre阴性对照进行比较。骨表型通过μCT测量进行,骨组织形态计量学,定量骨转换标志物CTX和1型前胶原前肽(P1NP),和三点弯曲试验。通过计数细胞核和Ki-67染色的细胞来测量BMSCs的增殖。体外,通过ALP染色确定成骨分化能力,以及通过检测成骨标志物的基因表达。来自PtencKO小鼠的BMSCs在功能上不同于对照BMSCs。在PtencKO小鼠来源的BMSCs中,成骨标志物增加,而Pten蛋白表达较低,Akt磷酸化增加。我们在PtencKO骨骼中检测到更高的小梁骨体积和皮质骨形态改变,骨骼和组织矿物质密度逐渐降低。PtencKO骨每个骨小梁表面显示出较少的破骨细胞和较多的成骨细胞(P=.00095),并且骨小梁形成率较高。生物力学分析显示,PtencKO股骨的骨强度(男性为P=.00012)和弹性显着提高。在细胞层面,与对照组相比,PtencKOBMSCs的增殖和成骨分化能力均显着增加。我们的发现表明,骨祖细胞中的Pten敲除通过增加小梁骨量来增加骨稳定性和弹性,并导致BMSCs的增殖和成骨分化增加。
    Bone development and remodeling are controlled by the phosphoinositide-3-kinase (Pi3k) signaling pathway. We investigated the effects of downregulation of phosphatase and tensin homolog (Pten), a negative regulator of Pi3k signaling, in a mouse model of Pten deficiency in preosteoblasts. We aimed to identify mechanisms that are involved in the regulation of bone turnover and are linked to bone disorders. Femora, tibiae, and bone marrow stromal cells (BMSCs) isolated from mice with a conditional deletion of Pten (Pten cKO) in Osterix/Sp7-expressing osteoprogenitor cells were compared to Cre-negative controls. Bone phenotyping was performed by μCT measurements, bone histomorphometry, quantification of bone turnover markers CTX and procollagen type 1 N propeptide (P1NP), and three-point bending test. Proliferation of BMSCs was measured by counting nuclei and Ki-67-stained cells. In vitro, osteogenic differentiation capacity was determined by ALP staining, as well as by detecting gene expression of osteogenic markers. BMSCs from Pten cKO mice were functionally different from control BMSCs. Osteogenic markers were increased in BMSCs derived from Pten cKO mice, while Pten protein expression was lower and Akt phosphorylation was increased. We detected a higher trabecular bone volume and an altered cortical bone morphology in Pten cKO bones with a progressive decrease in bone and tissue mineral density. Pten cKO bones displayed fewer osteoclasts and more osteoblasts (P = .00095) per trabecular bone surface and a higher trabecular bone formation rate. Biomechanical analysis revealed a significantly higher bone strength (P = .00012 for males) and elasticity of Pten cKO femora. On the cellular level, both proliferation and osteogenic differentiation capacity of Pten cKO BMSCs were significantly increased compared to controls. Our findings suggest that Pten knockout in osteoprogenitor cells increases bone stability and elasticity by increasing trabecular bone mass and leads to increased proliferation and osteogenic differentiation of BMSCs.
    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
    远程肿瘤破坏骨髓(BM)生态系统(BME),引起BM来源的免疫抑制细胞的过量生产。然而,潜在的机制仍然知之甚少。在这里,我们表征了乳腺癌和肺癌诱导的BME在肿瘤切除前后的变化。远程肿瘤逐渐导致骨原(OP)扩张,造血干细胞脱位,和CD41-粒细胞-单核细胞祖细胞(GMP)聚集。肿瘤夹带的BME的特征在于CD41-GMPs和OP之间的共定位。OP消融消除了这种作用,并减少了异常的骨髓过度生产。机械上,由肿瘤衍生的小细胞外囊泡携带的HTRA1上调OPs中的MMP-13,这反过来又诱导了造血程序的改变。重要的是,这些作用在术后持续存在,并继续损害抗肿瘤免疫力.MMP-13的条件性敲除或抑制加速免疫恢复并恢复免疫疗法的功效。因此,肿瘤诱导的全身效应是由OP-GMP串扰引发的,其持续时间超过肿瘤负荷,并且需要额外的治疗来逆转这些效果以获得最佳的治疗效果。
    Remote tumors disrupt the bone marrow (BM) ecosystem (BME), eliciting the overproduction of BM-derived immunosuppressive cells. However, the underlying mechanisms remain poorly understood. Herein, we characterized breast and lung cancer-induced BME shifts pre- and post-tumor removal. Remote tumors progressively lead to osteoprogenitor (OP) expansion, hematopoietic stem cell dislocation, and CD41- granulocyte-monocyte progenitor (GMP) aggregation. The tumor-entrained BME is characterized by co-localization between CD41- GMPs and OPs. OP ablation abolishes this effect and diminishes abnormal myeloid overproduction. Mechanistically, HTRA1 carried by tumor-derived small extracellular vesicles upregulates MMP-13 in OPs, which in turn induces the alterations in the hematopoietic program. Importantly, these effects persist post-surgery and continue to impair anti-tumor immunity. Conditional knockout or inhibition of MMP-13 accelerates immune reinstatement and restores the efficacies of immunotherapies. Therefore, tumor-induced systemic effects are initiated by OP-GMP crosstalk that outlasts tumor burden, and additional treatment is required to reverse these effects for optimal therapeutic efficacy.
    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
    RGS5(G蛋白信号调节因子5)是异源三聚体G蛋白α亚基的GTP酶激活剂,显示为周细胞的标记。骨髓基质细胞群(BMSCs)是非常异质的。间充质祖细胞的种群,支持造血的细胞和调节骨重塑的基质细胞最近被发现。骨膜和骨髓间充质干细胞(MSCs)参与骨折愈合,但是很难区分愈伤组织中细胞的数量。考虑到血管周围细胞发挥骨祖细胞的潜能,我们产生了一个RGS5转基因小鼠模型(Rgs5-CreER),当与Ai9报告动物(Rgs5/番茄)杂交时,适用于生长和损伤后的谱系追踪。流式细胞术分析和组织学证实CD31+内皮内存在Rgs5/番茄+细胞,CD45+造血细胞和CD31-CD45-间充质/血管周围细胞。他莫昔芬追踪显示在位于矿化基质和脉管系统之间的小梁内表达osterix的Rgs5/Tomato细胞扩增。长期追踪显示Rgs5/番茄+细胞的比例有助于表达骨钙蛋白的成熟成骨细胞。股骨骨折后,在BM腔内新形成的骨骼周围观察到Rgs5/番茄细胞,并表达osterix和骨钙蛋白,而骨膜内的贡献较低,仅限于成纤维细胞骨痂,很少有阳性软骨细胞。此外,BM损伤模型证实,RGS5-Cre标记的BMSCs群体在损伤过程中扩大并参与成骨。在稳态条件下,小梁区域内谱系追踪的RGS5细胞证明了在损伤模型中主要在BM壁龛内促进新骨形成的骨祖细胞能力。
    Regulator of G protein signaling 5 (RGS5) is a GTPase activator for heterotrimeric G-protein α-subunits, shown to be a marker of pericytes. Bone marrow stromal cell population (BMSCs) is heterogeneous. Populations of mesenchymal progenitors, cells supportive of hematopoiesis, and stromal cells regulating bone remodeling have been recently identified. Periosteal and bone marrow mesenchymal stem cells (MSCs) are participating in fracture healing, but it is difficult to distinguish the source of cells within the callus. Considering that perivascular cells exert osteoprogenitor potential, we generated an RGS5 transgenic mouse model (Rgs5-CreER) which when crossed with Ai9 reporter animals (Rgs5/Tomato), is suitable for lineage tracing during growth and post-injury. Flow cytometry analysis and histology confirmed the presence of Rgs5/Tomato+ cells within CD31+ endothelial, CD45+ hematopoietic, and CD31-CD45- mesenchymal/perivascular cells. A tamoxifen chase showed expansion of Rgs5/Tomato+ cells expressing osterix within the trabeculae positioned between mineralized matrix and vasculature. Long-term chase showed proportion of Rgs5/Tomato+ cells contributes to mature osteoblasts expressing osteocalcin. Following femoral fracture, Rgs5/Tomato+ cells are observed around newly formed bone within the BM cavity and expressed osterix and osteocalcin, while contribution within periosteum was low and limited to fibroblastic callus with very few positive chondrocytes. In addition, BM injury model confirmed that RGS5-Cre labels population of BMSCs expands during injury and participates in osteogenesis. Under homeostatic conditions, lineage-traced RGS5 cells within the trabecular area demonstrate osteoprogenitor capacity that in an injury model contributes to new bone formation primarily within the BM niche.
    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:哺乳动物颅骨由两个起源发育的扁平骨组成,神经嵴和中胚层。来自两种来源的细胞表现出相似的行为,但表达不同的转录组。有趣的是,两个起源共有的基因在发育中是否发挥着相似或不同的作用。在目前的研究中,我们研究了Pdgfra的作用,在神经c和中胚层中都有表达,在颅骨发育过程中的特定谱系。
    结果:我们发现在颅骨祖细胞中,需要Pdgfra来维持神经c细胞的正常增殖和迁移,但仅需要中胚层细胞的增殖。后来在颅骨成骨细胞中,我们发现Pdgfra对于神经c衍生细胞的增殖和分化都是必需的,但不能分化中胚层来源的细胞。我们还研究了Pdgfra与其他参与颅骨成骨细胞的信号传导途径之间的潜在相互作用,但在Pdgfra遗传模型中没有发现Wnt或Hh信号传导活性的显著改变。
    结论:Pdgfra是神经c细胞和中胚层细胞正常颅骨发育所必需的,但是这些谱系对Pdgfra活性的改变表现出不同的反应。本文受版权保护。保留所有权利。
    Mammalian calvarium is composed of flat bones developed from two origins, neural crest, and mesoderm. Cells from both origins exhibit similar behavior but express distinct transcriptomes. It is intriguing to ask whether genes shared by both origins play similar or distinct roles in development. In the present study, we have examined the role of Pdgfra, which is expressed in both neural crest and mesoderm, in specific lineages during calvarial development.
    We found that in calvarial progenitor cells, Pdgfra is needed to maintain normal proliferation and migration of neural crest cells but only proliferation of mesoderm cells. Later in calvarial osteoblasts, we found that Pdgfra is necessary for both proliferation and differentiation of neural crest-derived cells, but not for differentiation of mesoderm-derived cells. We also examined the potential interaction between Pdgfra and other signaling pathway involved in calvarial osteoblasts but did not identify significant alteration of Wnt or Hh signaling activity in Pdgfra genetic models.
    Pdgfra is required for normal calvarial development in both neural crest cells and mesoderm cells, but these lineages exhibit distinct responses to alteration of Pdgfra activity.
    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
    亲环蛋白D(CypD)促进线粒体通透性转换孔(MPTP)的开放,在细胞生理学和病理学中起关键作用。是的,因此,有利于细胞紧密调节CypD和MPTP,但对这种调节知之甚少。我们之前报道过CypD在各种组织的分化过程中下调而MPTP失活。在这里,我们识别BMP/Smad信号,差异化的主要驱动力,作为CypD基因的转录调节因子,ppif.使用间充质细胞系的成骨诱导作为BMP/Smad激活依赖性分化模型,我们表明CypD在这个过程中实际上被转录抑制。这种CypD下调的重要性由CypD通过功能获得对体外和小鼠模型中的骨生成的负面影响证明。总之,我们表征了BMP/Smad信号作为CypD表达的调节因子,并阐明了CypD下调在细胞分化过程中的作用.
    Cyclophilin D (CypD) promotes opening of the mitochondrial permeability transition pore (MPTP) which plays a key role in both cell physiology and pathology. It is, therefore, beneficial for cells to tightly regulate CypD and MPTP but little is known about such regulation. We have reported before that CypD is downregulated and MPTP deactivated during differentiation in various tissues. Herein, we identify BMP/Smad signaling, a major driver of differentiation, as a transcriptional regulator of the CypD gene, Ppif. Using osteogenic induction of mesenchymal lineage cells as a BMP/Smad activation-dependent differentiation model, we show that CypD is in fact transcriptionally repressed during this process. The importance of such CypD downregulation is evidenced by the negative effect of CypD \'rescue\' via gain-of-function on osteogenesis both in vitro and in a mouse model. In sum, we characterized BMP/Smad signaling as a regulator of CypD expression and elucidated the role of CypD downregulation during cell differentiation.
    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
    骨质疏松症的主要原因是耦合骨形成受损。机械上,破骨细胞来源的生长因子和骨来源的生长因子以前都有牵连.这里,我们假设破骨细胞骨吸收过程中骨钙的释放对于偶联骨形成至关重要。破骨细胞吸收使间质液钙从正常的1.8mM局部增加到5mM。MC3T3-E1骨祖细胞,在3.6mM钙培养基中培养,证明钙信号刺激成骨细胞增殖,分化,和移民。钙通道敲低研究涉及钙通道,Cav1.2,商店操作的钙入口(SOCE),和钙敏感受体(CaSR)调节骨细胞合成代谢活性。在3.6mM钙培养基中培养的MC3T3-E1细胞表达Wnt信号和生长因子血小板衍生生长因子(PDGF)的基因表达增加,血管内皮生长因子(VEGF),和骨形态发生蛋白2(BMP2)。我们的骨形成耦合模型,核因子-κΒ配体(RANKL)处理的小鼠颅骨的受体激活剂,通过显示osterix和骨钙蛋白的基因表达增加,证实了钙信号在偶联骨形成中的作用。严重的,双重免疫细胞化学显示,RANKL处理增加了osterix阳性细胞,并增加了每个osterix阳性细胞的Cav1.2和CaSR蛋白表达的荧光强度。上述数据证实破骨细胞释放的钙有助于调节偶联的骨形成。在基础钙培养基中培养的骨祖细胞中Cav1.2的CRISPR/Cas-9敲除导致下游成骨基因表达下降>80%,强调钙信号的影响。因此,钙信号是偶联骨形成的主要调节因子。
    A major cause of osteoporosis is impaired coupled bone formation. Mechanistically, both osteoclast-derived and bone-derived growth factors have been previously implicated. Here, we hypothesize that the release of bone calcium during osteoclastic bone resorption is essential for coupled bone formation. Osteoclastic resorption increases interstitial fluid calcium locally from the normal 1.8 mM up to 5 mM. MC3T3-E1 osteoprogenitor cells, cultured in a 3.6 mM calcium medium, demonstrated that calcium signaling stimulated osteogenic cell proliferation, differentiation, and migration. Calcium channel knockdown studies implicated calcium channels, Cav1.2, store-operated calcium entry (SOCE), and calcium-sensing receptor (CaSR) in regulating bone cell anabolic activities. MC3T3-E1 cells cultured in a 3.6 mM calcium medium expressed increased gene expression of Wnt signaling and growth factors platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), and bone morphogenic protein-2 (BMP 2). Our coupling model of bone formation, the receptor activator of nuclear factor-κΒ ligand (RANKL)-treated mouse calvaria, confirmed the role of calcium signaling in coupled bone formation by exhibiting increased gene expression for osterix and osteocalcin. Critically, dual immunocytochemistry showed that RANKL treatment increased osterix-positive cells and increased fluorescence intensity of Cav1.2 and CaSR protein expression per osterix-positive cell. The above data established that calcium released by osteoclasts contributed to the regulation of coupled bone formation. CRISPR/Cas-9 knockout of Cav1.2 in osteoprogenitor cells cultured in basal calcium medium caused a >80% decrease in the expression of downstream osteogenic genes, emphasizing the large magnitude of the effect of calcium signaling. Thus, calcium signaling is a major regulator of coupled bone formation.
    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:我们最近的研究表明,粘着斑蛋白Kindlin-2在间充质干细胞中发挥着至关重要的功能,成熟的成骨细胞和骨细胞控制小鼠早期骨骼发育和骨稳态。然而,Kindlin-2是否在骨祖细胞中发挥作用尚不清楚.
    方法:在表达Osterix(Osx)的细胞中缺乏Kindlin-2表达的小鼠(即,骨祖细胞)产生。微计算机断层扫描(μCT)分析,组织学,进行骨组织形态计量学和免疫组织化学,以确定Kindlin-2缺失对骨骼发育、骨量累积和体内平衡的影响.分离来自突变小鼠(Kindlin-2fl/fl;OsxCre)和对照同窝的骨髓基质细胞(BMSC),并确定其成骨细胞分化能力。
    结果:Kindlin-2在软骨内骨化过程中骨祖细胞中高表达。骨祖细胞中Kindlin-2表达的缺失损害了膜内和软骨内骨化。突变小鼠表现出多种严重的骨骼异常,包括未矿化的fontanel,肢体缩短和生长迟缓。骨祖细胞中Kindlin-2的缺失会损害生长板的发育,并大大延迟了长骨中次要骨化中心的形成。此外,成年突变小鼠表现出严重的低周转性骨质减少,骨形成急剧下降,超过了骨吸收。从突变小鼠分离的原代BMSCs表现出降低的成骨细胞分化能力。
    结论:我们的研究表明,Kinlind-2在骨祖细胞中的表达在调节小鼠骨骼生成、骨量积累和体内平衡方面具有重要作用。
    这项研究揭示了Kindlin-2通过其在骨祖细胞中的表达来控制软骨形成和骨量。我们可以为骨骼疾病的治疗定义一个新的治疗靶点,如软骨发育不良和骨质疏松症。
    BACKGROUND: Our recent studies demonstrate that the focal adhesion protein Kindlin-2 exerts crucial functions in the mesenchymal stem cells, mature osteoblasts and osteocytes in control of early skeletal development and bone homeostasis in mice. However, whether Kindlin-2 plays a role in osteoprogenitors remains unclear.
    METHODS: Mice lacking Kindlin-2 expression in osterix (Osx)-expressing cells (i.e., osteoprogenitors) were generated. Micro-computerized tomography (μCT) analyses, histology, bone histomorphometry and immunohistochemistry were performed to determine the effects of Kindlin-2 deletion on skeletal development and bone mass accrual and homeostasis. Bone marrow stromal cells (BMSCs) from mutant mice (Kindlin-2 fl/fl ; Osx Cre ) and control littermates were isolated and determined for their osteoblastic differentiation capacity.
    RESULTS: Kindlin-2 was highly expressed in osteoprogenitors during endochondral ossification. Deleting Kindlin-2 expression in osteoprogenitors impaired both intramembranous and endochondral ossifications. Mutant mice displayed multiple severe skeletal abnormalities, including unmineralized fontanel, limb shortening and growth retardation. Deletion of Kindlin-2 in osteoprogenitors impaired the growth plate development and largely delayed formation of the secondary ossification center in the long bones. Furthermore, adult mutant mice displayed a severe low-turnover osteopenia with a dramatic decrease in bone formation which exceeded that in bone resorption. Primary BMSCs isolated from mutant mice exhibited decreased osteoblastic differentiation capacity.
    CONCLUSIONS: Our study demonstrates an essential role of Kinlind-2 expression in osteoprogenitors in regulating skeletogenesis and bone mass accrual and homeostasis in mice.
    UNASSIGNED: This study reveals that Kindlin-2 through its expression in osteoprogenitor cells controls chondrogenesis and bone mass. We may define a novel therapeutic target for treatment of skeletal diseases, such as chondrodysplasia and osteoporosis.
    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
    Transforming growth factor beta (TGFβ) signaling plays an important role during osteogenesis. However, most research in this area focuses on cortical and trabecular bone, whereas alveolar bone is largely overlooked. To address the role of TGFβR2 (the key receptor for TGFβ signaling) during postnatal alveolar bone development, we conditionally deleted Tgfβr2 in early mesenchymal progenitors by crossing Gli1-Cre ERT2; Tgfβr2 flox/flox ; R26R tdTomato mice (named early cKO) or in osteoblasts by crossing 3.2kb Col1-Cre ERT2 ; Tgfβr2 flox/flox ; R26R tdTomato mice (named late cKO). Both cKO lines were induced at postnatal day 5 (P5) and mice were harvested at P28. Compared to the control littermates, early cKO mice exhibited significant reduction in alveolar bone mass and bone mineral density, with drastic defects in the periodontal ligament (PDL); conversely, the late cKO mice displayed very minor changes in alveolar bone. Mechanism studies showed a significant reduction in PCNA+ PDL cell numbers and OSX+ alveolar bone cell numbers, as well as disorganized PDL fibers with a great reduction in periostin (the most abundant extracellular matrix protein) on both mRNA and protein levels. We also showed a drastic reduction in β-catenin in the early cKO PDL and a great increase in SOST (a potent inhibitor of Wnt signaling). Based on these findings, we conclude that TGFβ signaling plays critical roles during early alveolar bone formation via the promotion of PDL mesenchymal progenitor proliferation and differentiation mechanisms.
    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
    Hematopoietic stem and progenitor cell (HSPC) engraftment after transplantation during anticancer treatment depends on support from the recipient bone marrow (BM) microenvironment. Here, by studying physiological homing of fetal HSPCs, we show the critical requirement of balanced local crosstalk within the skeletal niche for successful HSPC settlement in BM. Transgene-induced overproduction of vascular endothelial growth factor (VEGF) by osteoprogenitor cells elicits stromal and endothelial hyperactivation, profoundly impacting the stromal-vessel interface and vascular architecture. Concomitantly, HSPC homing and survival are drastically impaired. Transcriptome profiling, flow cytometry, and high-resolution imaging indicate alterations in perivascular and endothelial cell characteristics, vascular function and cellular metabolism, associated with increased oxidative stress within the VEGF-enriched BM environment. Thus, developmental HSPC homing to bone is controlled by local stromal-vascular integrity and the oxidative-metabolic status of the recipient milieu. Interestingly, irradiation of adult mice also induces stromal VEGF expression and similar osteo-angiogenic niche changes, underscoring that our findings may contribute targets for improving stem cell therapies.
    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