Skeletal stem cells

骨骼干细胞
  • 文章类型: Journal Article
    半月板撕裂后的自我修复能力较差,导致创伤后骨关节炎的发展。促进半月板损伤的修复仍然是一个巨大的挑战,尤其是在无血管区域。
    骨骼干细胞(SSC)衍生的外泌体(SSC-Exos)的局部递送将促进半月板愈合并防止继发性骨关节炎进展。
    对照实验室研究。
    从骨髓中分离SSC,并通过超速离心提取外泌体。通过体外细胞培养验证与外泌体孵育后的细胞迁移能力。在40只雄性Sprague-Dawley大鼠和20只雄性新西兰白兔的无血管区域进行了全层纵向内侧半月板撕裂,随机分为2组:用磷酸盐缓冲盐水(GCON)治疗的组和用外泌体(GExosome)治疗的组。这些治疗对半月板愈合和继发性骨关节炎的影响通过肉眼检查进行评估,生物力学测试,和组织学评估。进行体外细胞培养物的RNA测序以探索潜在的机制。
    外泌体被成功提取和鉴定。这些外泌体显著促进了体外细胞迁移能力(P<0.01)。GExosome表现出更大的细胞增殖和组织再生,分泌2型胶原,术后8周半月板修复评分明显高于GCON(P<0.05)。与GCON的半月板和关节软骨的退行性变化相反,GExosome中的半月板组织在半月板修复后8周时表现出正常形态的恢复,具有光滑有光泽的白色表面和更好的机械强度。GCON的变性评分和滑膜炎评分均明显高于GExosome(P<0.05)。与GCON相比,与细胞迁移相关的关键基因的表达,比如趋化因子家族,通过外泌体注射增强,导致细胞外基质表达上调,同时下调炎症相关基因如CD68和基质金属蛋白酶家族的表达。
    SSC-Exos的给药有效促进无血管区域的半月板愈合并改善继发性骨关节炎。这种作用可能归因于炎症调节,促进细胞迁移,和分泌细胞外基质成分。
    注射SSC-Exos代表了促进无血管区域半月板愈合的有希望的治疗选择。
    UNASSIGNED: The self-repair ability after meniscal tears is poor, leading to the development of posttraumatic osteoarthritis. Promoting the repair of meniscal injuries remains a great challenge, especially in the avascular region.
    UNASSIGNED: Local delivery of skeletal stem cell (SSC)-derived exosomes (SSC-Exos) would promote meniscal healing and prevent secondary osteoarthritis progression.
    UNASSIGNED: Controlled laboratory study.
    UNASSIGNED: SSCs were isolated from bone marrow and exosomes were extracted via ultracentrifugation. The cell migration capabilities after incubation with exosomes were validated through in vitro cell culture. Full-thickness longitudinal medial meniscal tears were performed in the avascular region of 40 male Sprague-Dawley rats and 20 male New Zealand White rabbits, which were randomly divided into 2 groups: group treated with phosphate-buffered saline (GCON) and group treated with exosomes (GExosome). The effects of these treatments on meniscal healing and secondary osteoarthritis were evaluated by gross inspection, biomechanical testing, and histological assessment. RNA sequencing of in vitro cell cultures was performed to explore the underlying mechanisms.
    UNASSIGNED: Exosomes were successfully extracted and identified. These exosomes significantly promoted cell migration capabilities in vitro (P < .01). The GExosome exhibited greater cell proliferation and tissue regeneration with type 2 collagen secretion, and a significantly higher meniscal repair score than that of the GCON at 8 weeks postoperatively (P < .05). In contrast to the degenerative changes in both the meniscus and articular cartilage of the GCON, meniscal tissue in the GExosome exhibited restoration of normal morphology with a smooth and glossy white surface and better mechanical strength at 8 weeks after meniscal repair. Both degeneration scores and synovitis scores were significantly higher in the GCON than in the GExosome (P < .05). Compared with the GCON, the expression of key genes related to cell migration, such as the chemokine family, was enhanced by exosome injection, leading to an upregulation of extracellular matrix expression while downregulating the expression of inflammation-related genes such as CD68 and the matrix metalloproteinase family.
    UNASSIGNED: The administration of SSC-Exos effectively promoted meniscal healing in the avascular region and ameliorated secondary osteoarthritis. The effect might be attributed to inflammation modulation, promotion of cell migration, and secretion of extracellular matrix components.
    UNASSIGNED: Injection of SSC-Exos represents a promising therapeutic option for promoting meniscal healing in the avascular region.
    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
    人类衰老与骨质流失有关,导致骨脆性和骨折风险增加。这主要是由于骨形成成骨细胞的功能与年龄相关的下降以及骨微环境中细胞衰老的加速。这里,我们详细讨论了与年龄相关的骨形成缺陷是由骨骼干细胞衰老引起的假设,因为它们是骨形成成骨细胞的主要来源,并影响骨微环境的组成。此外,这篇综述讨论了靶向细胞衰老作为治疗年龄相关性骨丢失的新兴方法的潜在策略.
    Human aging is linked to bone loss, resulting in bone fragility and an increased risk of fractures. This is primarily due to an age-related decline in the function of bone-forming osteoblastic cells and accelerated cellular senescence within the bone microenvironment. Here, we provide a detailed discussion of the hypothesis that age-related defective bone formation is caused by senescence of skeletal stem cells, as they are the main source of bone forming osteoblastic cells and influence the composition of bone microenvironment. Furthermore, this review discusses potential strategies to target cellular senescence as an emerging approach to treat age-related bone loss.
    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
    骨髓间充质干细胞(BMSCs)在增强肌腱-骨(T-B)愈合的应用中具有巨大的潜力。最近,据报道,骨骼干细胞(SSC)可以诱导骨骼和软骨再生。因此,SSC代表了基于细胞的疗法以改善T-B愈合的有希望的选择。在这项研究中,我们旨在比较MSCs和BMSCs对腱-骨愈合的治疗潜力.
    通过流式细胞术分离SSC和BMSCs,CCK-8法检测其增殖能力。成骨,软骨形成,通过定量实时聚合酶链反应(qRT-PCR)检测细胞中脂肪基因的表达。C57BL/6小鼠行单侧冈上肌腱脱离修复,然后将小鼠随机分为4组:对照组(未经任何处理的腱-骨界面),水凝胶组(向肌腱-骨界面施用空白水凝胶),水凝胶+BMSCs组(向肌腱-骨界面施用具有BMSCs的水凝胶),和水凝胶+SSC组(向肌腱-骨界面施用具有SSC的水凝胶)。组织学染色,微型计算机断层扫描(Micro-CT)扫描,生物力学测试,在手术后4周和8周进行qRT-PCR以测定T-B愈合。
    SSC显示出更多的细胞比例,表现出更强的乘法能力,与BMSCs相比,表达更高的成骨和成软骨标志物和更低的成脂标志物。体内试验,SSC组出现较好的成熟界面,其特点是软骨细胞较丰富,蛋白聚糖沉积较多,以及与其他组相比,在愈合部位新形成的骨更多,机械性能增加。qRT-PCR分析显示,SSC组中的愈合界面比其他组的界面表达更多的成骨和软骨形成必需的转录因子。
    总的来说,结果表明,在腱-骨愈合方面,SSCs优于BMSCs。
    这项研究提供了有价值的见解,即与BMSCs相比,SSC可能是增强T-B愈合的更有效的细胞疗法。
    UNASSIGNED: Bone marrow mesenchymal stem cells (BMSCs) have immense potential in applications for the enhancement of tendon-bone (T-B) healing. Recently, it has been well-reported that skeletal stem cells (SSCs) could induce bone and cartilage regeneration. Therefore, SSCs represent a promising choice for cell-based therapies to improve T-B healing. In this study, we aimed to compare the therapeutic potential of SSCs and BMSCs for tendon-bone healing.
    UNASSIGNED: SSCs and BMSCs were isolated by flow cytometry, and their proliferation ability was measured by CCK-8 assay. The osteogenic, chondrogenic, and adipogenic gene expression in cells was detected by quantitative real-time polymerase chain reaction (qRT-PCR). C57BL/6 mice underwent unilateral supraspinatus tendon detachment and repair, and the mice were then randomly allocated to 4 groups: control group (tendon-bone interface without any treatment), hydrogel group (administration of blank hydrogel into the tendon-bone interface), hydrogel + BMSCs group (administration of hydrogel with BMSCs into the tendon-bone interface), and hydrogel + SSCs group (administration of hydrogel with SSCs into the tendon-bone interface). Histological staining, Micro-computed tomography (Micro-CT) scanning, biomechanical testing, and qRT-PCR were performed to assay T-B healing at 4 and 8 weeks after surgery.
    UNASSIGNED: SSCs showed more cell proportion, exhibited stronger multiplication capacity, and expressed higher osteogenic and chondrogenic markers and lower adipogenic markers than BMSCs. In vivo assay, the SSCs group showed a better-maturated interface which was characterized by richer chondrocytes and more proteoglycan deposition, as well as more newly formed bone at the healing site and increased mechanical properties when compared to other there groups. qRT-PCR analysis revealed that the healing interface in the SSCs group expressed more transcription factors essential for osteogenesis and chondrogenesis than the interfaces in the other groups.
    UNASSIGNED: Overall, the results demonstrated the superior therapeutic potential of SSCs over BMSCs in tendon-bone healing.
    UNASSIGNED: This current study provides valuable insights that SSCs may be a more effective cell therapy for enhancing T-B healing compared to 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
    背景:骨骼干细胞(SSC)是骨骼发育所必需的,稳态,和修复。它们在再生医学方法中广泛应用的观点支持了这一领域的研究,尽管到目前为止诊所的结果还没有达到预期,可能也是由于内在的部分知识,潜在可行的SSC调节因素。其中,多效性细胞因子RANKL,在骨生物学中也有重要作用,是值得深入研究的候选人。
    方法:为了剖析RANKL细胞因子在SSC生物学中的作用,我们通过细胞荧光分选和分析来自不同骨骼区室的SSC群体,在缺乏Rankl(Rankl-/-)的小鼠中进行了SSC和下游祖细胞(SSPC)的离体表征。基因表达分析,和体外成骨分化。此外,我们评估了抗RANKL阻断抗体Denosumab(已批准用于病理性骨丢失患者的治疗)药物治疗对人健康受试者骨髓来源基质细胞(hBMSCs)成骨潜能的影响.
    结果:我们发现,不管骨化类型的骨,与野生型相比,Rankl-/-小鼠中的骨软骨SSC具有更高的频率和沿着骨软骨谱系的分化受损。Rankl-/-小鼠也具有来自血管周围SSC的定型骨软骨形成和脂肪形成祖细胞的频率增加。这些变化不是由于缺乏破骨细胞吸收(明确的骨硬化)引起的密度增加的特殊骨表型;它们没有在另一个石骨鼠模型中发现,即,oc/oc鼠标,因此不是由于石骨症本身。此外,Rankl-/-SSC和原代成骨细胞显示出降低的矿化能力。值得注意的是,与对照培养物相比,用Denosumab体外处理的hBMSCs具有降低的成骨能力。
    结论:我们首次提供了来自严重隐性石斑病小鼠模型的SSPCs特征。我们证明了鼠SSC中的Rankl遗传缺陷和hBMSCs中的功能阻断降低了其成骨潜力。因此,我们认为RANKL是具有翻译相关性的SSC特征的重要调控因子。
    BACKGROUND: Skeletal Stem Cells (SSCs) are required for skeletal development, homeostasis, and repair. The perspective of their wide application in regenerative medicine approaches has supported research in this field, even though so far results in the clinic have not reached expectations, possibly due also to partial knowledge of intrinsic, potentially actionable SSC regulatory factors. Among them, the pleiotropic cytokine RANKL, with essential roles also in bone biology, is a candidate deserving deep investigation.
    METHODS: To dissect the role of the RANKL cytokine in SSC biology, we performed ex vivo characterization of SSCs and downstream progenitors (SSPCs) in mice lacking Rankl (Rankl-/-) by means of cytofluorimetric sorting and analysis of SSC populations from different skeletal compartments, gene expression analysis, and in vitro osteogenic differentiation. In addition, we assessed the effect of the pharmacological treatment with the anti-RANKL blocking antibody Denosumab (approved for therapy in patients with pathological bone loss) on the osteogenic potential of bone marrow-derived stromal cells from human healthy subjects (hBMSCs).
    RESULTS: We found that, regardless of the ossification type of bone, osteochondral SSCs had a higher frequency and impaired differentiation along the osteochondrogenic lineage in Rankl-/- mice as compared to wild-type. Rankl-/- mice also had increased frequency of committed osteochondrogenic and adipogenic progenitor cells deriving from perivascular SSCs. These changes were not due to the peculiar bone phenotype of increased density caused by lack of osteoclast resorption (defined osteopetrosis); indeed, they were not found in another osteopetrotic mouse model, i.e., the oc/oc mouse, and were therefore not due to osteopetrosis per se. In addition, Rankl-/- SSCs and primary osteoblasts showed reduced mineralization capacity. Of note, hBMSCs treated in vitro with Denosumab had reduced osteogenic capacity compared to control cultures.
    CONCLUSIONS: We provide for the first time the characterization of SSPCs from mouse models of severe recessive osteopetrosis. We demonstrate that Rankl genetic deficiency in murine SSCs and functional blockade in hBMSCs reduce their osteogenic potential. Therefore, we propose that RANKL is an important regulatory factor of SSC features with translational relevance.
    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,增长,修复是复杂的过程,涉及各种细胞类型和相互作用,骨骼干细胞和祖细胞发挥着核心作用。最近的研究为介导膜内和软骨内骨发育的骨骼前体群体带来了新的见解。在以后的生活中,许多决定发育的细胞和分子机制在骨折后被重新激活,与强大的创伤诱导的信号线索触发各种出生后骨骼干/祖细胞(SSPC)居住在骨缺损附近。有趣的是,在这种伤害的背景下,目前的证据表明,SSPC和分化的骨骼细胞的命运可以相当灵活和动态,并且可以激活多种细胞来源以作为产生软骨细胞和/或成骨细胞的功能祖细胞。体内谱系追踪的联合实施,基于细胞表面标记的选择,单细胞分子分析,高分辨率原位成像极大地改善了我们对发育和修复性茎/祖细胞亚群的多样性和作用的认识,同时也揭示了它们动态的复杂性,层次结构,和关系。尽管目前还不完全了解,支持谱系灵活性和成骨细胞来源之间可能的可塑性的发现挑战了单一原始的经典教条,自我更新,多能干细胞驱动骨组织形成和再生从一个分层和严格单向分化树的顶点。我们在这里回顾该领域的状况和起源的最新发现,身份,骨骼祖细胞在骨骼发育和生长过程中的命运,讨论成人SSPC人群对骨折修复的贡献,并反思骨骼前体和分化细胞谱系之间的动力学和关系。进一步的研究旨在揭示SSPC的异质性和能力,以及决定其命运和运作的监管线索,将为临床转化为受损的骨折愈合和骨再生医学提供重要的新选择。
    骨骼祖细胞对骨骼发育和生长至关重要,因为它们提供细胞构建块(软骨细胞和成骨细胞),形成骨骼组成的软骨和骨组织。在成人生活中,骨折的发生重新激活了类似的组织形成机制,从创伤开始,引发位于骨缺损附近的各种出生后骨骼干/祖细胞(SSPC)分裂和迁移。这些细胞随后通过分化成成熟的软骨细胞和/或成骨细胞而产生功能性骨折修复细胞。近年来,各种先进的研究方法和新技术的结合使用极大地改善了我们对起源的认识,身份,命运,以及发育和修复性骨骼干细胞和祖细胞亚群的作用。同时,这项研究还揭示了它们动力学的相当复杂,多样性,层次结构,和关系,目前还没有完全理解。在这次审查中,我们讨论了该领域的状况以及骨骼干细胞和祖细胞介导骨骼发育的身份和作用的最新发现,增长,和修复。对这些细胞群的进一步研究,包括确定它们的确切性质,命运,和功能,以及如何收获和调节它们,对于开发治疗不愈合骨折的新疗法至关重要。
    Bone development, growth, and repair are complex processes involving various cell types and interactions, with central roles played by skeletal stem and progenitor cells. Recent research brought new insights into the skeletal precursor populations that mediate intramembranous and endochondral bone development. Later in life, many of the cellular and molecular mechanisms determining development are reactivated upon fracture, with powerful trauma-induced signaling cues triggering a variety of postnatal skeletal stem/progenitor cells (SSPCs) residing near the bone defect. Interestingly, in this injury context, the current evidence suggests that the fates of both SSPCs and differentiated skeletal cells can be considerably flexible and dynamic, and that multiple cell sources can be activated to operate as functional progenitors generating chondrocytes and/or osteoblasts. The combined implementation of in vivo lineage tracing, cell surface marker-based cell selection, single-cell molecular analyses, and high-resolution in situ imaging has strongly improved our insights into the diversity and roles of developmental and reparative stem/progenitor subsets, while also unveiling the complexity of their dynamics, hierarchies, and relationships. Albeit incompletely understood at present, findings supporting lineage flexibility and possibly plasticity among sources of osteogenic cells challenge the classical dogma of a single primitive, self-renewing, multipotent stem cell driving bone tissue formation and regeneration from the apex of a hierarchical and strictly unidirectional differentiation tree. We here review the state of the field and the newest discoveries in the origin, identity, and fates of skeletal progenitor cells during bone development and growth, discuss the contributions of adult SSPC populations to fracture repair, and reflect on the dynamism and relationships among skeletal precursors and differentiated cell lineages. Further research directed at unraveling the heterogeneity and capacities of SSPCs, as well as the regulatory cues determining their fate and functioning, will offer vital new options for clinical translation toward compromised fracture healing and bone regenerative medicine.
    Skeletal progenitor cells are crucial for bone development and growth, as they provide the cellular building blocks (chondrocytes and osteoblasts) that form the cartilage and bone tissues that the skeleton is composed of. In adult life, the occurrence of a bone fracture reactivates similar tissue-forming mechanisms, starting with the trauma triggering various postnatal skeletal stem/progenitor cells (SSPCs) residing near the bone defect to divide and migrate. These cells subsequently generate functional fracture-repairing cells by differentiating into mature chondrocytes and/or osteoblasts. In recent years, the combined use of various advanced research approaches and new techniques has strongly improved our insights into the origin, identity, fates, and roles of developmental and reparative skeletal stem cells and progenitor subsets. Concomitantly, this research also unveiled considerable complexity in their dynamics, diversity, hierarchies, and relationships, which is incompletely understood at present. In this review, we discuss the state of the field and the newest discoveries in the identity and roles of skeletal stem and progenitor cells mediating bone development, growth, and repair. Further research on these cell populations, including determining their exact nature, fate, and functioning, and how they can be harvested and regulated, is critical to develop new treatments for non-healing fractures.
    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
    干细胞保持在静止状态以长期维持和保存效力;该过程需要微调调节机制。在这项研究中,我们确定了沿着骨骼干细胞(SSC)的发育轨迹的表观遗传景观,Ptip(也称为Paxip1,Pax与转录激活域蛋白1相互作用)。我们的结果表明,Ptip是维持SSC的静止和效力所必需的,II型胶原(Col2)+祖细胞中Ptip的缺失导致SSC的异常活化和分化,受损的生长板形态发生,和长骨发育不良。我们还发现,Ptip通过在启动子区域抑制组蛋白H3K27ac,通过下调磷酸甘油酸激酶1(Pgk1)来抑制SSC的糖酵解。值得注意的是,尽管Ptip缺乏,但抑制糖酵解改善了SSC的功能。据我们所知,这是第一个基于Ptip建立表观遗传框架的研究,通过代谢控制保护骨骼干细胞的静止和效力。该框架有望改善基于SSC的骨发育障碍治疗。
    Stem cells remain in a quiescent state for long-term maintenance and preservation of potency; this process requires fine-tuning regulatory mechanisms. In this study, we identified the epigenetic landscape along the developmental trajectory of skeletal stem cells (SSCs) in skeletogenesis governed by a key regulator, Ptip (also known as Paxip1, Pax interaction with transcription-activation domain protein-1). Our results showed that Ptip is required for maintaining the quiescence and potency of SSCs, and loss of Ptip in type II collagen (Col2)+ progenitors causes abnormal activation and differentiation of SSCs, impaired growth plate morphogenesis, and long bone dysplasia. We also found that Ptip suppressed the glycolysis of SSCs through downregulation of phosphoglycerate kinase 1 (Pgk1) by repressing histone H3 lysine 27 acetylation (H3K27ac) at the promoter region. Notably, inhibition of glycolysis improved the function of SSCs despite Ptip deficiency. To the best of our knowledge, this is the first study to establish an epigenetic framework based on Ptip, which safeguards skeletal stem cell quiescence and potency through metabolic control. This framework is expected to improve SSC-based treatments of bone developmental disorders.
    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
    甲状旁腺激素(PTH)在维持钙稳态中起着关键作用,主要通过调节骨重建过程。它对骨骼的影响尤其取决于暴露的持续时间和频率。具体来说,PTH可以启动骨形成和再吸收,结果受PTH给药方式的影响:连续性或间歇性。在连续管理中,PTH倾向于促进骨吸收,可能是通过调节骨细胞内的某些基因。相反,间歇性暴露通常有利于骨形成,可能是通过短暂的基因激活。PTH的作用延伸到骨细胞活性的各个方面。它直接影响骨骼干细胞,成骨细胞谱系,骨细胞,T细胞,在骨骼生成中起关键作用。同时,它间接影响破骨细胞前体细胞和破骨细胞,对T细胞有直接影响,有助于其在骨吸收中的作用。尽管有这些见解,PTH在骨髓小生境中发挥作用的复杂机制尚不完全清楚.本文综述了PTH-分解代谢和合成代谢-对骨细胞的双重作用,强调参与这些过程的细胞和分子途径。这些因素在骨重建中的复杂相互作用强调需要进一步研究以充分理解PTH对骨骼健康的多方面影响。
    Parathyroid hormone (PTH) plays a pivotal role in maintaining calcium homeostasis, largely by modulating bone remodeling processes. Its effects on bone are notably dependent on the duration and frequency of exposure. Specifically, PTH can initiate both bone formation and resorption, with the outcome being influenced by the manner of PTH administration: continuous or intermittent. In continuous administration, PTH tends to promote bone resorption, possibly by regulating certain genes within bone cells. Conversely, intermittent exposure generally favors bone formation, possibly through transient gene activation. PTH\'s role extends to various aspects of bone cell activity. It directly influences skeletal stem cells, osteoblastic lineage cells, osteocytes, and T cells, playing a critical role in bone generation. Simultaneously, it indirectly affects osteoclast precursor cells and osteoclasts, and has a direct impact on T cells, contributing to its role in bone resorption. Despite these insights, the intricate mechanisms through which PTH acts within the bone marrow niche are not entirely understood. This article reviews the dual roles of PTH-catabolic and anabolic-on bone cells, highlighting the cellular and molecular pathways involved in these processes. The complex interplay of these factors in bone remodeling underscores the need for further investigation to fully comprehend PTH\'s multifaceted influence on bone health.
    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
    组织驻留干细胞对于发育和修复至关重要,在骨骼中,此功能由最近鉴定的骨骼干细胞(SSC)实现。然而,最近的工作已经确定SSC不是整体的,长骨头,颅面部位,脊柱由不同的干细胞形成。最近的研究已经利用技术,如荧光激活细胞分选(FACS),谱系追踪和单细胞测序以研究SSC在骨发育中的参与,稳态和疾病。这些研究使研究人员能够绘制SSC在身体不同部位和不同时间点的谱系承诺轨迹。此外,最近的研究揭示了SSCs在生理和病理条件下的特点。这篇综述的重点是讨论SSCs的时空分布,并通过总结最近的发现来增强我们对SSCs的多样性和可塑性的理解。
    Tissue-resident stem cells are essential for development and repair, and in the skeleton, this function is fulfilled by recently identified skeletal stem cells (SSCs). However, recent work has identified that SSCs are not monolithic, with long bones, craniofacial sites, and the spine being formed by distinct stem cells. Recent studies have utilized techniques such as fluorescence-activated cell sorting, lineage tracing, and single-cell sequencing to investigate the involvement of SSCs in bone development, homeostasis, and disease. These investigations have allowed researchers to map the lineage commitment trajectory of SSCs in different parts of the body and at different time points. Furthermore, recent studies have shed light on the characteristics of SSCs in both physiological and pathological conditions. This review focuses on discussing the spatiotemporal distribution of SSCs and enhancing our understanding of the diversity and plasticity of SSCs by summarizing recent discoveries.
    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
    骨组织为我们的身体提供结构支撑,内部骨髓(BM)充当造血器官。在BM组织内,两种类型的干细胞起着至关重要的作用:间充质干细胞(MSCs)(或骨骼干细胞)和造血干细胞(HSCs)。这些干细胞错综复杂地联系在一起,其中BM-MSC产生骨形成成骨细胞,并作为BM微环境中维持HSC的必要组分。尽管20世纪中叶提出了BM-MSC,由于缺乏分析干性的工具,他们的体内鉴定仍然难以捉摸,特别是自我更新和多能。为了应对这一挑战,正在采用基于Cre/loxP的细胞谱系追踪分析。这项技术促进了特定细胞的体内标记,能够追踪他们的血统,确定它们的干系,并更深入地了解负责维持硬组织的干细胞群的体内动力学。这篇综述深入研究了在LepR的影响下使用表达Cre的常用转基因小鼠进行的细胞系追踪研究。Gli1和Axin2基因。这些研究集中在跨越长骨和口腔/颌面硬组织的研究领域,提供对硬组织稳态至关重要的干细胞群体的体内动力学的见解。
    Bone tissue provides structural support for our bodies, with the inner bone marrow (BM) acting as a hematopoietic organ. Within the BM tissue, two types of stem cells play crucial roles: mesenchymal stem cells (MSCs) (or skeletal stem cells) and hematopoietic stem cells (HSCs). These stem cells are intricately connected, where BM-MSCs give rise to bone-forming osteoblasts and serve as essential components in the BM microenvironment for sustaining HSCs. Despite the mid-20th century proposal of BM-MSCs, their in vivo identification remained elusive owing to a lack of tools for analyzing stemness, specifically self-renewal and multipotency. To address this challenge, Cre/loxP-based cell lineage tracing analyses are being employed. This technology facilitated the in vivo labeling of specific cells, enabling the tracking of their lineage, determining their stemness, and providing a deeper understanding of the in vivo dynamics governing stem cell populations responsible for maintaining hard tissues. This review delves into cell lineage tracing studies conducted using commonly employed genetically modified mice expressing Cre under the influence of LepR, Gli1, and Axin2 genes. These studies focus on research fields spanning long bones and oral/maxillofacial hard tissues, offering insights into the in vivo dynamics of stem cell populations crucial for hard tissue homeostasis.
    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
    骨髓肥胖(BMA)是一个快速增长但非常年轻的研究领域,基于其与骨骼和代谢疾病的密切关系,正在受到全世界的关注。还有血液学和癌症.此外,越来越多的年轻科学家和学生目前正在他们的研究项目中积极致力于BMA。这些发展导致了国际骨髓肥胖协会(BMAS)的成立,以在全球推广BMA知识为目标,并培养对研究这一领域感兴趣的新一代研究人员。在BMAS支持的许多倡议中,有BMAS暑期学校,于2021年开幕,现在是第二版。2023年BMAS暑期学校的目标是通过传播BMA的最新进展来教育和培训学生。此外,2023年暑期学校提供了有关如何撰写补助金的建议,处理科学中的负面结果,开始一个实验室,以及通往学术界的替代道路的插图。早期职业研究人员与更多高级科学家之间进行了建设性和互动的讨论,从而激发了该活动。在这份报告中,我们强调关键时刻和从活动中吸取的教训。
    Bone marrow adiposity (BMA) is a rapidly growing yet very young research field that is receiving worldwide attention based on its intimate relationship with skeletal and metabolic diseases, as well as hematology and cancer. Moreover, increasing numbers of young scientists and students are currently and actively working on BMA within their research projects. These developments led to the foundation of the International Bone Marrow Adiposity Society (BMAS), with the goal to promote BMA knowledge worldwide, and to train new generations of researchers interested in studying this field. Among the many initiatives supported by BMAS, there is the BMAS Summer School, inaugurated in 2021 and now at its second edition. The aim of the BMAS Summer School 2023 was to educate and train students by disseminating the latest advancement on BMA. Moreover, Summer School 2023 provided suggestions on how to write grants, deal with negative results in science, and start a laboratory, along with illustrations of alternative paths to academia. The event was animated by constructive and interactive discussions between early-career researchers and more senior scientists. In this report, we highlight key moments and lessons learned from the event.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号