Mesonephros

Mesonephros
  • 文章类型: Journal Article
    世界范围内肾脏疾病的发病率一直在上升。因此,最近的研究集中在斑马鱼上,其快速发育和先天再生能力允许识别影响肾脏过程的因素。在这些鲜有研究的因子中,有细胞外基质(ECM)蛋白,如纤连蛋白(Fn),在各种组织中必不可少,但尚未在肾脏环境中进行评估。我们利用早期nat和han斑马鱼突变胚胎和载体成虫来研究Fn在肾脏发育和再生过程中的作用。基因座natter(nat)编码Fn,基因座han编码Hand2,这导致Fn沉积增加。我们的结果表明,Fn会影响发育过程中的身份维持和形态发生,并影响再生过程中新生成簇形成的条件。组织学分析显示,Fn突变体中的前肾结构被破坏,血细胞积累增加。尽管正常表达的规格标记(pax2,ATPα1a.1),结构异常明显。野生型和突变携带者之间的差异表明单倍体不足的情况。这些发现揭示了ECM在肾脏发育和再生中的新功能,对了解和治疗肾脏疾病有潜在的影响。
    Worldwide incidence of kidney diseases has been rising. Thus, recent research has focused on zebrafish, whose fast development and innate regeneration capacity allow identifying factors influencing renal processes. Among these poorly studied factors are extracellular matrix (ECM) proteins like Fibronectin (Fn) essential in various tissues but not yet evaluated in a renal context. We utilized early nat and han zebrafish mutant embryos and carrier adults to investigate Fn\'s role during kidney development and regeneration. The locus natter (nat) encodes Fn and the locus han encodes Hand2, which results in increased Fn deposition. Our results show that Fn impacts identity maintenance and morphogenesis during development and influences conditions for neonephrogenic cluster formation during regeneration. Histological analysis revealed disrupted pronephric structures and increased blood cell accumulation in Fn mutants. Despite normal expression of specification markers (pax2, ATPα1a.1), structural abnormalities were evident. Differences between wild-type and mutation-carriers suggest a haploinsufficiency scenario. These findings reveal a novel function for ECM in renal development and regeneration, with potential implications for understanding and treating kidney 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
    了解在胚胎发生过程中促进造血干细胞(HSC)规范的调节机制对于体外生成HSC很重要。巨核细胞从卵黄囊出现并产生血小板,参与多种生物过程,比如防止出血。然而,巨核细胞是否调节胚胎主动脉-性腺-中肾(AGM)区域的HSC发育尚不清楚.这里,我们使用血小板因子4(PF4)-Cre;Rosa-tdTomato细胞报告HSC发育小生境中巨核细胞的存在。Further,我们使用PF4-Cre;Rosa-DTA(DTA)耗竭模型来揭示巨核细胞控制小鼠胚胎中的HSC规格。巨核细胞缺乏阻断前HSC的生成和成熟并改变AGM处的HSC活性。此外,巨核细胞在OP9-DL1共培养系统中促进内皮到造血的转变。单细胞RNA测序将细胞表面标记CD226阳性的巨核细胞鉴定为通过分泌TNFSF14促进内皮细胞血源性命运的最高潜力亚群。在一条线上,TNFSF14治疗在巨核细胞耗尽的共培养物中拯救造血细胞功能。一起来看,巨核细胞促进前造血干细胞的产生和成熟,在胚胎造血过程中充当关键的微环境控制因子。
    Understanding the regulatory mechanisms facilitating hematopoietic stem cell (HSC) specification during embryogenesis is important for the generation of HSCs in vitro. Megakaryocyte emerged from the yolk sac and produce platelets, which are involved in multiple biological processes, such as preventing hemorrhage. However, whether megakaryocytes regulate HSC development in the embryonic aorta-gonad-mesonephros (AGM) region is unclear. Here, we use platelet factor 4 (PF4)-Cre;Rosa-tdTomato+ cells to report presence of megakaryocytes in the HSC developmental niche. Further, we use the PF4-Cre;Rosa-DTA (DTA) depletion model to reveal that megakaryocytes control HSC specification in the mouse embryos. Megakaryocyte deficiency blocks the generation and maturation of pre-HSCs and alters HSC activity at the AGM. Furthermore, megakaryocytes promote endothelial-to-hematopoietic transition in a OP9-DL1 coculture system. Single-cell RNA-sequencing identifies megakaryocytes positive for the cell surface marker CD226 as the subpopulation with highest potential in promoting the hemogenic fate of endothelial cells by secreting TNFSF14. In line, TNFSF14 treatment rescues hematopoietic cell function in megakaryocyte-depleted cocultures. Taken together, megakaryocytes promote production and maturation of pre-HSCs, acting as a critical microenvironmental control factor during embryonic hematopoiesis.
    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
    了解调节胚胎造血干细胞(HSC)发育的机制将有助于其再生。主动脉-性腺-中肾区域是从血源性内皮细胞(HEC)产生HSC的位点。虽然几个不同的监管机构参与了这一过程,目前尚不清楚巨自噬(自噬)是否在肝脏前期的造血中起作用。这里,根据LC3-RFP-EGFP小鼠模型,我们发现造血前体中存在不同的自噬状态,并与造血潜能相关。内皮细胞中特异性自噬相关基因5(Atg5)的缺失会破坏内皮细胞向造血转化(EHT),通过阻断自噬过程。使用组合方法,包括单细胞RNA测序(scRNA-seq),我们已经证实,Atg5缺失中断EHT的发育时间顺序,进一步影响前HSCI成熟,自噬可能通过核仁素途径影响HEC的生血潜能和前HSCI的形成。这些发现证明了自噬在造血前体的形成/成熟中的作用。
    An understanding of the mechanisms regulating embryonic hematopoietic stem cell (HSC) development would facilitate their regeneration. The aorta-gonad-mesonephros region is the site for HSC production from hemogenic endothelial cells (HEC). While several distinct regulators are involved in this process, it is not yet known whether macroautophagy (autophagy) plays a role in hematopoiesis in the pre-liver stage. Here, we show that different states of autophagy exist in hematopoietic precursors and correlate with hematopoietic potential based on the LC3-RFP-EGFP mouse model. Deficiency of autophagy-related gene 5 (Atg5) specifically in endothelial cells disrupts endothelial to hematopoietic transition (EHT), by blocking the autophagic process. Using combined approaches, including single-cell RNA-sequencing (scRNA-seq), we have confirmed that Atg5 deletion interrupts developmental temporal order of EHT to further affect the pre-HSC I maturation, and that autophagy influences hemogenic potential of HEC and the formation of pre-HSC I likely via the nucleolin pathway. These findings demonstrate a role for autophagy in the formation/maturation of hematopoietic precursors.
    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
    第一个造血干细胞和祖细胞(HSPCs)出现在中期妊娠小鼠胚胎的主动脉-性腺-中肾(AGM)区域。然而,其支持性间充质微环境的确切性质在很大程度上仍未被探索。这里,我们对激光显微解剖的主动脉组织在三个发育阶段的转录组和单个AGM细胞进行了分析。计算分析允许鉴定胚胎第11.5天AGM间充质内的几个细胞亚群,与显着存在尚未确定的亚群,其特征是与粘附或神经元功能有关的基因的双重表达。我们证实了这个细胞亚群作为神经间质细胞群的身份,通过形态学和谱系追踪分析。斑马鱼的功能丧失证实了Decorin,神经间质的特征性细胞外基质成分,对于HSPC开发至关重要。我们进一步证明了这种细胞群不仅仅来自神经c,因此,是AGM间充质的真正新颖亚群。
    The first hematopoietic stem and progenitor cells (HSPCs) emerge in the Aorta-Gonad-Mesonephros (AGM) region of the mid-gestation mouse embryo. However, the precise nature of their supportive mesenchymal microenvironment remains largely unexplored. Here, we profiled transcriptomes of laser micro-dissected aortic tissues at three developmental stages and individual AGM cells. Computational analyses allowed the identification of several cell subpopulations within the E11.5 AGM mesenchyme, with the presence of a yet unidentified subpopulation characterized by the dual expression of genes implicated in adhesive or neuronal functions. We confirmed the identity of this cell subset as a neuro-mesenchymal population, through morphological and lineage tracing assays. Loss of function in the zebrafish confirmed that Decorin, a characteristic extracellular matrix component of the neuro-mesenchyme, is essential for HSPC development. We further demonstrated that this cell population is not merely derived from the neural crest, and hence, is a bona fide novel subpopulation of the AGM mesenchyme.
    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
    造血干细胞(HSC)产生血液的所有必需细胞成分。支持HSC的基质细胞系遵循血管平滑肌细胞(vSMC)分化途径,这表明一些造血支持细胞源自vSMC前体。最近在主动脉-性腺-中肾(AGM)区域发现了这些周细胞样前体;然而,它们在体内造血发育中的作用仍然未知。这里,我们确定了NG2+Runx1+血管周细胞亚群,其显示了一个来自巩膜组的vSMC转录组特征.我们表明,在NG2+细胞中删除Runx1会损害体内造血发育,并导致周细胞/vSMC的转录变化,鼠AGM中的内皮细胞和造血细胞。重要的是,这种缺失还导致体内骨髓中HSC重建潜力的显著降低。这个缺陷是发育性的,因为在成人骨髓中未检测到NG2+Runx1+细胞,证明了在产生HSC的利基中存在专门的周细胞种群,独特的胚胎。
    Hematopoietic stem cells (HSCs) produce all essential cellular components of the blood. Stromal cell lines supporting HSCs follow a vascular smooth muscle cell (vSMC) differentiation pathway, suggesting that some hematopoiesis-supporting cells originate from vSMC precursors. These pericyte-like precursors were recently identified in the aorta-gonad-mesonephros (AGM) region; however, their role in the hematopoietic development in vivo remains unknown. Here, we identify a subpopulation of NG2+Runx1+ perivascular cells that display a sclerotome-derived vSMC transcriptomic profile. We show that deleting Runx1 in NG2+ cells impairs the hematopoietic development in vivo and causes transcriptional changes in pericytes/vSMCs, endothelial cells and hematopoietic cells in the murine AGM. Importantly, this deletion leads also to a significant reduction of HSC reconstitution potential in the bone marrow in vivo. This defect is developmental, as NG2+Runx1+ cells were not detected in the adult bone marrow, demonstrating the existence of a specialised pericyte population in the HSC-generating niche, unique to the embryo.
    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
    造血干细胞(HSC)从主动脉-性腺-和中肾(AGM)区域中的生血内皮(HE)发育,并与造血祖细胞(HPC)一起存在于主动脉内造血簇(IAHC)内。区分HSC和HPC的信号传导机制是未知的。Notch信号对动脉规范至关重要,IAHC形成和HSC活性,但是目前关于Notch如何将这些不同的命运分开的研究是不一致的。我们现在证明Notch活性在以下子集中最高,GFI1+,HSC引发的HE细胞,并且随着HSC成熟而逐渐丧失。我们发现,由于同一细胞表面上NOTCH1和JAG1相互作用水平的增加(顺式),导致NOTCH1受体被激活,因此HSC表型得以维持。IAHC中NOTCH1受体的强制激活激活造血分化程序。我们的结果表明,NOTCH1-JAG1顺式抑制保留了胚胎主动脉造血簇中的HSC表型。
    Hematopoietic stem cells (HSCs) develop from the hemogenic endothelium (HE) in the aorta- gonads-and mesonephros (AGM) region and reside within Intra-aortic hematopoietic clusters (IAHC) along with hematopoietic progenitors (HPC). The signalling mechanisms that distinguish HSCs from HPCs are unknown. Notch signaling is essential for arterial specification, IAHC formation and HSC activity, but current studies on how Notch segregates these different fates are inconsistent. We now demonstrate that Notch activity is highest in a subset of, GFI1 + , HSC-primed HE cells, and is gradually lost with HSC maturation. We uncover that the HSC phenotype is maintained due to increasing levels of NOTCH1 and JAG1 interactions on the surface of the same cell (cis) that renders the NOTCH1 receptor from being activated. Forced activation of the NOTCH1 receptor in IAHC activates a hematopoietic differentiation program. Our results indicate that NOTCH1-JAG1 cis-inhibition preserves the HSC phenotype in the hematopoietic clusters of the embryonic aorta.
    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
    在人类胎儿发育过程中,性别分化不仅发生在性腺中,而且发生在相邻的发育中的生殖道中。然而,虽然男性和女性胎儿性腺的细胞组成得到了很好的描述,相邻发育中的生殖道的特征仍然很差。这里,我们对男性和女性人类胎儿性腺以及邻近的发育中的生殖道进行了单细胞转录组学,突出性别分化过程中的形态和分子变化。我们验证了发育中的生殖道和性腺的不同细胞群,并比较了孕早期和中期的分子特征,以及两性之间,以确定保守性和性别特异性特征。一起,我们的研究提供了人类胎儿性别特异性性腺发生和性腺以外生殖道发育的见解。
    During human fetal development, sex differentiation occurs not only in the gonads but also in the adjacent developing reproductive tract. However, while the cellular composition of male and female human fetal gonads is well described, that of the adjacent developing reproductive tract remains poorly characterized. Here, we performed single-cell transcriptomics on male and female human fetal gonads together with the adjacent developing reproductive tract from first and second trimesters, highlighting the morphological and molecular changes during sex differentiation. We validated different cell populations of the developing reproductive tract and gonads and compared the molecular signatures between the first and second trimesters, as well as between sexes, to identify conserved and sex-specific features. Together, our study provides insights into human fetal sex-specific gonadogenesis and development of the reproductive tract beyond the gonads.
    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
    在主动脉-性腺-中肾(AGM)区域的卡内基阶段(CS)14-17期间,确定的人类造血干细胞(HSC)的出现是一个严格调节的过程。以前,我们在这一时期结束时(CS16/17)对人类AGM区进行了空间转录组分析,并鉴定了参与HSC发育的分泌因子.这里,我们扩展了我们的分析,以调查在整个HSC出现期间,背主动脉周围背腹极化信号的进展.我们的结果表明,从CS13到CS14过渡的腹侧信号复杂性急剧增加,与确定的HSC的首次出现相吻合。我们进一步观察到直至CS17的信号传导的阶段特异性变化,这可能是小鼠模型中描述的HSC逐步成熟的基础。数据丰富的资源也显示在在线界面中,从而可以对AGM区域空间定义域之间的分子相互作用进行计算机模拟分析。该资源对于研究人类HSC发育的潜在机制以及开发从多能干细胞产生临床相关HSC的体外方法的研究人员将特别感兴趣。
    The emergence of definitive human haematopoietic stem cells (HSCs) from Carnegie Stage (CS) 14 to CS17 in the aorta-gonad-mesonephros (AGM) region is a tightly regulated process. Previously, we conducted spatial transcriptomic analysis of the human AGM region at the end of this period (CS16/CS17) and identified secreted factors involved in HSC development. Here, we extend our analysis to investigate the progression of dorso-ventral polarised signalling around the dorsal aorta over the entire period of HSC emergence. Our results reveal a dramatic increase in ventral signalling complexity from the CS13-CS14 transition, coinciding with the first appearance of definitive HSCs. We further observe stage-specific changes in signalling up to CS17, which may underpin the step-wise maturation of HSCs described in the mouse model. The data-rich resource is also presented in an online interface enabling in silico analysis of molecular interactions between spatially defined domains of the AGM region. This resource will be of particular interest for researchers studying mechanisms underlying human HSC development as well as those developing in vitro methods for the generation of clinically relevant HSCs from pluripotent stem cells.
    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
    显示可用于识别/确认中肾/中肾样分化的免疫组织化学标记(MLD标记)包括甲状腺转录因子(TTF1),GATA结合蛋白3(GATA3),和分化簇10(CD10)。只有少数研究检查了子宫内膜子宫内膜样癌(EEC)中MLD标志物的表达水平。本研究旨在分析低度EECs中MLD标记表达的频率和模式。我们进行了免疫染色以检测50个低度EEC组织样品中的TTF1,GATA3和CD10表达,并评估了它们的染色比例和强度。9个肿瘤(18.0%)以不同的比例和强度表达至少一种MLD标记,这些肿瘤中的2个对2个MLD标志物(分别为TTF1/GATA3和GATA3/CD10)呈阳性。三个(6.0%)肿瘤在≤5%的肿瘤细胞中显示出中强核TTF1免疫反应性。五个肿瘤(10.0%)至少有中度核GATA3染色,其中3例染色比例≥15%。三个肿瘤(6.0%)是局灶性的(平均比例,15%)但CD10呈强阳性。我们的发现表明,EEC的子集可以表达一种或多种具有不同染色比例和强度的MLD标记。鉴于子宫中肾样腺癌的诊断应基于特征性组织学特征的组合来确定,独特的免疫表型,并证实了分子发现,病理学家不应仅基于MLD标记的局灶性免疫反应性的存在而排除EEC。了解EEC中MLD标志物的非典型表达模式有助于病理学家避免将EEC误诊为子宫中肾样腺癌。
    Immunohistochemical markers shown to be useful in identifying/confirming mesonephric/mesonephric-like differentiation (MLD markers) include thyroid transcription factor (TTF1), GATA-binding protein 3 (GATA3), and cluster of differentiation 10 (CD10). Only a few studies have examined the expression levels of MLD markers in endometrial endometrioid carcinomas (EECs). This study aimed to analyze the frequency and pattern of MLD marker expression in low-grade EECs. We performed immunostaining for the detection of TTF1, GATA3, and CD10 expression in 50 low-grade EEC tissue samples and evaluated their staining proportion and intensity. Nine tumors (18.0%) expressed at least one MLD marker in varying proportions and intensities, and 2 of these tumors were positive for 2 MLD markers (TTF1/GATA3 and GATA3/CD10, respectively). Three (6.0%) tumors showed moderate-to-strong nuclear TTF1 immunoreactivity in ≤5% of the tumor cells. Five tumors (10.0%) had at least moderate nuclear GATA3 staining, and three of them displayed a staining proportion of ≥15%. Three tumors (6.0%) were focal (mean proportion, 15%) but strongly positive for CD10. Our findings indicate that a subset of EEC can express one or more MLD markers with varying staining proportions and intensities. Given that a diagnosis of uterine mesonephric-like adenocarcinoma should be established based on a combination of characteristic histologic features, unique immunophenotypes, and confirmed molecular findings, pathologists should not exclude EEC based only on the presence of focal immunoreactivity for MLD markers. Awareness of the atypical expression patterns of MLD markers in EEC helps pathologists avoid misdiagnosing EEC as a uterine mesonephric-like adenocarcinoma.
    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
    发育造血由多个组成,部分重叠的造血波,产生胚胎发育所需的分化血细胞,同时建立一个未分化的造血干细胞(HSC)库用于出生后的生活。这种多层设计,其中活跃的造血通过不同的胚胎外和胚胎内组织迁移,这使得很难定义生成HSC与非自我更新祖细胞的路线图。尤其是在人类中。最近的单细胞研究有助于在功能测定不适合将其与祖细胞区分开的阶段鉴定稀有人类HSC。这种方法使得可以将人类HSC的起源追踪到主动脉-性腺-中肾区域中独特类型的动脉内皮,并记录了概念中HSC迁移和成熟的新基准。这些研究为HSC生成的复杂过程提供了新的见解,并提供了工具来告知体外努力,以通过不同的中胚层和内皮中间体复制从多能干细胞到HSC的生理发育历程。
    Developmental hematopoiesis consists of multiple, partially overlapping hematopoietic waves that generate the differentiated blood cells required for embryonic development while establishing a pool of undifferentiated hematopoietic stem cells (HSCs) for postnatal life. This multilayered design in which active hematopoiesis migrates through diverse extra and intraembryonic tissues has made it difficult to define a roadmap for generating HSCs vs non-self-renewing progenitors, especially in humans. Recent single-cell studies have helped in identifying the rare human HSCs at stages when functional assays are unsuitable for distinguishing them from progenitors. This approach has made it possible to track the origin of human HSCs to the unique type of arterial endothelium in the aorta-gonad-mesonephros region and document novel benchmarks for HSC migration and maturation in the conceptus. These studies have delivered new insights into the intricate process of HSC generation and provided tools to inform the in vitro efforts to replicate the physiological developmental journey from pluripotent stem cells via distinct mesodermal and endothelial intermediates to HSCs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号