Hepatocyte Growth Factor (HGF)

肝细胞生长因子 (HGF)
  • 文章类型: Journal Article
    一种新颖的支架设计已经创建,以加强组织工程和再生医学通过优化控制,肝细胞生长因子(HGF)的延长释放,内源性间充质干细胞的强大化学引诱物。我们提出了一种新的堆叠支架,它由三个不同的纤维蛋白凝胶层组成,每个都具有整合到基质中的HGF。该设计试图长时间保持HGF的再生特性,这是复杂组织再生所必需的。这些多层纤维蛋白凝胶已使用流变仪进行了机械评估,并使用D-二聚体ELISA研究了它们的降解行为。了解HGF从这种新型支架结构中释放的动力学对于了解HGF的长期持续生物活性至关重要。进行一系列基于细胞的测试以验证延长掺入后HGF的功能性。这些测试包括2光子显微镜使用鬼笔环肽染色检查细胞形态,支架-细胞相互作用的SEM分析,以及划痕和散射分析,以评估迁移和运动性。分析表明,除了支持HGF的生物活性外,新型堆叠支架还可促进组织再生的重要细胞过程。这种支架设计是为原位组织工程开发的。用身体作为生物反应器,支架应该从它们的小生境中招募间充质干细胞,从而结合HGF和MSCs的再生能力,促进组织重塑和伤口修复。
    A novel scaffold design has been created to enhance tissue engineering and regenerative medicine by optimizing the controlled, prolonged release of Hepatocyte Growth Factor (HGF), a powerful chemoattractant for endogenous mesenchymal stem cells. We present a new stacked scaffold that is made up of three different fibrin gel layers, each of which has HGF integrated into the matrix. The design attempts to preserve HGF\'s regenerative properties for long periods of time, which is necessary for complex tissue regeneration. These multi-layered fibrin gels have been mechanically evaluated using rheometry, and their degradation behavior has been studied using D-Dimer ELISA. Understanding the kinetics of HGF release from this novel scaffold configuration is essential for understanding HGF\'s long-term sustained bioactivity. A range of cell-based tests were carried out to verify the functionality of HGF following extended incorporation. These tests included 2-photon microscopy using phalloidin staining to examine cellular morphology, SEM analysis for scaffold-cell interactions, and scratch and scatter assays to assess migration and motility. The analyses show that the novel stacking scaffold promotes vital cellular processes for tissue regeneration in addition to supporting HGF\'s bioactivity. This scaffold design was developed for in situ tissue engineering. Using the body as a bioreactor, the scaffold should recruit mesenchymal stem cells from their niche, thus combining the regenerative abilities of HGF and MSCs to promote tissue remodeling and wound 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
    肝细胞生长因子(HGF)信号的正调节可能代表了阿尔茨海默病(AD)的一种有希望的治疗策略,基于其多模式神经营养,神经保护,和抗炎作用解决了神经变性的复杂病理生理学。Fosgonimeton是HGF系统的小分子阳性调节剂,已在痴呆的临床前模型中证明了神经营养和认知作用。在这里,我们评估fosgonimeton的神经保护潜力,或其活性代谢物,Fosgo-AM,在淀粉样β(Aβ)驱动的AD临床前模型中,提供对其行动模式的机械洞察。在用Aβ(Aβ1-42)攻击的原代大鼠皮层神经元中,fosgo-AM治疗显著改善神经元的存活,受保护的神经突网络,并减少tau过度磷酸化。细胞内事件的询问表明,用fosgo-AM处理的皮质神经元表现出线粒体氧化应激和细胞色素c释放的显着降低。Aβ损伤后,fosgo-AM显著增强前生存效应因子ERK和AKT的激活,GSK3β活性降低,涉及tau过度磷酸化的主要激酶之一。Fosgo-AM还减轻了Aβ诱导的Unc样激酶1(ULK1)和Beclin-1缺陷,表明对自噬的潜在影响。用fosgo-AM治疗保护皮质神经元免受谷氨酸兴奋性毒性,在存在AKT或MEK/ERK抑制剂的情况下,此类作用被消除。在体内,fosgonimeton给药导致脑室内Aβ25-35大鼠AD模型的功能改善,因为它在被动回避测试中显著挽救了认知功能。一起,我们的数据表明fosgonimeton能够抵消Aβ诱导的毒性机制。Fosgonimeton目前正在进行轻度至中度AD的临床试验(NCT04488419;NCT04886063)。
    Positive modulation of hepatocyte growth factor (HGF) signaling may represent a promising therapeutic strategy for Alzheimer\'s disease (AD) based on its multimodal neurotrophic, neuroprotective, and anti-inflammatory effects addressing the complex pathophysiology of neurodegeneration. Fosgonimeton is a small-molecule positive modulator of the HGF system that has demonstrated neurotrophic and pro-cognitive effects in preclinical models of dementia. Herein, we evaluate the neuroprotective potential of fosgonimeton, or its active metabolite, fosgo-AM, in amyloid-beta (Aβ)-driven preclinical models of AD, providing mechanistic insight into its mode of action. In primary rat cortical neurons challenged with Aβ (Aβ1-42), fosgo-AM treatment significantly improved neuronal survival, protected neurite networks, and reduced tau hyperphosphorylation. Interrogation of intracellular events indicated that cortical neurons treated with fosgo-AM exhibited a significant decrease in mitochondrial oxidative stress and cytochrome c release. Following Aβ injury, fosgo-AM significantly enhanced activation of pro-survival effectors ERK and AKT, and reduced activity of GSK3β, one of the main kinases involved in tau hyperphosphorylation. Fosgo-AM also mitigated Aβ-induced deficits in Unc-like kinase 1 (ULK1) and Beclin-1, suggesting a potential effect on autophagy. Treatment with fosgo-AM protected cortical neurons from glutamate excitotoxicity, and such effects were abolished in the presence of an AKT or MEK/ERK inhibitor. In vivo, fosgonimeton administration led to functional improvement in an intracerebroventricular Aβ25-35 rat model of AD, as it significantly rescued cognitive function in the passive avoidance test. Together, our data demonstrate the ability of fosgonimeton to counteract mechanisms of Aβ-induced toxicity. Fosgonimeton is currently in clinical trials for mild-to-moderate AD (NCT04488419; NCT04886063).
    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
    目的:临床研究发现牙周炎患者肝细胞生长因子(HGF)的高表达。研究表明,HGF在牙周炎中起着重要作用,与炎症有关,并调节牙周炎中牙槽骨的完整性。本研究旨在探讨HGF在实验性牙周炎进展中的作用及其机制。
    方法:我们使用丝线结扎在过表达HGF的转基因(HGF-Tg)和野生型C57BL/6J小鼠中诱导牙周炎。在基线和第7、14、21和28天通过显微计算机断层扫描成像评估HGF过表达对牙槽骨破坏的影响。我们通过酶联免疫吸附法和流式细胞术分析了牙周炎组织中的细胞因子(IL-6和TNF-α)和淋巴细胞。通过定量全身骨代谢标志物CTXI和PINP以及通过RNA测序涉及骨破坏的信号通路,进一步测试了HGF对牙槽骨破坏的作用。进行蛋白质印迹和免疫组织化学以进一步阐明所涉及的信号通路。
    结果:我们发现实验性牙周炎增加了牙周炎组织中HGF的产生;然而,HGF过表达的作用与疾病进展不一致.在牙周炎的早期阶段,HGF-Tg小鼠的牙周炎症和牙槽骨破坏明显低于野生型小鼠。在后期阶段,HGF-Tg小鼠表现出更高的炎症反应,并随着炎症的持续刺激而逐渐加重骨破坏。我们确定IL-17/RANKL/TRAF6通路是参与HGF对牙周炎进展的影响的信号通路。
    结论:HGF在实验性牙周炎的进展中发挥不同的作用,并在炎症晚期加速破骨细胞活性和骨破坏。
    OBJECTIVE: Clinical studies found high levels of hepatocyte growth factor (HGF) expression in patients with periodontitis. Studies suggest that HGF plays an important role in periodontitis, is involved in inflammation, and modulates alveolar bone integrity in periodontitis. This study aims to investigate the effects and mechanisms of HGF in the progression of experimental periodontitis.
    METHODS: We used silk thread ligation to induce periodontitis in HGF-overexpressing transgenic (HGF-Tg) and wild-type C57BL/6J mice. The effects of HGF overexpression on alveolar bone destruction were assessed by microcomputed tomography imaging at baseline and on days 7, 14, 21, and 28. We analyzed the cytokines (IL-6 and TNF-α) and lymphocytes in periodontitis tissues by enzyme-linked immunosorbent assay and flow cytometry. The effects of HGF on alveolar bone destruction were further tested by quantifying the systemic bone metabolism markers CTXI and PINP and by RNA sequencing for the signaling pathways involved in bone destruction. Western blotting and immunohistochemistry were performed to further elucidate the involved signaling pathways.
    RESULTS: We found that experimental periodontitis increased HGF production in periodontitis tissues; however, the effects of HGF overexpression were inconsistent with disease progression. In the early stage of periodontitis, periodontal inflammation and alveolar bone destruction were significantly lower in HGF-Tg mice than in wild-type mice. In the late stage, HGF-Tg mice showed higher inflammatory responses and progressively aggravated bone destruction with continued stimulation of inflammation. We identified the IL-17/RANKL/TRAF6 pathway as a signaling pathway involved in the HGF effects on the progression of periodontitis.
    CONCLUSIONS: HGF plays divergent effects in the progression of experimental periodontitis and accelerates osteoclastic activity and bone destruction in the late stage of inflammation.
    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
    脊髓损伤(SCI)可导致严重和永久性的神经损伤,神经元凋亡对受损脊髓的功能恢复有很大的抑制作用。人脐带间充质干细胞(hUC-MSCs)因其一系列优点,在脊髓损伤修复中具有巨大的潜力,包括抑制神经元凋亡和多分化。前者可能发挥重要作用。然而,与hUC-MSCs给药后神经元凋亡抑制相关的详细调控机制尚未阐明。在这项研究中,从接受hUC-MSC递送的SCI受试者收集的珍贵人脑脊液(CSF)样品的蛋白质组学分析显示肝细胞生长因子(HGF)在很大程度上参与SCI修复。此外,hUC-MSCs过表达HGF可以最大限度地减少活性氧,防止神经元凋亡,从而导致脊髓功能障碍的显著恢复。此外,HGF可以促进Akt/FoxO3a通路的磷酸化,减少活性氧,从而减少神经元凋亡。第一次,我们的研究表明,hUC-MSCs分泌的HGF通过磷酸化Akt/FoxO3a来修复SCI,从而抑制神经元凋亡。这项研究为有效治疗人类SCI提供了与药物选择相关的重要线索。
    Spinal cord injury (SCI) can cause severe and permanent neurological damage, and neuronal apoptosis could inhibit functional recovery of damaged spinal cord greatly. Human umbilical cord mesenchymal stem cells (hUC-MSCs) have great potential to repair SCI because of a series of advantages, including inhibition of neuronal apoptosis and multiple differentiation. The former may play an important role. However, the detailed regulatory mechanism associated with the inhibition of neuronal apoptosis after hUC-MSCs administration has not been elucidated. In this study, proteomics analysis of precious human cerebrospinal fluid (CSF) samples collected from SCI subjects receiving hUC-MSCs delivery indicated that hepatocyte growth factor (HGF) is largely involved in SCI repair. Furthermore, overexpression of HGF derived from hUC-MSCs could decrease reactive oxygen species to prevent neuron apoptosis to the maximum, and thus lead to significant recovery of spinal cord dysfunction. Moreover, HGF could promote phosphorylation of Akt/FoxO3a pathway to decrease reactive oxygen species to reduce neuron apoptosis. For the first time, our research revealed that HGF secreted by hUC-MSCs inhibits neuron apoptosis by phosphorylation of Akt/FoxO3a to repair SCI. This study provides important clues associated with drug selection for the effective treatment of SCI in humans.
    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
    机械扰动通过一系列事件触发常驻肌源性干细胞的激活以进入细胞周期,包括肝细胞生长因子(HGF)从其细胞外束缚释放并随后呈递至信号受体c-met。这里,我们证明随着年龄的增长,细胞外HGF经历酪氨酸残基(Y)硝化并失去c-met结合,从而扰乱肌肉稳态。生化研究表明,硝化/功能障碍对其他主要生长因子中的HGF具有特异性,其特征在于其在c-met结合域中Y198和Y250的位置。三个年龄组大鼠下肢肌肉的直接免疫荧光显微镜检查,为年龄相关的ECM结合的HGF硝化增加提供了直接的体内证据,在快速IIa和IIx肌纤维中优先染色抗硝化Y198和Y250-HGFmAb(内部培养)。总的来说,研究结果强调了HGF硝化对肌源性干细胞动力学的抑制作用,开创了一个有说服力的讨论,以更好地理解与年龄相关的肌肉萎缩和纤维化(包括肌肉减少症和虚弱)的再生受损。
    Mechanical perturbation triggers activation of resident myogenic stem cells to enter the cell cycle through a cascade of events including hepatocyte growth factor (HGF) release from its extracellular tethering and the subsequent presentation to signaling-receptor c-met. Here, we show that with aging, extracellular HGF undergoes tyrosine-residue (Y) nitration and loses c-met binding, thereby disturbing muscle homeostasis. Biochemical studies demonstrated that nitration/dysfunction is specific to HGF among other major growth factors and is characterized by its locations at Y198 and Y250 in c-met-binding domains. Direct-immunofluorescence microscopy of lower hind limb muscles from three age groups of rat, provided direct in vivo evidence for age-related increases in nitration of ECM-bound HGF, preferentially stained for anti-nitrated Y198 and Y250-HGF mAbs (raised in-house) in fast IIa and IIx myofibers. Overall, findings highlight inhibitory impacts of HGF nitration on myogenic stem cell dynamics, pioneering a cogent discussion for better understanding age-related muscle atrophy and impaired regeneration with fibrosis (including sarcopenia and frailty).
    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
    血管生成在外周动脉疾病(PAD)中起重要作用,血管生成相关蛋白可作为预后生物标志物。这项研究评估了血管生成相关蛋白预测与PAD相关的不良事件的潜力。
    这是一项病例对照研究。患有PAD的患者(n=250)和没有PAD的患者(n=125)提供了血液样本,并进行了三年的前瞻性随访。测定血浆中17种血管生成相关蛋白的浓度。主要不良肢体事件(MALE)的发生率,定义为大截肢或血管介入的复合材料,是主要结果。PAD状态恶化,定义为踝臂指数下降≥0.15,是次要结局.进行校正基线特征的多变量回归以确定血管生成相关蛋白在预测男性中的预测值。
    相对于没有PAD的患者,8种与血管生成相关的蛋白在PAD患者中差异表达。在52(14%)和83(22%)患者中观察到PAD状态和男性恶化,分别。肝细胞生长因子(HGF)是男性最可靠的预测因子(校正HR0.79,95%CI0.15-0.86)。与高HGF的个体相比,低HGF患者的三年男性自由度下降[66%vs88%,p=0.001],大截肢[93%对98%,p=0.023],血管介入[68%和88%,p=0.001],PAD状况恶化[81%对91%,p=0.006]。
    测量PAD患者的血浆HGF水平可以帮助识别可能受益于其他评估或治疗的与PAD相关的不良事件风险升高的患者。
    UNASSIGNED: Angiogenesis plays an important role in peripheral artery disease (PAD) and angiogenesis-related proteins may act as prognostic biomarkers. This study assesses the potential for angiogenesis-related proteins to predict adverse events associated with PAD.
    UNASSIGNED: This was a case-control study. Patients with PAD (n = 250) and without PAD (n = 125) provided blood samples and were followed prospectively for three years. Concentrations of 17 angiogenesis-related proteins were measured in plasma. The incidence of major adverse limb event (MALE), defined as a composite of major amputation or vascular intervention, was the primary outcome. Worsening PAD status, defined as a drop in ankle brachial index ≥ 0.15, was the secondary outcome. Multivariable regression adjusted for baseline characteristics was conducted to determine the prognostication value of angiogenesis-related proteins in predicting MALE.
    UNASSIGNED: Relative to patients without PAD, 8 proteins related to angiogenesis were expressed differentially in PAD patients. Worsening PAD status and MALE were observed in 52 (14%) and 83 (22%) patients, respectively. Hepatocyte growth factor (HGF) was the most reliable predictor of MALE (adjusted HR 0.79, 95% CI 0.15-0.86). Compared to individuals with high HGF, patients with low HGF had a decreased three-year freedom from MALE [66% vs 88%, p = 0.001], major amputation [93% vs 98%, p = 0.023], vascular intervention [68% vs 88%, p = 0.001], and worsening PAD status [81% vs 91%, p = 0.006].
    UNASSIGNED: Measuring plasma levels of HGF in individuals with PAD can assist in identifying patients at elevated risk of adverse events related to PAD who may benefit from additional evaluation or treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Comparative Study
    同种异体刺激的CD8+T细胞(CD8+T细胞),作为急性肝排斥反应(ARJ)的主要介质,由于死亡受体FAS介导的途径失活而对细胞凋亡不敏感,并且无法诱导耐受,最终导致急性移植物排斥反应。虽然他克莫司(FK506),临床上最常用的免疫抑制剂(IS),允许公差感应,它的使用是有限的,因为它的靶免疫细胞是未知的,并且它与恶性肿瘤的发病率增加有关,感染,和肾毒性,这对长期肝移植(LTx)结局有重大影响。暗刺鼠(DA)至Lewis大鼠LTx模型是众所周知的ARJ模型,因此选择用于本研究。我们显示肝细胞生长因子(HGF)(cHGF,含有促进HGF产生的主要形式)和重组HGF(h-rHGF)主要通过抑制cMet对FAS的拮抗作用和Fas三聚作用,通过FAS介导的凋亡途径对同种异体aCD8T细胞抑制发挥免疫调节作用,导致急性耐受诱导。我们还表明,通过用针对cMet(仅HGF受体)的中和抗体处理可以消除这种抑制。相比之下,我们在用FK506治疗的大鼠中未观察到这些作用。然而,我们观察到FK506的抗排斥作用主要是对同种异体刺激的CD4+T细胞(aCD4+T细胞)抑制和调节性T细胞(Treg)的促进,与HGF的机制相反。此外,探讨了HGF在FK506介导的肾毒性中的保护机制.因此,HGF作为耐受性诱导剂,无论是与FK506联合使用还是作为单一疗法,有较好的临床价值。这些T细胞亚群在其他生物系统中的其他作用以及这些领域的研究也将是有意义的。
    Allostimulated CD8+ T cells (aCD8+ T cells), as the main mediators of acute liver rejection (ARJ), are hyposensitive to apoptosis due to the inactivation of death receptor FAS-mediated pathways and fail to allow tolerance induction, eventually leading to acute graft rejection. Although tacrolimus (FK506), the most commonly used immunosuppressant (IS) in the clinic, allows tolerance induction, its use is limited because its target immune cells are unknown and it is associated with increased incidences of malignancy, infection, and nephrotoxicity, which substantially impact long-term liver transplantation (LTx) outcomes. The dark agouti (DA)-to-Lewis rat LTx model is a well-known ARJ model and was hence chosen for the present study. We show that both hepatocyte growth factor (HGF) (cHGF, containing the main form of promoting HGF production) and recombinant HGF (h-rHGF) exert immunoregulatory effects mainly on allogeneic aCD8+ T cell suppression through FAS-mediated apoptotic pathways by inhibiting cMet to FAS antagonism and Fas trimerization, leading to acute tolerance induction. We also showed that such inhibition can be abrogated by treatment with neutralizing antibodies against cMet (HGF-only receptor). In contrast, we did not observe these effects in rats treated with FK506. However, we observed that the effect of anti-rejection by FK506 was mainly on allostimulated CD4+ T cell (aCD4+ T cell) suppression and regulatory T cell (Treg) promotion, in contrast to the mechanism of HGF. In addition, the protective mechanism of HGF in FK506-mediated nephrotoxicity was addressed. Therefore, HGF as a tolerance inducer, whether used in combination with FK506 or as monotherapy, may have good clinical value. Additional roles of these T-cell subpopulations in other biological systems and studies in these fields will also be meaningful.
    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
    原发性卵巢功能不全(POI)引起的并发症,包括不孕症,骨质疏松,心血管疾病和抑郁症,严重影响女性患者的生活质量。尽管激素替代疗法(HRT)可以缓解一些长期并发症,目前仍没有标准的卵巢储备功能恢复治疗方法。目前,人脐带间充质干细胞(HUCMSC)移植在大鼠模型和临床上对POI均有较好的治疗效果。为了提高幼稚HUCMSC(HUCMSC-Null)治疗对POI的有效性,在POI卵巢中促进卵泡血管生成的外源基因肝细胞生长因子(HGF)用于修饰HUCMSC。随后,将HGF过表达的HUCMSC(HUCMSC-HGF)移植到化疗诱导的Sprague-Dawley(SD)大鼠的卵巢中,观察其对POI的改善效果及其相关机制。我们的结果表明,与POI和HUCMSC-Null治疗组相比,HUCMSC-HGF显著改善POI组卵巢储备功能,这可能归因于卵巢组织纤维化和颗粒细胞(GCs)凋亡的减少,以及HGF过表达介导的卵巢血管生成的增加。研究结果表明,HGF修饰的HUCMSC在挽救POI中的卵巢储备功能方面可能比单独的HUCMSC具有更好的能力。
    Complications caused by Primary ovarian insufficiency (POI), including infertility, osteoporosis, cardiovascular diseases and depression, severely affect the life quality of female patients. Although hormone replacement therapy (HRT) can alleviate some long-term complications, there is still no standard treatment for the restoration of ovarian reserve function. Currently, human umbilical cord mesenchymal stem cells (HUCMSC) transplantation showed considerable treatment effect for POI in both rat model and clinic. To improve the effectiveness of naïve HUCMSC (HUCMSC-Null) treatments on POI, an exogenous gene hepatocyte growth factor (HGF) which promotes follicular angiogenesis in POI ovaries was used to modify HUCMSC. Subsequently, HGF-overexpressed HUCMSC (HUCMSC-HGF) was transplanted into the ovaries of chemotherapy-induced POI Sprague-Dawley (SD) rats to observe the effectiveness on POI improvement and its related mechanisms. Our results showed that when compared with POI and HUCMSC-Null treatment group, HUCMSC-HGF significantly improved ovarian reserve function in POI group, which might be attributed to the decrease of ovarian tissue fibrosis and granulosa cells (GCs) apoptosis, and the increase of ovarian angiogenesis mediated by HGF over-expression. The findings suggest that HGF-modified HUCMSC may present a more superior capacity than HUCMSC alone for the rescue of ovarian reserve function in POI.
    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
    目的:由于化疗耐药,大多数患者无法治愈癌症。癌症相关成纤维细胞(CAFs)在癌症化疗耐药中起着至关重要的作用,但缺乏对化疗耐药肺癌的详细了解。在这项研究中,我们研究了程序性死亡配体1(PDL-1)作为CAF诱导的化疗耐药的潜在生物标志物,并评估了其在非小细胞肺癌(NSCLC)中的作用和潜在的化疗耐药机制.
    方法:对NSCLC中多个组织的基因表达谱进行系统搜索,以确定传统成纤维细胞生物标志物和CAF分泌的原瘤细胞因子的表达强度。通过ELISA分析CAFs中的PDL-1表达,西方印迹,和流式细胞术。人细胞因子阵列用于鉴定从CAF分泌的特定细胞因子。使用CRISPR/Cas9敲低和包括MTT在内的各种功能测定来评估PDL-1在NSCLC化学抗性中的作用。细胞入侵,球体形成,和细胞凋亡。使用具有活细胞成像和免疫组织化学的共植入异种移植小鼠模型进行体内实验。
    结果:我们证明化疗刺激的CAFs促进NSCLC细胞的致瘤和干细胞样特性,这有助于他们的化学抗性。随后,我们发现,PDL-1表达在化疗的CAFs中上调,并与不良预后相关.沉默PDL-1表达抑制CAFs促进干细胞样特性和肺癌细胞侵袭力的能力,有利于化学抗性。机械上,PDL-1在化疗治疗的CAFs中的上调导致肝细胞生长因子(HGF)分泌增加,刺激癌症进展,细胞入侵,和肺癌细胞的干性,同时抑制细胞凋亡。
    结论:我们的结果表明,PDL-1阳性CAFs通过分泌升高的HGF来调节NSCLC细胞的干细胞样特性,从而促进化学抗性。我们的发现支持PDL-1在CAF中作为化疗反应生物标志物,并作为化疗耐药NSCLC的药物递送和治疗靶点。
    A cure for cancer is out of reach for most patients due to chemoresistance. Cancer-associated fibroblasts (CAFs) play a vital role in cancer chemoresistance, but detailed understanding of the process particularly in chemoresistant lung cancer is lacking. In this study, we investigated programmed death-ligand 1 (PDL-1) as a potential biomarker for CAF-induced chemoresistance and evaluated its role and the underlying mechanisms of chemoresistance in non-small cell lung cancer (NSCLC).
    A systemic search of gene expression profiles of multiple tissues in NSCLC was carried out to determine the expression intensities of traditional fibroblast biomarkers and CAF-secreted protumorigenic cytokines. PDL-1 expression in CAFs was analyzed by ELISA, Western blotting, and flow cytometry. Human cytokine array was used to identify specific cytokines secreted from CAFs. Role of PDL-1 in NSCLC chemoresistance was assessed using CRISPR/Cas9 knockdown and various functional assays including MTT, cell invasion, sphere formation, and cell apoptosis. In vivo experiments were conducted using a co-implantation xenograft mouse model with live cell imaging and immunohistochemistry.
    We demonstrated that chemotherapy-stimulated CAFs promoted tumorigenic and stem cell-like properties of NSCLC cells, which contribute to their chemoresistance. Subsequently, we revealed that PDL-1 expression is upregulated in chemotherapy-treated CAFs and is associated with poor prognosis. Silencing PDL-1 expression suppressed CAFs\' ability to promote stem cell-like properties and invasiveness of lung cancer cells, favoring chemoresistance. Mechanistically, an upregulation of PDL-1 in chemotherapy-treated CAFs led to an increase in hepatocyte growth factor (HGF) secretion, which stimulates cancer progression, cell invasion, and stemness of lung cancer cells, while inhibiting apoptosis.
    Our results show that PDL-1-positive CAFs modulate stem cell-like properties of NSCLC cells by secreting elevated HGF, thereby promoting chemoresistance. Our finding supports PDL-1 in CAFs as a chemotherapy response biomarker and as a drug delivery and therapeutic target for chemoresistant NSCLC.
    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
    压力性溃疡(PU)随着全球老龄化而增加,但是没有有效的因果疗法。虽然间充质干细胞(MSCs)促进皮肤伤口愈合,MSCs的条件培养基(CM)对缺血再灌注损伤诱导的皮肤PU形成的影响研究甚少。为了解决这个问题,在这里,我们首先从人类脱落的乳牙(SHED)中建立了永生化的干细胞系。该细胞系具有优越的特性,因为它无限旺盛地生长,并且稳定且一致地分泌多种细胞因子。使用从永生化SHED细胞系获得的CM,我们使用两个磁性板研究了皮肤缺血再灌注小鼠模型对PU形成的治疗潜力。这是第一个研究表明来自永生化SHEDs的CM通过血管内皮生长因子和肝细胞生长因子促进血管生成和氧化应激抵抗而对PU形成发挥治疗作用。因此,MSC的CM具有有效的治疗作用,而这些疗法尚未在人类医学中实施。尽量满足法规对制造和质量控制的要求,有必要生产一贯安全有效的CM。干细胞的永生化可能是满足监管要求的突破之一,因此开辟了一条新的途径来创造一种新型的无细胞再生医学,尽管有必要对质量控制进行进一步调查。
    Pressure ulcers (PUs) are increasing with aging worldwide, but there is no effective causal therapy. Although mesenchymal stem cells (MSCs) promote cutaneous wound healing, the effects of the conditioned medium (CM) of MSCs on cutaneous PU formation induced by ischemia-reperfusion injury have been poorly investigated. To address this issue, herein, we first established an immortalized stem cell line from human exfoliated deciduous teeth (SHED). This cell line was revealed to have superior characteristics in that it grows infinitely and vigorously, and stably and consistently secretes a variety of cytokines. Using the CM obtained from the immortalized SHED cell line, we investigated the therapeutic potential on a cutaneous ischemia-reperfusion mouse model for PU formation using two magnetic plates. This is the first study to show that CM from immortalized SHEDs exerts therapeutic effects on PU formation by promoting angiogenesis and oxidative stress resistance through vascular endothelial growth factor and hepatocyte growth factor. Thus, the CM of MSCs has potent therapeutic effects, whereas these therapies have not been implemented in human medicine. To try to meet the regulatory requirements for manufacturing and quality control as much as possible, it is necessary to produce CM that is consistently safe and effective. The immortalization of stem cells could be one of the breakthroughs to meet the regulatory requirements and consequently open up a novel avenue to create a novel type of cell-free regenerative medicine, although further investigation into the quality control is warranted.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号