Immune Privilege

免疫特权
  • 文章类型: Journal Article
    历史上,中枢神经系统(CNS)被认为是“免疫特权”,拥有自己独特的免疫细胞群。这种免疫特权被认为是由严密的血脑屏障(BBB)和血脑脊液屏障(BCSFB)建立的。这阻止了外周免疫细胞及其分泌因子进入CNS实质。然而,最近的研究表明,在各种脑边界壁ni如脉络丛附近存在外周免疫细胞,颅骨骨髓(CBM),脑膜,和血管周围的空间。此外,新出现的证据表明,外周免疫细胞可能能够通过这些部位渗透到大脑中,并在神经退行性疾病中驱动神经元细胞死亡和病理进展中发挥重要作用。因此,在这次审查中,我们探讨脑边界免疫壁如何有助于神经退行性疾病的发病机制,如阿尔茨海默病(AD),帕金森病(PD),和多发性硬化症(MS)。然后,我们讨论了利用这些生态位的神经免疫潜力来改善这些衰弱性疾病的预后和治疗的几种新兴选择。
    Historically, the central nervous system (CNS) was regarded as \'immune-privileged\', possessing its own distinct immune cell population. This immune privilege was thought to be established by a tight blood-brain barrier (BBB) and blood-cerebrospinal-fluid barrier (BCSFB), which prevented the crossing of peripheral immune cells and their secreted factors into the CNS parenchyma. However, recent studies have revealed the presence of peripheral immune cells in proximity to various brain-border niches such as the choroid plexus, cranial bone marrow (CBM), meninges, and perivascular spaces. Furthermore, emerging evidence suggests that peripheral immune cells may be able to infiltrate the brain through these sites and play significant roles in driving neuronal cell death and pathology progression in neurodegenerative disease. Thus, in this review, we explore how the brain-border immune niches may contribute to the pathogenesis of neurodegenerative disorders such as Alzheimer\'s disease (AD), Parkinson\'s disease (PD), and multiple sclerosis (MS). We then discuss several emerging options for harnessing the neuroimmune potential of these niches to improve the prognosis and treatment of these debilitative disorders using novel insights from recent studies.
    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
    毛囊是一种动态的微型器官,具有特殊的周期和结构,具有多种细胞类型以形成毛发。过去几十年的研究已经研究了小鼠和人类皮肤中胚胎发育和成人毛发周期期间的形态发生和信号通路。特别是,毛囊干细胞和间充质利基作为关键参与者受到了主要关注,他们的角色和互动被大量揭示。尽管驻留和循环的免疫细胞会影响皮肤中的细胞功能和相互作用,对免疫细胞的研究主要受到疾病的关注,而不是发育或稳态。最近,许多研究表明,不同的免疫细胞作为毛囊的生态位的功能作用。这里,我们将回顾有关毛囊免疫生态位的最新发现,并提供有关毛发生长和疾病机制的见解。
    The hair follicle is a dynamic mini-organ that has specialized cycles and architectures with diverse cell types to form hairs. Previous studies for several decades have investigated morphogenesis and signaling pathways during embryonic development and adult hair cycles in both mouse and human skin. In particular, hair follicle stem cells and mesenchymal niches received major attention as key players, and their roles and interactions were heavily revealed. Although resident and circulating immune cells affect cellular function and interactions in the skin, research on immune cells has mainly received attention on diseases rather than development or homeostasis. Recently, many studies have suggested the functional roles of diverse immune cells as a niche for hair follicles. Here, we will review recent findings about immune niches for hair follicles and provide insight into mechanisms of hair growth and 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
    斑秃(AA)和白癜风是不同的,异质,和复杂的疾病实体,其特点是无疤痕的头皮终端脱发和皮肤色素损失,分别。在AA,炎症细胞浸润在靠近毛球(蜂群)的深层网状真皮中,而在白癜风中,炎性浸润在表皮和乳头状真皮中。免疫特权崩溃在AA发病机制中已被广泛研究,包括抑制免疫调节因子(例如,转化生长因子-β(TGF-β),程序性死亡配体1(PDL1),白细胞介素-10(IL-10),α-黑素细胞刺激素(α-MSH),和巨噬细胞迁移抑制因子(MIF)),并增强了整个毛囊中主要组织相容性复合物(MHC)的表达。然而,免疫特权崩溃在白癜风中的探索仍然较少。AA和白癜风都是自身免疫性疾病,在发病机制上有共同之处。包括浆细胞样树突状细胞(和干扰素-α(IFN-α)信号通路)和细胞毒性CD8+T淋巴细胞(和激活的IFN-γ信号通路)的参与。血液趋化因子C-X-C基序配体9(CXCL9)和CXCL10在两种疾病中均升高。导致AA和白癜风的常见因素包括氧化应激,自噬,2型细胞因子,和Wnt/β-catenin途径(例如,dickkopf1(DKK1))。这里,我们总结了AA和白癜风之间的共同点和区别,专注于他们的发病机制。
    Both alopecia areata (AA) and vitiligo are distinct, heterogenous, and complex disease entities, characterized by nonscarring scalp terminal hair loss and skin pigment loss, respectively. In AA, inflammatory cell infiltrates are in the deep reticular dermis close to the hair bulb (swarm of bees), whereas in vitiligo the inflammatory infiltrates are in the epidermis and papillary dermis. Immune privilege collapse has been extensively investigated in AA pathogenesis, including the suppression of immunomodulatory factors (e.g., transforming growth factor-β (TGF-β), programmed death-ligand 1 (PDL1), interleukin-10 (IL-10), α-melanocyte-stimulating hormone (α-MSH), and macrophage migration inhibitory factor (MIF)) and enhanced expression of the major histocompatibility complex (MHC) throughout hair follicles. However, immune privilege collapse in vitiligo remains less explored. Both AA and vitiligo are autoimmune diseases that share commonalities in pathogenesis, including the involvement of plasmacytoid dendritic cells (and interferon-α (IFN- α) signaling pathways) and cytotoxic CD8+ T lymphocytes (and activated IFN-γ signaling pathways). Blood chemokine C-X-C motif ligand 9 (CXCL9) and CXCL10 are elevated in both diseases. Common factors that contribute to AA and vitiligo include oxidative stress, autophagy, type 2 cytokines, and the Wnt/β-catenin pathway (e.g., dickkopf 1 (DKK1)). Here, we summarize the commonalities and differences between AA and vitiligo, focusing on their pathogenesis.
    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
    皮肤,人体最大的器官,是具有多种免疫细胞集合的高度免疫原性组织。高度多态性人类白细胞抗原(HLA)分子作为识别分子在协调免疫应答中具有中心作用。然而,特定器官内不同细胞类型之间的HLA基因表达模式,像皮肤一样,尚未彻底调查,基质细胞比免疫细胞吸引的注意力要少得多。为了说明皮肤中不同细胞类型的HLA表达谱,我们对皮肤数据集进行了单细胞RNA测序(scRNA-seq)分析,覆盖成人和胎儿皮肤,和毛囊作为皮肤附属物。我们通过检查正常成人皮肤数据集,揭示了不同皮肤群体之间HLA表达的变化。此外,我们基于经典HLAI类基因的表达评估了多个皮肤群体的潜在免疫原性,在所有细胞类型中都有很好的表现。此外,我们从15个受孕周(PCW)的胎儿中生成了发育中的皮肤的scRNA-seq数据,17PCW,和22PCW,描绘HLA基因在发育过程中各种细胞类型之间的细胞类型依赖性变异的动态表达。值得注意的是,伪时间轨迹分析揭示了血管内皮细胞进化过程中HLA基因的显著差异.此外,我们发现毛囊在单细胞分辨率下的免疫特权特性。我们的研究提出了一个全面的单细胞转录组景观的HLA基因在皮肤,这为HLA分子的变异提供了新的见解,并为同种异体皮肤移植提供了线索。
    Skin, the largest organ of body, is a highly immunogenic tissue with a diverse collection of immune cells. Highly polymorphic human leukocyte antigen (HLA) molecules have a central role in coordinating immune responses as recognition molecules. Nevertheless, HLA gene expression patterns among diverse cell types within a specific organ, like the skin, have yet to be thoroughly investigated, with stromal cells attracting much less attention than immune cells. To illustrate HLA expression profiles across different cell types in the skin, we performed single-cell RNA sequencing (scRNA-seq) analyses on skin datasets, covering adult and fetal skin, and hair follicles as the skin appendages. We revealed the variation in HLA expression between different skin populations by examining normal adult skin datasets. Moreover, we evaluated the potential immunogenicity of multiple skin populations based on the expression of classical HLA class I genes, which were well represented in all cell types. Furthermore, we generated scRNA-seq data of developing skin from fetuses of 15 post conception weeks (PCW), 17 PCW, and 22 PCW, delineating the dynamic expression of HLA genes with cell type-dependent variation among various cell types during development. Notably, the pseudotime trajectory analysis unraveled the significant variance in HLA genes during the evolution of vascular endothelial cells. Moreover, we uncovered the immune-privileged properties of hair follicles at single-cell resolution. Our study presents a comprehensive single-cell transcriptomic landscape of HLA genes in the skin, which provides new insights into variation in HLA molecules and offers a clue for allogeneic skin transplantation.
    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
    自身免疫促进细胞因子,白细胞介素-15(IL-15),通常被认为是斑秃(AA)的关键致病细胞因子。然而,rhIL-15促进人毛囊(HF)离体生长。我们已经询问IL-15及其受体(IL-15R)亚型的表达是否在人类AA中发生改变,以及在存在/不存在干扰素的情况下IL-15如何影响人类HF免疫特权(HF-IP)-γ(IFNγ),有据可查的关键AA致病细胞因子,以及体内实验性AA诱导后的毛发再生。定量免疫形态学显示,与健康对照皮肤相比,AA皮肤活检组织中卵泡周IL-15+T细胞的数量显着增加,而IL-15,IL-15R-α,在AAHFs中,毛球内的IL-15R-γ蛋白表达显着下调。在器官培养的人类头皮HFs中,rhIL-15显着降低MICA的毛球表达,AA发病机制中的关键“危险”信号,并增加了HF-IP守护者的产量,α-MSH.至关重要的是,离体,rhIL-15预防IFNγ诱导的HF-IP崩溃,通过IL-15Rα依赖性信号(如IL-15Rα沉默所记录)恢复崩溃的HF-IP,并保护AA预防性免疫抑制iNKT10细胞免受IFNγ诱导的凋亡。在SCID/米色小鼠体内的人头皮皮肤异种移植中实验性AA诱导后,rhIL-15甚至促进了头发的再生。我们的数据介绍了IL-15作为一种新的,功能重要的HF-IP监护人,其信号在AA患者中是组成型缺陷。我们的数据表明,选择性刺激卵泡内IL-15Rα信号可能成为AA管理的一种新的治疗方法,虽然在药理学上阻断它可能会阻碍HF-IP恢复和头发再生长,因此可能使HF更容易受到AA复发的影响。
    The autoimmunity-promoting cytokine, Interleukin-15 (IL-15), is often claimed to be a key pathogenic cytokine in alopecia areata (AA). Yet, rhIL-15 promotes human hair follicle (HF) growth ex vivo. We have asked whether the expression of IL-15 and its receptor (IL-15R) isoforms is altered in human AA and how IL-15 impacts on human HF immune privilege (HF-IP) in the presence/absence of interferon-γ (IFNγ), the well-documented key AA-pathogenic cytokine, as well as on hair regrowth after experimental AA induction in vivo. Quantitative immunohistomorphometry showed the number of perifollicular IL-15+ T cells in AA skin biopsies to be significantly increased compared to healthy control skin, while IL-15, IL-15Rα, and IL-15Rγ protein expression within the hair bulb were significantly down-regulated in AA HFs. In organ-cultured human scalp HFs, rhIL-15 significantly reduced hair bulb expression of MICA, the key \"danger\" signal in AA pathogenesis, and increased production of the HF-IP guardian, α-MSH. Crucially, ex vivo, rhIL-15 prevented IFNγ-induced HF-IP collapse, restored a collapsed HF-IP by IL-15Rα-dependent signaling (as documented by IL-15Rα-silencing), and protected AA-preventive immunoinhibitory iNKT10 cells from IFNγ-induced apoptosis. rhIL-15 even promoted hair regrowth after experimental AA induction in human scalp skin xenotransplants on SCID/beige mice in vivo. Our data introduce IL-15 as a novel, functionally important HF-IP guardian whose signaling is constitutively defective in scalp HFs of AA patients. Our data suggest that selective stimulation of intrafollicular IL-15Rα signaling could become a novel therapeutic approach in AA management, while blocking it pharmacologically may hinder both HF-IP restoration and hair re-growth and may thus make HFs more vulnerable to AA relapse.
    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
    尽管免疫疗法彻底改变了癌症治疗,仍然迫切需要提高疗效并确保其安全。正确的癌症理论和适当的科学方法赋予相关的癌症研究,并为患者护理提供有效和高效的药物与治疗开发。从这个角度来看,我们重新审视癌细胞相对于正常细胞的免疫特权的概念,以及在癌症干细胞和正常干细胞中。我们重新检查有效的免疫疗法是否由于其抗癌和/或免疫调节机制而有效。我们重新评估了为什么检查点抑制剂(CPIs)不相等。我们重新考虑是否可以将免疫疗法的效用归因于特定的癌症亚型,而将其归因于某些肿瘤/免疫区室。组件,和微环境。我们根据适当的科学理论,通过将抗PD1/L1与各种其他治疗方式相结合,提出了超越CPIs的免疫治疗方法。例如,癌症的干细胞起源,基于现有的临床证据,例如,随机临床试验。我们预测,癌症干细胞理论将有助于设计更好,更安全的免疫疗法,并在正确的时间为正确的癌症类型的正确患者选择其用途,以优化临床益处并最大程度地减少潜在的毒性作用和并发症。
    Although immunotherapy has revolutionized cancer care, there is still an urgent need to enhance its efficacy and ensure its safety. A correct cancer theory and proper scientific method empower pertinent cancer research and enable effective and efficient drug versus therapy development for patient care. In this perspective, we revisit the concept of immune privilege in a cancer cell versus normal cell, as well as in a cancer stem cell versus normal stem cell. We re-examine whether effective immunotherapies are efficacious due to their anti-cancer and/or immune modulatory mechanisms. We reassess why checkpoint inhibitors (CPIs) are not equal. We reconsider whether one can attribute the utility of immunotherapy to specific cancer subtypes and its futility to certain tumor/immune compartments, components, and microenvironments. We propose ways and means to advance immunotherapy beyond CPIs by combining anti-PD1/L1 with various other treatment modalities according to an appropriate scientific theory, e.g., stem cell origin of cancer, and based on available clinical evidence, e.g., randomized clinical trials. We predict that a stem cell theory of cancer will facilitate the design of better and safer immunotherapy with improved selection of its use for the right patient with the right cancer type at the right time to optimize clinical benefits and minimize potential toxic effects and complications.
    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
    持续性鼠诺如病毒株MNVCR6是人诺如病毒和肠道病毒持续性的模型。MNVCR6通过直接感染肠道簇细胞而引起慢性感染,罕见的化学感受上皮细胞。虽然MNVCR6诱导功能性MNV特异性CD8+T细胞,这些淋巴细胞无法清除感染。为了研究簇绒细胞如何促进免疫逃逸,我们通过将JEDI(仅EGFP诱导死亡)CD8+T细胞过继转移到Gfi1b-GFP簇细胞报告小鼠中来询问簇细胞与CD8+T细胞的相互作用。出乎意料的是,与Lgr5+肠干细胞和肠外簇细胞不同,一些肠簇细胞部分抵抗JEDICD8+T细胞介导的杀伤作用,尽管抗原递呈看似正常.当靶向肠簇细胞时,JEDICD8+T细胞主要采用T驻留记忆表型,效应子和细胞毒性能力降低,使簇绒细胞存活。JEDICD8+T细胞既不清除也不阻止结肠中的MNVCR6感染,病毒持续存在的部位,尽管靶向病毒非依赖性抗原。最终,我们表明肠簇状细胞对CD8+T细胞具有相对的抗性,不依赖于诺如病毒感染,代表一个免疫特权的生态位,可以被肠道微生物利用。
    The persistent murine norovirus strain MNVCR6 is a model for human norovirus and enteric viral persistence. MNVCR6 causes chronic infection by directly infecting intestinal tuft cells, rare chemosensory epithelial cells. Although MNVCR6 induces functional MNV-specific CD8+ T cells, these lymphocytes fail to clear infection. To examine how tuft cells promote immune escape, we interrogated tuft cell interactions with CD8+ T cells by adoptively transferring JEDI (just EGFP death inducing) CD8+ T cells into Gfi1b-GFP tuft cell reporter mice. Unexpectedly, some intestinal tuft cells partially resisted JEDI CD8+ T cell-mediated killing-unlike Lgr5+ intestinal stem cells and extraintestinal tuft cells-despite seemingly normal antigen presentation. When targeting intestinal tuft cells, JEDI CD8+ T cells predominantly adopted a T resident memory phenotype with decreased effector and cytotoxic capacity, enabling tuft cell survival. JEDI CD8+ T cells neither cleared nor prevented MNVCR6 infection in the colon, the site of viral persistence, despite targeting a virus-independent antigen. Ultimately, we show that intestinal tuft cells are relatively resistant to CD8+ T cells independent of norovirus infection, representing an immune-privileged niche that can be leveraged by enteric microbes.
    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
    免疫耐受在淋巴器官(LOs)中保持。尽管在非LO中存在复杂的免疫细胞网络,尚不清楚这些组织是否保持了自身耐受。我们开发了一种技术,将遗传重组限制为仅在皮肤中的调节性T细胞(Tregs)。皮肤Treg的选择性消耗导致T细胞介导的毛囊(HF)炎症。抑制不依赖于CTLA-4,而是依赖于皮肤Tregs的高亲和力白介素-2(IL-2)受体表达,完全以细胞外在方式运作。在HF干细胞(HFSC)驱动的自身免疫的新模型中,我们发现皮肤Tregs免疫保护HFSC生态位。最后,我们使用空间转录组学技术在一种罕见的以HFSC破坏和斑秃为特征的人类脱发中,在基质-HF界面上鉴定了异常的IL-2信号传导。总的来说,这些结果揭示了皮肤中Treg的基本生物学特性,并阐明了自身耐受的机制。
    Immune tolerance is maintained in lymphoid organs (LOs). Despite the presence of complex immune cell networks in non-LOs, it is unknown whether self-tolerance is maintained in these tissues. We developed a technique to restrict genetic recombination to regulatory T cells (Tregs) only in skin. Selective depletion of skin Tregs resulted in T cell-mediated inflammation of hair follicles (HFs). Suppression did not rely on CTLA-4, but instead on high-affinity interleukin-2 (IL-2) receptor expression by skin Tregs, functioning exclusively in a cell-extrinsic manner. In a novel model of HF stem cell (HFSC)-driven autoimmunity, we reveal that skin Tregs immunologically protect the HFSC niche. Finally, we used spatial transcriptomics to identify aberrant IL-2 signaling at stromal-HF interfaces in a rare form of human alopecia characterized by HFSC destruction and alopecia areata. Collectively, these results reveal the fundamental biology of Tregs in skin uncoupled from the systemic pool and elucidate a mechanism of self-tolerance.
    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
    目标:免疫特权器官(IPOs),例如中枢神经系统和睾丸,Zika病毒(ZIKV)感染浸润的CD8+T细胞后呈现致病性和炎症。我们的数据表明,CD8+T细胞在免疫特权器官中跟上病毒的增加和减少。此外,我们的研究提供了在解剖部位和ZIKV感染阶段与TCRα/β克隆型相关的组成和多样性的首次离体比较分析。我们表明,组织中绝大多数TCRα/β克隆型利用TRAV9N-3具有保守性。特异性趋化因子表达,包括Ccr2和Ccr5,被发现在大脑和睾丸内的E294-302特异性CD8+T细胞中选择性表达,表明感染后病毒特异性CD8+T细胞以IPO为导向的迁移。我们的研究增加了对不同IPOs中ZIKV感染后病毒特异性CD8+T细胞的抗病毒免疫学特征和趋化机制的见解。
    Zika virus (ZIKV) infection caused neurological complications and male infertility, leading to the accumulation of antigen-specific immune cells in immune-privileged organs (IPOs). Thus, it is important to understand the immunological responses to ZIKV in IPOs. We extensively investigated the ZIKV-specific T cell immunity in IPOs in Ifnar1-/- mice, based on an immunodominant epitope E294-302 tetramer. The distinct kinetics and functions of virus-specific CD8+ T cells infiltrated into different IPOs were characterized, with late elevation in the brain and spinal cord. Single epitope E294-302-specific T cells can account for 20-60% of the total CD8+ T cells in the brain, spinal cord, and testicle and persist for at least 90 days in the brain and spinal cord. The E294-302-specific TCRαβs within the IPOs are featured with the majority of clonotypes utilizing TRAV9N-3 paired with diverse TRBV chains, but with distinct αβ paired clonotypes in 7 and 30 days post-infection. Specific chemokine receptors, Ccr2 and Ccr5, were selectively expressed in the E294-302-specific CD8+ T cells within the brain and testicle, indicating an IPO-oriented migration of virus-specific CD8+ T cells after infection. Overall, this study adds to the understanding of virus-specific CD8+ T cell responses for controlling and clearing ZIKV infection in IPOs.IMPORTANCEThe immune-privileged organs (IPOs), such as the central nervous system and testicles, presented pathogenicity and inflammation after Zika virus (ZIKV) infection with infiltrated CD8+ T cells. Our data show that CD8+ T cells keep up with virus increases and decreases in immune-privileged organs. Furthermore, our study provides the first ex vivo comparative analyses of the composition and diversity related to TCRα/β clonotypes across anatomical sites and ZIKV infection phases. We show that the vast majority of TCRα/β clonotypes in tissues utilize TRAV9N-3 with conservation. Specific chemokine expression, including Ccr2 and Ccr5, was found to be selectively expressed in the E294-302-specific CD8+ T cells within the brain and testicle, indicating an IPO-oriented migration of the virus-specific CD8+ T cells after the infection. Our study adds insights into the anti-viral immunological characterization and chemotaxis mechanism of virus-specific CD8+ T cells after ZIKV infection in different IPOs.
    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
    癌症干细胞的免疫特权是一个众所周知且广泛研究的问题,因为肿瘤中这种细胞的存在与难治性有关,复发,和转移。越来越多的证据还表明,在非病理性干细胞中,除了它们对损伤因子的其他防御机制外,还存在免疫特权。病理和正常干细胞之间的这种相似性提出了为什么干细胞具有这种潜在危险特性的问题。通过免疫细胞之间的相互作用实现的自身免疫控制和再生的重要过程的调节,干细胞,在这项工作中回顾了它们的微环境作为干细胞免疫特权形成的原因。注意到干细胞和免疫细胞的调节之间的深度相互整合。考虑到干细胞相互调节的多样性和复杂性,他们的微环境,和免疫系统,我建议使用术语“茎系统”。
    Immune privileges of cancer stem cells is a well-known and widely studied problem, as presence of such cells in tumors is associated with refractoriness, recurrence, and metastasis. Accumulating evidence also suggests presence of immune privileges in non-pathological stem cells in addition to their other defense mechanisms against damaging factors. This similarity between pathological and normal stem cells raises the question of why stem cells have such a potentially dangerous property. Regulation of vital processes of autoimmunity control and regeneration realized through interactions between immune cells, stem cells, and their microenvironment are reviewed in this work as causes of formation of the stem cell immune privilege. Deep mutual integration between regulations of stem and immune cells is noted. Considering diversity and complexity of mutual regulation of stem cells, their microenvironment, and immune system, I suggest the term \"stem system\".
    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