Hepatocyte Nuclear Factor 1-alpha

肝细胞核因子 1 - α
  • 文章类型: Journal Article
    隐窝处的肠干细胞分裂并产生祖细胞,该祖细胞在转运扩增(TA)区中增殖并分化成各种成熟细胞类型。这里,我们表明转录因子ARID3A调节TA祖细胞的肠上皮细胞增殖和分化。ARID3A形成由TGF-β和WNT介导的从绒毛尖端到上隐窝的表达梯度。Arid3a的肠特异性缺失减少隐窝增殖,主要在TA细胞中。大量和单细胞转录组学分析显示Arid3acKO肠中的肠细胞增加和分泌分化减少,伴随着两个细胞谱系的丰富的上绒毛基因签名。我们发现,缺乏Arid3a的肠道中上皮分化的增强是由HNF1和HNF4的结合和转录增加引起的。最后,我们表明,Arid3a的丢失会损害辐照诱导的再生,并伴有持续的细胞死亡和重编程。我们的发现暗示Arid3a具有微调TA祖细胞的增殖分化动力学的功能,这对损伤诱导的再生至关重要。
    Intestinal stem cells at the crypt divide and give rise to progenitor cells that proliferate and differentiate into various mature cell types in the transit-amplifying (TA) zone. Here, we showed that the transcription factor ARID3A regulates intestinal epithelial cell proliferation and differentiation at the TA progenitors. ARID3A forms an expression gradient from the villus tip to the upper crypt mediated by TGF-β and WNT. Intestinal-specific deletion of Arid3a reduces crypt proliferation, predominantly in TA cells. Bulk and single-cell transcriptomic analysis shows increased enterocyte and reduced secretory differentiation in the Arid3a cKO intestine, accompanied by enriched upper-villus gene signatures of both cell lineages. We find that the enhanced epithelial differentiation in the Arid3a-deficient intestine is caused by increased binding and transcription of HNF1 and HNF4. Finally, we show that loss of Arid3a impairs irradiation-induced regeneration with sustained cell death and reprogramming. Our findings imply that Arid3a functions to fine-tune the proliferation-differentiation dynamics at the TA progenitors, which are essential for injury-induced regeneration.
    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
    背景:抗PD-1抗体已经彻底改变了癌症免疫疗法,因为它们能够在一定比例的患者中诱导持久的完全缓解。当前的研究努力正在尝试鉴定生物标志物和合适的组合伙伴以预测或进一步改善免疫检查点抑制剂的活性。抗体-细胞因子融合是一类显示出增强其他免疫疗法的抗癌特性的潜力的药物。外生A-纤连蛋白(EDA-FN),它在大多数实体和血液肿瘤中表达,但在健康的成人组织中几乎检测不到,是在疾病部位递送细胞因子的有吸引力的靶标。
    方法:在这项工作中,我们描述了一种新的基于白细胞介素7的融合蛋白的产生和表征,该融合蛋白靶向EDA-FN,称为F8(scDb)-IL7。产物由与人IL-7融合的单链双抗体(scDb)形式的FN的选择性剪接EDA特异性的F8抗体组成。
    结果:F8(scDb)-IL7在体外有效刺激人外周血单核细胞。此外,与IL2融合蛋白相比,该产物显着增加CD8T细胞上T细胞因子1(TCF-1)的表达。TCF-1已成为影响抗肿瘤免疫应答的持久性和效力的关键转录因子。在临床前癌症模型中,当与抗PD-1组合时,F8(scDb)-IL7表现出有效的单剂活性并根除肉瘤病变。
    结论:我们的结果为探索F8(scDb)-IL7与抗PD-1抗体联合治疗癌症患者提供了理论基础。
    BACKGROUND: Anti-PD-1 antibodies have revolutionized cancer immunotherapy due to their ability to induce long-lasting complete remissions in a proportion of patients. Current research efforts are attempting to identify biomarkers and suitable combination partners to predict or further improve the activity of immune checkpoint inhibitors. Antibody-cytokine fusions are a class of pharmaceuticals that showed the potential to boost the anticancer properties of other immunotherapies. Extradomain A-fibronectin (EDA-FN), which is expressed in most solid and hematological tumors but is virtually undetectable in healthy adult tissues, is an attractive target for the delivery of cytokine at the site of the disease.
    METHODS: In this work, we describe the generation and characterization of a novel interleukin-7-based fusion protein targeting EDA-FN termed F8(scDb)-IL7. The product consists of the F8 antibody specific to the alternatively spliced EDA of FN in the single-chain diabody (scDb) format fused to human IL-7.
    RESULTS: F8(scDb)-IL7 efficiently stimulates human peripheral blood mononuclear cells in vitro. Moreover, the product significantly increases the expression of T Cell Factor 1 (TCF-1) on CD8+T cells compared with an IL2-fusion protein. TCF-1 has emerged as a pivotal transcription factor that influences the durability and potency of immune responses against tumors. In preclinical cancer models, F8(scDb)-IL7 demonstrates potent single-agent activity and eradicates sarcoma lesions when combined with anti-PD-1.
    CONCLUSIONS: Our results provide the rationale to explore the combination of F8(scDb)-IL7 with anti-PD-1 antibodies for the treatment of patients with cancer.
    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
    肝细胞核因子-1(HNF1α)是多种肿瘤进展的转录因子。然而,关于HNF1α活性的机制知之甚少。本研究旨在探讨HNF1α在结直肠癌(CRC)进展中的作用及其分子机制。CRC标本中HNF1α表达上调,高表达与CRC患者预后不良相关。HNF1α敲低和过表达抑制和促进增殖,CRC细胞在体外和体内的迁移和侵袭。机械上,HNF1α增加己糖激酶结构域组分1(HKDC1)启动子的转录活性,从而激活AKT/AMPK信号。同时,HKDC1上调对增殖很重要,HKDC1基因敲除后可显著逆转CRC细胞的增殖、迁移和侵袭,HNF1α过表达诱导的迁移和侵袭。一起来看,HNF1α通过与HKDC1结合并激活AKT/AMPK信号参与CRC的进展和转移。靶向HNF1α可能是CRC患者的潜在治疗策略。
    The hepatocyte nuclear factor-1 (HNF1ɑ) is a transcription factor that contributes to several kinds of cancer progression. However, very little is known regarding the mechanisms underlying the activity of HNF1ɑ. We aimed to explore the role of HNF1ɑ in the progress of colorectal cancer (CRC) and elucidate its molecular mechanism. HNF1ɑ expression was upregulated in CRC samples and high expression of HNF1ɑ was associated with poor prognosis of CRC patients. HNF1α knockdown and overexpression inhibited and promoted proliferation, migration and invasion of CRC cells both in vitro and in vivo respectively. Mechanistically, HNF1ɑ increased the transcriptional activity of hexokinase domain component 1(HKDC1)promoter, thus activated AKT/AMPK signaling. Meanwhile, HKDC1 upregulation was important for the proliferation, migration and invasion of CRC cells and knockdown of HKDC1 significantly reversed the proliferation, migration and invasion induced by HNF1α overexpression. Taken together, HNF1ɑ contributes to CRC progression and metastasis through binding to HKDC1 and activating AKT/AMPK signaling. Targeting HNF1ɑ could be a potential therapeutic strategy for CRC patients.
    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
    背景:黑色素瘤,最致命的皮肤癌,随着检查点阻断免疫疗法(CBI)的出现,经历了变革性的治疗转变。了解浸润肿瘤的免疫细胞的复杂网络并协调黑色素瘤细胞的控制和对CBI的反应目前至关重要。有证据强调了组织驻留记忆(TRM)CD8T细胞和经典的1型树突状细胞(cDC1)在癌症保护中的重要性。转录组学研究还支持TCF7+(编码TCF1)T细胞的存在,作为最重要的免疫治疗反应,尽管关于是否存在TCF1+TRMT细胞存在不确定性,因为有证据表明TCF1下调组织滞留激活。
    方法:我们使用多重免疫荧光和光谱流式细胞术评估两个黑色素瘤患者队列中的TRMCD8T细胞和cDC1:一个未接受免疫治疗,另一个接受免疫治疗。第一个队列在诊断后2年无疾病或有转移的患者之间进行划分,而第二个队列在CBI应答者和无应答者之间进行划分。
    结果:我们的研究确定了两个CD8+TRM亚群,TCF1+和TCF1-,与黑色素瘤保护相关。TCF1+TRM细胞显示IFN-γ和Ki67的高表达,而TCF1-TRM细胞显示细胞毒性分子的高表达。在转移性患者中,TRM子集经历了标记表达的转变,TCF1-亚群显示耗尽标志物的表达增加。我们观察到cDC1和TRM之间存在密切的空间相关性,TCF1+TRM/cDC1对富集在基质中,TCF1-TRM/cDC1对富集在肿瘤区域中。值得注意的是,这些TCF1-TRMs表达细胞毒性分子并与凋亡的黑色素瘤细胞相关。TCF1+和TCF1-TRM子集,与cDC1一起,证明与CBI反应相关。
    结论:我们的研究支持TRMCD8T细胞和cDC1在黑色素瘤保护中的重要性,同时也强调了功能上独特的TCF1和TCF1-TRM亚群的存在,对黑素瘤控制和CBI反应都至关重要。
    BACKGROUND: Melanoma, the most lethal form of skin cancer, has undergone a transformative treatment shift with the advent of checkpoint blockade immunotherapy (CBI). Understanding the intricate network of immune cells infiltrating the tumor and orchestrating the control of melanoma cells and the response to CBI is currently of utmost importance. There is evidence underscoring the significance of tissue-resident memory (TRM) CD8 T cells and classic dendritic cell type 1 (cDC1) in cancer protection. Transcriptomic studies also support the existence of a TCF7+ (encoding TCF1) T cell as the most important for immunotherapy response, although uncertainty exists about whether there is a TCF1+TRM T cell due to evidence indicating TCF1 downregulation for tissue residency activation.
    METHODS: We used multiplexed immunofluorescence and spectral flow cytometry to evaluate TRM CD8 T cells and cDC1 in two melanoma patient cohorts: one immunotherapy-naive and the other receiving immunotherapy. The first cohort was divided between patients free of disease or with metastasis 2 years postdiagnosis while the second between CBI responders and non-responders.
    RESULTS: Our study identifies two CD8+TRM subsets, TCF1+ and TCF1-, correlating with melanoma protection. TCF1+TRM cells show heightened expression of IFN-γ and Ki67 while TCF1- TRM cells exhibit increased expression of cytotoxic molecules. In metastatic patients, TRM subsets undergo a shift in marker expression, with the TCF1- subset displaying increased expression of exhaustion markers. We observed a close spatial correlation between cDC1s and TRMs, with TCF1+TRM/cDC1 pairs enriched in the stroma and TCF1- TRM/cDC1 pairs in tumor areas. Notably, these TCF1- TRMs express cytotoxic molecules and are associated with apoptotic melanoma cells. Both TCF1+ and TCF1- TRM subsets, alongside cDC1, prove relevant to CBI response.
    CONCLUSIONS: Our study supports the importance of TRM CD8 T cells and cDC1 in melanoma protection while also highlighting the existence of functionally distinctive TCF1+ and TCF1- TRM subsets, both crucial for melanoma control and CBI response.
    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
    CD8+T细胞必须在不同的肿瘤微环境中持续存在并发挥作用以发挥其作用。因此,了解常见的基本表达程序可以更好地为下一代免疫疗法提供信息.我们应用了一种可推广的矩阵分解算法,该算法将共享和上下文特定的表达程序从不同的数据集中恢复到来自132例7例人类癌症患者的33,161个CD8T细胞的单细胞RNA测序(scRNA-seq)汇编。我们的元单细胞分析揭示了一个泛癌症T细胞功能障碍程序,该程序可预测黑色素瘤对检查点阻断的临床无反应,并突出显示CXCR6作为慢性活化T细胞的泛癌症标志物。Cxcr6被AP-1反式激活并被TCF1抑制。使用鼠标模型,我们显示CD8+T细胞中的Cxcr6缺失增加PD1+TIM3+细胞的凋亡,抑制CD28信号,并危及肿瘤生长控制。我们的研究揭示了TCF1:CXCR6轴,可以抵消PD1介导的CD8细胞反应抑制,并且对于有效的抗肿瘤免疫至关重要。
    CD8+ T cells must persist and function in diverse tumor microenvironments to exert their effects. Thus, understanding common underlying expression programs could better inform the next generation of immunotherapies. We apply a generalizable matrix factorization algorithm that recovers both shared and context-specific expression programs from diverse datasets to a single-cell RNA sequencing (scRNA-seq) compendium of 33,161 CD8+ T cells from 132 patients with seven human cancers. Our meta-single-cell analyses uncover a pan-cancer T cell dysfunction program that predicts clinical non-response to checkpoint blockade in melanoma and highlights CXCR6 as a pan-cancer marker of chronically activated T cells. Cxcr6 is trans-activated by AP-1 and repressed by TCF1. Using mouse models, we show that Cxcr6 deletion in CD8+ T cells increases apoptosis of PD1+TIM3+ cells, dampens CD28 signaling, and compromises tumor growth control. Our study uncovers a TCF1:CXCR6 axis that counterbalances PD1-mediated suppression of CD8+ cell responses and is essential for effective anti-tumor immunity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    目的:探讨儿童青少年单基因糖尿病的临床特点及分子基础。
    方法:对2020年1月至2023年3月在宁波市妇女儿童医院确诊为糖尿病的116例儿童青少年的临床表现和实验室资料进行回顾性分析。对21例疑似单基因糖尿病患儿进行了全外显子组测序和线粒体基因测序。
    结果:共诊断出10例单基因糖尿病,所有这些都是成熟型年轻糖尿病(MODY)。6例MODY2是由于GCK基因突变,1例MODY3是由于HNF1A基因突变,2例MODY12是由于ABCC8基因突变,MODY131例因KCNJ11基因突变。10例MODY患者中有9例没有典型的糖尿病症状。MODY组糖尿病家族史明显高于T1DM和T2DM组(P<0.05)。MODY组BMI高于T1DM组(P<0.05)。初始血糖水平低于T1DM组(P<0.05),与T2DM组比拟差别无统计学意义。MODY组空腹C肽水平高于T1DM组(P<0.05),与T2DM组比拟差别无统计学意义。MODY组糖化血红蛋白低于T1DM和T2DM组(P<0.05)。
    结论:在这项研究中,MODY占儿童和青少年单基因糖尿病的大多数,常见的突变是与MODY2相关的GCK基因。MODY患儿血糖和糖化血红蛋白略有升高,而胰岛细胞功能仍然存在,临床表现和实验室检查与2型糖尿病患者有重叠.WES和线粒体基因测序可以明确单基因糖尿病的病因,便于精准治疗。
    OBJECTIVE: To explore the clinical characteristics and molecular basis for children and adolescents with monogenic diabetes.
    METHODS: A retrospective analysis was carried out for the clinical manifestations and laboratory data of 116 children and adolescents diagnosed with diabetes at Ningbo Women and Children\'s Hospital from January 2020 to March 2023. Whole exome sequencing and mitochondrial gene sequencing were carried out on 21 children with suspected monogenic diabetes.
    RESULTS: A total of 10 cases of monogenic diabetes were diagnosed, all of which were Maturity-onset Diabetes Of the Young (MODY). Six cases of MODY2 were due to GCK gene mutations, 1 case of MODY3 was due to HNF1A gene mutation, 2 cases of MODY12 were due to ABCC8 gene mutations, and 1 case of MODY13 was due to KCNJ11 gene mutation. Nine of the 10 patients with MODY had no typical symptoms of diabetes. A family history of diabetes was significantly more common in the MODY group compared with the T1DM and T2DM groups (P < 0.05). The BMI of the MODY group was higher than that of the T1DM group (P < 0.05). The initial blood glucose level was lower than that of the T1DM group (P < 0.05), and there was no significant difference compared with the T2DM group. The fasting C-peptide level of the MODY group was higher than that of the T1DM group (P < 0.05), and there was no significant difference compared with the T2DM group. Glycosylated hemoglobin of the MODY group was lower than both the T1DM and T2DM groups (P < 0.05).
    CONCLUSIONS: In this study, MODY has accounted for the majority of monogenic diabetes among children and adolescents, and the common mutations were those of the GCK gene in association with MODY2. Blood glucose and glycosylated hemoglobin of children with MODY were slightly increased, whilst the islet cell function had remained, and the clinical manifestations and laboratory tests had overlapped with those of type 2 diabetes. WES and mitochondrial gene sequencing can clarify the etiology of monogenic diabetes and facilitate precise treatment.
    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
    不同的自然杀伤(NK)样CD57+和PD-1+CD8+耗尽样T细胞群(Tex)都与自身免疫性1型糖尿病(T1D)患者的有益免疫疗法反应有关。这些细胞类型之间的起源和关系知之甚少。在这里,我们表明,虽然PD-1+和CD57+Tex群体在表观遗传上相似,CD57+Tex细胞显示抑制性杀伤细胞免疫球蛋白样受体(iKIR)和其他NK细胞基因的独特增加的染色质可及性。PD-1+和CD57+Tex还显示抑制性受体(IR)和iKIR的相互表达,伴随着Tcf1和Tbet转录因子靶位点的染色质可及性,分别。CD57+Tex显示未被重视的基因表达异质性,并与PD-1+Tex共享克隆关系,这些细胞沿着四个相互关联的谱系轨迹分化:Tex-PD-1+,Tex-CD57+,Tex-分支,和Tex-Fluid.我们的发现证明了人类自身免疫性疾病中Tex样人群之间的新关系,并表明调节常见的前体人群可能会增强对自身免疫性疾病治疗的反应。
    Distinct Natural Killer (NK)-like CD57+ and PD-1+ CD8+ exhausted-like T cell populations (Tex) have both been linked to beneficial immunotherapy response in autoimmune type 1 diabetes (T1D) patients. The origins and relationships between these cell types are poorly understood. Here we show that while PD-1+ and CD57+ Tex populations are epigenetically similar, CD57+ Tex cells display unique increased chromatin accessibility of inhibitory Killer Cell Immunoglobulin-like Receptor (iKIR) and other NK cell genes. PD-1+ and CD57+ Tex also show reciprocal expression of Inhibitory Receptors (IRs) and iKIRs accompanied by chromatin accessibility of Tcf1 and Tbet transcription factor target sites, respectively. CD57+ Tex show unappreciated gene expression heterogeneity and share clonal relationships with PD-1+ Tex, with these cells differentiating along four interconnected lineage trajectories: Tex-PD-1+, Tex-CD57+, Tex-Branching, and Tex-Fluid. Our findings demonstrate new relationships between Tex-like populations in human autoimmune disease and suggest that modulating common precursor populations may enhance response to autoimmune disease 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: Letter
    暂无摘要。
    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
    HNF4A和HNF1A编码对胰腺和肝脏的发育和功能重要的转录因子。这两个基因的突变都与年轻人的成年糖尿病(MODY)和2型糖尿病(T2D)风险直接相关。为了更好地定义HNF4A和HNF1A的多效性基因调控作用,我们使用ChIP-Seq生成了其在胰腺和肝细胞中的结合靶标的全面全基因组图谱。HNF4A被发现结合和调节已知(ACY3,HAAO,HNF1A,MAP3K11)和先前未识别的(ABCD3、CDKN2AIP、USH1C,VIL1)以组织依赖性方式定位。功能随访强调了HAAO和USH1C作为β细胞功能调节剂的潜在作用。与功能丧失型HNF4A/MODY1变体I271fs不同,发现T2D相关的HNF4A变体(rs1800961)激活AKAP1,GAD2和HOPX基因表达,可能是由于DNA结合亲和力的变化。我们还发现HNF1A结合并调节β细胞中的GPR39表达。总的来说,我们的研究为揭示可能有助于β细胞或肝细胞(dys)功能的HNF4A和HNF1A的下游分子靶标提供了丰富的资源,并建立了基因发现和功能验证的框架。
    HNF4A and HNF1A encode transcription factors that are important for the development and function of the pancreas and liver. Mutations in both genes have been directly linked to Maturity Onset Diabetes of the Young (MODY) and type 2 diabetes (T2D) risk. To better define the pleiotropic gene regulatory roles of HNF4A and HNF1A, we generated a comprehensive genome-wide map of their binding targets in pancreatic and hepatic cells using ChIP-Seq. HNF4A was found to bind and regulate known (ACY3, HAAO, HNF1A, MAP3K11) and previously unidentified (ABCD3, CDKN2AIP, USH1C, VIL1) loci in a tissue-dependent manner. Functional follow-up highlighted a potential role for HAAO and USH1C as regulators of beta cell function. Unlike the loss-of-function HNF4A/MODY1 variant I271fs, the T2D-associated HNF4A variant (rs1800961) was found to activate AKAP1, GAD2 and HOPX gene expression, potentially due to changes in DNA-binding affinity. We also found HNF1A to bind to and regulate GPR39 expression in beta cells. Overall, our studies provide a rich resource for uncovering downstream molecular targets of HNF4A and HNF1A that may contribute to beta cell or hepatic cell (dys)function, and set up a framework for gene discovery and functional validation.
    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T细胞在溃疡性结肠炎(UC)发病机制中的作用,我们分析了从UC患者和对照组分离的结肠T细胞.在这里,我们确定了结肠CD4+和CD8+T淋巴细胞亚群的基因表达谱类似于茎样祖细胞,先前在几种自身免疫性疾病的小鼠模型中报道。与同一患者的非发炎区域相比,发炎区域的干细胞样T细胞增加。此外,TCR序列分析显示干细胞样T细胞与促炎T细胞呈克隆相关,表明他们参与了驱动炎症的持续效应。使用小鼠过继转移结肠炎模型,我们证明了CD4+T细胞缺乏BCL-6或TCF1,转录因子,促进T细胞的干细胞,结肠T细胞减少,致病性减弱。我们的结果建立了干细胞样T细胞群和UC发病机制之间的强关联。强调了针对该人群改善临床结局的潜力。
    To understand the role of T cells in the pathogenesis of ulcerative colitis (UC), we analyzed colonic T cells isolated from patients with UC and controls. Here we identified colonic CD4+ and CD8+ T lymphocyte subsets with gene expression profiles resembling stem-like progenitors, previously reported in several mouse models of autoimmune disease. Stem-like T cells were increased in inflamed areas compared to non-inflamed regions from the same patients. Furthermore, TCR sequence analysis indicated stem-like T cells were clonally related to proinflammatory T cells, suggesting their involvement in sustaining effectors that drive inflammation. Using an adoptive transfer colitis model in mice, we demonstrated that CD4+ T cells deficient in either BCL-6 or TCF1, transcription factors that promote T cell stemness, had decreased colon T cells and diminished pathogenicity. Our results establish a strong association between stem-like T cell populations and UC pathogenesis, highlighting the potential of targeting this population to improve clinical outcomes.
    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