LCP1

LCP1
  • 文章类型: Journal Article
    目的:三阴性乳腺癌(TNBC)因缺乏ER而具有侵袭性和治疗挑战,PR,和HER2受体。我们的工作强调LCP1(淋巴细胞胞浆蛋白1)的预后价值,在细胞过程和免疫细胞活动中起着至关重要的作用,预测结果并指导TNBC的治疗。
    方法:我们探索了LCP1作为TNBC的潜在生物标志物,并研究了LCP1的mRNA和蛋白质表达水平。我们调查了不同的数据库,包括GTEX,TCGA,GEO,cBioPortal和Kaplan-Meier绘图仪。对TNBC和良性肿瘤样本进行免疫组织化学检查,以检查LCP1与患者临床特征和巨噬细胞标记的关系。我们还评估了生存率,免疫细胞浸润,以及使用各种生物信息学工具与LCP1相关的药物敏感性。
    结果:结果表明,与邻近的正常组织相比,TNBC组织中LCP1的表达更高。然而,LCP1的高表达与TNBC患者的良好生存结局显著相关.富集分析显示与LCP1共表达的基因在各种免疫过程中显著富集。LCP1与静息树突状细胞浸润呈正相关,M1巨噬细胞,和记忆CD4T细胞,与M2巨噬细胞呈负相关。进一步的分析表明,高水平的LCP1与接受免疫治疗的癌症患者的生存结果增加之间存在联系。
    结论:LCP1可作为TNBC的潜在诊断和预后生物标志物,这与免疫细胞浸润密切相关,特别是M1和M2巨噬细胞。我们的发现可能为TNBC患者的免疫治疗策略提供有价值的见解。
    OBJECTIVE: Triple-Negative Breast Cancer (TNBC) is known for its aggressiveness and treatment challenges due to the absence of ER, PR, and HER2 receptors. Our work emphasizes the prognostic value of LCP1 (Lymphocyte cytosolic protein 1), which plays a crucial role in cell processes and immune cell activity, to predict outcomes and guide treatments in TNBC.
    METHODS: We explored LCP1 as a potential biomarker in TNBC and investigated the mRNA and protein expression levels of LCP1. We investigated different databases, including GTEX, TCGA, GEO, cBioPortal and Kaplan-Meier Plotter. Immunohistochemistry on TNBC and benign tumor samples was performed to examine LCP1\'s relationship with patient clinical characteristics and macrophage markers. We also assessed survival rates, immune cell infiltration, and drug sensitivity related to LCP1 using various bioinformatics tools.
    RESULTS: The results indicated that LCP1 expression was higher in TNBC tissues compared to adjacent normal tissues. However, high expression of LCP1 was significantly associated with favorable survival outcomes in patients with TNBC. Enrichment analysis revealed that genes co-expressed with LCP1 were significantly enriched in various immune processes. LCP1 showed a positive correlation with the infiltration of resting dendritic cells, M1 macrophages, and memory CD4 T cells, and a negative correlation with M2 macrophages. Further analysis suggested a link between high levels of LCP1 and increased survival outcomes in cancer patients receiving immunotherapy.
    CONCLUSIONS: LCP1 may serve as a potential diagnostic and prognostic biomarker for TNBC, which was closely associated with immune cell infiltration, particularly M1 and M2 macrophages. Our findings may provide valuable insights into immunotherapeutic strategies for TNBC patients.
    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
    持续砷暴露与前列腺癌之间的联系已经建立。然而,砷肿瘤发生的确切机制尚不清楚。这里,我们使用人前列腺基质永生化细胞(WPMY-1)连续暴露于1和2μM亚砷酸盐29周来鉴定恶性表型并探索潜在的分子机制。不出所料,持续低剂量亚砷酸盐暴露导致WPMY-1细胞的恶性表型。定量蛋白质组学鉴定了517种差异表达蛋白(DEP),其中变化最显著的蛋白质(如LCP1和DDX58等。)和生物信息学分析集中在细胞骨架的调节上,细胞粘附,和移民。Further,细胞实验表明,持续的亚砷酸盐暴露改变了细胞骨架结构,增强细胞粘附能力,并提高了活性氧(ROS)的水平,ATM,p-ATM,p-ERK1/2和LCP1蛋白。N-乙酰半胱氨酸(NAC)治疗拮抗LCP1蛋白的增加,LCP1敲低部分恢复了砷引起的F-肌动蛋白组织。总的来说,结果表明,ROS-ATM-ERK1/2信号通路参与了LCP1的激活,导致细胞骨架改变。这些改变被认为在亚砷酸盐触发的肿瘤微环境细胞获得的恶性表型中起重要作用。这可能提供潜在的生物标志物与前列腺癌的治疗意义。
    The connection between continuous arsenic exposure and prostate cancer is already established. However, the exact mechanisms of arsenic tumorigenesis are far from clear. Here, we employed human prostate stromal immortalized cells (WPMY-1) continuous exposure to 1 and 2 μM arsenite for 29 weeks to identify the malignant phenotype and explore the underlying molecular mechanism. As expected, continuous low-dose arsenite exposure led to the malignant phenotype of WPMY-1 cells. Quantitative proteomics identified 517 differentially expressed proteins (DEPs), of which the most remarkably changed proteins (such as LCP1 and DDX58, etc.) and the bioinformatic analysis were focused on the regulation of cytoskeleton, cell adhesion, and migration. Further, cell experiments showed that continuous arsenite exposure altered cytoskeleton structure, enhanced cell adhesive capability, and raised the levels of reactive oxygen species (ROS), ATM, p-ATM, p-ERK1/2, and LCP1 proteins. N-acetylcysteine (NAC) treatment antagonized the increase of LCP1 proteins, and LCP1 knockdown partially restored F-actin organization caused by arsenic. Overall, the results demonstrated that ROS-ATM-ERK1/2 signaling pathway was involved in the activation of LCP1, leading to cytoskeleton alterations. These alterations are believed to play a significant role in arsenite-triggered tumor microenvironment cell-acquired malignant phenotype, which could provide potential biomarkers with therapeutic implications for prostate 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
    脑梗死(CI),也被称为缺血性中风,具有较高的发病率和死亡率。本研究的目的是探讨淋巴细胞胞浆蛋白1(LCP1)在CI进展中的潜在作用和机制。采用大脑中动脉阻塞(MCAO)处理的大鼠和氧葡萄糖剥夺/复氧(OGD/R)刺激的PC12细胞建立体内和体外CI模型。CCK-8法和流式细胞术检测细胞增殖和凋亡,分别。进行免疫沉淀和蛋白质印迹以测试LCP1的乳酸化水平。用环己酰亚胺处理细胞以测定LCP1的蛋白质稳定性。用商业试剂盒测定葡萄糖摄取和乳酸盐产生。通过海马评估细胞外酸化速率。结果表明,LCP1在MCAO大鼠中上调,OGD/R刺激PC12细胞。LCP1敲低可显著降低神经评分,MCAO大鼠的梗死体积和脑含水量。此外,LCP1敲低可促进OGD/R刺激的PC12细胞的活力,降低细胞凋亡率。此外,在脑梗死患者体内和体外,LCP1的整体乳酸化和乳酸化水平显著增强.2-DG治疗显着降低了它。总之,抑制糖酵解降低了LCP1的乳酸化水平,并导致LCP1降解,最终缓解了CI进展。
    Cerebral infarction (CI), also known as ischemic stroke, has a high incidence rate and mortality rate. The purpose of this study was to investigate the potential effect and mechanism of Lymphocyte cytosolic protein 1 (LCP1) in the CI progression. The middle cerebral artery occlusion (MCAO) treated rats and oxygen-glucose deprivation/reoxygenation (OGD/R) stimulated PC12 cells were used to establish CI model in vivo and in vitro. The cell proliferation and apoptosis was determined by CCK-8 assay and flow cytometry, respectively. Immunoprecipitation and western blot was performed to test the lactylation levels of LCP1. The cells were treated with cycloheximide to determined the protein stability of LCP1. The glucose uptake and lactate production was determined with commercial kits. The extracellular acidification rate were evaluated by Seahorse. The results showed that LCP1 was upregulated in the MCAO rats and OGD/R stimulated PC12 cells. LCP1 knockdown dramatically decreased the neurological score, infarct volume and the brain water content of MCAO rats. Besides, LCP1 knockdown promoted the cell viability while decreased the apoptosis rate of the OGD/R stimulated PC12 cells. Additionally, the global lactylation and lactylation levels of LCP1 was prominently enhanced in vivo and in vitro in cerebral infarction. 2-DG treatment prominently decreased it. In conclusion, inhibiting the glycolysis decreased the lactylation levels of LCP1 and resulted in the degradation of LCP1, which eventually relieved the CI progression.
    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:结直肠癌是全球第三大流行癌症。右侧结肠癌患者的预后通常比患有左侧结肠癌的患者差。对于左侧结肠癌与右侧结肠癌在肿瘤浸润免疫细胞(TIIC)和相关基因方面的差异,目前尚不清楚。
    方法:癌症基因组图谱提供了关于结肠腺癌的RNA-seq数据和临床信息。我们进行了单样品基因集富集分析(ssGSEA)以量化浸润组织的24种免疫细胞的水平。基于单变量Cox回归分析,确定了与存活相关的免疫细胞类型.加权基因共表达网络分析(WGCNA)用于鉴定与位置和关键免疫细胞相关的枢纽基因。基于检索相互作用基因的搜索工具(STRING),预测了hub基因之间的相互作用潜力。使用最小绝对收缩和选择算子(LASSO)鉴定通过免疫浸润影响结果的集线器基因。然后,我们使用TISIDB数据库(肿瘤-免疫系统相互作用的存储门户)来验证hub基因与免疫细胞浸润之间的相关性.最后,进行免疫组织化学测定以确定关键TIIC和癌细胞表达的蛋白质水平。
    结果:身体右侧的结肠癌比左侧的结肠癌具有更高水平的骨髓来源的抑制细胞(MDSC)。有三个关键基因:LCP1,ITGB2和IKZF1。发现它们的表达与不良预后和MDSC浸润水平增加有关。免疫组织化学研究证实了这些发现。
    结论:与左侧结肠癌相比,右侧结肠癌的MDSC浸润率更高。COAD结果与MDSC渗透的变化相关,因此LCP1、ITGB2和IKZF1可能成为免疫治疗的新靶点。
    BACKGROUND: Colorectal cancer is the third most prevalent cancer worldwide. A right-sided colon cancer patient typically has a worse prognosis than one who has a left-sided colon cancer. There is an unclear understanding of how left-sided colon cancer differs from right-sided colon cancer in tumor-infiltrating immune cells (TIICs) and relevant genes.
    METHODS: The Cancer Genome Atlas provided RNA-seq data and clinical information regarding colon adenocarcinoma. We conducted a single-sample gene set enrichment analysis (ssGSEA) to quantify the level of 24 immune cells infiltrating the tissues. Based on an analysis of univariate Cox regression, immune cell types associated with survival were identified. Weighted gene co-expression network analysis (WGCNA) was used to identify hub genes related to location and critical immune cells. Based on the Search Tool for the Retrieval of Interacting Genes (STRING), interaction potential was predicted among the hub genes. Hub genes that influence outcomes through immune infiltration were identified using the least absolute shrinkage and selection operator (LASSO). Then, we used the TISIDB database (a repository portal for tumor-immune system interactions) to validate the correlation between hub genes and immune cell infiltration. Finally, immunohistochemical assays were conducted to determine the levels of proteins expressed by critical TIICs and cancer cells.
    RESULTS: Colon cancers on the right side of the body had higher levels of myeloid-derived suppressor cells (MDSCs) than on the left side. There were three key genes: LCP1, ITGB2, and IKZF1. It was found that their expression was linked to poor prognosis and an increased level of MDSC infiltration. An immunohistochemical study confirmed these findings.
    CONCLUSIONS: There is a higher rate of MDSC infiltration in right-sided colon cancer when compared with left-sided colon cancer. COAD outcomes are associated with changes in MDSC infiltration, and therefore LCP1, ITGB2, and IKZF1 may be novel targets for immunotherapy.
    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:胃癌(GC)仍然是一个重要的健康问题,并伴随着大量的发病率和死亡率。Chidamide是一种新型的口服组蛋白去乙酰化酶(HDAC)抑制剂,已被证明对不同类型的血液和实体瘤具有抗肿瘤作用。然而,西达本胺耐药的潜在机制仍未得到充分表征。
    方法:我们建立了西达丁胺抗性GC细胞系,AGSChiR和MGC803ChiR,并研究了通过细胞存活的毒理学效应,体外集落形成和流式细胞术测定,和体内皮下异种移植模型。然后进行RNA序列以筛选AGS和AGSChiR细胞之间的西达本胺抗性相关基因。通过功能的增益和丧失分析探讨了淋巴细胞胞浆蛋白1(LCP1)在西达本胺抗性中的作用。
    结果:我们发现,与西达胺抗性细胞系相比,野生型GC细胞系中的西达胺显着抑制细胞增殖并以浓度依赖性方式诱导细胞凋亡。转录组分析,定量RT-PCR,蛋白质印迹数据显示,与亲本细胞相比,LCP1在AGSChiR细胞中上调。LCP1的过表达赋予和敲低LCP1减弱了GC细胞的西达胺抗性。西达胺对LCP1的表观遗传抑制可能是LCP1对西达胺抗性的贡献的可能原因。
    结论:这些发现表明LCP1可能在GC中发挥西达本胺抗性作用,提示LCP1可能是GC联合西达胺治疗的潜在靶点。
    BACKGROUND: Gastric cancer (GC) remains a significant health problem and carries with it substantial morbidity and mortality. Chidamide is a novel and orally administered histone deacetylase (HDAC) inhibitor and has been demonstrated its anti-tumor efficacy on different kinds of hematological and solid tumors. However, the underlying mechanism of chidamide resistance is still poorly characterized.
    METHODS: We established chidamide resistant GC cell lines, AGS ChiR and MGC803 ChiR and investigated the toxicologic effects through cell survival, colony formation and flow cytometry assays in vitro, and a subcutaneous xenograft model in vivo. RNA-sequence was then performed to screen chidamide resistance-associated genes between AGS and AGS ChiR cells. The role of Lymphocyte cytosolic protein 1 (LCP1) in chidamide resistance was explored by gain- and loss-of-function analyses.
    RESULTS: We found that chidamide significantly inhibited cell proliferation and induced the apoptosis in a concentration-dependent manner in wild-type GC cell lines as compared to chidamide resistant cell lines. The transcriptomic profiling, quantitative RT-PCR, and western blot data revealed that LCP1 was upregulated in AGS ChiR cells compared with parental cells. Overexpression of LCP1 conferred and knockdown of LCP1 attenuated the chidamide resistance of GC cells. Epigenetic derepression of LCP1 by chidamide may be a possible reason for the contribution of LCP1 to chidamide resistance.
    CONCLUSIONS: These findings illustrated that LCP1 may play a chidamide resistance role in GC, suggesting that LCP1 could be a potential target for the therapy of GC combined with chidamide.
    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
    发炎的滑膜在关节破坏中起主要作用,其特征在于免疫细胞浸润和成纤维细胞增殖。这项蛋白质组学研究通过分析滑膜活检在整个不同关节炎关节疾病中呈现组织学炎症连续体,在分子水平上考虑了炎症过程。膝关节滑膜活检来自骨关节炎(OA;n=9),慢性焦磷酸盐关节病(CPPA;n=7)或类风湿性关节炎(RA;n=8)患者。使用基于滑膜增生的半定量量表确定组织学炎症评分,淋巴细胞,浆细胞,中性粒细胞和巨噬细胞浸润。通过液相色谱-质谱(LC-MS/MS)进行蛋白质组分析。通过免疫组织化学证实差异表达的蛋白质。在通过LC-MS/MS鉴定和定量的1871种蛋白质中,10种蛋白质(LAP3,MANF,LCP1,CTSZ,PTPRC,DNAJB11,EML4,SCARA5,EIF3K,C1orf123)在三个疾病组中的至少一个的滑膜中差异表达(RA,OA和CPPA)。在RA中检测到七种第一蛋白质的表达显着增加,并与组织学炎症评分相关。因此,蛋白质组学是一种强大的工具,可以为通常用于滑膜炎表征的经典组织学提供分子模式。除LCP1、CTSZ和PTPRC外,从未在人类滑膜炎中描述过所有蛋白质。
    An inflamed synovial membrane plays a major role in joint destruction and is characterized by immune cells infiltration and fibroblast proliferation. This proteomic study considers the inflammatory process at the molecular level by analyzing synovial biopsies presenting a histological inflammatory continuum throughout different arthritis joint diseases. Knee synovial biopsies were obtained from osteoarthritis (OA; n = 9), chronic pyrophosphate arthropathy (CPPA; n = 7) or rheumatoid arthritis (RA; n = 8) patients. The histological inflammatory score was determined using a semi-quantitative scale based on synovial hyperplasia, lymphocytes, plasmocytes, neutrophils and macrophages infiltration. Proteomic analysis was performed by liquid chromatography-mass spectrometry (LC-MS/MS). Differentially expressed proteins were confirmed by immunohistochemistry. Out of the 1871 proteins identified and quantified by LC-MS/MS, 10 proteins (LAP3, MANF, LCP1, CTSZ, PTPRC, DNAJB11, EML4, SCARA5, EIF3K, C1orf123) were differentially expressed in the synovial membrane of at least one of the three disease groups (RA, OA and CPPA). Significant increased expression of the seven first proteins was detected in RA and correlated to the histological inflammatory score. Proteomics is therefore a powerful tool that provides a molecular pattern to the classical histology usually applied for synovitis characterization. Except for LCP1, CTSZ and PTPRC, all proteins have never been described in human synovitis.
    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
    Increased browning of white adipocytes (beiging) is considered a promising therapeutic strategy to fight obesity and its associated metabolic complications. However, the molecular mechanism modulating brown and beige fat-mediated thermogenesis is not fully elucidated. Here, we identified the lymphocyte cytosolic protein 1 (LCP1) as a factor that obstructs fat browning in white adipocytes. LCP1 plays a vital role in non-hematopoietic malignancies, and is also a well-known tumor biomarker; however, evidence regarding its function in adipocytes remains to be elucidated. The current study explores the physiological role of LCP1 in cultured 3T3-L1 white adipocytes, by applying the loss-of-function study using siRNA. Induction of fat browning by LCP1 depletion was evidenced by evaluating the gene and protein expression levels of brown fat-associated markers through real-time qRT-PCR and immunoblot analysis, respectively. We observed that deficiency of LCP1 promotes mitochondrial biogenesis, and significantly enhances expressions of the core brown fat-specific genes (Cd137, Cidea, Cited1, Tbx1, and Tmem26) and proteins (PGC-1α, PRDM16, and UCP1). In addition, deficiency of LCP1 promotes lipid catabolism as well as suppresses adipogenesis and lipogenesis. Loss of LCP1 also ameliorates cellular stress by downregulating JNK and c-JUN in adipocytes, and stimulates apoptosis. A mechanistic study revealed that deficiency of LCP1 induces browning in white adipocytes, independently via β3-AR and the ERK signaling pathway. The current data reveals a previously unknown mechanism of LCP1 in browning of white adipocytes, and highlights the potential of LCP1 as a pharmacotherapeutic target for treating obesity and other metabolic disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    BACKGROUND: Previous studies have identified LCP1 as a diagnostic and prognostic marker in several cancers. However, the role of LCP1 in gastric cancer (GC) and its effect on tumor immune infiltration remain unclear.
    OBJECTIVE: The aim was to explore the role of LCP1 in GC and its effect on tumor immune infiltration.
    METHODS: We explored the expression of LCP1 relative to clinicopathology in GC patients by bioinformatics analysis and immunohistochemistry. Using cBioportal database, we analyzed the characteristic genetic variations of LCP1 in GC. In addition, we evaluated the correlation between LCP1 expression and tumor-infiltrating lymphocytes (TILs) using R software, TIMER and TISIDB databases. Finally, we analyzed the biological functions in which LCP1 may participate and the signaling pathways it may regulate.
    RESULTS: Here, we showed that LCP1 expression is significantly correlated with tumor aggressiveness and poor prognosis in GC patients. Additionally, the results indicated that LCP1 was associated with TILs, including both immunosuppressive and immunosupportive cells, and was strongly correlated with various immune marker sets in GC. GSEA analysis demonstrated that LCP1 expression played an important role in lymphocyte formation and immune reaction.
    CONCLUSIONS: LCP1 may be a potential prognostic biomarker for GC patients and a marker for tumor immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    肌动蛋白细胞骨架成束和网络的动态组织是由多种肌动蛋白结合蛋白协调的。其中,肌动蛋白束蛋白L-plastin通常在造血细胞中表达,它参与了免疫反应。然而,L-激酶也经常在非造血起源的恶性癌细胞中异位表达,并且甚至被认为是癌症进展的标志物。翻译后修饰调节L-激酶活性。特别是,L-plastinSer5磷酸化已被证明对于白细胞中的免疫应答以及癌细胞的侵袭和转移形成是重要的。本章讨论了L-plastin的生理和病理作用,特别关注L-plastinSer5磷酸化对蛋白质功能的重要性。将引发Ser5磷酸化L-激酶作为生物标志物和/或治疗靶标的潜在用途。
    The dynamic organization of the actin cytoskeleton into bundles and networks is orchestrated by a large variety of actin-binding proteins. Among them, the actin-bundling protein L-plastin is normally expressed in hematopoietic cells, where it is involved in the immune response. However, L-plastin is also often ectopically expressed in malignant cancer cells of non-hematopoietic origin and is even considered as a marker for cancer progression. Post-translational modification modulates L-plastin activity. In particular, L-plastin Ser5 phosphorylation has been shown to be important for the immune response in leukocytes as well as for invasion and metastasis formation of carcinoma cells. This chapter discusses the physiological and pathological role of L-plastin with a special focus on the importance of L-plastin Ser5 phosphorylation for the protein functions. The potential use of Ser5 phosphorylated L-plastin as a biomarker and/or therapeutic target will be evoked.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Hypereosinophilia (HE) is caused by a variety of disorders, ranging from parasite infections to autoimmune diseases and cancer. Only a small proportion of HE cases are clonal malignancies, and one of these, the group of eosinophilia-associated tyrosine kinase fusion-driven neoplasms, is sensitive to tyrosine kinase inhibitors, while most subtypes lack specific treatment. Eosinophil functions are highly dependent on actin polymerization, promoting priming, shape change, and infiltration of inflamed tissues. Therefore, we investigated the role of the actin-binding protein lymphocyte cytosolic protein 1 (LCP1) in malignant and nonmalignant eosinophil differentiation. We use the protein kinase C-β (PKCβ) selective inhibitor enzastaurin (Enza) to dephosphorylate and inactivate LCP1 in FIP1L1-platelet-derived growth factor receptor α (PDGFRA)-positive Eol-1 cells, and this was associated with reduced proliferation, metabolic activity, and colony formation as well as enhanced apoptosis and impaired migration. While Enza did not alter FIP1L1-PDGFRA-induced signal transducer and activator of transcription 3 (STAT3), STAT5, and ERK1/2 phosphorylation, it inhibited STAT1Tyr701 and AKTSer473 (but not AKTThr308 ) phosphorylation, and short hairpin RNA knockdown experiments confirmed that this process was mediated by LCP1 and associated mammalian target of rapamycin complex 2 (mTORC2) activity loss. Homeobox protein HoxB8 immortalized murine bone marrow cells showed impaired eosinophilic differentiation upon Enza treatment or LCP1 knockdown. Furthermore, Enza treatment of primary HE samples reduced eosinophil differentiation and survival. In conclusion, our data show that HE involves active LCP1, which interacts with mTOR and triggers mTORC2 activity, and that the PKCβ inhibitor Enza as well as targeting of LCP1 may provide a novel treatment approach to hypereosinophilic disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号