首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到19条相似文献,搜索用时 218 毫秒
1.
2.
马宁  赵砚  康续  潘珊 《病毒学报》2021,37(1):153-158
由于具有相同的传播途径,人类免疫缺陷病毒(Human immunodeficiency virus,HIV)和丙型肝炎病毒(Hepatitis C virus,HCV)共感染非常普遍,但是关于合并感染的程度,两种病毒之间的相互关系,在艾滋病抗逆转录病毒治疗(Antiretroviral therapy,ART)前后,HCV合并感染对HIV患者免疫细胞恢复的影响仍不明确。为了通过分析CD4+和CD8+T淋巴细胞数的变化,以了解辽宁省HIV/HCV共感染者ART后免疫恢复的情况,本研究从辽宁省艾滋病抗病毒治疗数据库中筛选符合要求的HIV感染者和HIV/HCV共感染者,收集感染者基本人口学资料及HCV抗体检测结果、HIV/HCV共感染途径等资料。采用t检验或卡方检验进行组间比较,采用Kaplan-Meier乘积极限法绘制生存分析函数图。结果显示,本研究共纳入HIV感染者12742人,HIV/HCV共感染者340人。HIV感染者和HIV/HCV共感染者的不同人口学特征均差异显著(P<0.001)。HIV感染和HIV/HCV共感染者ART治疗后CD4+细胞数和CD4+/CD8+比值显著升高(P<0.05),CD8+细胞数比ART前显著下降(P<0.05)。HIV/HCV共感染者随着ART时长,CD4+T淋巴细胞数恢复情况始终显著低于HIV感染者(P<0.05)。生存分析曲线表明,HCV/HIV共感染者从艾滋病诊断开始随着ART的治疗CD4+细胞恢复情况显著低于HIV感染者,Log-Rank检验统计量为4.483(P=0.034)。本研究揭示,HCV感染对ART患者CD4+和CD8+T淋巴细胞的恢复有影响。ART后HIV/HCV共感染者中CD4+T淋巴细胞计数的改善低于HIV单一感染者,并且单一感染患者对ART的反应比合并感染患者更好。因此,建议在启动ART之前,对每个感染HIV的患者进行HCV抗体筛查。  相似文献   

3.
目的:探讨扶正化瘀胶囊联合微生态制剂和恩替卡韦治疗乙型肝炎病毒(HBV)感染失代偿期肝硬化的临床效果。方法:选取2015年6月~2019年6月期间我院收治的HBV感染失代偿期肝硬化患者73例,根据随机数字表法分为对照组(n=36)和研究组(n=37),对照组患者予以微生态制剂和恩替卡韦治疗,研究组则在对照组的基础上联合扶正化瘀胶囊治疗,比较两组患者疗效、T淋巴细胞亚群、肝功能[天冬氨酸氨基转移酶(AST)、总胆红素(TBIL)]以及肝纤维化指标[透明质酸(HA)、层黏连蛋白(LN)],记录两组乙肝表面e抗原(HBeAg)、乙肝病毒基因(HBV-DNA)转阴情况,记录两组不良反应发生情况。结果:研究组治疗6个月后的临床总有效率为91.89%(34/37),高于对照组的72.22%(26/36)(P0.05)。两组治疗6个月后AST、TBIL、HA、LN、CD8+~均下降,且研究组低于对照组(P0.05)。两组治疗6个月后CD4+~、CD4+~/CD8+~升高,且研究组高于对照组(P0.05)。两组治疗6个月后HBeAg、HBV-DNA转阴率比较无统计学差异(P0.05)。两组不良反应发生率对比未见统计学差异(P0.05)。结论:扶正化瘀胶囊联合微生态制剂和恩替卡韦治疗HBV感染失代偿期肝硬化疗效显著,虽在HbeAg、HBV-DNA转阴率方面未见明显改善,但可有效改善肝功能,减轻肝纤维化,提高机体免疫功能,且不增加不良反应发生率,安全性较好。  相似文献   

4.
目的检测传染性单核细胞增多症(IM)患者及非本病的发热患者外周血T淋巴细胞亚群,探讨EB病毒感染导致的传染性单核细胞增多症的免疫反应机制及与非本病发热患者的鉴别诊断价值,同时通过检测患者血液中的EBV-DNA载量加以确证。方法选取确诊为传染性单核细胞增多症的患者30例(IM组),非传染性单核细胞增多症的发热患者30例(对照组)。应用流式细胞技术检测患者外周血中淋巴细胞亚群,同时应用荧光定量PCR技术检测血清EBV-DNA载量,采用卡方检验及Mann-Whitney检验进行组间差异分析。结果 IM组与对照组患者T淋巴细胞亚群相比,CD8~+T淋巴细胞所占比例显著升高(Z=1.776,P0.001),CD4~+T淋巴细胞所占比例降低(F=4.008,P0.050),同时CD4~+/CD8~+比例明显下降(Z=6.653,P0.001)。IM组EBV-DNA病毒载量显著高于对照组,差异具有统计学意义(P0.050)。结论传染性单核细胞增多症患者外周血T淋巴细胞亚群主要以CD4~+T淋巴细胞及CD8~+T淋巴细胞的变化为主,T淋巴细胞亚群结合血清EBV-DNA病毒载量的联合检测对IM及非本病的发热患者具有鉴别诊断价值。  相似文献   

5.
目的探讨慢性丙型肝炎(HCV)患者CD4+CD25+Treg细胞对外周血树突状细胞的作用。方法对35例HCV患者和35例健康对照各抽取外周血,采用密度梯度离心法分离外周血单个核细胞(PBMC),采用特异性免疫磁珠分选获得CD4+CD25+Treg细胞,并体外诱导培养获得树突状细胞(DCs);将CD4+CD25+Treg细胞与DC共培养5d后,采用流式细胞仪检测DC表面标志CD83、CD80、HLADR的表达,同时酶联免疫吸附法检测上清液中IL-10和TGF-β含量。结果与健康对照组相比,HCV患者CD83、CD80和HLA-DR的表达均显著下降,差异有统计学意义(P0.01);HCV组患者CD4+CD25+Treg分泌IL-10和TGF-β的水平均高于健康组(P0.01)。结论 HCV患者外周血Treg细胞能够抑制DC的成熟,细胞因子参与了免疫应答的调节。  相似文献   

6.
目的:检测系统性红斑狼疮(systemic lupus erythematosus,SLE)合并带状疱疹患者外周血CD4~+CD28~+和CD4~+CD25~+Fox P3~+调节性T细胞的表达及相关性,探讨其在SLE合并带状疱疹发病中的临床意义。方法:采用流式细胞术检测30例SLE患者、30例SLE合并带状疱疹患者及30例健康对照者外周血中CD4~+/CD8~+T淋巴细胞亚群表面CD28的表达及CD4~+CD25~+Fox P3~+Treg细胞的表达水平,并分析SLE合并带状疱疹患者外周血CD4~+CD28~+和CD4~+CD25~+Fox P3~+调节性T细胞表达的相关性。结果:SLE合并带状疱疹组患者急性期外周血CD4~+T淋巴细胞比率、绝对计数显著降低,CD4~+、CD8~+T淋巴细胞表面的CD28表达下调,CD4~+CD25~+Fox P3~+Treg细胞水平显著高于SLE组及健康对照组,SLE合并带状疱疹组患者外周血CD4~+CD25~+Fox P3~+Treg水平与CD4~+CD28~+水平成负相关(P均0.05)。结论:SLE合并带状疱疹患者CD4~+、CD8~+T细胞活化异常,CD4~+CD25~+Fox P3~+Treg细胞可能参与抑制了T细胞的活化。  相似文献   

7.
目的:探讨人类免疫缺陷病毒(HIV)感染患者血清白细胞介素-2(IL-2)、白细胞介素-16(IL-16)水平和CD4细胞计数的相关性。方法:选择2017年3月至2018年3月我院接诊的50例HIV感染患者作为观察组及同期于我院进行体检的健康人群40例作为对照组。检测和比较两组血清IL-2、IL-16与CD4细胞计数,并分析观察组血清IL-2、IL-16与CD4细胞计数的相关性。结果:观察组患者血清IL-2、IL-16水平及CD4细胞计数显著低于对照组,差异具有统计学意义(P0.05);Pearson相关分析结果显示观察组患者血清IL-2、IL-16水平与CD4细胞计数呈显著正相关(r=0.514,r=0.499,P0.05)。结论:HIV感染患者血清IL-2、IL-16水平显著下调,并与CD4细胞计数呈显著正相关,可在一定程度上反映HIV感染的严重度。  相似文献   

8.
目的:研究急性冠脉综合征(ACS)患者CD4+T淋巴细胞亚群的变化及临床意义。方法:收集2013年3月-2014年3月入住我院首次行冠脉成形术(PCI)治疗的ACS患者88例以及造影结果正常患者28例。分别于冠脉造影时取冠脉血流式细胞技术检测CD4+T细胞亚群Th1/Th2、Th17/Treg比例表达变化。结果:与对照组相比,ACS组患者冠脉血中Th1、Th17细胞占CD4+T淋巴细胞的百分比显著升高,而Th1、Treg细胞占CD4+T淋巴细胞的百分比显著降低,差异具有统计学意义(P0.05)。结论:ACS患者体内免疫反应增强,CD4+T淋巴细胞不同功能亚群均参与了动脉粥样硬化(AS)的发生发展,Th1、Th17可促进AS的进展和不稳定性,而Th1、Treg作用相反。  相似文献   

9.
目的分析人类免疫缺陷病毒(Human immunodeficiency virus,HIV)阴性隐球菌性脑膜炎(Cryptococcal meningitis,CM)患者外周血淋巴细胞亚群改变特点,深化对隐球菌病发病机制的认识。方法筛选HIV阴性CM患者31例和健康献血员21例进行淋巴细胞亚群分析,CM患者根据是否存在免疫抑制基础疾病分为两个亚组,比较3组研究对象淋巴细胞亚群中B、NK、CD_4~+T、CD_8~+T细胞亚群,CD_4~+T及CD_8~+T细胞的第二信号受体CD28表达比例及CD_8~+T细胞激活标记物HLA-DR、CD38的表达水平。结果有基础病CM组B、NK、CD_4~+T和CD_8~+T 4种亚群中位数依次为56×10~6/L、86×10~6/L、218×10~6/L、164×10~6/L,显著低于健康对照组的223×10~6/L、280×10~6/L、695×10~6/L、521×10~6/L(P值均0.001),同时存在CD_8~+T细胞激活亚群比例较健康对照显著升高。无基础病CM患者上述4种细胞亚群中位数依次为128×10~6/L、128×10~6/L、567×10~6/L、527×10~6/L,除CD_8~+T细胞计数水平与健康对照相似以外,B(P=0.02)、NK(P=0.002)和CD_4~+T细胞(P=0.03)计数均低于健康对照,其CD_8~+T细胞激活亚群比例与健康对照组相似。CD_4~+T和CD_8~+T细胞的CD28表达水平在3组间未见显著差异。结论 HIV阴性CM患者,无论是否存在免疫抑制基础疾病,外周血B、NK及CD_4~+T 3种细胞计数可出现同时减少。  相似文献   

10.
目的:探讨人腺病毒55型感染肺部病变与外周血淋巴细胞改变的关系及致病意义。方法:以50例经胸部CT证实为腺病毒肺炎的患者为研究对象(肺炎组),调查其外周血细胞及T淋巴细胞亚群变化情况,并与同期腺病毒55型感染未出现肺部病变的患者30例(非肺炎组)对照;亚组分析50例肺炎组中多肺叶病变与单肺叶病变患者之间T淋巴细胞变化的差异。结果:与非肺炎组患者相比,肺炎组患者急性期外周血淋巴细胞比例明显降低、单核细胞比例明显升高(P0.01);肺炎组CD3+T淋巴细胞比例、CD3+CD4+T淋巴细胞比例及CD4/CD8比值较非肺炎组均有明显降低(P0.01)。亚组分析显示,肺炎组患者肺部病变范围不同,T淋巴细胞亚群的变化亦有差异,多肺叶病变组患者CD3+CD8+T淋巴细胞比例较单肺叶病变组明显升高、CD4/CD8比值较单肺叶病变组明显降低(P0.05)。结论:HAdv-55感染引起肺炎病变时,宿主存在明显的细胞免疫功能受损,且与肺部病变程度有一定正相关。  相似文献   

11.
The hepatitis B virus (HBV) and the human immunodeficiency virus type 1 (HIV-1) can infect cells of the lymphatic system. It is unknown whether HIV-1 co-infection impacts infection of peripheral blood mononuclear cell (PBMC) subsets by the HBV. Aims To compare the detection of HBV genomes and HBV sequences in unsorted PBMCs and subsets (i.e., CD4+ T, CD8+ T, CD14+ monocytes, CD19+ B, CD56+ NK cells) in HBV mono-infected vs. HBV/HIV-1 co-infected individuals. Methods Total PBMC and subsets isolated from 14 HBV mono-infected (4/14 before and after anti-HBV therapy) and 6 HBV/HIV-1 co-infected individuals (5/6 consistently on dual active anti-HBV/HIV therapy) were tested for HBV genomes, including replication indicative HBV covalently closed circular (ccc)-DNA, by nested PCR/nucleic hybridization and/or quantitative PCR. In CD4+, and/or CD56+ subsets from two HBV monoinfected cases, the HBV polymerase/overlapping surface region was analyzed by next generation sequencing. Results All analyzed whole PBMC from HBV monoinfected and HBV/HIV coinfected individuals were HBV genome positive. Similarly, HBV DNA was detected in all target PBMC subsets regardless of antiviral therapy, but was absent from the CD4+ T cell subset from all HBV/HIV-1 positive cases (P<0.04). In the CD4+ and CD56+ subset of 2 HBV monoinfected cases on tenofovir therapy, mutations at residues associated with drug resistance and/or immune escape (i.e., G145R) were detected in a minor percentage of the population. Summary HBV genomes and drug resistant variants were detectable in PBMC subsets from HBV mono-infected individuals. The HBV replicates in PBMC subsets of HBV/HIV-1 patients except the CD4+ T cell subpopulation.  相似文献   

12.
13.

Objective

The aim of our study was to assess a possible association between plasma inflammatory biomarkers (CRP, IL-6, soluble CD14) and the extent of fibrosis or cirrhosis using a FibroScan® in HIV/HCV co-infected patients.

Methods

This cross-sectional study assessed 60 HIV/HCV co-infected patients who had paired plasma samples and FibroScan® values available. All included patients were controlled for HIV infection (HIV-1 RNA <50 copies/mL) and had detectable HCV RNA levels. Levels of three biomarkers were measured in all samples using commercial ELISA kits. Multivariate logistic regression models identified factors associated with the METAVIR stages of fibrosis (F0–F2 vs. F3–F4).

Results

In univariate logistic regression analyses, in addition to sCD14 (odds ratio [OR] = 3.23, 95% confidence interval [95%CI] = 1.30–7.97, P = 0.01), aspartate aminotransferase (AST), alanine aminotransferase, platelet counts, and CD4 cell counts were associated with the stage of liver fibrosis and, thus, were introduced into the model. However, only AST (OR = 1.06, 95%CI = 1.02–1.10, P = 0.0009) was independently associated with F3–F4 stage liver fibrosis.

Conclusions

In our study of HIV/HCV co-infected patients, sCD14 plasma level, a biomarker of monocyte activation, was not independently associated with the F3–F4 stage of liver fibrosis. We hypothesize that the higher levels of inflammation markers observed in HIV/HCV co-infected patients, compared to HCV mono-infected patients, prevent this association being observed within this population.  相似文献   

14.

Background

Monocyte-derived macrophages critically perpetuate inflammatory responses after liver injury as a prerequisite for organ fibrosis. Experimental murine models identified an essential role for the CCR2-dependent infiltration of classical Gr1/Ly6C+ monocytes in hepatic fibrosis. Moreover, the monocyte-related chemokine receptors CCR1 and CCR5 were recently recognized as important fibrosis modulators in mice. In humans, monocytes consist of classical CD14+CD16 and non-classical CD14+CD16+ cells. We aimed at investigating the relevance of monocyte subpopulations for human liver fibrosis, and hypothesized that ‘non-classical’ monocytes critically exert inflammatory as well as profibrogenic functions in patients during liver disease progression.

Methodology/Principal Findings

We analyzed circulating monocyte subsets from freshly drawn blood samples of 226 patients with chronic liver disease (CLD) and 184 healthy controls by FACS analysis. Circulating monocytes were significantly expanded in CLD-patients compared to controls with a marked increase of the non-classical CD14+CD16+ subset that showed an activated phenotype in patients and correlated with proinflammatory cytokines and clinical progression. Correspondingly, CD14+CD16+ macrophages massively accumulated in fibrotic/cirrhotic livers, as evidenced by immunofluorescence and FACS. Ligands of monocyte-related chemokine receptors CCR2, CCR1 and CCR5 were expressed at higher levels in fibrotic and cirrhotic livers, while CCL3 and CCL4 were also systemically elevated in CLD-patients. Isolated monocyte/macrophage subpopulations were functionally characterized regarding cytokine/chemokine expression and interactions with primary human hepatic stellate cells (HSC) in vitro. CD14+CD16+ monocytes released abundant proinflammatory cytokines. Furthermore, CD14+CD16+, but not CD14+CD16 monocytes could directly activate collagen-producing HSC.

Conclusions/Significance

Our data demonstrate the expansion of CD14+CD16+ monocytes in the circulation and liver of CLD-patients upon disease progression and suggest their functional contribution to the perpetuation of intrahepatic inflammation and profibrogenic HSC activation in liver cirrhosis. The modulation of monocyte-subset recruitment into the liver via chemokines/chemokine receptors and their subsequent differentiation may represent promising approaches for therapeutic interventions in human liver fibrosis.  相似文献   

15.

Background and Aims

Hepatitis C Virus (HCV)-related liver disease progresses more rapidly in individuals co-infected with Human Immunodeficiency Virus-1 (HIV), although the underlying immunologic mechanisms are unknown. We examined whether HIV-specific T-cells are identified in the liver of HCV/HIV co-infected individuals and promote liver inflammation through bystander immune responses.

Methods

Ex-vivo intra-hepatic lymphocytes from HCV mono-infected and HCV/HIV co-infected individuals were assessed for immune responses to HIV and HCV antigens by polychromatic flow cytometry.

Results

HCV/HIV liver biopsies had similar frequencies of lymphocytes but lower percentages of CD4+ T-cells compared to HCV biopsies. In co-infection, intra-hepatic HIV-specific CD8+ and CD4+ T-cells producing IFN-γ and TNF-α were detected and were comparable in frequency to those that were HCV-specific. In co-infected individuals, viral-specific CD8+ T-cells produced more of the fibrogenic cytokine, TNF-α. In both mono- and co-infected individuals, intra-hepatic HCV-specific T-cells were poorly functional compared to HIV-specific T-cells. In co-infection, HAART was not associated with a reconstitution of intra-hepatic CD4+ T-cells and was associated with reduction in both HIV and HCV-specific intra-hepatic cytokine responses.

Conclusion

The accumulation of functional HIV-specific T-cells in the liver during HCV/HIV co-infection may represent a bystander role for HIV in inducing faster progression of liver disease.  相似文献   

16.
The effects of highly active antiretroviral therapy (HAART) on progression of hepatic fibrosis in HIV-hepatitis C virus (HCV) co-infection are not well understood. Deaths from liver diseases have risen in the post-HAART era, yet some cross-sectional studies have suggested that HAART use is associated with improved fibrosis rates. In a retrospective cohort of 533 HIV mono-infected and 127 HIV/HCV co-infected patients, followed between January 1991 and July 2005 at a university-based HIV clinic, we investigated the relationship between cumulative HAART exposure and hepatic fibrosis, as measured by the aspartate aminotransferase-to-platelet ratio index (APRI). We used a novel methodological approach to estimate the dose-response relationship of the effect of HAART exposure on APRI. HAART was associated with increasing APRI over time in HIV/HCV co-infected patients suggesting that they may be experiencing cumulative hepatotoxicity from antiretrovirals. The estimated median change (95% confidence interval) in APRI per one year of HAART intake was of −0.46% (−1.61% to 0.71%) in HIV mono-infected compared to 2.54% (−1.77% to 7.03%) in HIV/HCV co-infected patients. Similar results were found when the direct effect of HAART intake since the last visit was estimated on the change in APRI. HAART use associated is with increased APRI in patients with HIV/HCV co-infection. Therefore treatment for HCV infection may be required to slow the growing epidemic of end-stage liver disease in this population.  相似文献   

17.
Liver disease is a leading cause of mortality among HIV-infected persons in the highly active anti-retroviral therapy (HAART) era. Hepatitis C Virus (HCV) co-infection is prevalent in, and worsened by HIV; consequently many co-infected persons require liver transplantation (LT). Despite the need, post-LT outcomes are poor in co-infection. We examined predictors of outcomes post-LT. Immunologic biomarkers of immune activation, microbial translocation, and Th1/Th2 skewing were measured pre-LT in participants enrolled in a cohort of HIV infected persons requiring solid organ transplant (HIVTR). Predictive biomarkers were analyzed in Cox-proportional hazards models; multivariate models included known predictors of outcome and biomarkers from univariate analyses. Sixty-nine HIV-HCV co-infected persons with available pre-LT samples were tested: median (IQR) CD4+ T-cell count was 286 (210–429) cells mm-3; 6 (9%) had detectable HIV RNA. Median (IQR) follow-up was 2.1 (0.7–4.0) years, 29 (42%) people died, 35 (51%) had graft loss, 22 (32%) were treated for acute rejection, and 14 (20%) had severe recurrent HCV. In multivariate models, sCD14 levels were significantly lower in persons with graft loss post-LT (HR 0.10 [95%CI 0.02–0.68]). IL-10 levels were higher in persons with rejection (HR 2.10 [95%CI 1.01–4.34]). No markers predicted severe recurrent HCV. Monocyte activation pre-LT may be mechanistically linked to graft health in HIV-HCV co-infection.  相似文献   

18.
The mechanism by which HIV and HCV cooperatively accelerate hepatocyte damage is not clearly understood; however, each virus affects the TRAIL: TRAIL-receptor system. We, therefore, questioned whether the independent effects of HCV and HIV combine to synergistically result in TRAIL dependent hepatocyte killing. We describe that Huh7 hepatocytes treated with HIV gp120 results in both increase TRAIL-R2 expression and an acquired sensitivity to TRAIL mediated killing. Moreover HCV infection and HCV core expression alone in Huh7 cells upregulates TRAIL. Co-incubation of HIV gp120 primed hepatocytes with HCV core expressing hepatocytes results in the selective death of the HIV gp120 primed hepatocytes that is selectively blocked by TRAIL-R2-Fc fusion protein. Liver biopsies from HIV mono-infected patients have increased TRAIL-R2; biopsies from HCV infected patients have increased TRAIL, while co-infected liver biopsies have increased PARP cleavage within hepatocytes indicating enhanced apoptosis. These findings suggest a pathogenic model to understand why HIV/HCV co-infection accelerates liver injury.  相似文献   

19.
In immuno-competent individuals, the natural course of chronic hepatitis C virus (HCV) infection is highly variable and 5%-30% of patients develop cirrhosis over 20 years. Co-infection with HCV and human immunodeficiency virus (HIV) is an important prognostic factor and associated with more frequent and accelerated progression to cirrhosis. Until recently HIV/AIDS-related complications were life limiting in patients co-infected with HCV; the introduction of highly active antiretroviral treatment (HAART) and the better prognosis of HIV infection has made HCV-related complications an emerging health problem in HCV/HIV coinfected individuals. Treatment of chronic HCV infection has also evolved since the introduction of interferon-alpha. Recently, introduction of pegylated interferon-alpha (peginterferon-alpha) has resulted in an increase in sustained virus clearance rates of up to 80% in selected genotypes and patient populations. The safety and efficacy of modern anti HCV treatment regimens - based on peginterferon-alpha in combination with ribavirin - was evaluated in 4 controlled trials. Sustained clearance of hepatitis C virus can be achieved in up to 35% of patients with HIV/HCV co-infection, and novel HCV treatment regimens based on peginterferon-alpha have no negative effect on the control of HIV disease. In conclusion, if HIV infection is well controlled and CD4+ cell counts >100/mm3, treatment of chronic hepatitis C with peginterferon in combination with ribavirin is safe and should be given for 48 weeks regardless of the HCV genotype. Introduction of peginterferon-alpha has significantly improved adherence to treatment and treatment efficacy; in particular sustained virologic response in patients with HCV genotype 1 or 4 infection improved, but sustained viral clearance in only 7%-38% of patients infected with genotype I and 4 cannot be the final step in development of effective treatments in patients with HCV/HIV co-infection.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号