Pub Date : 2024-11-04eCollection Date: 2024-01-01DOI: 10.3389/fimmu.2024.1463655
Xue-Mei You, Fei-Chen Lu, Fan-Rong Li, Feng-Juan Zhao, Rong-Rui Huo
Objective: We aimed to characterize quality of life (QOL) trajectories among patients with intermediate and advanced hepatocellular carcinoma patients treated with immunotherapy.
Methods: Barcelona Clinic Liver Cancer (BCLC) stage B-C HCC patients receiving immunotherapy at Guangxi Medical University Cancer Hospital were included. Trajectories of QOL, assessed using the Functional Assessment of Cancer Therapy-Hepatobiliary (FACT-Hep) questionnaire, were identified through iterative estimations of group-based trajectory models. Associations with trajectory group membership were analyzed using multivariable multinomial logistic regression.
Results: Three trajectory groups were identified (n=156): excellent (35.3%), poor (43.6%), and deteriorating (21.1%) QOL. The deteriorating trajectory group reported a mean QOL score of 124.79 (95% CI, 116.58-133.00), but then declined significantly at month-2 (estimated QOL score 98.67 [95% CI, 84.33-113.00]), and the lowest mean score is reached at month-6 (estimated QOL score 16.58 [95% CI, 0-46.07]). Factors associated with membership to the deteriorating group included no drinking (odds ratio [OR] vs yes [95% CI], 3.70 [1.28-11.11]), no received radiotherapy (OR vs yes [95% CI], 8.33 [1.41-50.00]), diabetes (OR vs no [95% CI], 6.83 [1.57-29.73]), and extrahepatic metastasis (OR vs no [95% CI], 3.08 [1.07-8.87]). Factors associated with membership to the poor group also included body mass index ≤24.0 kg/m2 (OR vs no [95% CI], 4.49 [1.65-12.22]).
Conclusions: This latent-class analysis identified a high-risk cluster of patients with severe, persistent post-immunotherapy QOL deterioration. Screening relevant patient-level characteristics may inform tailored interventions to mitigate the detrimental impact of immunotherapy and preserve QOL.
目的:我们旨在描述接受免疫疗法的中晚期肝细胞癌患者的生活质量(QOL)轨迹:我们旨在描述接受免疫治疗的中晚期肝细胞癌患者的生活质量(QOL)轨迹:方法:纳入在广西医科大学附属肿瘤医院接受免疫治疗的巴塞罗那临床肝癌(BCLC)B-C期HCC患者。通过迭代估计基于组的轨迹模型,确定了使用癌症治疗肝胆功能评估(FACT-Hep)问卷评估的QOL轨迹。采用多变量多项式逻辑回归分析了与轨迹组成员的关联:结果:确定了三个轨迹组(n=156):QOL 优秀组(35.3%)、QOL 较差组(43.6%)和QOL 恶化组(21.1%)。恶化轨迹组的平均 QOL 得分为 124.79(95% CI,116.58-133.00),但在第 2 个月时显著下降(估计 QOL 得分为 98.67 [95% CI,84.33-113.00]),在第 6 个月时达到最低平均分(估计 QOL 得分为 16.58 [95% CI,0-46.07])。与病情恶化组相关的因素包括不饮酒(几率比[OR] vs 是[95% CI],3.70 [1.28-11.11])、未接受放疗(OR vs 是[95% CI],8.33 [1.41-50.00])、糖尿病(OR vs 否[95% CI],6.83 [1.57-29.73])和肝外转移(OR vs 否[95% CI],3.08 [1.07-8.87])。贫困组的相关因素还包括体重指数≤24.0 kg/m2(OR vs no [95% CI],4.49 [1.65-12.22]):该潜类分析确定了免疫治疗后QOL持续严重恶化的高危患者群。筛查患者的相关特征可为有针对性的干预措施提供信息,以减轻免疫疗法的不利影响并保护QOL。
{"title":"Dynamics trajectory of patient-reported quality of life and its associated risk factors among hepatocellular carcinoma patients receiving immune checkpoint inhibitors: a prospective cohort study.","authors":"Xue-Mei You, Fei-Chen Lu, Fan-Rong Li, Feng-Juan Zhao, Rong-Rui Huo","doi":"10.3389/fimmu.2024.1463655","DOIUrl":"10.3389/fimmu.2024.1463655","url":null,"abstract":"<p><strong>Objective: </strong>We aimed to characterize quality of life (QOL) trajectories among patients with intermediate and advanced hepatocellular carcinoma patients treated with immunotherapy.</p><p><strong>Methods: </strong>Barcelona Clinic Liver Cancer (BCLC) stage B-C HCC patients receiving immunotherapy at Guangxi Medical University Cancer Hospital were included. Trajectories of QOL, assessed using the Functional Assessment of Cancer Therapy-Hepatobiliary (FACT-Hep) questionnaire, were identified through iterative estimations of group-based trajectory models. Associations with trajectory group membership were analyzed using multivariable multinomial logistic regression.</p><p><strong>Results: </strong>Three trajectory groups were identified (n=156): excellent (35.3%), poor (43.6%), and deteriorating (21.1%) QOL. The deteriorating trajectory group reported a mean QOL score of 124.79 (95% CI, 116.58-133.00), but then declined significantly at month-2 (estimated QOL score 98.67 [95% CI, 84.33-113.00]), and the lowest mean score is reached at month-6 (estimated QOL score 16.58 [95% CI, 0-46.07]). Factors associated with membership to the deteriorating group included no drinking (odds ratio [OR] <i>vs</i> yes [95% CI], 3.70 [1.28-11.11]), no received radiotherapy (OR <i>vs</i> yes [95% CI], 8.33 [1.41-50.00]), diabetes (OR <i>vs</i> no [95% CI], 6.83 [1.57-29.73]), and extrahepatic metastasis (OR <i>vs</i> no [95% CI], 3.08 [1.07-8.87]). Factors associated with membership to the poor group also included body mass index ≤24.0 kg/m<sup>2</sup> (OR vs no [95% CI], 4.49 [1.65-12.22]).</p><p><strong>Conclusions: </strong>This latent-class analysis identified a high-risk cluster of patients with severe, persistent post-immunotherapy QOL deterioration. Screening relevant patient-level characteristics may inform tailored interventions to mitigate the detrimental impact of immunotherapy and preserve QOL.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"15 ","pages":"1463655"},"PeriodicalIF":5.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570585/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142670238","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-04eCollection Date: 2024-01-01DOI: 10.3389/fimmu.2024.1486627
Shenghao Xue, Yumeng Lin, Haoran Chen, Zhengyu Yang, Junting Zha, Xuan Jiang, Zhongyu Han, Ke Wang
Autophagy is a highly conserved cellular self-digestive process that underlies the maintenance of cellular homeostasis. Autophagy is classified into three types: macrophage, chaperone-mediated autophagy (CMA) and microphagy, which maintain cellular homeostasis through different mechanisms. Altered autophagy regulation affects the progression of various skin diseases, including psoriasis (PA), systemic lupus erythematosus (SLE), vitiligo, atopic dermatitis (AD), alopecia areata (AA) and systemic sclerosis (SSc). In this review, we review the existing literature focusing on three mechanisms of autophagy, namely macrophage, chaperone-mediated autophagy and microphagy, as well as the roles of autophagy in the above six dermatological disorders in order to aid in further studies in the future.
{"title":"Mechanisms of autophagy and their implications in dermatological disorders.","authors":"Shenghao Xue, Yumeng Lin, Haoran Chen, Zhengyu Yang, Junting Zha, Xuan Jiang, Zhongyu Han, Ke Wang","doi":"10.3389/fimmu.2024.1486627","DOIUrl":"10.3389/fimmu.2024.1486627","url":null,"abstract":"<p><p>Autophagy is a highly conserved cellular self-digestive process that underlies the maintenance of cellular homeostasis. Autophagy is classified into three types: macrophage, chaperone-mediated autophagy (CMA) and microphagy, which maintain cellular homeostasis through different mechanisms. Altered autophagy regulation affects the progression of various skin diseases, including psoriasis (PA), systemic lupus erythematosus (SLE), vitiligo, atopic dermatitis (AD), alopecia areata (AA) and systemic sclerosis (SSc). In this review, we review the existing literature focusing on three mechanisms of autophagy, namely macrophage, chaperone-mediated autophagy and microphagy, as well as the roles of autophagy in the above six dermatological disorders in order to aid in further studies in the future.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"15 ","pages":"1486627"},"PeriodicalIF":5.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570406/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667660","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-04eCollection Date: 2024-01-01DOI: 10.3389/fimmu.2024.1477991
Laura Mora-Bitria, Becca Asquith
In addition to their central role during innate responses, NK cells regulate adaptive immunity through various mechanisms. A wide array of innate receptors has been involved in the NK cell regulatory function. However, the clinical implications of these regulatory pathways are poorly understood. Here, we review the experimental evidence on the effects of NK cells on T cells and their positive and negative consequences for disease outcome during T cell responses in humans.
NK 细胞除了在先天性免疫反应中发挥核心作用外,还通过各种机制调节适应性免疫。多种先天受体参与了 NK 细胞的调节功能。然而,人们对这些调节途径的临床意义还知之甚少。在此,我们回顾了有关 NK 细胞对 T 细胞的影响及其对人类 T 细胞反应过程中疾病结果的积极和消极影响的实验证据。
{"title":"Germline natural killer cell receptors modulating the T cell response.","authors":"Laura Mora-Bitria, Becca Asquith","doi":"10.3389/fimmu.2024.1477991","DOIUrl":"10.3389/fimmu.2024.1477991","url":null,"abstract":"<p><p>In addition to their central role during innate responses, NK cells regulate adaptive immunity through various mechanisms. A wide array of innate receptors has been involved in the NK cell regulatory function. However, the clinical implications of these regulatory pathways are poorly understood. Here, we review the experimental evidence on the effects of NK cells on T cells and their positive and negative consequences for disease outcome during T cell responses in humans.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"15 ","pages":"1477991"},"PeriodicalIF":5.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570266/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667652","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Due to the unique heterogeneity of neuroblastoma, its treatment and prognosis are closely related to the biological behavior of the tumor. However, the effect of the tumor immune microenvironment on neuroblastoma needs to be investigated, and there is a lack of biomarkers to reflect the condition of the tumor immune microenvironment.
Methods: The GEO Database was used to download transcriptome data (both training dataset and test dataset) on neuroblastoma. Immunity scores were calculated for each sample using ssGSEA, and hierarchical clustering was used to categorize the samples into high and low immunity groups. Subsequently, the differences in clinicopathological characteristics and treatment between the different groups were examined. Three machine learning algorithms (LASSO, SVM-RFE, and Random Forest) were used to screen biomarkers and synthesize their function in neuroblastoma.
Results: In the training set, there were 362 samples in the immunity_L group and 136 samples in the immunity_H group, with differences in age, MYCN status, etc. Additionally, the tumor microenvironment can also affect the therapeutic response of neuroblastoma. Six characteristic genes (BATF, CXCR3, GIMAP5, GPR18, ISG20, and IGHM) were identified by machine learning, and these genes are associated with multiple immune-related pathways and immune cells in neuroblastoma.
Conclusions: BATF, CXCR3, GIMAP5, GPR18, ISG20, and IGHM may serve as biomarkers that reflect the conditions of the immune microenvironment of neuroblastoma and hold promise in guiding neuroblastoma treatment.
{"title":"Identification of novel markers for neuroblastoma immunoclustering using machine learning.","authors":"Longguo Zhang, Huixin Li, Fangyan Sun, Qiuping Wu, Leigang Jin, Aimin Xu, Jiarui Chen, Ranyao Yang","doi":"10.3389/fimmu.2024.1446273","DOIUrl":"10.3389/fimmu.2024.1446273","url":null,"abstract":"<p><strong>Background: </strong>Due to the unique heterogeneity of neuroblastoma, its treatment and prognosis are closely related to the biological behavior of the tumor. However, the effect of the tumor immune microenvironment on neuroblastoma needs to be investigated, and there is a lack of biomarkers to reflect the condition of the tumor immune microenvironment.</p><p><strong>Methods: </strong>The GEO Database was used to download transcriptome data (both training dataset and test dataset) on neuroblastoma. Immunity scores were calculated for each sample using ssGSEA, and hierarchical clustering was used to categorize the samples into high and low immunity groups. Subsequently, the differences in clinicopathological characteristics and treatment between the different groups were examined. Three machine learning algorithms (LASSO, SVM-RFE, and Random Forest) were used to screen biomarkers and synthesize their function in neuroblastoma.</p><p><strong>Results: </strong>In the training set, there were 362 samples in the immunity_L group and 136 samples in the immunity_H group, with differences in age, MYCN status, etc. Additionally, the tumor microenvironment can also affect the therapeutic response of neuroblastoma. Six characteristic genes (BATF, CXCR3, GIMAP5, GPR18, ISG20, and IGHM) were identified by machine learning, and these genes are associated with multiple immune-related pathways and immune cells in neuroblastoma.</p><p><strong>Conclusions: </strong>BATF, CXCR3, GIMAP5, GPR18, ISG20, and IGHM may serve as biomarkers that reflect the conditions of the immune microenvironment of neuroblastoma and hold promise in guiding neuroblastoma treatment.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"15 ","pages":"1446273"},"PeriodicalIF":5.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570813/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667619","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-04eCollection Date: 2024-01-01DOI: 10.3389/fimmu.2024.1446672
Sturla M Grøndal, Magnus Blø, Linn I H Nilsson, Austin J Rayford, Akil Jackson, Gro Gausdal, James B Lorens
Introduction: The incidence of chronic kidney disease (CKD) is increasing, in parallel with risk factors including obesity and diabetes mellitus. AXL plays a central role in CKD, providing a rationale to evaluate clinical AXL targeting agents.
Methods: To determine the efficacy and underlying molecular mechanisms of AXL inhibition in CKD, we employed a murine unilateral ureteral obstruction (UUO) model preventively treated with a selective AXL kinase inhibitor (bemcentinib) during disease progression. We isolated kidneys at an early (3 days) or late (15 days) timepoint and profiled the cell populations using mass cytometry.
Results: Preventive treatment with bemcentinib significantly attenuated fibrosis in the UUO model. The anti-fibrotic effect correlated with a decrease in mesangial cells and inhibition of innate immune cell infiltration, while the proportion of epithelial cells increased. We mapped AXL expression to a unique network of cells in the kidney: mesangial cells, pericytes, macrophages and dendritic cells.
Discussion: We propose that AXL targeting affects an important cellular interaction network underlying fibrotic progression. These results support the clinical application of AXL targeting agents to treat CKD.
导言:随着肥胖和糖尿病等风险因素的增加,慢性肾脏病(CKD)的发病率也在增加。AXL 在 CKD 中起着核心作用,这为评估临床 AXL 靶向药物提供了依据:为了确定 AXL 抑制剂在 CKD 中的疗效和潜在分子机制,我们采用了一种小鼠单侧输尿管梗阻(UUO)模型,在疾病进展期间使用选择性 AXL 激酶抑制剂(bemcentinib)进行预防性治疗。我们在早期(3 天)或晚期(15 天)分离了肾脏,并使用质谱细胞计数法分析了细胞群:结果:贝门替尼的预防性治疗可显著减轻UUO模型的纤维化。抗纤维化效果与间质细胞的减少和先天性免疫细胞浸润的抑制有关,而上皮细胞的比例则有所增加。我们将 AXL 的表达映射到肾脏中独特的细胞网络:间质细胞、周细胞、巨噬细胞和树突状细胞:我们认为,AXL靶向作用会影响纤维化进程中一个重要的细胞相互作用网络。这些结果支持临床应用 AXL 靶向药物治疗慢性肾脏病。
{"title":"Targeting AXL cellular networks in kidney fibrosis.","authors":"Sturla M Grøndal, Magnus Blø, Linn I H Nilsson, Austin J Rayford, Akil Jackson, Gro Gausdal, James B Lorens","doi":"10.3389/fimmu.2024.1446672","DOIUrl":"10.3389/fimmu.2024.1446672","url":null,"abstract":"<p><strong>Introduction: </strong>The incidence of chronic kidney disease (CKD) is increasing, in parallel with risk factors including obesity and diabetes mellitus. AXL plays a central role in CKD, providing a rationale to evaluate clinical AXL targeting agents.</p><p><strong>Methods: </strong>To determine the efficacy and underlying molecular mechanisms of AXL inhibition in CKD, we employed a murine unilateral ureteral obstruction (UUO) model preventively treated with a selective AXL kinase inhibitor (bemcentinib) during disease progression. We isolated kidneys at an early (3 days) or late (15 days) timepoint and profiled the cell populations using mass cytometry.</p><p><strong>Results: </strong>Preventive treatment with bemcentinib significantly attenuated fibrosis in the UUO model. The anti-fibrotic effect correlated with a decrease in mesangial cells and inhibition of innate immune cell infiltration, while the proportion of epithelial cells increased. We mapped AXL expression to a unique network of cells in the kidney: mesangial cells, pericytes, macrophages and dendritic cells.</p><p><strong>Discussion: </strong>We propose that AXL targeting affects an important cellular interaction network underlying fibrotic progression. These results support the clinical application of AXL targeting agents to treat CKD.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"15 ","pages":"1446672"},"PeriodicalIF":5.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570270/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667726","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-04eCollection Date: 2024-01-01DOI: 10.3389/fimmu.2024.1491481
Ella Barnett, Snehal Kaginkar, Kimberly Schmitt, Leila Remling-Mulder, Ramesh Akkina
Nonhuman primate (NHP) models employing simian/simian-human immunodeficiency viruses (SIV/SHIVs) played a major role in the study of HIV pathogenesis, latency, and cure studies in a preclinical setting. However, it took many years to arrive at the current effective triple drug ARV regimen against SIV due to the genetic differences with that of HIVs. Since new combinations of drugs will be used in the evolving HIV cure studies, a small animal model would be ideal to determine their efficacy against the commonly used SIVs such as SIVmac239 to triage ineffective drugs prior to their application in NHPs. We recently determined that humanized mice (hu-mice) with a transplanted human immune system are permissive to SIVmac strains in addition to HIVs. Based on this novel finding, here we evaluated the utility of this dual-purpose hu-mouse model to test different ART regimens against SIVmac239. Infected mice showing chronic viremia were treated with a combination anti-retroviral treatment (cART) regimen consisting of emtricitabine/elvitegravir/tenofovir disoproxil fumarate (FTC/EVG/TDF). Full viral suppression was seen for several weeks in SIVmac239-infected and treated mice similar to that seen with HIV-1 BaL virus used as a control. However, viral rebound was eventually observed in SIVmac239 infected mice during the treatment period, suggesting viral escape compared to HIV-1 BaL with which viral suppression was fully sustained. Next, a cART regimen consisting of emtricitabine/bictegravir/tenofovir alafenamide fumarate (FTC/BIC/TAF) was similarly evaluated. Our results showed that this ARV regimen was fully effective in rapidly suppressing both SIVmac239 and HIV-1 BaL. Complete viral suppression was maintained until treatment interruption after which viral loads rebounded. These findings highlight the utility of humanized mice for in vivo screening of new combinations of ARV compounds against various SIVs prior to employing them in NHPs. In addition to identifying new effective cART regimens against SIVs, this model would also be amenable to evaluating immunotherapeutic strategies using broadly neutralizing antibodies, LRAs and novel therapeutics in comparative cure studies of SIV and HIV.
采用猿/猿人免疫缺陷病毒(SIV/SHIVs)的非人灵长类(NHP)模型在临床前研究艾滋病发病机制、潜伏期和治愈研究中发挥了重要作用。然而,由于 SIV 与 HIV 在基因上的差异,目前针对 SIV 的有效三联抗逆转录病毒药物方案需要多年时间才能形成。由于新的药物组合将用于不断发展的艾滋病治愈研究中,因此理想的做法是使用小动物模型来确定它们对常用 SIV(如 SIVmac239)的疗效,以便在将无效药物用于 NHPs 之前对其进行分流。我们最近发现,移植了人类免疫系统的人源化小鼠(hu-mice)除了能抵抗 HIV 外,还能抵抗 SIVmac 株。基于这项新发现,我们在此评估了这种两用胡鼠模型的实用性,以测试针对 SIVmac239 的不同抗逆转录病毒疗法。对出现慢性病毒血症的感染小鼠采用由恩曲他滨/埃替拉韦/富马酸替诺福韦二吡呋酯(FTC/EVG/TDF)组成的联合抗逆转录病毒治疗(cART)方案进行治疗。感染 SIVmac239 并接受治疗的小鼠在数周内出现了完全的病毒抑制,与作为对照的 HIV-1 BaL 病毒的抑制效果相似。然而,在治疗期间,SIVmac239 感染的小鼠最终出现了病毒反弹,这表明与完全维持病毒抑制的 HIV-1 BaL 相比,SIVmac239 感染的小鼠出现了病毒逃逸。接下来,我们对一种由恩曲他滨/比克替韦/富马酸替诺福韦阿拉非酰胺(FTC/BIC/TAF)组成的 cART 方案进行了类似的评估。我们的研究结果表明,这种抗逆转录病毒疗法对快速抑制 SIVmac239 和 HIV-1 BaL 完全有效。完全的病毒抑制可维持到治疗中断,之后病毒载量会反弹。这些发现凸显了人源化小鼠在体内筛选抗各种 SIV 的抗逆转录病毒化合物新组合的实用性,然后再将其用于 NHPs。除了确定针对 SIVs 的新的有效 cART 方案外,该模型还适合在 SIV 和 HIV 的比较治愈研究中评估使用广谱中和抗体、LRA 和新型疗法的免疫治疗策略。
{"title":"A dual-purpose humanized mouse model for testing antiviral strategies against both SIV and HIV.","authors":"Ella Barnett, Snehal Kaginkar, Kimberly Schmitt, Leila Remling-Mulder, Ramesh Akkina","doi":"10.3389/fimmu.2024.1491481","DOIUrl":"10.3389/fimmu.2024.1491481","url":null,"abstract":"<p><p>Nonhuman primate (NHP) models employing simian/simian-human immunodeficiency viruses (SIV/SHIVs) played a major role in the study of HIV pathogenesis, latency, and cure studies in a preclinical setting. However, it took many years to arrive at the current effective triple drug ARV regimen against SIV due to the genetic differences with that of HIVs. Since new combinations of drugs will be used in the evolving HIV cure studies, a small animal model would be ideal to determine their efficacy against the commonly used SIVs such as SIVmac239 to triage ineffective drugs prior to their application in NHPs. We recently determined that humanized mice (hu-mice) with a transplanted human immune system are permissive to SIVmac strains in addition to HIVs. Based on this novel finding, here we evaluated the utility of this dual-purpose hu-mouse model to test different ART regimens against SIVmac239. Infected mice showing chronic viremia were treated with a combination anti-retroviral treatment (cART) regimen consisting of emtricitabine/elvitegravir/tenofovir disoproxil fumarate (FTC/EVG/TDF). Full viral suppression was seen for several weeks in SIVmac239-infected and treated mice similar to that seen with HIV-1 BaL virus used as a control. However, viral rebound was eventually observed in SIVmac239 infected mice during the treatment period, suggesting viral escape compared to HIV-1 BaL with which viral suppression was fully sustained. Next, a cART regimen consisting of emtricitabine/bictegravir/tenofovir alafenamide fumarate (FTC/BIC/TAF) was similarly evaluated. Our results showed that this ARV regimen was fully effective in rapidly suppressing both SIVmac239 and HIV-1 BaL. Complete viral suppression was maintained until treatment interruption after which viral loads rebounded. These findings highlight the utility of humanized mice for <i>in vivo</i> screening of new combinations of ARV compounds against various SIVs prior to employing them in NHPs. In addition to identifying new effective cART regimens against SIVs, this model would also be amenable to evaluating immunotherapeutic strategies using broadly neutralizing antibodies, LRAs and novel therapeutics in comparative cure studies of SIV and HIV.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"15 ","pages":"1491481"},"PeriodicalIF":5.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570277/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667613","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-04eCollection Date: 2024-01-01DOI: 10.3389/fimmu.2024.1429817
Xingwang Wang, Hao Hu, Guangning Yan, Bo Zheng, Jinxia Luo, Jianyong Fan
Background: Dermatomyositis (DM) is an autoimmune disease that primarily affects the skin and muscles. It can lead to increased mortality, particularly when patients develop associated malignancies or experience fatal complications such as pulmonary fibrosis. Identifying reliable biomarkers is essential for the early diagnosis and treatment of DM. This study aims to identify and validate pivotal diagnostic biomarker for DM through integrated bioinformatics analysis and clinical sample validation.
Methods: Gene expression datasets GSE46239 and GSE142807 from the Gene Expression Omnibus (GEO) database were merged for analysis. Differentially expressed genes (DEGs) were identified and subjected to enrichment analysis. Advanced machine learning methods were utilized to further pinpoint hub genes. Weighted gene co-expression network analysis (WGCNA) was also conducted to discover key gene modules. Subsequently, we derived intersection gene from these methods. The diagnostic performance of the candidate biomarker was evaluated using analysis with dataset GSE128314 and confirmed by immunohistochemistry (IHC) in skin lesion biopsy specimens. The CIBERSORT algorithm was used to analyze immune cell infiltration patterns in DM, then the association between the hub gene and immune cells was investigated. Gene set enrichment analysis (GSEA) was performed to understand the biomarker's biological functions. Finally, the drug-gene interactions were predicted using the DrugRep server.
Results: Interferon-stimulated gene 15 (ISG15) was identified by intersecting DEGs, advanced machine learning-selected genes and key module genes from WGCNA. ROC analysis showed ISG15 had a high Area under the curve (AUC) of 0.950. IHC findings confirmed uniformly positive expression of ISG15, particularly in perivascular regions and lymphocytes, contrasting with universally negative expression in controls. Further analysis revealed that ISG15 is involved in abnormalities in various immune cells and inflammation-related pathways. We also predicted three drugs targeting ISG15, supported by molecular docking studies.
Conclusion: Our study identifies ISG15 as a highly specific diagnostic biomarker for DM, ISG15 may be closely related to the pathogenesis of DM, demonstrating promising potential for clinical application.
{"title":"Identification and validation of interferon-stimulated gene 15 as a biomarker for dermatomyositis by integrated bioinformatics analysis and machine learning.","authors":"Xingwang Wang, Hao Hu, Guangning Yan, Bo Zheng, Jinxia Luo, Jianyong Fan","doi":"10.3389/fimmu.2024.1429817","DOIUrl":"10.3389/fimmu.2024.1429817","url":null,"abstract":"<p><strong>Background: </strong>Dermatomyositis (DM) is an autoimmune disease that primarily affects the skin and muscles. It can lead to increased mortality, particularly when patients develop associated malignancies or experience fatal complications such as pulmonary fibrosis. Identifying reliable biomarkers is essential for the early diagnosis and treatment of DM. This study aims to identify and validate pivotal diagnostic biomarker for DM through integrated bioinformatics analysis and clinical sample validation.</p><p><strong>Methods: </strong>Gene expression datasets GSE46239 and GSE142807 from the Gene Expression Omnibus (GEO) database were merged for analysis. Differentially expressed genes (DEGs) were identified and subjected to enrichment analysis. Advanced machine learning methods were utilized to further pinpoint hub genes. Weighted gene co-expression network analysis (WGCNA) was also conducted to discover key gene modules. Subsequently, we derived intersection gene from these methods. The diagnostic performance of the candidate biomarker was evaluated using analysis with dataset GSE128314 and confirmed by immunohistochemistry (IHC) in skin lesion biopsy specimens. The CIBERSORT algorithm was used to analyze immune cell infiltration patterns in DM, then the association between the hub gene and immune cells was investigated. Gene set enrichment analysis (GSEA) was performed to understand the biomarker's biological functions. Finally, the drug-gene interactions were predicted using the DrugRep server.</p><p><strong>Results: </strong>Interferon-stimulated gene 15 (ISG15) was identified by intersecting DEGs, advanced machine learning-selected genes and key module genes from WGCNA. ROC analysis showed ISG15 had a high Area under the curve (AUC) of 0.950. IHC findings confirmed uniformly positive expression of ISG15, particularly in perivascular regions and lymphocytes, contrasting with universally negative expression in controls. Further analysis revealed that ISG15 is involved in abnormalities in various immune cells and inflammation-related pathways. We also predicted three drugs targeting ISG15, supported by molecular docking studies.</p><p><strong>Conclusion: </strong>Our study identifies ISG15 as a highly specific diagnostic biomarker for DM, ISG15 may be closely related to the pathogenesis of DM, demonstrating promising potential for clinical application.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"15 ","pages":"1429817"},"PeriodicalIF":5.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570269/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667618","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: The Epithelial-Mesenchymal Transition (EMT) is a very important process involved in cancer invasion and metastasis. Additionally, the Cathepsin K (CTSK) gene is closely related to the degradation of the extracellular matrix, which is a critical component of the EMT. The purpose of this study was to determine the relationships between EMT-related genes and immune cell infiltration and their prognostic value in Thyroid carcinoma (THCA). The effect of the CTSK gene on the aggressive biological features of THCA was assessed.
Methods: Within the framework of the present study, the THCA cohort was analyzed in detail based on data obtained from The TCGA database in the context of the EMT. The TCGA-THCA cohort was then divided into two groups, namely, high- and low-risk groups, based on the calculated EMT scores. Finally, based on the findings from the Weighted Gene Co-Expression Network Analysis (WGCNA) algorithm, LASSO regression analysis, and Kaplan-Meier plotter, we selected five genes (CTSK, C3ORF80, FBLN2, PRELP and SRPX2) associated with patient prognosis. Furthermore, this study examined the presence of various immune cells within the THCA samples using three distinct algorithms, namely ssGSEA, xCell, and MCPcounter. Additional studies have been conducted to establish the roles of CTSK in THCA cell proliferation and migration using various assays, such as CCK8, colony formation, EdU proliferation, Transwell migration and wound healing assays. Additionally, the involvement of CTSK in the regulation of various EMT-related markers was confirmed using Western blot analysis.
Results: Based on EMT scores, TCGA-THCA patients were further divided into two groups, and the study revealed that patients in the high-risk group had a worse prognosis than those in the low-risk group. Among the five genes linked to the prognostic value of EMT (CTSK, C3ORF80, FBLN2, PRELP, and SRPX2), CTSK exhibited notably elevated expression in the high-risk cohort. This group also exhibited pronounced immune cell infiltration, with a marked correlation observed between CTSK expression and the levels of macrophages, MDSCs, and various T-cell subtypes. Furthermore, in vitro studies demonstrated that reducing CTSK expression led to significant reductions in THCA cell viability; clonogenic, proliferative, motility and migratory capacities; and the expression of key EMT-related proteins, including N-cadherin, vimentin, slug, and snail.
Conclusion: Our results suggest that the expression of CTSK, a gene associated with the EMT, may be associated with THCA onset and progression and thus may serve as a promising prognostic biomarker.
{"title":"Prognostic value of EMT-related genes and immune cell infiltration in thyroid carcinoma.","authors":"Shuping Wu, Yu Liu, Yu Zeng, Xianhui Ruan, Mei Tao, Wenrong Lin, Chang Liu, Hongbin Chen, Hui Liu, Yu Wu","doi":"10.3389/fimmu.2024.1463258","DOIUrl":"10.3389/fimmu.2024.1463258","url":null,"abstract":"<p><strong>Background: </strong>The Epithelial-Mesenchymal Transition (EMT) is a very important process involved in cancer invasion and metastasis. Additionally, the Cathepsin K (CTSK) gene is closely related to the degradation of the extracellular matrix, which is a critical component of the EMT. The purpose of this study was to determine the relationships between EMT-related genes and immune cell infiltration and their prognostic value in Thyroid carcinoma (THCA). The effect of the CTSK gene on the aggressive biological features of THCA was assessed.</p><p><strong>Methods: </strong>Within the framework of the present study, the THCA cohort was analyzed in detail based on data obtained from The TCGA database in the context of the EMT. The TCGA-THCA cohort was then divided into two groups, namely, high- and low-risk groups, based on the calculated EMT scores. Finally, based on the findings from the Weighted Gene Co-Expression Network Analysis (WGCNA) algorithm, LASSO regression analysis, and Kaplan-Meier plotter, we selected five genes (CTSK, C3ORF80, FBLN2, PRELP and SRPX2) associated with patient prognosis. Furthermore, this study examined the presence of various immune cells within the THCA samples using three distinct algorithms, namely ssGSEA, xCell, and MCPcounter. Additional studies have been conducted to establish the roles of CTSK in THCA cell proliferation and migration using various assays, such as CCK8, colony formation, EdU proliferation, Transwell migration and wound healing assays. Additionally, the involvement of CTSK in the regulation of various EMT-related markers was confirmed using Western blot analysis.</p><p><strong>Results: </strong>Based on EMT scores, TCGA-THCA patients were further divided into two groups, and the study revealed that patients in the high-risk group had a worse prognosis than those in the low-risk group. Among the five genes linked to the prognostic value of EMT (CTSK, C3ORF80, FBLN2, PRELP, and SRPX2), CTSK exhibited notably elevated expression in the high-risk cohort. This group also exhibited pronounced immune cell infiltration, with a marked correlation observed between CTSK expression and the levels of macrophages, MDSCs, and various T-cell subtypes. Furthermore, <i>in vitro</i> studies demonstrated that reducing CTSK expression led to significant reductions in THCA cell viability; clonogenic, proliferative, motility and migratory capacities; and the expression of key EMT-related proteins, including N-cadherin, vimentin, slug, and snail.</p><p><strong>Conclusion: </strong>Our results suggest that the expression of CTSK, a gene associated with the EMT, may be associated with THCA onset and progression and thus may serve as a promising prognostic biomarker.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"15 ","pages":"1463258"},"PeriodicalIF":5.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570584/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667722","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-04eCollection Date: 2024-01-01DOI: 10.3389/fimmu.2024.1475528
Qicen Yao, Wenfeng Tan, Feihu Bai
Systemic sclerosis (SSc) is a rare and highly heterogeneous chronic autoimmune disease characterized by multi-organ and tissue fibrosis, often accompanied by a poor prognosis and high mortality rates. The primary pathogenic mechanisms of SSc are considered to involve tissue fibrosis, autoimmune dysfunction, and microvascular abnormalities. Recent studies have shed light on the gut microbiota (GM) and metabolites in SSc patients, revealing their association with gastrointestinal symptoms and disease phenotypes. However, further elucidation is needed on the specific mechanisms underlying the interactions between GM, metabolites, and the immune system and their roles in the pathogenesis of SSc. This review outlines the characteristics of GM and metabolites in SSc patients, exploring their interrelationships and analyzing their correlations with the clinical phenotypes of SSc. The findings indicate that while the α-diversity of GM in SSc patients resembles that of healthy individuals, notable differences exist in the β-diversity and the abundance of specific bacterial genera, which are closely linked to gastrointestinal symptoms. Moreover, alterations in the levels of amino acids and lipid metabolites in SSc patients are prominently observed and significantly associated with clinical phenotypes. Furthermore, this review delves into the potential immunopathological mechanisms of GM and metabolites in SSc, emphasizing the critical role of interactions between GM, metabolites, and the immune system in comprehending the immunopathological processes of SSc. These insights may offer new scientific evidence for the development of future treatment strategies.
{"title":"Gut microbiome and metabolomics in systemic sclerosis: feature, link and mechanisms.","authors":"Qicen Yao, Wenfeng Tan, Feihu Bai","doi":"10.3389/fimmu.2024.1475528","DOIUrl":"10.3389/fimmu.2024.1475528","url":null,"abstract":"<p><p>Systemic sclerosis (SSc) is a rare and highly heterogeneous chronic autoimmune disease characterized by multi-organ and tissue fibrosis, often accompanied by a poor prognosis and high mortality rates. The primary pathogenic mechanisms of SSc are considered to involve tissue fibrosis, autoimmune dysfunction, and microvascular abnormalities. Recent studies have shed light on the gut microbiota (GM) and metabolites in SSc patients, revealing their association with gastrointestinal symptoms and disease phenotypes. However, further elucidation is needed on the specific mechanisms underlying the interactions between GM, metabolites, and the immune system and their roles in the pathogenesis of SSc. This review outlines the characteristics of GM and metabolites in SSc patients, exploring their interrelationships and analyzing their correlations with the clinical phenotypes of SSc. The findings indicate that while the α-diversity of GM in SSc patients resembles that of healthy individuals, notable differences exist in the β-diversity and the abundance of specific bacterial genera, which are closely linked to gastrointestinal symptoms. Moreover, alterations in the levels of amino acids and lipid metabolites in SSc patients are prominently observed and significantly associated with clinical phenotypes. Furthermore, this review delves into the potential immunopathological mechanisms of GM and metabolites in SSc, emphasizing the critical role of interactions between GM, metabolites, and the immune system in comprehending the immunopathological processes of SSc. These insights may offer new scientific evidence for the development of future treatment strategies.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"15 ","pages":"1475528"},"PeriodicalIF":5.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570262/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667582","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-04eCollection Date: 2024-01-01DOI: 10.3389/fimmu.2024.1481296
Tiantian Yang, Wanru Luo, Jie Yu, Huiping Zhang, Meichun Hu, Jun Tian
As an immune-related tumor type, bladder cancer has been attracting much attention in the study of its markers. In recent years, researchers have made rapid progress in the study of immune-related markers for bladder cancer. Studies have shown that immune-related markers play an important role in the diagnosis, prognosis assessment and treatment of bladder cancer. In addition, the detection of immune-related markers can also be used to evaluate the efficacy of immunotherapy and predict the treatment response of patients. Therefore, in depth study of the expression of immune-related markers in bladder cancer and their application in the clinic is of great significance and is expected to provide new breakthroughs for individualized treatment of bladder cancer. Future studies will focus more on how to detect immune-related markers with low cost and high accuracy, as well as develop new immunotherapeutic strategies to bring better therapeutic outcomes to bladder cancer patients.
{"title":"Bladder cancer immune-related markers: diagnosis, surveillance, and prognosis.","authors":"Tiantian Yang, Wanru Luo, Jie Yu, Huiping Zhang, Meichun Hu, Jun Tian","doi":"10.3389/fimmu.2024.1481296","DOIUrl":"10.3389/fimmu.2024.1481296","url":null,"abstract":"<p><p>As an immune-related tumor type, bladder cancer has been attracting much attention in the study of its markers. In recent years, researchers have made rapid progress in the study of immune-related markers for bladder cancer. Studies have shown that immune-related markers play an important role in the diagnosis, prognosis assessment and treatment of bladder cancer. In addition, the detection of immune-related markers can also be used to evaluate the efficacy of immunotherapy and predict the treatment response of patients. Therefore, in depth study of the expression of immune-related markers in bladder cancer and their application in the clinic is of great significance and is expected to provide new breakthroughs for individualized treatment of bladder cancer. Future studies will focus more on how to detect immune-related markers with low cost and high accuracy, as well as develop new immunotherapeutic strategies to bring better therapeutic outcomes to bladder cancer patients.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"15 ","pages":"1481296"},"PeriodicalIF":5.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570592/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667634","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}