首页 > 最新文献

Retrovirology最新文献

英文 中文
A gut check: understanding the interplay of the gastrointestinal microbiome and the developing immune system towards the goal of pediatric HIV remission. 肠道检查:了解胃肠道微生物组与发育中免疫系统的相互作用,以实现小儿艾滋病缓解的目标。
IF 2.7 3区 医学 Q3 VIROLOGY Pub Date : 2024-10-18 DOI: 10.1186/s12977-024-00648-9
Nicole Soo, Omotayo Farinre, Ann Chahroudi, Saikat Boliar, Ria Goswami

Despite the efficacy of antiretroviral therapy (ART) in reducing the global incidence of vertical HIV transmissions, more than 120,000 children are still infected with the virus each year. Since ART cannot clear the HIV reservoir that is established soon after infection, children living with HIV (CLWH) are forced to rely on therapy for their lives and suffer from long-term drug-related complications. Pediatric HIV infection, like adult infection, is associated with gut microbial dysbiosis, loss of gut epithelial integrity, bacterial translocation, CD4 + T cell depletion, systemic immune activation, and viral reservoir establishment. However, unlike in adults, HIV that is vertically acquired by infants interacts with a gut microbiome that is continuously evolving while concomitantly shaping the infant's immune ontogeny. Therefore, to determine whether there may be interventions that target the HIV reservoir through microbiome-directed approaches, understanding the complex tripartite interactions between the transmitted HIV, the maturing gut microbiome, and the developing immune system during early life is crucial. Importantly, early life is the time when the gut microbiome of an individual is highly dynamic, and this temporal development of the gut microbiome plays a crucial role in educating the maturing immune system of a child. Therefore, manipulation of the gut microbiome of CLWH to a phenotype that can reduce HIV persistence by fostering an antiviral immune system might be an opportune strategy to achieve ART-free viral suppression in CLWH. This review summarizes the current state of knowledge on the vertical transmission of HIV, the developing gut microbiome of CLWH, and the immune landscape of pediatric elite controllers, and explores the prospect of employing microbial modulation as a potential therapeutic approach to achieve ART-free viral suppression in the pediatric population.

尽管抗逆转录病毒疗法(ART)能有效降低全球艾滋病垂直传播的发病率,但每年仍有超过 12 万名儿童感染病毒。由于抗逆转录病毒疗法无法清除感染后不久形成的艾滋病病毒库,感染艾滋病病毒的儿童(CLWH)不得不终身依赖治疗,并长期遭受与药物相关的并发症的折磨。小儿艾滋病病毒感染与成人感染一样,与肠道微生物菌群失调、肠道上皮完整性丧失、细菌易位、CD4 + T 细胞耗竭、全身免疫激活和病毒库建立有关。然而,与成人不同的是,婴儿垂直感染的艾滋病病毒与肠道微生物群相互作用,而肠道微生物群在不断演变的同时也在塑造婴儿的免疫本体。因此,要确定是否有可能通过以微生物组为导向的方法针对艾滋病病毒库进行干预,了解生命早期传播的艾滋病病毒、成熟的肠道微生物组和发育中的免疫系统之间复杂的三方相互作用至关重要。重要的是,生命早期是个体肠道微生物组高度动态的时期,而肠道微生物组的这种时间性发展在教育儿童发育成熟的免疫系统方面起着至关重要的作用。因此,通过培养抗病毒免疫系统将儿童慢性淋巴细胞白血病患者的肠道微生物组调控到可减少艾滋病病毒持续存在的表型,可能是实现儿童慢性淋巴细胞白血病患者无抗病毒疗法病毒抑制的一个恰当策略。本综述总结了目前有关 HIV 垂直传播、CLWH 正在发育的肠道微生物组和儿科精英控制者免疫系统的知识,并探讨了将微生物调节作为一种潜在的治疗方法在儿科人群中实现无抗逆转录病毒疗法病毒抑制的前景。
{"title":"A gut check: understanding the interplay of the gastrointestinal microbiome and the developing immune system towards the goal of pediatric HIV remission.","authors":"Nicole Soo, Omotayo Farinre, Ann Chahroudi, Saikat Boliar, Ria Goswami","doi":"10.1186/s12977-024-00648-9","DOIUrl":"https://doi.org/10.1186/s12977-024-00648-9","url":null,"abstract":"<p><p>Despite the efficacy of antiretroviral therapy (ART) in reducing the global incidence of vertical HIV transmissions, more than 120,000 children are still infected with the virus each year. Since ART cannot clear the HIV reservoir that is established soon after infection, children living with HIV (CLWH) are forced to rely on therapy for their lives and suffer from long-term drug-related complications. Pediatric HIV infection, like adult infection, is associated with gut microbial dysbiosis, loss of gut epithelial integrity, bacterial translocation, CD4 + T cell depletion, systemic immune activation, and viral reservoir establishment. However, unlike in adults, HIV that is vertically acquired by infants interacts with a gut microbiome that is continuously evolving while concomitantly shaping the infant's immune ontogeny. Therefore, to determine whether there may be interventions that target the HIV reservoir through microbiome-directed approaches, understanding the complex tripartite interactions between the transmitted HIV, the maturing gut microbiome, and the developing immune system during early life is crucial. Importantly, early life is the time when the gut microbiome of an individual is highly dynamic, and this temporal development of the gut microbiome plays a crucial role in educating the maturing immune system of a child. Therefore, manipulation of the gut microbiome of CLWH to a phenotype that can reduce HIV persistence by fostering an antiviral immune system might be an opportune strategy to achieve ART-free viral suppression in CLWH. This review summarizes the current state of knowledge on the vertical transmission of HIV, the developing gut microbiome of CLWH, and the immune landscape of pediatric elite controllers, and explores the prospect of employing microbial modulation as a potential therapeutic approach to achieve ART-free viral suppression in the pediatric population.</p>","PeriodicalId":21123,"journal":{"name":"Retrovirology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11490017/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142473594","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
High level of genomic divergence in orf-I p12 and hbz genes of HTLV-1 subtype-C in Central Australia. 澳大利亚中部地区 HTLV-1 亚型-C 的 orf-I p12 和 hbz 基因存在高度基因组差异。
IF 2.7 3区 医学 Q3 VIROLOGY Pub Date : 2024-07-17 DOI: 10.1186/s12977-024-00647-w
Ashley Hirons, David Yurick, Natasha Jansz, Paula Ellenberg, Genoveffa Franchini, Lloyd Einsiedel, Georges Khoury, Damian F J Purcell

Background: Human T cell lymphotropic virus type 1 (HTLV-1) infection remains a largely neglected public health problem, particularly in resource-poor areas with high burden of communicable and non-communicable diseases, such as some remote populations in Central Australia where an estimated 37% of adults are infected with HTLV-1. Most of our understanding of HTLV-1 infection comes from studies of the globally spread subtype-A (HTLV-1a), with few molecular studies reported with the Austral-Melanesian subtype-C (HTLV-1c) predominant in the Indo-Pacific and Oceania regions.

Results: Using a primer walking strategy and direct sequencing, we constructed HTLV-1c genomic consensus sequences from 22 First Nations participants living with HTLV-1c in Central Australia. Phylogenetic and pairwise analysis of this subtype-C proviral gDNA showed higher levels of genomic divergence in comparison to previously published HTLV-1a genomes. While the overall genomic homology between subtypes was 92.5%, the lowest nucleotide and amino acid sequence identity occurred near the 3' end of the proviral genome coding regulatory genes, especially overlapping hbz (85.37%, 77.46%, respectively) and orf-I product p12 (82.00%, 70.30%, respectively). Strikingly, the HTLV-1c genomic consensus sequences uniformly showed a defective translation start codon for the immune regulatory proteins p12/p8 encoded by the HTLV-1A orf-I. Deletions in the proviral genome were detected in many subjects, particularly in the structural gag, pol and env genes. Similarly, using a droplet digital PCR assay measuring the copies of gag and tax per reference host genome, we quantitatively confirmed that provirus retains the tax gene region at higher levels than gag.

Conclusions: Our genomic analysis of HTLV-1c in Central Australia in conjunction with earlier Melanesian HTLV-1c sequences, elucidate substantial differences with respect to the globally spread HTLV-1a. Future studies should address the impact these genomic differences have on infection and the regionally distinctive frequency of associated pulmonary disease. Understanding the host and virus subtype factors which contribute to the differential morbidity observed, is crucial for the development of much needed therapeutics and vaccine strategies against this highly endemic infection in remote First Nations communities in Central Australia.

背景:人类 T 细胞淋巴细胞病毒 1 型(HTLV-1)感染在很大程度上仍是一个被忽视的公共卫生问题,尤其是在传染性和非传染性疾病负担较重的资源贫乏地区,如澳大利亚中部的一些偏远地区,估计有 37% 的成年人感染了 HTLV-1。我们对 HTLV-1 感染的了解大多来自于对全球传播的亚型-A(HTLV-1a)的研究,而对印度洋-太平洋和大洋洲地区流行的澳大拉西亚-美拉尼西亚亚型-C(HTLV-1c)的分子研究报道很少:结果:我们采用引物游走策略和直接测序方法,从澳大利亚中部 22 名感染 HTLV-1c 的原住民参与者中构建了 HTLV-1c 基因组共识序列。与之前发表的 HTLV-1a 基因组相比,该亚型-C 前病毒 gDNA 的系统发育和配对分析表明了更高程度的基因组差异。虽然亚型之间的总体基因组同源性为 92.5%,但核苷酸和氨基酸序列的最低一致性出现在前病毒基因组编码调控基因的 3' 端附近,尤其是重叠的 hbz(分别为 85.37%和 77.46%)和 orf-I 产物 p12(分别为 82.00%和 70.30%)。引人注目的是,HTLV-1c 基因组共识序列一致显示 HTLV-1A orf-I 编码的免疫调节蛋白 p12/p8 的翻译起始密码子有缺陷。在许多受试者中检测到了病毒基因组的缺失,尤其是结构性 gag、pol 和 env 基因。同样,通过使用液滴数字 PCR 检测法测量每个参考宿主基因组中 gag 和 tax 的拷贝数,我们定量地证实了前病毒保留 tax 基因区域的水平高于 gag:结论:我们对澳大利亚中部 HTLV-1c 的基因组分析与早期美拉尼西亚 HTLV-1c 序列相结合,阐明了与全球传播的 HTLV-1a 的实质性差异。未来的研究应探讨这些基因组差异对感染的影响,以及不同地区相关肺病的发病率。了解导致不同发病率的宿主和病毒亚型因素,对于针对澳大利亚中部偏远原住民社区的这种高流行性感染开发急需的疗法和疫苗策略至关重要。
{"title":"High level of genomic divergence in orf-I p12 and hbz genes of HTLV-1 subtype-C in Central Australia.","authors":"Ashley Hirons, David Yurick, Natasha Jansz, Paula Ellenberg, Genoveffa Franchini, Lloyd Einsiedel, Georges Khoury, Damian F J Purcell","doi":"10.1186/s12977-024-00647-w","DOIUrl":"10.1186/s12977-024-00647-w","url":null,"abstract":"<p><strong>Background: </strong>Human T cell lymphotropic virus type 1 (HTLV-1) infection remains a largely neglected public health problem, particularly in resource-poor areas with high burden of communicable and non-communicable diseases, such as some remote populations in Central Australia where an estimated 37% of adults are infected with HTLV-1. Most of our understanding of HTLV-1 infection comes from studies of the globally spread subtype-A (HTLV-1a), with few molecular studies reported with the Austral-Melanesian subtype-C (HTLV-1c) predominant in the Indo-Pacific and Oceania regions.</p><p><strong>Results: </strong>Using a primer walking strategy and direct sequencing, we constructed HTLV-1c genomic consensus sequences from 22 First Nations participants living with HTLV-1c in Central Australia. Phylogenetic and pairwise analysis of this subtype-C proviral gDNA showed higher levels of genomic divergence in comparison to previously published HTLV-1a genomes. While the overall genomic homology between subtypes was 92.5%, the lowest nucleotide and amino acid sequence identity occurred near the 3' end of the proviral genome coding regulatory genes, especially overlapping hbz (85.37%, 77.46%, respectively) and orf-I product p12 (82.00%, 70.30%, respectively). Strikingly, the HTLV-1c genomic consensus sequences uniformly showed a defective translation start codon for the immune regulatory proteins p12/p8 encoded by the HTLV-1A orf-I. Deletions in the proviral genome were detected in many subjects, particularly in the structural gag, pol and env genes. Similarly, using a droplet digital PCR assay measuring the copies of gag and tax per reference host genome, we quantitatively confirmed that provirus retains the tax gene region at higher levels than gag.</p><p><strong>Conclusions: </strong>Our genomic analysis of HTLV-1c in Central Australia in conjunction with earlier Melanesian HTLV-1c sequences, elucidate substantial differences with respect to the globally spread HTLV-1a. Future studies should address the impact these genomic differences have on infection and the regionally distinctive frequency of associated pulmonary disease. Understanding the host and virus subtype factors which contribute to the differential morbidity observed, is crucial for the development of much needed therapeutics and vaccine strategies against this highly endemic infection in remote First Nations communities in Central Australia.</p>","PeriodicalId":21123,"journal":{"name":"Retrovirology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11253349/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141627530","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
In situ analysis of neuronal injury and neuroinflammation during HIV-1 infection. 对 HIV-1 感染期间神经元损伤和神经炎症的原位分析
IF 2.7 3区 医学 Q3 VIROLOGY Pub Date : 2024-07-01 DOI: 10.1186/s12977-024-00644-z
Jenna B Honeycutt, Angela Wahl, Jacob K Files, Alexis F League, Barkha J Yadav-Samudrala, J Victor Garcia, Sylvia Fitting

Background: Since the introduction of combination antiretroviral therapy (cART) the brain has become an important human immunodeficiency virus (HIV) reservoir due to the relatively low penetration of many drugs utilized in cART into the central nervous system (CNS). Given the inherent limitations of directly assessing acute HIV infection in the brains of people living with HIV (PLWH), animal models, such as humanized mouse models, offer the most effective means of studying the effects of different viral strains and their impact on HIV infection in the CNS. To evaluate CNS pathology during HIV-1 infection in the humanized bone marrow/liver/thymus (BLT) mouse model, a histological analysis was conducted on five CNS regions, including the frontal cortex, hippocampus, striatum, cerebellum, and spinal cord, to delineate the neuronal (MAP2ab, NeuN) and neuroinflammatory (GFAP, Iba-1) changes induced by two viral strains after 2 weeks and 8 weeks post-infection.

Results: Findings reveal HIV-infected human cells in the brain of HIV-infected BLT mice, demonstrating HIV neuroinvasion. Further, both viral strains, HIV-1JR-CSF and HIV-1CH040, induced neuronal injury and astrogliosis across all CNS regions following HIV infection at both time points, as demonstrated by decreases in MAP2ab and increases in GFAP fluorescence signal, respectively. Importantly, infection with HIV-1JR-CSF had more prominent effects on neuronal health in specific CNS regions compared to HIV-1CH040 infection, with decreasing number of NeuN+ neurons, specifically in the frontal cortex. On the other hand, infection with HIV-1CH040 demonstrated more prominent effects on neuroinflammation, assessed by an increase in GFAP signal and/or an increase in number of Iba-1+ microglia, across CNS regions.

Conclusion: These findings demonstrate that CNS pathology is widespread during acute HIV infection. However, neuronal loss and the magnitude of neuroinflammation in the CNS is strain dependent indicating that strains of HIV cause differential CNS pathologies.

背景:自从引入联合抗逆转录病毒疗法(cART)以来,由于许多用于 cART 的药物在中枢神经系统(CNS)中的渗透率相对较低,因此大脑已成为重要的人类免疫缺陷病毒(HIV)库。鉴于直接评估 HIV 感染者(PLWH)大脑中急性 HIV 感染的固有局限性,人源化小鼠模型等动物模型为研究不同病毒株的作用及其对中枢神经系统 HIV 感染的影响提供了最有效的方法。为了评估人源化骨髓/肝脏/胸腺(BLT)小鼠模型在感染HIV-1期间的中枢神经系统病理变化,我们对包括额叶皮层、海马、纹状体、小脑和脊髓在内的五个中枢神经系统区域进行了组织学分析,以确定感染后2周和8周后两种病毒株诱导的神经元(MAP2ab、NeuN)和神经炎症(GFAP、Iba-1)变化:结果:研究结果显示,HIV 感染 BLT 小鼠的大脑中存在受 HIV 感染的人类细胞,这表明 HIV 已入侵神经系统。此外,两种病毒株(HIV-1JR-CSF 和 HIV-1CH040)在感染 HIV 后的两个时间点都会诱发中枢神经系统各区域的神经元损伤和星形胶质细胞增多,这分别表现为 MAP2ab 的减少和 GFAP 荧光信号的增加。重要的是,与感染 HIV-1CH040 相比,感染 HIV-1JR-CSF 对特定中枢神经系统区域神经元健康的影响更为显著,NeuN+神经元的数量减少,尤其是在额叶皮层。另一方面,HIV-1CH040感染对中枢神经系统各区域神经炎症的影响更为显著,具体表现为GFAP信号增加和/或Iba-1+小胶质细胞数量增加:这些研究结果表明,中枢神经系统病理变化在艾滋病病毒急性感染期间十分普遍。然而,中枢神经系统中神经元的损失和神经炎症的程度与菌株有关,这表明艾滋病毒菌株会导致不同的中枢神经系统病理变化。
{"title":"In situ analysis of neuronal injury and neuroinflammation during HIV-1 infection.","authors":"Jenna B Honeycutt, Angela Wahl, Jacob K Files, Alexis F League, Barkha J Yadav-Samudrala, J Victor Garcia, Sylvia Fitting","doi":"10.1186/s12977-024-00644-z","DOIUrl":"10.1186/s12977-024-00644-z","url":null,"abstract":"<p><strong>Background: </strong>Since the introduction of combination antiretroviral therapy (cART) the brain has become an important human immunodeficiency virus (HIV) reservoir due to the relatively low penetration of many drugs utilized in cART into the central nervous system (CNS). Given the inherent limitations of directly assessing acute HIV infection in the brains of people living with HIV (PLWH), animal models, such as humanized mouse models, offer the most effective means of studying the effects of different viral strains and their impact on HIV infection in the CNS. To evaluate CNS pathology during HIV-1 infection in the humanized bone marrow/liver/thymus (BLT) mouse model, a histological analysis was conducted on five CNS regions, including the frontal cortex, hippocampus, striatum, cerebellum, and spinal cord, to delineate the neuronal (MAP2ab, NeuN) and neuroinflammatory (GFAP, Iba-1) changes induced by two viral strains after 2 weeks and 8 weeks post-infection.</p><p><strong>Results: </strong>Findings reveal HIV-infected human cells in the brain of HIV-infected BLT mice, demonstrating HIV neuroinvasion. Further, both viral strains, HIV-1<sub>JR-CSF</sub> and HIV-1<sub>CH040</sub>, induced neuronal injury and astrogliosis across all CNS regions following HIV infection at both time points, as demonstrated by decreases in MAP2ab and increases in GFAP fluorescence signal, respectively. Importantly, infection with HIV-1<sub>JR-CSF</sub> had more prominent effects on neuronal health in specific CNS regions compared to HIV-1<sub>CH040</sub> infection, with decreasing number of NeuN<sup>+</sup> neurons, specifically in the frontal cortex. On the other hand, infection with HIV-1<sub>CH040</sub> demonstrated more prominent effects on neuroinflammation, assessed by an increase in GFAP signal and/or an increase in number of Iba-1<sup>+</sup> microglia, across CNS regions.</p><p><strong>Conclusion: </strong>These findings demonstrate that CNS pathology is widespread during acute HIV infection. However, neuronal loss and the magnitude of neuroinflammation in the CNS is strain dependent indicating that strains of HIV cause differential CNS pathologies.</p>","PeriodicalId":21123,"journal":{"name":"Retrovirology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11215835/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141470443","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Comparative analysis of retroviral Gag-host cell interactions: focus on the nuclear interactome. 逆转录病毒 Gag-宿主细胞相互作用的比较分析:聚焦核相互作用组。
IF 2.7 3区 医学 Q3 VIROLOGY Pub Date : 2024-06-19 DOI: 10.1186/s12977-024-00645-y
Gregory S Lambert, Breanna L Rice, Rebecca J Kaddis Maldonado, Jordan Chang, Leslie J Parent

Retroviruses exploit host proteins to assemble and release virions from infected cells. Previously, most studies focused on interacting partners of retroviral Gag proteins that localize to the cytoplasm or plasma membrane. Given that several full-length Gag proteins have been found in the nucleus, identifying the Gag-nuclear interactome has high potential for novel findings involving previously unknown host processes. Here we systematically compared nuclear factors identified in published HIV-1 proteomic studies and performed our own mass spectrometry analysis using affinity-tagged HIV-1 and RSV Gag proteins mixed with nuclear extracts. We identified 57 nuclear proteins in common between HIV-1 and RSV Gag, and a set of nuclear proteins present in our analysis and ≥ 1 of the published HIV-1 datasets. Many proteins were associated with nuclear processes which could have functional consequences for viral replication, including transcription initiation/elongation/termination, RNA processing, splicing, and chromatin remodeling. Examples include facilitating chromatin remodeling to expose the integrated provirus, promoting expression of viral genes, repressing the transcription of antagonistic cellular genes, preventing splicing of viral RNA, altering splicing of cellular RNAs, or influencing viral or host RNA folding or RNA nuclear export. Many proteins in our pulldowns common to RSV and HIV-1 Gag are critical for transcription, including PolR2B, the second largest subunit of RNA polymerase II (RNAPII), and LEO1, a PAF1C complex member that regulates transcriptional elongation, supporting the possibility that Gag influences the host transcription profile to aid the virus. Through the interaction of RSV and HIV-1 Gag with splicing-related proteins CBLL1, HNRNPH3, TRA2B, PTBP1 and U2AF1, we speculate that Gag could enhance unspliced viral RNA production for translation and packaging. To validate one putative hit, we demonstrated an interaction of RSV Gag with Mediator complex member Med26, required for RNA polymerase II-mediated transcription. Although 57 host proteins interacted with both Gag proteins, unique host proteins belonging to each interactome dataset were identified. These results provide a strong premise for future functional studies to investigate roles for these nuclear host factors that may have shared functions in the biology of both retroviruses, as well as functions specific to RSV and HIV-1, given their distinctive hosts and molecular pathology.

逆转录病毒利用宿主蛋白组装病毒并从感染细胞中释放病毒。以前,大多数研究侧重于逆转录病毒 Gag 蛋白在细胞质或质膜上的相互作用伙伴。鉴于在细胞核中发现了几种全长的 Gag 蛋白,鉴定 Gag 与核的相互作用组很有可能发现涉及以前未知的宿主过程的新发现。在这里,我们系统地比较了已发表的 HIV-1 蛋白质组学研究中发现的核因子,并使用亲和标记的 HIV-1 和 RSV Gag 蛋白与核提取物混合进行了我们自己的质谱分析。我们在 HIV-1 和 RSV Gag 之间发现了 57 个共同的核蛋白,在我们的分析中发现了一组核蛋白,而在已发表的 HIV-1 数据集中发现的核蛋白≥1 个。许多蛋白质与核过程有关,这些过程可能会对病毒复制产生功能性影响,包括转录启动/延长/终止、RNA 处理、剪接和染色质重塑。例如,促进染色质重塑以暴露整合的前病毒、促进病毒基因的表达、抑制拮抗细胞基因的转录、阻止病毒 RNA 的剪接、改变细胞 RNA 的剪接、或影响病毒或宿主 RNA 折叠或 RNA 核输出。在我们与 RSV 和 HIV-1 Gag 共同的 pulldowns 中,许多蛋白质对转录至关重要,包括 RNA 聚合酶 II (RNAPII) 的第二大亚基 PolR2B 和 LEO1,LEO1 是 PAF1C 复合物成员,可调节转录伸长,这支持了 Gag 影响宿主转录概况以帮助病毒的可能性。通过 RSV 和 HIV-1 Gag 与剪接相关蛋白 CBLL1、HNRNPH3、TRA2B、PTBP1 和 U2AF1 的相互作用,我们推测 Gag 可能会增强未剪接病毒 RNA 的产生,从而促进翻译和包装。为了验证一个推测的结果,我们证明了 RSV Gag 与 Mediator 复合体成员 Med26 的相互作用,Med26 是 RNA 聚合酶 II 介导的转录所必需的。虽然有 57 种宿主蛋白与两种 Gag 蛋白发生了相互作用,但每个相互作用组数据集中都发现了独特的宿主蛋白。这些结果为今后的功能研究提供了一个强有力的前提,以研究这些核宿主因子在两种逆转录病毒生物学中可能具有的共同功能,以及 RSV 和 HIV-1 因其独特的宿主和分子病理学而具有的特定功能。
{"title":"Comparative analysis of retroviral Gag-host cell interactions: focus on the nuclear interactome.","authors":"Gregory S Lambert, Breanna L Rice, Rebecca J Kaddis Maldonado, Jordan Chang, Leslie J Parent","doi":"10.1186/s12977-024-00645-y","DOIUrl":"10.1186/s12977-024-00645-y","url":null,"abstract":"<p><p>Retroviruses exploit host proteins to assemble and release virions from infected cells. Previously, most studies focused on interacting partners of retroviral Gag proteins that localize to the cytoplasm or plasma membrane. Given that several full-length Gag proteins have been found in the nucleus, identifying the Gag-nuclear interactome has high potential for novel findings involving previously unknown host processes. Here we systematically compared nuclear factors identified in published HIV-1 proteomic studies and performed our own mass spectrometry analysis using affinity-tagged HIV-1 and RSV Gag proteins mixed with nuclear extracts. We identified 57 nuclear proteins in common between HIV-1 and RSV Gag, and a set of nuclear proteins present in our analysis and ≥ 1 of the published HIV-1 datasets. Many proteins were associated with nuclear processes which could have functional consequences for viral replication, including transcription initiation/elongation/termination, RNA processing, splicing, and chromatin remodeling. Examples include facilitating chromatin remodeling to expose the integrated provirus, promoting expression of viral genes, repressing the transcription of antagonistic cellular genes, preventing splicing of viral RNA, altering splicing of cellular RNAs, or influencing viral or host RNA folding or RNA nuclear export. Many proteins in our pulldowns common to RSV and HIV-1 Gag are critical for transcription, including PolR2B, the second largest subunit of RNA polymerase II (RNAPII), and LEO1, a PAF1C complex member that regulates transcriptional elongation, supporting the possibility that Gag influences the host transcription profile to aid the virus. Through the interaction of RSV and HIV-1 Gag with splicing-related proteins CBLL1, HNRNPH3, TRA2B, PTBP1 and U2AF1, we speculate that Gag could enhance unspliced viral RNA production for translation and packaging. To validate one putative hit, we demonstrated an interaction of RSV Gag with Mediator complex member Med26, required for RNA polymerase II-mediated transcription. Although 57 host proteins interacted with both Gag proteins, unique host proteins belonging to each interactome dataset were identified. These results provide a strong premise for future functional studies to investigate roles for these nuclear host factors that may have shared functions in the biology of both retroviruses, as well as functions specific to RSV and HIV-1, given their distinctive hosts and molecular pathology.</p>","PeriodicalId":21123,"journal":{"name":"Retrovirology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11186191/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141427479","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Retroviral PBS-segment sequence and structure: Orchestrating early and late replication events. 逆转录病毒 PBS 片段序列和结构:协调早期和晚期复制事件
IF 2.7 3区 医学 Q2 Medicine Pub Date : 2024-06-17 DOI: 10.1186/s12977-024-00646-x
Xiao Heng, Amanda Paz Herrera, Zhenwei Song, Kathleen Boris-Lawrie

An essential regulatory hub for retroviral replication events, the 5' untranslated region (UTR) encodes an ensemble of cis-acting replication elements that overlap in a logical manner to carry out divergent RNA activities in cells and in virions. The primer binding site (PBS) and primer activation sequence initiate the reverse transcription process in virions, yet overlap with structural elements that regulate expression of the complex viral proteome. PBS-segment also encompasses the attachment site for Integrase to cut and paste the 3' long terminal repeat into the host chromosome to form the provirus and purine residues necessary to execute the precise stoichiometry of genome-length transcripts and spliced viral RNAs. Recent genetic mapping, cofactor affinity experiments, NMR and SAXS have elucidated that the HIV-1 PBS-segment folds into a three-way junction structure. The three-way junction structure is recognized by the host's nuclear RNA helicase A/DHX9 (RHA). RHA tethers host trimethyl guanosine synthase 1 to the Rev/Rev responsive element (RRE)-containing RNAs for m7-guanosine Cap hyper methylation that bolsters virion infectivity significantly. The HIV-1 trimethylated (TMG) Cap licenses specialized translation of virion proteins under conditions that repress translation of the regulatory proteins. Clearly host-adaption and RNA shapeshifting comprise the fundamental basis for PBS-segment orchestrating both reverse transcription of virion RNA and the nuclear modification of m7G-Cap for biphasic translation of the complex viral proteome. These recent observations, which have exposed even greater complexity of retroviral RNA biology than previously established, are the impetus for this article. Basic research to fully comprehend the marriage of PBS-segment structures and host RNA binding proteins that carry out retroviral early and late replication events is likely to expose an immutable virus-specific therapeutic target to attenuate retrovirus proliferation.

5' 非翻译区(UTR)是逆转录病毒复制活动的重要调控枢纽,它编码了一系列顺式复制元件,这些元件以合理的方式重叠在一起,在细胞和病毒中执行不同的 RNA 活动。引物结合位点(PBS)和引物激活序列启动了病毒中的反转录过程,但又与调控复杂病毒蛋白质组表达的结构元件重叠。PBS 片段还包括整合酶的附着位点,用于将 3' 长末端重复序列剪切并粘贴到宿主染色体上,以形成前病毒和嘌呤残基,这些残基是执行基因组长转录本和剪接病毒 RNA 精确配比所必需的。最近的基因图谱、辅因子亲和性实验、核磁共振(NMR)和 SAXS 阐明,HIV-1 PBS 片段折叠成一个三向连接结构。宿主的核 RNA 螺旋酶 A/DHX9 (RHA) 能识别这种三向连接结构。RHA 将宿主的三甲基鸟苷合成酶 1 与含 Rev/Rev 反应元件 (RRE) 的 RNA 绑在一起,进行 m7-鸟苷帽超甲基化,从而显著增强病毒的感染力。在抑制调控蛋白翻译的条件下,HIV-1 三甲基化(TMG)Cap 允许病毒蛋白进行专门翻译。显然,宿主适应和 RNA 变形是 PBS 片段协调病毒 RNA 逆转录和 m7G-Cap 核修饰以实现复杂病毒蛋白组双相翻译的根本基础。这些最新观察结果揭示了逆转录病毒 RNA 生物学的复杂性,其复杂程度甚至超过了以前的研究成果,这也是本文写作的动力所在。通过基础研究来充分理解 PBS 片段结构与宿主 RNA 结合蛋白的结合,从而完成逆转录病毒的早期和晚期复制事件,很可能会发现一个永恒不变的病毒特异性治疗靶点,以减弱逆转录病毒的增殖。
{"title":"Retroviral PBS-segment sequence and structure: Orchestrating early and late replication events.","authors":"Xiao Heng, Amanda Paz Herrera, Zhenwei Song, Kathleen Boris-Lawrie","doi":"10.1186/s12977-024-00646-x","DOIUrl":"10.1186/s12977-024-00646-x","url":null,"abstract":"<p><p>An essential regulatory hub for retroviral replication events, the 5' untranslated region (UTR) encodes an ensemble of cis-acting replication elements that overlap in a logical manner to carry out divergent RNA activities in cells and in virions. The primer binding site (PBS) and primer activation sequence initiate the reverse transcription process in virions, yet overlap with structural elements that regulate expression of the complex viral proteome. PBS-segment also encompasses the attachment site for Integrase to cut and paste the 3' long terminal repeat into the host chromosome to form the provirus and purine residues necessary to execute the precise stoichiometry of genome-length transcripts and spliced viral RNAs. Recent genetic mapping, cofactor affinity experiments, NMR and SAXS have elucidated that the HIV-1 PBS-segment folds into a three-way junction structure. The three-way junction structure is recognized by the host's nuclear RNA helicase A/DHX9 (RHA). RHA tethers host trimethyl guanosine synthase 1 to the Rev/Rev responsive element (RRE)-containing RNAs for m7-guanosine Cap hyper methylation that bolsters virion infectivity significantly. The HIV-1 trimethylated (TMG) Cap licenses specialized translation of virion proteins under conditions that repress translation of the regulatory proteins. Clearly host-adaption and RNA shapeshifting comprise the fundamental basis for PBS-segment orchestrating both reverse transcription of virion RNA and the nuclear modification of m7G-Cap for biphasic translation of the complex viral proteome. These recent observations, which have exposed even greater complexity of retroviral RNA biology than previously established, are the impetus for this article. Basic research to fully comprehend the marriage of PBS-segment structures and host RNA binding proteins that carry out retroviral early and late replication events is likely to expose an immutable virus-specific therapeutic target to attenuate retrovirus proliferation.</p>","PeriodicalId":21123,"journal":{"name":"Retrovirology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11181671/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141420619","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
HIV-1 with gag processing defects activates cGAS sensing. 存在凝集素处理缺陷的 HIV-1 可激活 cGAS 传感。
IF 3.3 3区 医学 Q2 Medicine Pub Date : 2024-05-23 DOI: 10.1186/s12977-024-00643-0
Rebecca P Sumner, Henry Blest, Meiyin Lin, Carlos Maluquer de Motes, Greg J Towers

Background: Detection of viruses by host pattern recognition receptors induces the expression of type I interferon (IFN) and IFN-stimulated genes (ISGs), which suppress viral replication. Numerous studies have described HIV-1 as a poor activator of innate immunity in vitro. The exact role that the viral capsid plays in this immune evasion is not fully understood.

Results: To better understand the role of the HIV-1 capsid in sensing we tested the effect of making HIV-1 by co-expressing a truncated Gag that encodes the first 107 amino acids of capsid fused with luciferase or GFP, alongside wild type Gag-pol. We found that unlike wild type HIV-1, viral particles produced with a mixture of wild type and truncated Gag fused to luciferase or GFP induced a potent IFN response in THP-1 cells and macrophages. Innate immune activation by Gag-fusion HIV-1 was dependent on reverse transcription and DNA sensor cGAS, suggesting activation of an IFN response by viral DNA. Further investigation revealed incorporation of the Gag-luciferase/GFP fusion proteins into viral particles that correlated with subtle defects in wild type Gag cleavage and a diminished capacity to saturate restriction factor TRIM5α, likely due to aberrant particle formation. We propose that expression of the Gag fusion protein disturbs the correct cleavage and maturation of wild type Gag, yielding viral particles that are unable to effectively shield viral DNA from detection by innate sensors including cGAS.

Conclusions: These data highlight the crucial role of capsid in innate evasion and support growing literature that disruption of Gag cleavage and capsid formation induces a viral DNA- and cGAS-dependent innate immune response. Together these data demonstrate a protective role for capsid and suggest that antiviral activity of capsid-targeting antivirals may benefit from enhanced innate and adaptive immunity in vivo.

背景:宿主模式识别受体对病毒的检测会诱导 I 型干扰素(IFN)和 IFN 刺激基因(ISGs)的表达,从而抑制病毒复制。大量研究表明,HIV-1 在体外对先天性免疫的激活作用很弱。目前还不完全清楚病毒外壳在这种免疫逃避中的确切作用:为了更好地了解 HIV-1 病毒噬菌体在感知中的作用,我们测试了通过将编码噬菌体前 107 个氨基酸的截短 Gag 与荧光素酶或 GFP 融合后与野生型 Gag-pol 共同表达来制造 HIV-1 的效果。我们发现,与野生型HIV-1不同,用野生型和与荧光素酶或GFP融合的截短Gag混合产生的病毒颗粒能诱导THP-1细胞和巨噬细胞产生强效的IFN反应。融合了 Gag 的 HIV-1 对先天性免疫的激活依赖于反转录和 DNA 传感器 cGAS,这表明病毒 DNA 激活了 IFN 反应。进一步研究发现,Gag-荧光素酶/GFP融合蛋白与病毒颗粒的结合与野生型Gag裂解的微小缺陷和限制因子TRIM5α的饱和能力有关,这可能是由于颗粒形成异常所致。我们认为,Gag融合蛋白的表达干扰了野生型Gag的正确裂解和成熟,产生的病毒颗粒不能有效地保护病毒DNA不被包括cGAS在内的先天传感器检测到:这些数据强调了噬菌体在先天逃避中的关键作用,并支持了越来越多的文献,即破坏 Gag 的裂解和噬菌体的形成会诱发依赖于病毒 DNA 和 cGAS 的先天免疫反应。这些数据共同证明了噬菌体的保护作用,并表明噬菌体靶向抗病毒药物的抗病毒活性可能得益于体内先天和适应性免疫的增强。
{"title":"HIV-1 with gag processing defects activates cGAS sensing.","authors":"Rebecca P Sumner, Henry Blest, Meiyin Lin, Carlos Maluquer de Motes, Greg J Towers","doi":"10.1186/s12977-024-00643-0","DOIUrl":"10.1186/s12977-024-00643-0","url":null,"abstract":"<p><strong>Background: </strong>Detection of viruses by host pattern recognition receptors induces the expression of type I interferon (IFN) and IFN-stimulated genes (ISGs), which suppress viral replication. Numerous studies have described HIV-1 as a poor activator of innate immunity in vitro. The exact role that the viral capsid plays in this immune evasion is not fully understood.</p><p><strong>Results: </strong>To better understand the role of the HIV-1 capsid in sensing we tested the effect of making HIV-1 by co-expressing a truncated Gag that encodes the first 107 amino acids of capsid fused with luciferase or GFP, alongside wild type Gag-pol. We found that unlike wild type HIV-1, viral particles produced with a mixture of wild type and truncated Gag fused to luciferase or GFP induced a potent IFN response in THP-1 cells and macrophages. Innate immune activation by Gag-fusion HIV-1 was dependent on reverse transcription and DNA sensor cGAS, suggesting activation of an IFN response by viral DNA. Further investigation revealed incorporation of the Gag-luciferase/GFP fusion proteins into viral particles that correlated with subtle defects in wild type Gag cleavage and a diminished capacity to saturate restriction factor TRIM5α, likely due to aberrant particle formation. We propose that expression of the Gag fusion protein disturbs the correct cleavage and maturation of wild type Gag, yielding viral particles that are unable to effectively shield viral DNA from detection by innate sensors including cGAS.</p><p><strong>Conclusions: </strong>These data highlight the crucial role of capsid in innate evasion and support growing literature that disruption of Gag cleavage and capsid formation induces a viral DNA- and cGAS-dependent innate immune response. Together these data demonstrate a protective role for capsid and suggest that antiviral activity of capsid-targeting antivirals may benefit from enhanced innate and adaptive immunity in vivo.</p>","PeriodicalId":21123,"journal":{"name":"Retrovirology","volume":null,"pages":null},"PeriodicalIF":3.3,"publicationDate":"2024-05-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11112816/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141080567","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Inhibition of caspase pathways limits CD4+ T cell loss and restores host anti-retroviral function in HIV-1 infected humanized mice with augmented lymphoid tissue 抑制 Caspase 通路可限制 CD4+ T 细胞的损失,并恢复淋巴组织增大的 HIV-1 感染人源化小鼠宿主的抗逆转录病毒功能
IF 3.3 3区 医学 Q2 Medicine Pub Date : 2024-05-02 DOI: 10.1186/s12977-024-00641-2
Alex J. Holloway, Tais B. Saito, Kubra F. Naqvi, Matthew B. Huante, Xiuzhen Fan, Joshua G. Lisinicchia, Benjamin B. Gelman, Janice J. Endsley, Mark A. Endsley
The study of HIV infection and pathogenicity in physical reservoirs requires a biologically relevant model. The human immune system (HIS) mouse is an established model of HIV infection, but defects in immune tissue reconstitution remain a challenge for examining pathology in tissues. We utilized exogenous injection of the human recombinant FMS-like tyrosine kinase 3 ligand (rFLT-3 L) into the hematopoietic stem cell (HSC) cord blood HIS mouse model to significantly expand the total area of lymph node (LN) and the number of circulating human T cells. The results enabled visualization and quantification of HIV infectivity, CD4 T cell depletion and other measures of pathogenesis in the secondary lymphoid tissues of the spleen and LN. Treatment with the Caspase-1/4 inhibitor VX-765 limited CD4+ T cell loss in the spleen and reduced viral load in both the spleen and axillary LN. In situ hybridization further demonstrated a decrease in viral RNA in both the spleen and LN. Transcriptomic analysis revealed that in vivo inhibition of caspase-1/4 led to an upregulation in host HIV restriction factors including SAMHD1 and APOBEC3A. These findings highlight the use of rFLT-3 L to augment human immune system characteristics in HIS mice to support investigations of HIV pathogenesis and test host directed therapies, though further refinements are needed to further augment LN architecture and cellular populations. The results further provide in vivo evidence of the potential to target inflammasome pathways as an avenue of host-directed therapy to limit immune dysfunction and virus replication in tissue compartments of HIV+ persons.
研究艾滋病毒在物理储库中的感染和致病性需要一个与生物相关的模型。人类免疫系统(HIS)小鼠是一种成熟的艾滋病病毒感染模型,但免疫组织重建的缺陷仍然是研究组织病理学的一个挑战。我们利用向造血干细胞(HSC)脐带血人类免疫系统小鼠模型中外源性注射人类重组FMS样酪氨酸激酶3配体(rFLT-3 L)的方法,显著扩大了淋巴结(LN)的总面积和循环人类T细胞的数量。研究结果实现了脾脏和淋巴结二级淋巴组织中艾滋病毒感染性、CD4 T细胞耗竭及其他致病因素的可视化和量化。Caspase-1/4抑制剂VX-765的治疗限制了脾脏中CD4+ T细胞的损失,并降低了脾脏和腋窝淋巴结中的病毒载量。原位杂交进一步表明,脾脏和LN中的病毒RNA均有所减少。转录组分析表明,体内抑制 caspase-1/4 导致宿主 HIV 限制因子(包括 SAMHD1 和 APOBEC3A)上调。这些发现强调了利用 rFLT-3 L 增强 HIS 小鼠的人类免疫系统特征,以支持对 HIV 发病机制的研究和测试宿主导向疗法,不过还需要进一步改进,以进一步增强 LN 结构和细胞群。这些结果进一步提供了体内证据,证明以炎性体通路为靶点作为宿主导向疗法的一种途径,有可能限制 HIV 感染者组织中的免疫功能障碍和病毒复制。
{"title":"Inhibition of caspase pathways limits CD4+ T cell loss and restores host anti-retroviral function in HIV-1 infected humanized mice with augmented lymphoid tissue","authors":"Alex J. Holloway, Tais B. Saito, Kubra F. Naqvi, Matthew B. Huante, Xiuzhen Fan, Joshua G. Lisinicchia, Benjamin B. Gelman, Janice J. Endsley, Mark A. Endsley","doi":"10.1186/s12977-024-00641-2","DOIUrl":"https://doi.org/10.1186/s12977-024-00641-2","url":null,"abstract":"The study of HIV infection and pathogenicity in physical reservoirs requires a biologically relevant model. The human immune system (HIS) mouse is an established model of HIV infection, but defects in immune tissue reconstitution remain a challenge for examining pathology in tissues. We utilized exogenous injection of the human recombinant FMS-like tyrosine kinase 3 ligand (rFLT-3 L) into the hematopoietic stem cell (HSC) cord blood HIS mouse model to significantly expand the total area of lymph node (LN) and the number of circulating human T cells. The results enabled visualization and quantification of HIV infectivity, CD4 T cell depletion and other measures of pathogenesis in the secondary lymphoid tissues of the spleen and LN. Treatment with the Caspase-1/4 inhibitor VX-765 limited CD4+ T cell loss in the spleen and reduced viral load in both the spleen and axillary LN. In situ hybridization further demonstrated a decrease in viral RNA in both the spleen and LN. Transcriptomic analysis revealed that in vivo inhibition of caspase-1/4 led to an upregulation in host HIV restriction factors including SAMHD1 and APOBEC3A. These findings highlight the use of rFLT-3 L to augment human immune system characteristics in HIS mice to support investigations of HIV pathogenesis and test host directed therapies, though further refinements are needed to further augment LN architecture and cellular populations. The results further provide in vivo evidence of the potential to target inflammasome pathways as an avenue of host-directed therapy to limit immune dysfunction and virus replication in tissue compartments of HIV+ persons.","PeriodicalId":21123,"journal":{"name":"Retrovirology","volume":null,"pages":null},"PeriodicalIF":3.3,"publicationDate":"2024-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140840089","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: YTHDC1 regulates distinct post-integration steps of HIV-1 replication and is important for viral infectivity 更正:YTHDC1调节HIV-1复制过程中不同的整合后步骤,对病毒的感染性非常重要
IF 3.3 3区 医学 Q2 Medicine Pub Date : 2024-04-30 DOI: 10.1186/s12977-024-00642-1
Sarah N’Da Konan, Emmanuel Ségéral, Fabienne Bejjani, Maryam Bendoumou, Mélissa Ait Said, Sarah Gallois-Montbrun, Stéphane Emiliani

Following publication of the original article [1], we have been notified that within the Abstract, the word “writer” needs to be replaced with “reader”.

The original article has been corrected.

  1. N’Da Konan et al. Retrovirology. 2022;19:4. https://doi.org/10.1186/s12977-022-00589-1

Download references

Authors and Affiliations

  1. Institut Cochin, INSERM, CNRS, Université de Paris, 75014, Paris, France

    Sarah N’Da Konan, Emmanuel Ségéral, Fabienne Bejjani, Mélissa Ait Said, Sarah Gallois-Montbrun & Stéphane Emiliani

  2. Ser vice of Molecular Virology, Department of Molecular Biology, Université Libre de Bruxelles, 6041, Gosselies, Belgium

    Maryam Bendoumou

Authors
  1. Sarah N’Da KonanView author publications

    You can also search for this author in PubMed Google Scholar

  2. Emmanuel SégéralView author publications

    You can also search for this author in PubMed Google Scholar

  3. Fabienne BejjaniView author publications

    You can also search for this author in PubMed Google Scholar

  4. Maryam BendoumouView author publications

    You can also search for this author in PubMed Google Scholar

  5. Mélissa Ait SaidView author publications

    You can also search for this author in PubMed Google Scholar

  6. Sarah Gallois-MontbrunView author publications

    You can also search for this author in PubMed Google Scholar

  7. Stéphane EmilianiView author publications

    You can also search for this author in PubMed Google Scholar

Corresponding authors

Correspondence to Sarah Gallois-Montbrun or Stéphane Emiliani.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

The online version of the original article can be found at https://doi.org/10.1186/s12977-022-00589-1.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line

原文[1]发表后,我们接到通知,摘要中的 "作者 "一词需改为 "读者"。2022;19:4. https://doi.org/10.1186/s12977-022-00589-1下载参考文献作者及单位法国巴黎大学柯钦研究所、法国国家科学研究中心、法国国家科学研究中心,75014,巴黎,法国Sarah N'Da Konan, Emmanuel Ségéral, Fabienne Bejjani, Mélissa Ait Said, Sarah Gallois-Montbrun &;Stéphane EmilianiSer vice of Molecular Virology, Department of Molecular Biology, Université Libre de Bruxelles, 6041, Gosselies、比利时Maryam Bendoumou作者Sarah N'Da Konan查看作者发表的文章您也可以在PubMed Google Scholar中搜索该作者Emmanuel Ségéral查看作者发表的文章您也可以在PubMed Google Scholar中搜索该作者Fabienne Bejjani查看作者发表的文章您也可以在PubMed Google Scholar中搜索该作者您也可以在 PubMed Google Scholar中搜索该作者Maryam Bendoumou查看作者发表的论文您也可以在 PubMed Google Scholar中搜索该作者Mélissa Ait Said查看作者发表的论文您也可以在 PubMed Google Scholar中搜索该作者Sarah Gallois-Montbrun查看作者发表的作品您也可以在PubMed Google Scholar中搜索该作者Stéphane Emiliani查看作者发表的作品您也可以在PubMed Google Scholar中搜索该作者通信作者Sarah Gallois-Montbrun或Stéphane Emiliani。出版者注Springer Nature对已出版地图中的管辖权主张和机构隶属关系保持中立。原文的在线版本可在以下网址找到:https://doi.org/10.1186/s12977-022-00589-1.Open Access 本文采用知识共享署名 4.0 国际许可协议进行许可,该协议允许以任何媒介或格式使用、共享、改编、分发和复制,只要您适当注明原作者和来源,提供知识共享许可协议的链接,并说明是否进行了修改。本文中的图片或其他第三方材料均包含在文章的知识共享许可协议中,除非在材料的署名栏中另有说明。如果材料未包含在文章的知识共享许可协议中,且您打算使用的材料不符合法律规定或超出许可使用范围,您需要直接从版权所有者处获得许可。要查看该许可的副本,请访问 http://creativecommons.org/licenses/by/4.0/。除非在数据的信用行中另有说明,否则知识共享公共领域专用免责声明 (http://creativecommons.org/publicdomain/zero/1.0/) 适用于本文提供的数据。转载与许可引用本文Konan, S.N., Ségéral, E., Bejjani, F. et al. Correction:YTHDC1调控HIV-1复制的不同整合后步骤,对病毒的感染性很重要。Retrovirology 21, 9 (2024). https://doi.org/10.1186/s12977-024-00642-1Download citationPublished: 30 April 2024DOI: https://doi.org/10.1186/s12977-024-00642-1Share this articleAnyone you share the following link with will be able to read this content:Get shareable linkSorry, a shareable link is not currently available for this article.Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative.
{"title":"Correction: YTHDC1 regulates distinct post-integration steps of HIV-1 replication and is important for viral infectivity","authors":"Sarah N’Da Konan, Emmanuel Ségéral, Fabienne Bejjani, Maryam Bendoumou, Mélissa Ait Said, Sarah Gallois-Montbrun, Stéphane Emiliani","doi":"10.1186/s12977-024-00642-1","DOIUrl":"https://doi.org/10.1186/s12977-024-00642-1","url":null,"abstract":"<p>Following publication of the original article [1], we have been notified that within the Abstract, the word “writer” needs to be replaced with “reader”.</p><p>The original article has been corrected.</p><ol data-track-component=\"outbound reference\"><li data-counter=\"1.\"><p>N’Da Konan et al. Retrovirology. 2022;19:4. https://doi.org/10.1186/s12977-022-00589-1</p></li></ol><p>Download references<svg aria-hidden=\"true\" focusable=\"false\" height=\"16\" role=\"img\" width=\"16\"><use xlink:href=\"#icon-eds-i-download-medium\" xmlns:xlink=\"http://www.w3.org/1999/xlink\"></use></svg></p><h3>Authors and Affiliations</h3><ol><li><p>Institut Cochin, INSERM, CNRS, Université de Paris, 75014, Paris, France</p><p>Sarah N’Da Konan, Emmanuel Ségéral, Fabienne Bejjani, Mélissa Ait Said, Sarah Gallois-Montbrun &amp; Stéphane Emiliani</p></li><li><p>Ser vice of Molecular Virology, Department of Molecular Biology, Université Libre de Bruxelles, 6041, Gosselies, Belgium</p><p>Maryam Bendoumou</p></li></ol><span>Authors</span><ol><li><span>Sarah N’Da Konan</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Emmanuel Ségéral</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Fabienne Bejjani</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Maryam Bendoumou</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Mélissa Ait Said</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Sarah Gallois-Montbrun</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Stéphane Emiliani</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li></ol><h3>Corresponding authors</h3><p>Correspondence to Sarah Gallois-Montbrun or Stéphane Emiliani.</p><h3>Publisher’s Note</h3><p>Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.</p><p>The online version of the original article can be found at https://doi.org/10.1186/s12977-022-00589-1.</p><p><b>Open Access</b> This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line ","PeriodicalId":21123,"journal":{"name":"Retrovirology","volume":null,"pages":null},"PeriodicalIF":3.3,"publicationDate":"2024-04-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140840617","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
HERV-W upregulation expression in bipolar disorder and schizophrenia: unraveling potential links to systemic immune/inflammation status 双相情感障碍和精神分裂症中的 HERV-W 上调表达:揭示与全身免疫/炎症状态的潜在联系
IF 3.3 3区 医学 Q2 Medicine Pub Date : 2024-04-22 DOI: 10.1186/s12977-024-00640-3
Sara Coelho Rangel, Michelly Damasceno da Silva, Décio Gilberto Natrielli Filho, Samuel Nascimento Santos, Jonatas Bussador do Amaral, Jefferson Russo Victor, Kevin Cezar Nascimento Silva, Izabela Dorota Tuleta, Carolina Nunes França, Marina Tiemi Shio, Lucas Melo Neves, André Luis Lacerda Bachi, Luiz Henrique da Silva Nali
Bipolar disorder (BD) and schizophrenia (SZ) are the two main mental disorders with unknown etiology that significantly impact individuals’ quality of life. The potential pro-inflammatory role in their pathogenesis is postulated and Human Endogenous Retrovirus W (HERV-W) is an emerging candidate to modulate this pathogenic finding. HERVs, ancient retroviruses in the human genome, may play roles in inflammation and disease pathogenesis. Despite HERVs’ involvement in autoimmune diseases, their influence on mental disorders remains underexplored. Therefore, the aim of this study was to assess the level of HERV-W-env expression and the systemic inflammatory profile through the concentration of IL-2, IL-4, IL-6, IL-10, TNF-α and INF-γ cytokines in BD and SZ patients. All participants showed HERV-W-env expression, but its expression was higher in mental disorder patients (p < 0.01) than in control. When separated, SZ individuals exhibited higher HERV-W expression than the control group (p < 0.01). Higher serum levels of TNF-α and IL-10 were found in BD (p = 0.0001 and p = 0.001, respectively) and SZ (p = 0.01) and p = 0.01, respectively) than in the control group, while SZ showed decreased levels IFN-γ and IL-2 as compared to controls (p = 0.05) and BD patients (p = 0.05), respectively. Higher TNF-α/IL-4 and TNF-α/IL-10 ratios, and lower IFN-γ/IL-10 were observed in BD and SZ patients than controls. Significant negative correlation between HERV-W-env expression and IL-10 (r=-0.47 p < 0.05), as well as positive correlations between HERV-W-env expression and TNF-α/IL-10 or IFN-γ/IL-10 ratios (r = 0.48 p < 0.05 and r = 0.46 p < 0.05, respectively) were found in BD patients. These findings suggest not only a potential link between HERV-W-env expression both in BD and SZ, but also a possible involvement of systemic inflammatory status in BD patients.
躁郁症(BD)和精神分裂症(SZ)是两种病因不明的主要精神疾病,严重影响患者的生活质量。人类内源性逆转录病毒 W(HERV-W)被认为在这两种疾病的发病机制中起着潜在的促炎症作用,而人类内源性逆转录病毒 W 是调节这一发病机制的新兴候选病毒。HERVs 是人类基因组中古老的逆转录病毒,可能在炎症和疾病发病机制中发挥作用。尽管 HERVs 参与了自身免疫性疾病,但其对精神疾病的影响仍未得到充分探索。因此,本研究旨在通过检测 BD 和 SZ 患者体内 IL-2、IL-4、IL-6、IL-10、TNF-α 和 INF-γ 细胞因子的浓度,评估 HERV-W-env 的表达水平和全身炎症状况。所有参与者都有 HERV-W-env 表达,但精神障碍患者的 HERV-W-env 表达高于对照组(p < 0.01)。如果分开来看,精神分裂症患者的 HERV-W 表达高于对照组(p < 0.01)。与对照组相比,BD(分别为 p = 0.0001 和 p = 0.001)和 SZ(分别为 p = 0.01 和 p = 0.01)患者血清中的 TNF-α 和 IL-10 水平较高;与对照组(p = 0.05)和 BD 患者(p = 0.05)相比,SZ 患者血清中的 IFN-γ 和 IL-2 水平较低。与对照组相比,BD 和 SZ 患者的 TNF-α/IL-4 和 TNF-α/IL-10 比率较高,而 IFN-γ/IL-10 比率较低。在 BD 患者中,HERV-W-env 表达与 IL-10 呈显著负相关(r=-0.47 p < 0.05),HERV-W-env 表达与 TNF-α/IL-10 或 IFN-γ/IL-10 比率呈正相关(分别为 r = 0.48 p < 0.05 和 r = 0.46 p < 0.05)。这些发现不仅表明 HERV-W-env 表达在 BD 和 SZ 中存在潜在联系,而且可能与 BD 患者的全身炎症状态有关。
{"title":"HERV-W upregulation expression in bipolar disorder and schizophrenia: unraveling potential links to systemic immune/inflammation status","authors":"Sara Coelho Rangel, Michelly Damasceno da Silva, Décio Gilberto Natrielli Filho, Samuel Nascimento Santos, Jonatas Bussador do Amaral, Jefferson Russo Victor, Kevin Cezar Nascimento Silva, Izabela Dorota Tuleta, Carolina Nunes França, Marina Tiemi Shio, Lucas Melo Neves, André Luis Lacerda Bachi, Luiz Henrique da Silva Nali","doi":"10.1186/s12977-024-00640-3","DOIUrl":"https://doi.org/10.1186/s12977-024-00640-3","url":null,"abstract":"Bipolar disorder (BD) and schizophrenia (SZ) are the two main mental disorders with unknown etiology that significantly impact individuals’ quality of life. The potential pro-inflammatory role in their pathogenesis is postulated and Human Endogenous Retrovirus W (HERV-W) is an emerging candidate to modulate this pathogenic finding. HERVs, ancient retroviruses in the human genome, may play roles in inflammation and disease pathogenesis. Despite HERVs’ involvement in autoimmune diseases, their influence on mental disorders remains underexplored. Therefore, the aim of this study was to assess the level of HERV-W-env expression and the systemic inflammatory profile through the concentration of IL-2, IL-4, IL-6, IL-10, TNF-α and INF-γ cytokines in BD and SZ patients. All participants showed HERV-W-env expression, but its expression was higher in mental disorder patients (p < 0.01) than in control. When separated, SZ individuals exhibited higher HERV-W expression than the control group (p < 0.01). Higher serum levels of TNF-α and IL-10 were found in BD (p = 0.0001 and p = 0.001, respectively) and SZ (p = 0.01) and p = 0.01, respectively) than in the control group, while SZ showed decreased levels IFN-γ and IL-2 as compared to controls (p = 0.05) and BD patients (p = 0.05), respectively. Higher TNF-α/IL-4 and TNF-α/IL-10 ratios, and lower IFN-γ/IL-10 were observed in BD and SZ patients than controls. Significant negative correlation between HERV-W-env expression and IL-10 (r=-0.47 p < 0.05), as well as positive correlations between HERV-W-env expression and TNF-α/IL-10 or IFN-γ/IL-10 ratios (r = 0.48 p < 0.05 and r = 0.46 p < 0.05, respectively) were found in BD patients. These findings suggest not only a potential link between HERV-W-env expression both in BD and SZ, but also a possible involvement of systemic inflammatory status in BD patients.","PeriodicalId":21123,"journal":{"name":"Retrovirology","volume":null,"pages":null},"PeriodicalIF":3.3,"publicationDate":"2024-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140636431","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The cell biology of HIV-1 latency and rebound HIV-1 潜伏期和反弹的细胞生物学
IF 3.3 3区 医学 Q2 Medicine Pub Date : 2024-04-05 DOI: 10.1186/s12977-024-00639-w
Uri Mbonye, Jonathan Karn
Transcriptionally latent forms of replication-competent proviruses, present primarily in a small subset of memory CD4+ T cells, pose the primary barrier to a cure for HIV-1 infection because they are the source of the viral rebound that almost inevitably follows the interruption of antiretroviral therapy. Over the last 30 years, many of the factors essential for initiating HIV-1 transcription have been identified in studies performed using transformed cell lines, such as the Jurkat T-cell model. However, as highlighted in this review, several poorly understood mechanisms still need to be elucidated, including the molecular basis for promoter-proximal pausing of the transcribing complex and the detailed mechanism of the delivery of P-TEFb from 7SK snRNP. Furthermore, the central paradox of HIV-1 transcription remains unsolved: how are the initial rounds of transcription achieved in the absence of Tat? A critical limitation of the transformed cell models is that they do not recapitulate the transitions between active effector cells and quiescent memory T cells. Therefore, investigation of the molecular mechanisms of HIV-1 latency reversal and LRA efficacy in a proper physiological context requires the utilization of primary cell models. Recent mechanistic studies of HIV-1 transcription using latently infected cells recovered from donors and ex vivo cellular models of viral latency have demonstrated that the primary blocks to HIV-1 transcription in memory CD4+ T cells are restrictive epigenetic features at the proviral promoter, the cytoplasmic sequestration of key transcription initiation factors such as NFAT and NF-κB, and the vanishingly low expression of the cellular transcription elongation factor P-TEFb. One of the foremost schemes to eliminate the residual reservoir is to deliberately reactivate latent HIV-1 proviruses to enable clearance of persisting latently infected cells—the “Shock and Kill” strategy. For “Shock and Kill” to become efficient, effective, non-toxic latency-reversing agents (LRAs) must be discovered. Since multiple restrictions limit viral reactivation in primary cells, understanding the T-cell signaling mechanisms that are essential for stimulating P-TEFb biogenesis, initiation factor activation, and reversing the proviral epigenetic restrictions have become a prerequisite for the development of more effective LRAs.
具有转录潜伏形式的复制能力前病毒主要存在于一小部分记忆 CD4+ T 细胞中,是治愈 HIV-1 感染的主要障碍,因为它们是抗逆转录病毒疗法中断后几乎不可避免的病毒反弹的源头。在过去 30 年中,利用转化细胞系(如 Jurkat T 细胞模型)进行的研究发现了启动 HIV-1 转录所必需的许多因素。然而,正如本综述所强调的那样,仍有几种不甚明了的机制有待阐明,包括转录复合体启动子近端暂停的分子基础以及从 7SK snRNP 传递 P-TEFb 的详细机制。此外,HIV-1 转录的核心悖论仍未解决:在没有 Tat 的情况下,最初几轮转录是如何实现的?转化细胞模型的一个关键局限是它们不能再现活跃效应细胞和静止记忆 T 细胞之间的转变。因此,在适当的生理背景下研究 HIV-1 潜伏期逆转和 LRA 效力的分子机制需要利用原始细胞模型。最近利用从供体和病毒潜伏期体外细胞模型中回收的潜伏感染细胞进行的 HIV-1 转录机理研究表明,记忆 CD4+ T 细胞中 HIV-1 转录的主要障碍是前病毒启动子的限制性表观遗传特征、NFAT 和 NF-κB 等关键转录启动因子的细胞质封存以及细胞转录延伸因子 P-TEFb 的低表达。消除残余病毒库的最重要方案之一是故意重新激活潜伏的 HIV-1 病毒,以清除持续潜伏的感染细胞--"冲击和杀灭 "策略。要使 "冲击和杀灭 "有效,必须发现有效、无毒的潜伏逆转剂(LRA)。由于病毒在原代细胞中的再活化受到多重限制,因此了解刺激 P-TEFb 生物发生、启动因子活化和逆转前病毒表观遗传限制所必需的 T 细胞信号机制已成为开发更有效 LRA 的先决条件。
{"title":"The cell biology of HIV-1 latency and rebound","authors":"Uri Mbonye, Jonathan Karn","doi":"10.1186/s12977-024-00639-w","DOIUrl":"https://doi.org/10.1186/s12977-024-00639-w","url":null,"abstract":"Transcriptionally latent forms of replication-competent proviruses, present primarily in a small subset of memory CD4+ T cells, pose the primary barrier to a cure for HIV-1 infection because they are the source of the viral rebound that almost inevitably follows the interruption of antiretroviral therapy. Over the last 30 years, many of the factors essential for initiating HIV-1 transcription have been identified in studies performed using transformed cell lines, such as the Jurkat T-cell model. However, as highlighted in this review, several poorly understood mechanisms still need to be elucidated, including the molecular basis for promoter-proximal pausing of the transcribing complex and the detailed mechanism of the delivery of P-TEFb from 7SK snRNP. Furthermore, the central paradox of HIV-1 transcription remains unsolved: how are the initial rounds of transcription achieved in the absence of Tat? A critical limitation of the transformed cell models is that they do not recapitulate the transitions between active effector cells and quiescent memory T cells. Therefore, investigation of the molecular mechanisms of HIV-1 latency reversal and LRA efficacy in a proper physiological context requires the utilization of primary cell models. Recent mechanistic studies of HIV-1 transcription using latently infected cells recovered from donors and ex vivo cellular models of viral latency have demonstrated that the primary blocks to HIV-1 transcription in memory CD4+ T cells are restrictive epigenetic features at the proviral promoter, the cytoplasmic sequestration of key transcription initiation factors such as NFAT and NF-κB, and the vanishingly low expression of the cellular transcription elongation factor P-TEFb. One of the foremost schemes to eliminate the residual reservoir is to deliberately reactivate latent HIV-1 proviruses to enable clearance of persisting latently infected cells—the “Shock and Kill” strategy. For “Shock and Kill” to become efficient, effective, non-toxic latency-reversing agents (LRAs) must be discovered. Since multiple restrictions limit viral reactivation in primary cells, understanding the T-cell signaling mechanisms that are essential for stimulating P-TEFb biogenesis, initiation factor activation, and reversing the proviral epigenetic restrictions have become a prerequisite for the development of more effective LRAs.","PeriodicalId":21123,"journal":{"name":"Retrovirology","volume":null,"pages":null},"PeriodicalIF":3.3,"publicationDate":"2024-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140576218","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Retrovirology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1