PLoS PathogensPub Date : 2024-09-26eCollection Date: 2024-09-01DOI: 10.1371/journal.ppat.1012600
Gang Ye, Fan Bu, Ruangang Pan, Alise Mendoza, Ge Yang, Benjamin Spiller, Brian E Wadzinski, Lanying Du, Stanley Perlman, Bin Liu, Fang Li
{"title":"Structure-guided in vitro evolution of nanobodies targeting new viral variants.","authors":"Gang Ye, Fan Bu, Ruangang Pan, Alise Mendoza, Ge Yang, Benjamin Spiller, Brian E Wadzinski, Lanying Du, Stanley Perlman, Bin Liu, Fang Li","doi":"10.1371/journal.ppat.1012600","DOIUrl":"10.1371/journal.ppat.1012600","url":null,"abstract":"<p><p>A major challenge in antiviral antibody therapy is keeping up with the rapid evolution of viruses. Our research shows that nanobodies - single-domain antibodies derived from camelids - can be rapidly re-engineered to combat new viral strains through structure-guided in vitro evolution. Specifically, for viral mutations occurring at nanobody-binding sites, we introduce randomized amino acid sequences into nanobody residues near these mutations. We then select nanobody variants that effectively bind to the mutated viral target from a phage display library. As a proof of concept, we used this approach to adapt Nanosota-3, a nanobody originally identified to target the receptor-binding domain (RBD) of early Omicron subvariants, making it highly effective against recent Omicron subvariants. Remarkably, this adaptation process can be completed in less than two weeks, allowing drug development to keep pace with viral evolution and provide timely protection to humans.</p>","PeriodicalId":48999,"journal":{"name":"PLoS Pathogens","volume":null,"pages":null},"PeriodicalIF":5.5,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11460708/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142330627","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
PLoS PathogensPub Date : 2024-09-26eCollection Date: 2024-09-01DOI: 10.1371/journal.ppat.1012551
Olga A Nev, Márcia David-Palma, Joseph Heitman, Alistair J P Brown, Marco A Coelho
{"title":"Fungal pathogens and symbionts: Living off the fat of the land.","authors":"Olga A Nev, Márcia David-Palma, Joseph Heitman, Alistair J P Brown, Marco A Coelho","doi":"10.1371/journal.ppat.1012551","DOIUrl":"https://doi.org/10.1371/journal.ppat.1012551","url":null,"abstract":"","PeriodicalId":48999,"journal":{"name":"PLoS Pathogens","volume":null,"pages":null},"PeriodicalIF":5.5,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11426481/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142330617","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
PLoS PathogensPub Date : 2024-09-26eCollection Date: 2024-09-01DOI: 10.1371/journal.ppat.1012250
Kam Pui Tam, Jia Xie, Rex Kwok Him Au-Yeung, Alan K S Chiang
{"title":"Combination of bortezomib and venetoclax targets the pro-survival function of LMP-1 and EBNA-3C of Epstein-Barr virus in spontaneous lymphoblastoid cell lines.","authors":"Kam Pui Tam, Jia Xie, Rex Kwok Him Au-Yeung, Alan K S Chiang","doi":"10.1371/journal.ppat.1012250","DOIUrl":"10.1371/journal.ppat.1012250","url":null,"abstract":"<p><p>Epstein-Barr virus (EBV) manipulates the ubiquitin-proteasome system and regulators of Bcl-2 family to enable the persistence of the virus and survival of the host cells through the expression of viral proteins in distinct latency patterns. We postulate that the combination of bortezomib (proteasome inhibitor) and venetoclax (Bcl-2 inhibitor) [bort/venetoclax] will cause synergistic killing of post-transplant lymphoproliferative disorder (PTLD) through targeting the pro-survival function of latent viral proteins such as latent membrane protein-1 (LMP-1) and EBV nuclear antigen-3C (EBNA-3C). Bort/venetoclax could synergistically kill spontaneous lymphoblastoid cell lines (sLCLs) derived from patients with PTLD and EBV-associated hemophagocytic lymphohistiocytosis by inducing DNA damage response, apoptosis and G1-S cell cycle arrest in a ROS-dependent manner. Bortezomib potently induced the expression of Noxa, a pro-apoptotic initiator and when combined with venetoclax, inhibited Mcl-1 and Bcl-2 simultaneously. Bortezomib prevented LMP-1 induced proteasomal degradation of IκBα leading to the suppression of the NF-κB signaling pathway. Bortezomib also rescued Bcl-6 from EBNA-3C mediated proteasomal degradation thus maintaining the repression of cyclin D1 and Bcl-2 causing G1-S arrest and apoptosis. Concurrently, venetoclax inhibited Bcl-2 upregulated by either LMP-1 or EBNA-3C. Bort/venetoclax decreased the expression of phosphorylated p65 and Bcl-2 at serine 70 thereby suppressing the NF-κB signaling pathway and promoting apoptosis, respectively. These data corroborated the marked suppression of the growth of xenograft of sLCL in SCID mice (p<0.001). Taken together, the combination of bortezomib and venetoclax targets the pro-survival function of LMP-1 and EBNA-3C of Epstein-Barr virus in spontaneous lymphoblastoid cell lines.</p>","PeriodicalId":48999,"journal":{"name":"PLoS Pathogens","volume":null,"pages":null},"PeriodicalIF":5.5,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11481030/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142330603","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
PLoS PathogensPub Date : 2024-09-26eCollection Date: 2024-09-01DOI: 10.1371/journal.ppat.1012599
Zuxin Liang, Chunhui Li, Xiaohua Gong, Guoguo Ye, Yushan Jiang, Huiping Shi, Abid Hussain, Mengyuan Zhao, Mengjun Li, Yuxin Tian, Wei Zhao, Yang Yang, Yuanyu Huang, Chenguang Shen, Minghui Yang
{"title":"Development of Glycan-masked SARS-CoV-2 RBD vaccines against SARS-related coronaviruses.","authors":"Zuxin Liang, Chunhui Li, Xiaohua Gong, Guoguo Ye, Yushan Jiang, Huiping Shi, Abid Hussain, Mengyuan Zhao, Mengjun Li, Yuxin Tian, Wei Zhao, Yang Yang, Yuanyu Huang, Chenguang Shen, Minghui Yang","doi":"10.1371/journal.ppat.1012599","DOIUrl":"10.1371/journal.ppat.1012599","url":null,"abstract":"<p><p>Emerging and recurrent infectious diseases caused by coronaviruses remain a significant public health concern. Here, we present a targeted approach to elicit antibodies capable of neutralizing SARS-CoV-2 variants and other SARS-related coronaviruses. By introducing amino acid mutations at mutation-prone sites, we engineered glycosylation modifications to the Receptor Binding Domain (RBD) of SARS-CoV-2, thereby exposing more conserved, yet less accessible epitopes. We developed both messenger RNA (mRNA) and recombination subunit vaccines using these engineered-RBDs (M1, M2) and the wild-type RBD as immunogens. The engineered-RBD vaccines elicited robust neutralizing responses against various SARS-CoV-2 variants as well as SARS-CoV and WIV1-CoV, and conferred protection in mice challenged with the XBB.1.16 strain. Furthermore, We highlighted that glycan masking is a decisive factor in antibody binding changes and RBD-conserved antibody response. Additionally, the glycan-engineered RBD mRNA vaccines stimulated stronger cell-mediated immune responses. Our glycan modification strategy significantly enhances broad-spectrum neutralizing efficacy and cellular immunity, providing valuable insights for the development of vaccines against a wide range of SARS-related coronaviruses.</p>","PeriodicalId":48999,"journal":{"name":"PLoS Pathogens","volume":null,"pages":null},"PeriodicalIF":5.5,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11460674/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142330604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
PLoS PathogensPub Date : 2024-09-26eCollection Date: 2024-09-01DOI: 10.1371/journal.ppat.1012581
Zheng Yu, Varun Sasidharan-Nair, Thalea Buchta, Agnes Bonifacius, Fawad Khan, Beate Pietzsch, Hosein Ahmadi, Michael Beckstette, Jana Niemz, Philipp Hilgendorf, Philip Mausberg, Andreas Keller, Christine Falk, Dirk H Busch, Kilian Schober, Luka Cicin-Sain, Fabian Müller, Melanie M Brinkmann, Britta Eiz-Vesper, Stefan Floess, Jochen Huehn
{"title":"DNA methylation profiling identifies TBKBP1 as potent amplifier of cytotoxic activity in CMV-specific human CD8+ T cells.","authors":"Zheng Yu, Varun Sasidharan-Nair, Thalea Buchta, Agnes Bonifacius, Fawad Khan, Beate Pietzsch, Hosein Ahmadi, Michael Beckstette, Jana Niemz, Philipp Hilgendorf, Philip Mausberg, Andreas Keller, Christine Falk, Dirk H Busch, Kilian Schober, Luka Cicin-Sain, Fabian Müller, Melanie M Brinkmann, Britta Eiz-Vesper, Stefan Floess, Jochen Huehn","doi":"10.1371/journal.ppat.1012581","DOIUrl":"10.1371/journal.ppat.1012581","url":null,"abstract":"<p><p>Epigenetic mechanisms stabilize gene expression patterns during CD8+ T cell differentiation. Although adoptive transfer of virus-specific T cells is clinically applied to reduce the risk of virus infection or reactivation in immunocompromised individuals, the DNA methylation pattern of virus-specific CD8+ T cells is largely unknown. Hence, we here performed whole-genome bisulfite sequencing of cytomegalovirus-specific human CD8+ T cells and found that they display a unique DNA methylation pattern consisting of 79 differentially methylated regions (DMRs) when compared to memory CD8+ T cells. Among the top demethylated DMRs in cytomegalovirus-specific CD8+ T cells was TBKBP1, coding for TBK-binding protein 1 that can interact with TANK-binding kinase 1 (TBK1) and mediate pro-inflammatory responses in innate immune cells downstream of intracellular virus sensing. Since TBKBP1 has not yet been reported in T cells, we aimed to unravel its role in virus-specific CD8+ T cells. TBKBP1 demethylation in terminal effector CD8+ T cells correlated with higher TBKBP1 expression at both mRNA and protein level, independent of alternative splicing of TBKBP1 transcripts. Notably, the distinct DNA methylation patterns in CD8+ T cell subsets was stable upon long-term in vitro culture. TBKBP1 overexpression resulted in enhanced TBK1 phosphorylation upon stimulation of CD8+ T cells and significantly improved their virus neutralization capacity. Collectively, our data demonstrate that TBKBP1 modulates virus-specific CD8+ T cell responses and could be exploited as therapeutic target to improve adoptive T cell therapies.</p>","PeriodicalId":48999,"journal":{"name":"PLoS Pathogens","volume":null,"pages":null},"PeriodicalIF":5.5,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11460711/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142330605","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
PLoS PathogensPub Date : 2024-09-26eCollection Date: 2024-09-01DOI: 10.1371/journal.ppat.1012576
Meilin Li, Xinyuan Liu, Dingkun Peng, Meng Yao, Tao Wang, Yijing Wang, Hongwei Cao, Yanjin Wang, Jingwen Dai, Rui Luo, Hao Deng, Jiaqi Li, Yuzi Luo, Yongfeng Li, Yuan Sun, Su Li, Hua-Ji Qiu, Lian-Feng Li
{"title":"The I7L protein of African swine fever virus is involved in viral pathogenicity by antagonizing the IFN-γ-triggered JAK-STAT signaling pathway through inhibiting the phosphorylation of STAT1.","authors":"Meilin Li, Xinyuan Liu, Dingkun Peng, Meng Yao, Tao Wang, Yijing Wang, Hongwei Cao, Yanjin Wang, Jingwen Dai, Rui Luo, Hao Deng, Jiaqi Li, Yuzi Luo, Yongfeng Li, Yuan Sun, Su Li, Hua-Ji Qiu, Lian-Feng Li","doi":"10.1371/journal.ppat.1012576","DOIUrl":"10.1371/journal.ppat.1012576","url":null,"abstract":"<p><p>Cell-passage-adapted strains of African swine fever virus (ASFV) typically exhibit substantial genomic alterations and attenuated virulence in pigs. We have indicated that the human embryonic kidney (HEK293T) cells-adapted ASFV strain underwent genetic alterations and the I7L gene in the right variable region was deleted compared with the ASFV HLJ/2018 strain (ASFV-WT). A recent study has revealed that the deletion of the I7L-I11L genes results in attenuation of virulent ASFV in vivo, but the underlying mechanism remains largely unknown. Therefore, we hypothesized that the deletion of the I7L gene may be related to the pathogenicity of ASFV in pigs. We generated the I7L gene-deleted ASFV mutant (ASFV-ΔI7L) and found that the I7L gene deletion does not influence the replication of ASFV in primary porcine alveolar macrophages (PAMs). Using transcriptome sequencing analysis, we identified that the differentially expressed genes in the PAMs infected with ASFV-ΔI7L were mainly involved in antiviral immune responses induced by interferon gamma (IFN-γ) compared with those in the ASFV-WT-infected PAMs. Meanwhile, we further confirmed that the I7L protein (pI7L) suppressed the IFN-γ-triggered JAK-STAT signaling pathway. Mechanistically, pI7L interacts with STAT1 and inhibits its phosphorylation and homodimerization, which depends on the tyrosine at position 98 (Y98) of pI7L, thereby preventing the nuclear translocation of STAT1 and leading to the decreased production of IFN-γ-stimulated genes. Importantly, ASFV-ΔI7L exhibited reduced replication and virulence compared with ASFV-WT in pigs, likely due to the increased production of IFN-γ-stimulated genes, indicating that pI7L is involved in the virulence of ASFV. Taken together, our findings demonstrate that pI7L is associated with pathogenicity and antagonizes the IFN-γ-triggered JAK-STAT signaling pathway via inhibiting the phosphorylation and homodimerization of STAT1 depending on the Y98 residue of pI7L and the Src homology 2 domain of STAT1, which provides more information for understanding the immunoevasion strategies and designing the live attenuated vaccines against ASFV infection.</p>","PeriodicalId":48999,"journal":{"name":"PLoS Pathogens","volume":null,"pages":null},"PeriodicalIF":5.5,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11460700/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142330628","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
PLoS PathogensPub Date : 2024-09-25eCollection Date: 2024-09-01DOI: 10.1371/journal.ppat.1012550
Pinyi Yang, Xiaoli Wu, Hang Shang, Zixian Sun, Zhiying Wang, Zidan Song, Hong Yuan, Fei Deng, Shu Shen, Yu Guo, Nan Zhang
{"title":"Molecular mechanism and structure-guided humanization of a broadly neutralizing antibody against SFTSV.","authors":"Pinyi Yang, Xiaoli Wu, Hang Shang, Zixian Sun, Zhiying Wang, Zidan Song, Hong Yuan, Fei Deng, Shu Shen, Yu Guo, Nan Zhang","doi":"10.1371/journal.ppat.1012550","DOIUrl":"https://doi.org/10.1371/journal.ppat.1012550","url":null,"abstract":"<p><p>Severe fever with thrombocytopenia syndrome virus (SFTSV) is a novel tick-borne bunyavirus that causes severe fever with thrombocytopenia syndrome (SFTS), with a high mortality rate of up to 30%. The envelope glycoproteins of SFTSV, glycoprotein N (Gn) and glycoprotein C (Gc), facilitate the recognition of host receptors and the process of membrane fusion, allowing the virus to enter host cells. We previously reported a monoclonal antibody, mAb 40C10, capable of neutralizing different genotypes of SFTSV and SFTSV-related viruses. However, the specific neutralization mechanism is poorly understood. In this study, we elucidated the high-resolution structure of the SFTSV Gn head domain in complex with mAb 40C10, confirming that the binding epitope in the domain I region of SFTSV Gn, and it represented that a novel binding epitope of SFTSV Gn was identified. Through in-depth structural and sequence analyses, we found that the binding sites of mAb 40C10 are relatively conserved among different genotypes of SFTSV and SFTSV-related Heartland virus and Guertu virus, elucidating the molecular mechanism underlying the broad-spectrum neutralizing activity of mAb 40C10. Furthermore, we humanized of mAb 40C10, which is originally of murine origin, to reduce its immunogenicity. The resulting nine humanized antibodies maintained potent affinity and neutralizing activity. One of the humanized antibodies exhibited neutralizing activity at picomolar IC50 values and demonstrated effective therapeutic and protective effects in a mouse infection model. These findings provide a novel target for the future development of SFTSV vaccines or drugs and establish a foundation for the research and development of antibody therapeutics for clinical applications.</p>","PeriodicalId":48999,"journal":{"name":"PLoS Pathogens","volume":null,"pages":null},"PeriodicalIF":5.5,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11423973/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142330618","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
PLoS PathogensPub Date : 2024-09-25eCollection Date: 2024-09-01DOI: 10.1371/journal.ppat.1012578
Andreea Waltmann, Jacqueline T Balthazar, Afrin A Begum, Nancy Hua, Ann E Jerse, William M Shafer, Marcia M Hobbs, Joseph A Duncan
{"title":"Experimental genital tract infection demonstrates Neisseria gonorrhoeae MtrCDE efflux pump is not required for in vivo human infection and identifies gonococcal colonization bottleneck.","authors":"Andreea Waltmann, Jacqueline T Balthazar, Afrin A Begum, Nancy Hua, Ann E Jerse, William M Shafer, Marcia M Hobbs, Joseph A Duncan","doi":"10.1371/journal.ppat.1012578","DOIUrl":"10.1371/journal.ppat.1012578","url":null,"abstract":"<p><p>The MtrCDE efflux pump of Neisseria gonorrhoeae exports a wide range of antimicrobial compounds that the gonococcus encounters at mucosal surfaces during colonization and infection and is a known gonococcal virulence factor. Here, we evaluate the role of this efflux pump system in strain FA1090 during in vivo human male urethral infection with N. gonorrhoeae using a controlled human infection model. With the strategy of competitive infections initiated with mixtures of wild-type FA1090 and an isogenic mutant FA1090 strain that does not contain a functional MtrCDE pump, we found that the presence of the efflux pump is not required for an infection to be established in the human male urethra. This finding contrasts with previous studies of in vivo infection in the lower genital tract of female mice, which demonstrated that mutant gonococci of a different strain (FA19) lacking a functional MtrCDE pump had a significantly reduced fitness compared to their wild-type parental FA19 strain. To determine if these conflicting results are due to strain or human vs. mouse differences, we conducted a series of systematic competitive infections in female mice with the same FA1090 strains as in humans, and with FA19 strains, including mutants that do not assemble a functional MtrCDE efflux pump. Our results indicate the fitness advantage provided by the MtrCDE efflux pump during infection of mice is strain dependent. Owing to the equal fitness of the two FA1090 strains in men, our experiments also demonstrated the presence of a colonization bottleneck of N. gonorrhoeae in the human male urethra, which may open a new area of inquiry into N. gonorrhoeae infection dynamics and control. TRIAL REGISTRATION. Clinicaltrials.gov NCT03840811.</p>","PeriodicalId":48999,"journal":{"name":"PLoS Pathogens","volume":null,"pages":null},"PeriodicalIF":5.5,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11457995/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142330615","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
PLoS PathogensPub Date : 2024-09-24eCollection Date: 2024-09-01DOI: 10.1371/journal.ppat.1012470
Zicheng Hu, Paola Cinque, Ameet Dravid, Lars Hagberg, Aylin Yilmaz, Henrik Zetterberg, Dietmar Fuchs, Johanna Gostner, Kaj Blennow, Serena S Spudich, Laura Kincer, Shuntai Zhou, Sarah Beth Joseph, Ronald Swanstrom, Richard W Price, Magnus Gisslén
{"title":"Changes in cerebrospinal fluid proteins across the spectrum of untreated and treated chronic HIV-1 infection.","authors":"Zicheng Hu, Paola Cinque, Ameet Dravid, Lars Hagberg, Aylin Yilmaz, Henrik Zetterberg, Dietmar Fuchs, Johanna Gostner, Kaj Blennow, Serena S Spudich, Laura Kincer, Shuntai Zhou, Sarah Beth Joseph, Ronald Swanstrom, Richard W Price, Magnus Gisslén","doi":"10.1371/journal.ppat.1012470","DOIUrl":"10.1371/journal.ppat.1012470","url":null,"abstract":"<p><p>Using the Olink Explore 1536 platform, we measured 1,463 unique proteins in 303 cerebrospinal fluid (CSF) specimens from four clinical centers contributed by uninfected controls and 12 groups of people living with HIV-1 infection representing the spectrum of progressive untreated and treated chronic infection. We present three initial analyses of these measurements: an overview of the CSF protein features of the sample; correlations of the CSF proteins with CSF HIV-1 RNA and neurofilament light chain protein (NfL) concentrations; and comparison of CSF proteins in HIV-associated dementia (HAD) and neurosymptomatic CSF escape (NSE). These reveal a complex but coherent picture of CSF protein changes with highest concentrations of many proteins during CNS injury in the HAD and NSE groups and variable protein changes across the course of systemic HIV-1 progression that included two common patterns, designated as lymphoid and myeloid patterns, related to principal involvement of their underlying inflammatory cell lineages. Antiretroviral therapy reduced CSF protein perturbations, though not always to control levels. The dataset of these CSF protein measurements, along with background clinical information, is posted online. Extended studies of this unique dataset will supplement this report to provide more detailed characterization of the dynamic impact of HIV-1 infection on the CSF proteome across the spectrum of HIV-1 infection, advancing the mechanistic understanding of HIV-1-related CNS pathobiology.</p>","PeriodicalId":48999,"journal":{"name":"PLoS Pathogens","volume":null,"pages":null},"PeriodicalIF":5.5,"publicationDate":"2024-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11469498/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142330602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}