Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Immunol ; 25(2): 218-225, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38212464

RESUMO

Long COVID (LC) occurs after at least 10% of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, yet its etiology remains poorly understood. We used 'omic" assays and serology to deeply characterize the global and SARS-CoV-2-specific immunity in the blood of individuals with clear LC and non-LC clinical trajectories, 8 months postinfection. We found that LC individuals exhibited systemic inflammation and immune dysregulation. This was evidenced by global differences in T cell subset distribution implying ongoing immune responses, as well as by sex-specific perturbations in cytolytic subsets. LC individuals displayed increased frequencies of CD4+ T cells poised to migrate to inflamed tissues and exhausted SARS-CoV-2-specific CD8+ T cells, higher levels of SARS-CoV-2 antibodies and a mis-coordination between their SARS-CoV-2-specific T and B cell responses. Our analysis suggested an improper crosstalk between the cellular and humoral adaptive immunity in LC, which can lead to immune dysregulation, inflammation and clinical symptoms associated with this debilitating condition.


Assuntos
COVID-19 , SARS-CoV-2 , Feminino , Masculino , Humanos , Síndrome de COVID-19 Pós-Aguda , Linfócitos T CD8-Positivos , Imunidade Humoral , Anticorpos Antivirais , Inflamação
2.
Cell Rep ; 42(11): 113285, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37910505

RESUMO

Deciphering the mechanisms underlying viral persistence is critical to achieving a cure for human immunodeficiency virus (HIV) infection. Here, we implement a systems approach to discover molecular signatures of HIV latently infected CD4+ T cells, identifying the immunosuppressive, adenosine-producing ectonucleotidase CD73 as a key surface marker of latent cells. Hypoxic conditioning, reflecting the lymphoid tissue microenvironment, increases the frequency of CD73+ CD4+ T cells and promotes HIV latency. Transcriptomic profiles of CD73+ CD4+ T cells favor viral quiescence, immune evasion, and cell survival. CD73+ CD4+ T cells are capable of harboring a functional HIV reservoir and reinitiating productive infection ex vivo. CD73 or adenosine receptor blockade facilitates latent HIV reactivation in vitro, mechanistically linking adenosine signaling to viral quiescence. Finally, tissue imaging of lymph nodes from HIV-infected individuals on antiretroviral therapy reveals spatial association between CD73 expression and HIV persistence in vivo. Our findings warrant development of HIV-cure strategies targeting the hypoxia-CD73-adenosine axis.


Assuntos
Infecções por HIV , HIV-1 , Humanos , Adenosina/metabolismo , Linfócitos T CD4-Positivos , Ativação Viral , Latência Viral/fisiologia , Replicação Viral/fisiologia
3.
bioRxiv ; 2023 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-37333294

RESUMO

Progress in understanding long COVID and developing effective therapeutics is hampered in part by the lack of suitable animal models. Here we used ACE2-transgenic mice recovered from Omicron (BA.1) infection to test for pulmonary and behavioral post-acute sequelae. Through in-depth phenotyping by CyTOF, we demonstrate that naïve mice experiencing a first Omicron infection exhibit profound immune perturbations in the lung after resolving acute infection. This is not observed if mice were first vaccinated with spike-encoding mRNA. The protective effects of vaccination against post-acute sequelae were associated with a highly polyfunctional SARS-CoV-2-specific T cell response that was recalled upon BA.1 breakthrough infection but not seen with BA.1 infection alone. Without vaccination, the chemokine receptor CXCR4 was uniquely upregulated on multiple pulmonary immune subsets in the BA.1 convalescent mice, a process previously connected to severe COVID-19. Taking advantage of recent developments in machine learning and computer vision, we demonstrate that BA.1 convalescent mice exhibited spontaneous behavioral changes, emotional alterations, and cognitive-related deficits in context habituation. Collectively, our data identify immunological and behavioral post-acute sequelae after Omicron infection and uncover a protective effect of vaccination against post-acute pulmonary immune perturbations.

4.
bioRxiv ; 2023 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-36798286

RESUMO

Long COVID (LC), a type of post-acute sequelae of SARS-CoV-2 infection (PASC), occurs after at least 10% of SARS-CoV-2 infections, yet its etiology remains poorly understood. Here, we used multiple "omics" assays (CyTOF, RNAseq/scRNAseq, Olink) and serology to deeply characterize both global and SARS-CoV-2-specific immunity from blood of individuals with clear LC and non-LC clinical trajectories, 8 months following infection and prior to receipt of any SARS-CoV-2 vaccine. Our analysis focused on deep phenotyping of T cells, which play important roles in immunity against SARS-CoV-2 yet may also contribute to COVID-19 pathogenesis. Our findings demonstrate that individuals with LC exhibit systemic inflammation and immune dysregulation. This is evidenced by global differences in T cell subset distribution in ways that imply ongoing immune responses, as well as by sex-specific perturbations in cytolytic subsets. Individuals with LC harbored increased frequencies of CD4+ T cells poised to migrate to inflamed tissues, and exhausted SARS-CoV-2-specific CD8+ T cells. They also harbored significantly higher levels of SARS-CoV-2 antibodies, and in contrast to non-LC individuals, exhibited a mis-coordination between their SARS-CoV-2-specific T and B cell responses. RNAseq/scRNAseq and Olink analyses similarly revealed immune dysregulatory mechanisms, along with non-immune associated perturbations, in individuals with LC. Collectively, our data suggest that proper crosstalk between the humoral and cellular arms of adaptive immunity has broken down in LC, and that this, perhaps in the context of persistent virus, leads to the immune dysregulation, inflammation, and clinical symptoms associated with this debilitating condition.

5.
Elife ; 112022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35787792

RESUMO

High-parameter single-cell phenotyping has enabled in-depth classification and interrogation of immune cells, but to date has not allowed for glycan characterization. Here, we develop CyTOF-Lec as an approach to simultaneously characterize many protein and glycan features of human immune cells at the single-cell level. We implemented CyTOF-Lec to compare glycan features between different immune subsets from blood and multiple tissue compartments, and to characterize HIV-infected cell cultures. Using bioinformatics approaches to distinguish preferential infection of cellular subsets from viral-induced remodeling, we demonstrate that HIV upregulates the levels of cell-surface fucose and sialic acid in a cell-intrinsic manner, and that memory CD4+ T cells co-expressing high levels of fucose and sialic acid are highly susceptible to HIV infection. Sialic acid levels were found to distinguish memory CD4+ T cell subsets expressing different amounts of viral entry receptors, pro-survival factors, homing receptors, and activation markers, and to play a direct role in memory CD4+ T cells' susceptibility to HIV infection. The ability of sialic acid to distinguish memory CD4+ T cells with different susceptibilities to HIV infection was experimentally validated through sorting experiments. Together, these results suggest that HIV remodels not only cellular proteins but also glycans, and that glycan expression can differentiate memory CD4+ T cells with vastly different susceptibility to HIV infection.


Living cells have a sugar coating. These sugars include molecules called glycans, which help cells interact with the outside world. The types of sugars on cells can affect their properties, including potentially their susceptibility to infection by viruses, such as the human immunodeficiency virus, HIV. To date, most research examining cells susceptible to HIV has focused on cell surface proteins, not sugars. To study these proteins, researchers had previously covered them in metal-studded antibodies (which stick to proteins) and used a technique called cytometry time of flight, or CyTOF for short, to quantify the levels of these proteins on the surface of cells susceptible to HIV. Adapting this tool to investigate sugars could answer questions about HIV infection. For example, does the virus prefer to infect cells coated in certain sugar molecules? And does it change the pattern of sugars on the surface of the cells it infects? Ma et al. adapted CyTOF to use molecules called lectins (which stick to sugars) in conjunction with the metal-studded antibodies. This made it possible to simultaneously measure the levels of 34 different proteins and 5 different types of sugars on individual cells. The pattern of sugars on the surface of cells from the immune system differed depending on what tissues the cells came from, and what types of cells they were. The results showed that HIV preferred to infect memory CD4 T cells with high levels of two types of sugar: fucose and sialic acid. Furthermore, during infection, the levels of both these sugars increased. Current treatments for HIV keep virus levels low but do not cure the infection. Further research could determine whether sugars have a role to play in HIV persistence. It is possible that the sugar patterns preferred by the virus help it to avoid detection. A clearer understanding of cell surface sugars could lead to sugar-targeting drugs that kill infected cells.


Assuntos
Infecções por HIV , HIV-1 , Linfócitos T CD4-Positivos , Suscetibilidade a Doenças , Fucose , Glicômica , HIV-1/fisiologia , Humanos , Ácido N-Acetilneuramínico , Polissacarídeos
6.
Nature ; 607(7918): 351-355, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35584773

RESUMO

SARS-CoV-2 Delta and Omicron are globally relevant variants of concern. Although individuals infected with Delta are at risk of developing severe lung disease, infection with Omicron often causes milder symptoms, especially in vaccinated individuals1,2. The question arises of whether widespread Omicron infections could lead to future cross-variant protection, accelerating the end of the pandemic. Here we show that without vaccination, infection with Omicron induces a limited humoral immune response in mice and humans. Sera from mice overexpressing the human ACE2 receptor and infected with Omicron neutralize only Omicron, but not other variants of concern, whereas broader cross-variant neutralization was observed after WA1 and Delta infections. Unlike WA1 and Delta, Omicron replicates to low levels in the lungs and brains of infected animals, leading to mild disease with reduced expression of pro-inflammatory cytokines and diminished activation of lung-resident T cells. Sera from individuals who were unvaccinated and infected with Omicron show the same limited neutralization of only Omicron itself. By contrast, Omicron breakthrough infections induce overall higher neutralization titres against all variants of concern. Our results demonstrate that Omicron infection enhances pre-existing immunity elicited by vaccines but, on its own, may not confer broad protection against non-Omicron variants in unvaccinated individuals.


Assuntos
COVID-19 , Proteção Cruzada , SARS-CoV-2 , Vacinação , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , COVID-19/virologia , Vacinas contra COVID-19/administração & dosagem , Proteção Cruzada/imunologia , Citocinas , Humanos , Camundongos , SARS-CoV-2/classificação , SARS-CoV-2/imunologia , Vacinação/estatística & dados numéricos
7.
medRxiv ; 2022 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-35075459

RESUMO

SARS-CoV-2 Delta and Omicron strains are the most globally relevant variants of concern (VOCs). While individuals infected with Delta are at risk to develop severe lung disease 1 , Omicron infection causes less severe disease, mostly upper respiratory symptoms 2,3 . The question arises whether rampant spread of Omicron could lead to mass immunization, accelerating the end of the pandemic. Here we show that infection with Delta, but not Omicron, induces broad immunity in mice. While sera from Omicron-infected mice only neutralize Omicron, sera from Delta-infected mice are broadly effective against Delta and other VOCs, including Omicron. This is not observed with the WA1 ancestral strain, although both WA1 and Delta elicited a highly pro-inflammatory cytokine response and replicated to similar titers in the respiratory tracts and lungs of infected mice as well as in human airway organoids. Pulmonary viral replication, pro-inflammatory cytokine expression, and overall disease progression are markedly reduced with Omicron infection. Analysis of human sera from Omicron and Delta breakthrough cases reveals effective cross-variant neutralization induced by both viruses in vaccinated individuals. Together, our results indicate that Omicron infection enhances preexisting immunity elicited by vaccines, but on its own may not induce broad, cross-neutralizing humoral immunity in unvaccinated individuals.

8.
J Immunol ; 207(5): 1344-1356, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34389625

RESUMO

CD8+ T cells can potentiate long-lived immunity against COVID-19. We screened longitudinally-sampled convalescent human donors against SARS-CoV-2 tetramers and identified a participant with an immunodominant response against residues 322 to 311 of nucleocapsid (Nuc322-331), a peptide conserved in all variants of concern reported to date. We conducted 38-parameter cytometry by time of flight on tetramer-identified Nuc322-331-specific CD8+ T cells and on CD4+ and CD8+ T cells recognizing the entire nucleocapsid and spike proteins, and took 32 serological measurements. We discovered a coordination of the Nuc322-331-specific CD8+ T response with both the CD4+ T cell and Ab pillars of adaptive immunity. Over the approximately six month period of convalescence monitored, we observed a slow and progressive decrease in the activation state and polyfunctionality of Nuc322-331-specific CD8+ T cells, accompanied by an increase in their lymph node-homing and homeostatic proliferation potential. These results suggest that following a typical case of mild COVID-19, SARS-CoV-2-specific CD8+ T cells not only persist but continuously differentiate in a coordinated fashion well into convalescence into a state characteristic of long-lived, self-renewing memory.


Assuntos
Linfócitos T CD8-Positivos/imunologia , COVID-19/imunologia , Convalescença , SARS-CoV-2/imunologia , Linfócitos T CD8-Positivos/patologia , Humanos , Estudos Longitudinais
9.
bioRxiv ; 2021 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-33948597

RESUMO

CD8+ T cells are important antiviral effectors that can potentiate long-lived immunity against COVID-19, but a detailed characterization of these cells has been hampered by technical challenges. We screened 21 well-characterized, longitudinally-sampled convalescent donors that recovered from mild COVID-19 against a collection of SARS-CoV-2 tetramers, and identified one participant with an immunodominant response against Nuc322-331, a peptide that is conserved in all the SARS-CoV-2 variants-of-concern reported to date. We conducted 38-parameter CyTOF phenotyping on tetramer-identified Nuc322-331-specific CD8+ T cells, and on CD4+ and CD8+ T cells recognizing the entire nucleocapsid and spike proteins from SARS-CoV-2, and took 32 serological measurements on longitudinal specimens from this participant. We discovered a coordination of the Nuc322-331-specific CD8+ T response with both the CD4+ T cell and antibody pillars of adaptive immunity. Nuc322-331-specific CD8+ T cells were predominantly central memory T cells, but continually evolved over a ~6-month period of convalescence. We observed a slow and progressive decrease in the activation state and polyfunctionality of the Nuc322-331-specific CD8+ T cells, accompanied by an increase in their lymph-node homing and homeostatic proliferation potential. These results suggest that following a typical case of mild COVID-19, SARS-CoV-2-specific CD8+ T cells not only persist but continuously differentiate in a coordinated fashion well into convalescence, into a state characteristic of long-lived, self-renewing memory.

10.
J Virol ; 95(13): e0221120, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33883226

RESUMO

Merkel cell polyomavirus (MCPyV) infects most of the human population asymptomatically, but in rare cases it leads to a highly aggressive skin cancer called Merkel cell carcinoma (MCC). MCC incidence is much higher in aging and immunocompromised populations. The epidemiology of MCC suggests that dysbiosis between the host immune response and the MCPyV infectious cycle could contribute to the development of MCPyV-associated MCC. Insufficient restriction of MCPyV by normal cellular processes, for example, could promote the incidental oncogenic MCPyV integration events and/or entry into the original cell of MCC. Progress toward understanding MCPyV biology has been hindered by its narrow cellular tropism. Our discovery that primary human dermal fibroblasts (HDFs) support MCPyV infection has made it possible to closely model cellular responses to different stages of the infectious cycle. The present study reveals that the onset of MCPyV replication and early gene expression induces an inflammatory cytokine and interferon-stimulated gene (ISG) response. The cGAS-STING pathway, in coordination with NF-κB, mediates induction of this innate immune gene expression program. Further, silencing of cGAS or NF-κB pathway factors led to elevated MCPyV replication. We also discovered that the PYHIN protein IFI16 localizes to MCPyV replication centers but does not contribute to the induction of ISGs. Instead, IFI16 upregulates inflammatory cytokines in response to MCPyV infection by an alternative mechanism. The work described herein establishes a foundation for exploring how changes to the skin microenvironment induced by aging or immunodeficiency might alter the fate of MCPyV and its host cell to encourage carcinogenesis. IMPORTANCE MCC has a high rate of mortality and an increasing incidence. Immune-checkpoint therapies have improved the prognosis of patients with metastatic MCC. Still, a significant proportion of the patients fail to respond to immune-checkpoint therapies or have a medical need for iatrogenic immune-suppression. A greater understanding of MCPyV biology could inform targeted therapies for MCPyV-associated MCC. Moreover, cellular events preceding MCC oncogenesis remain largely unknown. The present study aims to explore how MCPyV interfaces with innate immunity during its infectious cycle. We describe how MCPyV replication and/or transcription elicit an innate immune response via cGAS-STING, NF-κB, and IFI16. We also explore the effects of this response on MCPyV replication. Our findings illustrate how healthy cellular conditions may allow low-level infection that evades immune destruction until highly active replication is restricted by host responses. Conversely, pathological conditions could result in unbridled MCPyV replication that licenses MCC tumorigenesis.


Assuntos
Citocinas/imunologia , Fibroblastos/imunologia , Imunidade Inata/imunologia , Poliomavírus das Células de Merkel/imunologia , Pele/imunologia , Sistemas CRISPR-Cas/genética , Carcinoma de Célula de Merkel/patologia , Células Cultivadas , Citocinas/biossíntese , Fibroblastos/virologia , Células HEK293 , Humanos , Imunidade Inata/genética , Interferons/biossíntese , Interferons/imunologia , Proteínas de Membrana/genética , Poliomavírus das Células de Merkel/crescimento & desenvolvimento , NF-kappa B/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleotidiltransferases/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Infecções por Polyomavirus/imunologia , Pele/citologia , Infecções Tumorais por Vírus/imunologia
11.
Cell Rep ; 35(4): 109038, 2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33910003

RESUMO

Relatively little is known about features of T cells targeted by HIV in vivo. By applying bioinformatics analysis to mass cytometry (CyTOF)-phenotyped specimens from individuals with viremia and in-vitro-infected cells from uninfected donors, we provide an atlas of the phenotypes of in vivo and in vitro HIV-susceptible cells. T helper 17 (Th17) and α4ß1+ cells are preferentially targeted in vivo, whereas T effector memory (Tem), T transitional memory (Ttm), Th1, and Th1/Th17 subsets are targeted in vitro. Multiple proteins-including chemokine and cytokine receptors-are remodeled by HIV in vivo, and these changes are mostly recapitulated in vitro. HIV remodels cells to a T follicular helper (Tfh) phenotype. Using clustering, we uncover a subset of CD29-expressing, Tem-like cells that are highly susceptible to infection in vivo and in vitro and experimentally confirm that susceptibility. These studies provide an in-depth look at features of HIV-susceptible cells in individuals with viremia and demonstrate that some-but not all-HIV-susceptible cells identified in vitro effectively model in vivo susceptibility.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , HIV-1/genética , Memória Imunológica/genética , Subpopulações de Linfócitos T/metabolismo , Humanos
12.
Elife ; 92020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32990219

RESUMO

The latent reservoir is a major barrier to HIV cure. As latently infected cells cannot be phenotyped directly, the features of the in vivo reservoir have remained elusive. Here, we describe a method that leverages high-dimensional phenotyping using CyTOF to trace latently infected cells reactivated ex vivo to their original pre-activation states. Our results suggest that, contrary to common assumptions, the reservoir is not randomly distributed among cell subsets, and is remarkably conserved between individuals. However, reservoir composition differs between tissues and blood, as do cells successfully reactivated by different latency reversing agents. By selecting 8-10 of our 39 original CyTOF markers, we were able to isolate highly purified populations of unstimulated in vivo latent cells. These purified populations were highly enriched for replication-competent and intact provirus, transcribed HIV, and displayed clonal expansion. The ability to isolate unstimulated latent cells from infected individuals enables previously impossible studies on HIV persistence.


There is no cure for the human immunodeficiency virus infection (HIV), but anti-retroviral drugs allow infected people to keep the virus at bay and lead a normal life. These drugs suppress the growth of HIV, but they do not eliminate the virus. If the treatment is interrupted, the virus bounces back within weeks in most individuals. HIV can start growing again because it hides within particular immune cells, called T cells. These infected cells stay in the infected person's body for their whole life in a dormant or "latent" state, and represent the main barrier to an HIV cure. If these cells could be eliminated or prevented from producing more virus without daily treatment, then HIV could be cured. The fact that HIV hides inside T cells has been known for a long time, but it has remained unclear exactly what kinds of T cells the virus prefers. One challenge to characterizing latently-infected cells is that there is no single protein made by them that is not also made by uninfected T cells. The latently-infected T cells are also very rare: HIV mainly attaches to a protein called CD4, but only one in about a million T cells with CD4 contain the virus. To figure out which CD4-carrying T cells in a patient sample are latently infected, the cells are extracted from the patient's body and 'reactivated' so the virus will start growing again. Unfortunately, the mixture of drugs used to reactivate the T cells changes the cells and the proteins they are producing, which obscures the features the latently-infected T cells had before reactivation. Neidleman, Luo et al. developed a new approach to trace the infected, reactivated T cells back to their state before reactivation. Using computational methods and a laboratory technique called mass cytometry, the levels of approximately 40 different proteins were measured in millions of T cells from people living with HIV. These experiments provided an 'atlas' of overall T cell features onto which each reactivated cell could be mapped. The population of latently-infected T cells exhibited common features among all the participants. Selecting a few of the most abundant proteins on the surface of the latently-infected cells allowed these cells to be physically separated from all other immune cells. In the future, this relatively pure population of infected T cells could be used to study how HIV can persist for many decades. The 'map' of these cells' features will provide a valuable resource for HIV researchers and might enable the discovery of new drugs to eliminate the latent T cells.


Assuntos
Linfócitos T CD4-Positivos , Infecções por HIV , HIV-1 , Latência Viral/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/virologia , Separação Celular , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/patogenicidade , Humanos , Imunofenotipagem , Espectrometria de Massas , Provírus
13.
Cell Rep Med ; 1(6): 100081, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32839763

RESUMO

Convalescing coronavirus disease 2019 (COVID-19) patients mount robust T cell responses against SARS-CoV-2, suggesting an important role of T cells in viral clearance. To date, the phenotypes of SARS-CoV-2-specific T cells remain poorly defined. Using 38-parameter CyTOF, we phenotyped longitudinal specimens of SARS-CoV-2-specific CD4+ and CD8+ T cells from nine individuals who recovered from mild COVID-19. SARS-CoV-2-specific CD4+ T cells were exclusively Th1 cells and predominantly Tcm cells with phenotypic features of robust helper function. SARS-CoV-2-specific CD8+ T cells were predominantly Temra cells in a state of less terminal differentiation than most Temra cells. Subsets of SARS-CoV-2-specific T cells express CD127, can proliferate homeostatically, and can persist for over 2 months. Our results suggest that long-lived and robust T cell immunity is generated following natural SARS-CoV-2 infection and support an important role of SARS-CoV-2-specific T cells in host control of COVID-19.

14.
Cell Rep ; 32(5): 107991, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32755584

RESUMO

A comprehensive understanding of the phenotype of persistent HIV-infected cells, transcriptionally active and/or transcriptionally inactive, is imperative for developing a cure. The relevance of cell-surface glycosylation to HIV persistence has never been explored. We characterize the relationship between cell-surface glycomic signatures and persistent HIV transcription in vivo. We find that the cell surface of CD4+ T cells actively transcribing HIV, despite suppressive therapy, harbors high levels of fucosylated carbohydrate ligands, including the cell extravasation mediator Sialyl-LewisX (SLeX), compared with HIV-infected transcriptionally inactive cells. These high levels of SLeX are induced by HIV transcription in vitro and are maintained after therapy in vivo. Cells with high-SLeX are enriched with markers associated with HIV susceptibility, signaling pathways that drive HIV transcription, and pathways involved in leukocyte extravasation. We describe a glycomic feature of HIV-infected transcriptionally active cells that not only differentiates them from their transcriptionally inactive counterparts but also may affect their trafficking abilities.


Assuntos
Terapia Antirretroviral de Alta Atividade , Linfócitos T CD4-Positivos/metabolismo , HIV/genética , Antígeno Sialil Lewis X/metabolismo , Transcrição Gênica , Linfócitos T CD4-Positivos/imunologia , Carboidratos/química , Linhagem Celular , Membrana Celular/metabolismo , Fucose/metabolismo , Glicômica , Glicosilação , Infecções por HIV/imunologia , Humanos , Memória Imunológica , Ligantes , Ativação Linfocitária/imunologia
15.
bioRxiv ; 2020 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-32577663

RESUMO

Convalescing COVID-19 patients mount robust T cell responses against SARS-CoV-2, suggesting an important role for T cells in viral clearance. To date, the phenotypes of SARS-CoV-2-specific T cells remain poorly defined. Using 38-parameter CyTOF, we phenotyped longitudinal specimens of SARS-CoV-2-specific CD4+ and CD8+ T cells from nine individuals who recovered from mild COVID-19. SARS-CoV-2-specific CD4+ T cells were exclusively Th1 cells, and predominantly Tcm with phenotypic features of robust helper function. SARS-CoV-2-specific CD8+ T cells were predominantly Temra cells in a state of less terminal differentiation than most Temra cells. Subsets of SARS-CoV-2-specific T cells express CD127, can homeostatically proliferate, and can persist for over two months. Our results suggest that long-lived and robust T cell immunity is generated following natural SARS-CoV-2 infection, and support an important role for SARS-CoV-2-specific T cells in host control of COVID-19.

16.
Elife ; 92020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32452381

RESUMO

The female reproductive tract (FRT) is the most common site of infection during HIV transmission to women, but viral remodeling complicates characterization of cells targeted for infection. Here, we report extensive phenotypic analyses of HIV-infected endometrial cells by CyTOF, and use a 'nearest neighbor' bioinformatics approach to trace cells to their original pre-infection phenotypes. Like in blood, HIV preferentially targets memory CD4+ T cells in the endometrium, but these cells exhibit unique phenotypes and sustain much higher levels of infection. Genital cell remodeling by HIV includes downregulating TCR complex components and modulating chemokine receptor expression to promote dissemination of infected cells to lymphoid follicles. HIV also upregulates the anti-apoptotic protein BIRC5, which when blocked promotes death of infected endometrial cells. These results suggest that HIV remodels genital T cells to prolong viability and promote viral dissemination and that interfering with these processes might reduce the likelihood of systemic viral spread.


Assuntos
Linfócitos T CD4-Positivos/virologia , Endométrio/virologia , Infecções por HIV/virologia , HIV/fisiologia , Interações Hospedeiro-Patógeno , Adulto , Endométrio/citologia , Feminino , Infecções por HIV/metabolismo , Humanos , Pessoa de Meia-Idade , Survivina/metabolismo , Adulto Jovem
17.
Virology ; 513: 91-97, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29040829

RESUMO

Bowman-Birk inhibitor (BBI) is a soybean-derived protease inhibitor that has anti-inflammation and anti-HIV effect. Here, we further investigated the anti-HIV action of BBI in macrophages, focusing on its effect on viral entry. We found that BBI could significantly block HIV entry into macrophages. Investigation of the mechanism(s) of the BBI action on HIV inhibition showed that BBI down-regulated the expression of CD4 receptor (as much as 80%) and induced the production of the CC chemokines (up to 60 folds at protein level) in macrophages. This inhibitory effect of BBI on HIV entry could be blocked by the neutralization antibodies to CC chemokines. These findings indicate that BBI may have therapeutic potential as a viral entry inhibitor for the prevention and treatment of HIV infection.


Assuntos
Inibidores da Fusão de HIV/metabolismo , HIV/efeitos dos fármacos , HIV/fisiologia , Macrófagos/virologia , Inibidor da Tripsina de Soja de Bowman-Birk/metabolismo , Internalização do Vírus/efeitos dos fármacos , Antígenos CD4/biossíntese , Células Cultivadas , Quimiocinas/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos
18.
Proc Natl Acad Sci U S A ; 114(27): E5352-E5361, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28630312

RESUMO

The epigenetic reader BRD4 plays a vital role in transcriptional regulation, cellular growth control, and cell-cycle progression. Dysregulation of BRD4 function has been implicated in the pathogenesis of a wide range of cancers. However, how BRD4 is regulated to maintain its normal function in healthy cells and how alteration of this process leads to cancer remain poorly understood. In this study, we discovered that BRD4 is hyperphosphorylated in NUT midline carcinoma and identified CDK9 as a potential kinase mediating BRD4 hyperphosphorylation. Disruption of BRD4 hyperphosphorylation using both chemical and molecular inhibitors led to the repression of BRD4 downstream oncogenes and abrogation of cellular transformation. BRD4 hyperphosphorylation is also observed in other cancers displaying enhanced BRD4 oncogenic activity. Our study revealed a mechanism that may regulate BRD4 biological function through phosphorylation, which, when dysregulated, could lead to oncogenesis. Our finding points to strategies to target the aberrant BRD4 signaling specifically for cancer intervention.


Assuntos
Carcinoma/genética , Carcinoma/metabolismo , Quinase 9 Dependente de Ciclina/química , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Fatores de Transcrição/química , Células A549 , Carcinogênese , Carcinoma/tratamento farmacológico , Proteínas de Ciclo Celular , Transformação Celular Neoplásica , Ensaios de Seleção de Medicamentos Antitumorais , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Proteínas de Neoplasias , Proteínas de Fusão Oncogênica/genética , Oncogenes , Fosforilação , Transdução de Sinais
19.
J Acquir Immune Defic Syndr ; 75(2): 232-240, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28328549

RESUMO

BACKGROUND: Human semen contains a factor that can enhance HIV infection up to 10-fold in cultures. This factor is termed semen-derived enhancer of virus infection (SEVI) and is composed of proteolytic fragments (PAP248-286) from prostatic acid phosphatase in semen. In this study, we examined whether macaque SEVI can facilitate simian immunodeficiency virus (SIV) or chimeric simian/human immunodeficiency virus (SHIV) infection. We also studied the effect of epigallocatechin gallate (EGCG) on macaque SEVI-mediated SIV or SHIV enhancement. METHODS: SIV or SHIV was mixed with different concentrations of macaque SEVI in the presence or absence of EGCG. The mixture was added to cultures of TZM-bl cells or macaque PBMCs. The effect of EGCG on macaque SEVI was measured by Congo-red staining assay and thioflavin T (ThT) fluorescence assay and was visualized by a transmission electron microscope. RESULTS: We identified that there is one amino acid difference at the site of 277 between human PAP248-286 and macaque PAP248-286. Macaque SEVI significantly enhanced SIV or SHIV infection of TZM-bl cells and macaque PBMCs. EGCG could block macaque SEVI-mediated enhancement of SIV or SHIV infection. Mechanistically, EGCG could degrade the formation of macaque SEVI amyloid fibrils that facilitates HIV attachment to the target cells. CONCLUSIONS: The finding that macaque SEVI could enhance SIV or SHIV infection indicates the possibility to use the macaque SEVI in vivo studies with the macaque models. In addition, future studies are necessary to examine whether EGCG can be used as an effective microbicide for preventing SIV or SHIV mucosal transmission.


Assuntos
Catequina/análogos & derivados , Inibidores de Proteases/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/transmissão , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Vírus da Imunodeficiência Símia/patogenicidade , Animais , Catequina/farmacologia , Macaca , Masculino , Mucosa/efeitos dos fármacos , Mucosa/virologia , Ligação Proteica , Sêmen/efeitos dos fármacos , Sêmen/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Ligação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...