Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Infect Dis ; 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38573164

RESUMO

Dysbiosis of the vaginal microbiome poses a serious risk for sexual HIV-1 transmission. Prevotella spp. are abundant during vaginal dysbiosis and associated with enhanced HIV-1 susceptibility; however, underlying mechanisms remain unclear. Here, we investigated the direct effect of vaginal bacteria on HIV-1 susceptibility of vaginal CD4+ T cells. Notably, pre-exposure to Prevotella timonensis enhanced HIV-1 uptake by vaginal T cells, leading to increased viral fusion and enhanced virus production. Pre-exposure to antiretroviral inhibitors abolished Prevotella timonensis-enhanced infection. Hence, our study shows that the vaginal microbiome directly affects mucosal CD4+ T cell susceptibility, emphasising importance of vaginal dysbiosis diagnosis and treatment.

2.
Nat Commun ; 15(1): 2465, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38548722

RESUMO

Chronic HIV-1 infection is characterized by T-cell dysregulation that is partly restored by antiretroviral therapy. Autophagy is a critical regulator of T-cell function. Here, we demonstrate a protective role for autophagy in HIV-1 disease pathogenesis. Targeted analysis of genetic variation in core autophagy gene ATG16L1 reveals the previously unidentified rs6861 polymorphism, which correlates functionally with enhanced autophagy and clinically with improved survival of untreated HIV-1-infected individuals. T-cells carrying ATG16L1 rs6861(TT) genotype display improved antiviral immunity, evidenced by increased proliferation, revamped immune responsiveness, and suppressed exhaustion/immunosenescence features. In-depth flow-cytometric and transcriptional profiling reveal T-helper-cell-signatures unique to rs6861(TT) individuals with enriched regulation of pro-inflammatory networks and skewing towards immunoregulatory phenotype. Therapeutic enhancement of autophagy recapitulates the rs6861(TT)-associated T-cell traits in non-carriers. These data underscore the in vivo relevance of autophagy for longer-lasting T-cell-mediated HIV-1 control, with implications towards development of host-directed antivirals targeting autophagy to restore immune function in chronic HIV-1 infection.


Assuntos
Infecções por HIV , HIV-1 , Humanos , HIV-1/genética , Proteínas Relacionadas à Autofagia/genética , Polimorfismo Genético , Autofagia/genética , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética
3.
Emerg Microbes Infect ; 12(1): 2195020, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36951188

RESUMO

SARS-CoV-2, the causative virus of COVID-19, continues to threaten global public health. COVID-19 is a multi-organ disease, causing not only respiratory distress, but also extrapulmonary manifestations, including gastrointestinal symptoms with SARS-CoV-2 RNA shedding in stool long after respiratory clearance. Despite global vaccination and existing antiviral treatments, variants of concern are still emerging and circulating. Of note, new Omicron BA.5 sublineages both increasingly evade neutralizing antibodies and demonstrate an increased preference for entry via the endocytic entry route. Alternative to direct-acting antivirals, host-directed therapies interfere with host mechanisms hijacked by viruses, and enhance cell-mediated resistance with a reduced likelihood of drug resistance development. Here, we demonstrate that the autophagy-blocking therapeutic berbamine dihydrochloride robustly prevents SARS-CoV-2 acquisition by human intestinal epithelial cells via an autophagy-mediated BNIP3 mechanism. Strikingly, berbamine dihydrochloride exhibited pan-antiviral activity against Omicron subvariants BA.2 and BA.5 at nanomolar potency, providing a proof of concept for the potential for targeting autophagy machinery to thwart infection of current circulating SARS-CoV-2 subvariants. Furthermore, we show that autophagy-blocking therapies limited virus-induced damage to intestinal barrier function, affirming the therapeutic relevance of autophagy manipulation to avert the intestinal permeability associated with acute COVID-19 and post-COVID-19 syndrome. Our findings underscore that SARS-CoV-2 exploits host autophagy machinery for intestinal dissemination and indicate that repurposed autophagy-based antivirals represent a pertinent therapeutic option to boost protection and ameliorate disease pathogenesis against current and future SARS-CoV-2 variants of concern.


Assuntos
COVID-19 , Hepatite C Crônica , Humanos , SARS-CoV-2 , Antivirais/farmacologia , Síndrome Pós-COVID-19 Aguda , RNA Viral , Anticorpos Neutralizantes , Autofagia , Anticorpos Antivirais , Glicoproteína da Espícula de Coronavírus , Proteínas de Membrana
4.
Sci Rep ; 13(1): 3283, 2023 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-36841916

RESUMO

Vaginal inflammation increases the risk for sexual HIV-1 transmission but underlying mechanisms remain unclear. In this study we assessed the impact of immune activation on HIV-1 susceptibility of primary human vaginal Langerhans cells (LCs). Vaginal LCs isolated from human vaginal tissue expressed a broad range of TLRs and became activated after exposure to both viral and bacterial TLR ligands. HIV-1 replication was restricted in immature vaginal LCs as only low levels of infection could be detected. Notably, activation of immature vaginal LCs by bacterial TLR ligands increased HIV-1 infection, whereas viral TLR ligands were unable to induce HIV-1 replication in vaginal LCs. Furthermore, mature vaginal LCs transmitted HIV-1 to CD4 T cells. This study emphasizes the role for vaginal LCs in protection against mucosal HIV-1 infection, which is abrogated upon activation. Moreover, our data suggest that bacterial STIs can increase the risk of HIV-1 acquisition in women.


Assuntos
Infecções por HIV , Soropositividade para HIV , HIV-1 , Infecções Sexualmente Transmissíveis , Humanos , Feminino , Células de Langerhans , HIV-1/fisiologia , Ligantes
5.
Viruses ; 13(2)2021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-33669846

RESUMO

Human immunodeficiency virus-1 (HIV-1) persists as a global health concern, with an incidence rate of approximately 2 million, and estimated global prevalence of over 35 million. Combination antiretroviral treatment is highly effective, but HIV-1 patients that have been treated still suffer from chronic inflammation and residual viral replication. It is therefore paramount to identify therapeutically efficacious strategies to eradicate viral reservoirs and ultimately develop a cure for HIV-1. It has been long accepted that the restriction factor tripartite motif protein 5 isoform alpha (TRIM5α) restricts HIV-1 infection in a species-specific manner, with rhesus macaque TRIM5α strongly restricting HIV-1, and human TRIM5α having a minimal restriction capacity. However, several recent studies underscore human TRIM5α as a cell-dependent HIV-1 restriction factor. Here, we present an overview of the latest research on human TRIM5α and propose a novel conceptualization of TRIM5α as a restriction factor with a varied portfolio of antiviral functions, including mediating HIV-1 degradation through autophagy- and proteasome-mediated mechanisms, and acting as a viral sensor and effector of antiviral signaling. We have also expanded on the protective antiviral roles of autophagy and outline the therapeutic potential of autophagy modulation to intervene in chronic HIV-1 infection.


Assuntos
Autofagia , Infecções por HIV/imunologia , Infecções por HIV/fisiopatologia , HIV-1/fisiologia , Proteínas com Motivo Tripartido/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Fatores de Restrição Antivirais , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/genética , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/genética , Replicação Viral
6.
Sci Rep ; 11(1): 4767, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33637808

RESUMO

Current direct-acting antiviral therapies are highly effective in suppressing HIV-1 replication. However, mucosal inflammation undermines prophylactic treatment efficacy, and HIV-1 persists in long-lived tissue-derived dendritic cells (DCs) and CD4+ T cells of treated patients. Host-directed strategies are an emerging therapeutic approach to improve therapy outcomes in infectious diseases. Autophagy functions as an innate antiviral mechanism by degrading viruses in specialized vesicles. Here, we investigated the impact of pharmaceutically enhancing autophagy on HIV-1 acquisition and viral replication. To this end, we developed a human tissue infection model permitting concurrent analysis of HIV-1 cellular targets ex vivo. Prophylactic treatment with autophagy-enhancing drugs carbamazepine and everolimus promoted HIV-1 restriction in skin-derived CD11c+ DCs and CD4+ T cells. Everolimus also decreased HIV-1 susceptibility to lab-adapted and transmitted/founder HIV-1 strains, and in vaginal Langerhans cells. Notably, we observed cell-specific effects of therapeutic treatment. Therapeutic rapamycin treatment suppressed HIV-1 replication in tissue-derived CD11c+ DCs, while all selected drugs limited viral replication in CD4+ T cells. Strikingly, both prophylactic and therapeutic treatment with everolimus or rapamycin reduced intestinal HIV-1 productive infection. Our findings highlight host autophagy pathways as an emerging target for HIV-1 therapies, and underscore the relevancy of repurposing clinically-approved autophagy drugs to suppress mucosal HIV-1 replication.


Assuntos
Fármacos Anti-HIV/farmacologia , Autofagia/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , HIV-1/fisiologia , Humanos , Mucosa/efeitos dos fármacos , Mucosa/virologia
7.
Mucosal Immunol ; 14(3): 743-750, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33568786

RESUMO

Semen is important in determining HIV-1 susceptibility but it is unclear how it affects virus transmission during sexual contact. Mucosal Langerhans cells (LCs) are the first immune cells to encounter HIV-1 during sexual contact and have a barrier function as LCs are restrictive to HIV-1. As semen from people living with HIV-1 contains complement-opsonized HIV-1, we investigated the effect of complement on HIV-1 dissemination by human LCs in vitro and ex vivo. Notably, pre-treatment of HIV-1 with semen enhanced LC infection compared to untreated HIV-1 in the ex vivo explant model. Infection of LCs and transmission to target cells by opsonized HIV-1 was efficiently inhibited by blocking complement receptors CR3 and CR4. Complement opsonization of HIV-1 enhanced uptake, fusion, and integration by LCs leading to an increased transmission of HIV-1 to target cells. However, in the absence of both CR3 and CR4, C-type lectin receptor langerin was able to restrict infection of complement-opsonized HIV-1. These data suggest that complement enhances HIV-1 infection of LCs by binding CR3 and CR4, thereby bypassing langerin and changing the restrictive nature of LCs into virus-disseminating cells. Targeting complement factors might be effective in preventing HIV-1 transmission.


Assuntos
Infecções por HIV/imunologia , HIV-1/fisiologia , Células de Langerhans/imunologia , Sêmen/imunologia , Anticorpos Bloqueadores/metabolismo , Antígenos CD/metabolismo , Linhagem Celular , Ativação do Complemento , Transmissão de Doença Infecciosa , Infecções por HIV/transmissão , HIV-1/patogenicidade , Interações Hospedeiro-Parasita , Humanos , Evasão da Resposta Imune , Integrina alfaXbeta2/metabolismo , Lectinas Tipo C/metabolismo , Antígeno de Macrófago 1/metabolismo , Lectinas de Ligação a Manose/metabolismo , Opsonização , Sêmen/virologia
8.
Viruses ; 12(11)2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-33238561

RESUMO

Animal models and cell lines are invaluable for virology research and host-pathogen interaction studies. However, it is increasingly evident that these models are not sufficient to fully understand human viral diseases. With the advent of three-dimensional organotypic cultures, it is now possible to study viral infections in the human context. This perspective explores the potential of these organotypic cultures, also known as organoids, for virology research, antiviral testing, and shaping the virology landscape.


Assuntos
Interações Hospedeiro-Patógeno , Organoides/virologia , Pesquisa , Técnicas de Cultura de Tecidos , Virologia/métodos , Humanos , Modelos Biológicos
9.
Int J Mol Sci ; 21(21)2020 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-33114346

RESUMO

Hijacking and manipulation of host cell biosynthetic pathways by human enveloped viruses are essential for the viral lifecycle. Flaviviridae members, including hepatitis C, dengue and Zika viruses, extensively manipulate host lipid metabolism, underlining the importance of lipid droplets (LDs) in viral infection. LDs are dynamic cytoplasmic organelles that can act as sequestration platforms for a unique subset of host and viral proteins. Transient recruitment and mobilization of proteins to LDs during viral infection impacts host-cell biological properties, LD functionality and canonical protein functions. Notably, recent studies identified LDs in the nucleus and also identified that LDs are transported extracellularly via an autophagy-mediated mechanism, indicating a novel role for autophagy in Flaviviridae infections. These developments underline an unsuspected diversity and localization of LDs and potential moonlighting functions of LD-associated proteins during infection. This review summarizes recent breakthroughs concerning the LD hijacking activities of hepatitis C, dengue and Zika viruses and potential roles of cytoplasmic, nuclear and extracellular LD-associated viral proteins during infection.


Assuntos
Flaviviridae/patogenicidade , Gotículas Lipídicas/metabolismo , Proteínas Virais/metabolismo , Animais , Autofagia , Núcleo Celular/metabolismo , Vírus da Dengue/metabolismo , Vírus da Dengue/patogenicidade , Espaço Extracelular/metabolismo , Flaviviridae/metabolismo , Hepacivirus/metabolismo , Hepacivirus/patogenicidade , Humanos , Gotículas Lipídicas/virologia , Zika virus/metabolismo , Zika virus/patogenicidade
10.
Front Immunol ; 11: 503, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32292405

RESUMO

Sexually transmitted Hepatitis C virus (HCV) infections and high reinfections are a major concern amongst men who have sex with men (MSM) living with HIV-1 and HIV-negative MSM. Immune activation and/or HIV-1 coinfection enhance HCV susceptibility via sexual contact, suggesting that changes in immune cells or external factors are involved in increased susceptibility. Activation of anal mucosal Langerhans cells (LCs) has been implicated in increased HCV susceptibility as activated but not immature LCs efficiently retain and transmit HCV to other cells. However, the underlying molecular mechanism of transmission remains unclear. Here we identified the Heparan Sulfate Proteoglycan Syndecan 4 as the molecular switch, controlling HCV transmission by LCs. Syndecan 4 was highly upregulated upon activation of LCs and interference with Heparan Sulfate Proteoglycans or silencing of Syndecan 4 abrogated HCV transmission. These data strongly suggest that Syndecan 4 mediates HCV transmission by activated LCs. Notably, our data also identified the C-type lectin receptor langerin as a restriction factor for HCV infection and transmission. Langerin expression abrogated HCV infection in HCV permissive cells, whereas langerin expression on the Syndecan 4 expressing cell line strongly decreased HCV transmission to a target hepatoma cell line. These data suggest that the balanced interplay between langerin restriction and Syndecan 4 transmission determines HCV dissemination. Silencing of langerin enhanced HCV transmission whereas silencing Syndecan 4 on activated LCs decreased transmission. Blocking Heparan Sulfate Proteoglycans abrogated HCV transmission by LCs ex vivo identifying Heparan Sulfate Proteoglycans and Syndecan 4 as potential targets to prevent sexual transmission of HCV. Thus, our data strongly suggest that the interplay between receptors promotes or restricts transmission and further indicate that Syndecan 4 is the molecular switch controlling HCV susceptibility after sexual contact.


Assuntos
Antígenos CD/metabolismo , Infecções por HIV/metabolismo , HIV-1/fisiologia , Hepacivirus/fisiologia , Hepatite C/metabolismo , Células de Langerhans/fisiologia , Lectinas Tipo C/metabolismo , Lectinas de Ligação a Manose/metabolismo , Infecções Sexualmente Transmissíveis/metabolismo , Sindecana-4/metabolismo , Antígenos CD/genética , Diferenciação Celular , Linhagem Celular , Coinfecção , Transmissão de Doença Infecciosa , Homossexualidade Masculina , Humanos , Lectinas Tipo C/genética , Masculino , Lectinas de Ligação a Manose/genética , RNA Interferente Pequeno/genética , Sindecana-4/genética , Regulação para Cima
11.
J Reprod Immunol ; 138: 103085, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32004804

RESUMO

Dysbiosis of the vaginal microbiome as a result of overgrowth of anaerobic bacteria leads to bacterial vaginosis (BV) which is associated with increased inflammation in the genital mucosa. Moreover, BV increases susceptibility to sexual transmitted infections (STIs) and is associated with adverse pregnancy outcomes. It remains unclear how specific vaginal aerobic and anaerobic bacteria affect health and disease. We selected different vaginal bacteria ranging from true commensals to species associated with dysbiosis and investigated their effects on activation of dendritic cells (DCs). Commensal Lactobacilli crispatus did not induce DC maturation nor led to production of pro-inflammatory cytokines. In contrast, BV-associated bacteria Megasphaera elsdenii and Prevotella timonensis induced DC maturation and increased levels of pro-inflammatory cytokines. Notably, DCs stimulated with Prevotella timonensis suppressed Th2 responses and induced Th1 skewing, typically associated with preterm birth. In contrast, Lactobacillus crispatus and Megasphaera elsdenii did not affect Th cell polarization. These results strongly indicate that the interaction of vaginal bacteria with mucosal DCs determines mucosal inflammation and we have identified the anaerobic bacterium Prevotella timonensis as a strong inducer of inflammatory responses. Specifically targeting these inflammation-inducing bacteria might be a therapeutic strategy to prevent BV and associated risks in STI susceptibility and preterm birth.


Assuntos
Células Dendríticas/imunologia , Disbiose/complicações , Megasphaera elsdenii/imunologia , Prevotella/imunologia , Vaginose Bacteriana/imunologia , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/metabolismo , Disbiose/imunologia , Disbiose/microbiologia , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Leucócitos Mononucleares , Prevotella/isolamento & purificação , Cultura Primária de Células , Vagina/citologia , Vagina/imunologia , Vagina/microbiologia , Vaginose Bacteriana/microbiologia
12.
PLoS One ; 14(12): e0226651, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31856198

RESUMO

A single HIV-1 variant establishes infection of the host after sexual contact. Identifying the phenotypic characteristics of these Transmitted Founder (T/F) viruses is important to understand the restriction mechanisms during transmission. Langerhans cells (LCs) are the mucosal dendritic cell subset that has been shown to have a protective role in HIV-1 transmission. Immature LCs efficiently capture and degrade HIV-1 via langerin-mediated restriction. Here we have investigated the capacity of T/F HIV-1 strains to infect mucosal Langerhans cells (LCs). Notably, most T/F variants efficiently infected immature LCs derived from skin and vaginal tissue in contrast to chronic HIV-1 laboratory strains. Next we screened a panel of T/F viruses and their matched 6-month consensus sequence viruses. Interestingly most T/F variants infected immature LCs whereas donor-matched 6-month consensus sequence viruses had lost the ability to infect LCs. However, we also identified 6-month consensus sequence viruses that had retained an ability to infect LCs similar to that of the donor-matched T/F virus. Moreover, some T/F viruses and 6-month consensus sequence viruses were unable to infect immature LCs. Further analyses indicated that T/F viruses are less sensitive to langerin-mediated restriction. These data suggest that T/F HIV-1 variants have the ability to infect immature LCs, which will facilitate transmission.


Assuntos
HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Células de Langerhans/virologia , Antígenos CD/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Células de Langerhans/imunologia , Lectinas Tipo C/metabolismo , Lectinas de Ligação a Manose/metabolismo
14.
Curr Opin Virol ; 22: 112-119, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28113112

RESUMO

Innate sensing of HIV-1 by dendritic cells (DCs) initiates cell-intrinsic signalling programs that direct virus restriction and antiviral defenses. These responses include the production of type I interferon (IFN) and a large number of IFN-stimulated genes (ISGs) with a broad spectrum of antiviral effector functions. Initial interactions of HIV-1 at the mucosal surfaces with DC-expressed innate immune factors including cGAS, TRIM5α and SAMHD1 are predictive of viraemia, inflammation and disease pathogenesis. Here, we review the molecular basis of HIV-1 sensing in the two major mucosal DC subsets, that is, epithelial Langerhans cells and subepithelial CD11c+ conventional DCs. We discuss the concerted actions of the host restriction factors and innate sensors as well as viral evasion mechanisms in determining HIV-1 susceptibility to infection and directing antiviral adaptive immune responses.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , HIV-1/imunologia , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Imunidade Inata , Humanos , Fatores Imunológicos/metabolismo
15.
Nat Immunol ; 18(2): 225-235, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28024153

RESUMO

The mechanisms by which human immunodeficiency virus 1 (HIV-1) avoids immune surveillance by dendritic cells (DCs), and thereby prevents protective adaptive immune responses, remain poorly understood. Here we showed that HIV-1 actively arrested antiviral immune responses by DCs, which contributed to efficient HIV-1 replication in infected individuals. We identified the RNA helicase DDX3 as an HIV-1 sensor that bound abortive HIV-1 RNA after HIV-1 infection and induced DC maturation and type I interferon responses via the signaling adaptor MAVS. Notably, HIV-1 recognition by the C-type lectin receptor DC-SIGN activated the mitotic kinase PLK1, which suppressed signaling downstream of MAVS, thereby interfering with intrinsic host defense during HIV-1 infection. Finally, we showed that PLK1-mediated suppression of DDX3-MAVS signaling was a viral strategy that accelerated HIV-1 replication in infected individuals.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Dendríticas/virologia , Infecções por HIV/imunologia , HIV-1/fisiologia , Evasão da Resposta Imune , Imunidade , Macrófagos/virologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Extratos Celulares , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Estudos de Coortes , RNA Helicases DEAD-box/metabolismo , Células Dendríticas/imunologia , Regulação Viral da Expressão Gênica , Células HEK293 , Infecções por HIV/virologia , Interações Hospedeiro-Patógeno/genética , Humanos , Interferon beta/sangue , Macrófagos/imunologia , Polimorfismo de Nucleotídeo Único , RNA Viral/imunologia , RNA Viral/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais , Carga Viral/genética
16.
Nature ; 540(7633): 448-452, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27919079

RESUMO

The most prevalent route of HIV-1 infection is across mucosal tissues after sexual contact. Langerhans cells (LCs) belong to the subset of dendritic cells (DCs) that line the mucosal epithelia of vagina and foreskin and have the ability to sense and induce immunity to invading pathogens. Anatomical and functional characteristics make LCs one of the primary targets of HIV-1 infection. Notably, LCs form a protective barrier against HIV-1 infection and transmission. LCs restrict HIV-1 infection through the capture of HIV-1 by the C-type lectin receptor Langerin and subsequent internalization into Birbeck granules. However, the underlying molecular mechanism of HIV-1 restriction in LCs remains unknown. Here we show that human E3-ubiquitin ligase tri-partite-containing motif 5α (TRIM5α) potently restricts HIV-1 infection of LCs but not of subepithelial DC-SIGN+ DCs. HIV-1 restriction by TRIM5α was thus far considered to be reserved to non-human primate TRIM5α orthologues, but our data strongly suggest that human TRIM5α is a cell-specific restriction factor dependent on C-type lectin receptor function. Our findings highlight the importance of HIV-1 binding to Langerin for the routeing of HIV-1 into the human TRIM5α-mediated restriction pathway. TRIM5α mediates the assembly of an autophagy-activating scaffold to Langerin, which targets HIV-1 for autophagic degradation and prevents infection of LCs. By contrast, HIV-1 binding to DC-SIGN+ DCs leads to disassociation of TRIM5α from DC-SIGN, which abrogates TRIM5α restriction. Thus, our data strongly suggest that restriction by human TRIM5α is controlled by C-type-lectin-receptor-dependent uptake of HIV-1, dictating protection or infection of human DC subsets. Therapeutic interventions that incorporate C-type lectin receptors and autophagy-targeting strategies could thus provide cell-mediated resistance to HIV-1 in humans.


Assuntos
Antígenos CD/metabolismo , Autofagia , Proteínas de Transporte/metabolismo , HIV-1/fisiologia , Células de Langerhans/metabolismo , Células de Langerhans/virologia , Lectinas Tipo C/metabolismo , Lectinas de Ligação a Manose/metabolismo , Receptores de HIV/metabolismo , Fatores de Restrição Antivirais , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Infecções por HIV/transmissão , HIV-1/imunologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade nas Mucosas , Células de Langerhans/citologia , Células de Langerhans/imunologia , Receptores de Superfície Celular/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
17.
Dev Comp Immunol ; 50(2): 146-54, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25681740

RESUMO

Common carp thrombocytes account for 30-40% of peripheral blood leukocytes and are abundant in the healthy animals' spleen, the thrombopoietic organ. We show that, ex vivo, thrombocytes from healthy carp express a large number of immune-relevant genes, among which several cytokines and Toll-like receptors, clearly pointing at immune functions of carp thrombocytes. Few studies have described the role of fish thrombocytes during infection. Carp are natural host to two different but related protozoan parasites, Trypanoplasma borreli and Trypanosoma carassii, which reside in the blood and tissue fluids. We used the two parasites to undertake controlled studies on the role of fish thrombocytes during these infections. In vivo, but only during infection with T. borreli, thrombocytes were massively depleted from the blood and spleen leading to severe thrombocytopenia. Ex vivo, addition of nitric oxide induced a clear and rapid apoptosis of thrombocytes from healthy carp, supporting a role for nitric oxide-mediated control of immune-relevant thrombocytes during infection with T. borreli. The potential advantage for parasites to selectively deplete the host of thrombocytes via nitric oxide-induced apoptosis is discussed.


Assuntos
Apoptose/imunologia , Plaquetas/imunologia , Carpas/imunologia , Óxido Nítrico/metabolismo , Trypanosoma/imunologia , Animais , Plaquetas/parasitologia , Carpas/parasitologia , Citocinas/biossíntese , Citocinas/genética , Citocinas/imunologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/parasitologia , Citometria de Fluxo , Trombocitopenia/imunologia , Trombocitopenia/parasitologia , Receptores Toll-Like/biossíntese , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
18.
Retrovirology ; 11: 52, 2014 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-24990163

RESUMO

BACKGROUND: Sexual transmission is the main route of HIV-1 infection and the CCR5-using (R5) HIV-1 is predominantly transmitted, even though CXCR4-using (X4) HIV-1 is often abundant in chronic HIV-1 patients. The mechanisms underlying this tropism selection are unclear. Mucosal Langerhans cells (LCs) are the first immune cells to encounter HIV-1 and here we investigated the role of LCs in selection of R5 HIV-1 using an ex vivo epidermal and vaginal transmission models. RESULTS: Immature LCs were productively infected by X4 as well as R5 HIV-1. However, only R5 but not X4 viruses were selectively transmitted by immature LCs to T cells. Transmission of HIV-1 was depended on de novo production of HIV-1 in LCs, since it could be inhibited by CCR5 fusion inhibitors as well as reverse transcription inhibitors. Notably, the activation state of LCs affected the restriction in X4 HIV-1 transmission; immune activation by TNF facilitated transmission of X4 as well as R5 HIV-1. CONCLUSIONS: These data suggest that LCs play a crucial role in R5 selection and that immature LCs effectively restrict X4 at the level of transmission.


Assuntos
Infecções por HIV/transmissão , HIV-1/fisiologia , Células de Langerhans/fisiologia , Receptores CXCR4/fisiologia , Humanos , Células de Langerhans/virologia , Receptores CXCR4/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Replicação Viral
19.
Retrovirology ; 11: 123, 2014 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-25551286

RESUMO

BACKGROUND: Human Langerhans cells (LCs) reside in foreskin and vaginal mucosa and are the first immune cells to interact with HIV-1 during sexual transmission. LCs capture HIV-1 through the C-type lectin receptor langerin, which routes the virus into Birbeck granules (BGs), thereby preventing HIV-1 infection. BGs are langerin-positive organelles exclusively present in LCs, however, their origin and function are unknown. RESULTS: Here, we not only show that langerin and caveolin-1 co-localize at the cell membrane and in vesicles but also that BGs are langerin/caveolin-1-positive vesicles are linked to the lysosomal degradation pathway in LCs. Moreover, inhibition of caveolar endocytosis in primary LCs abrogated HIV-1 sequestering into langerin(+) caveolar structures. Notably, both inhibition of caveolar uptake and silencing of caveolar structure protein caveolin-1 resulted in increased HIV-1 integration and subsequent infection. In contrast, inhibition of clathrin-mediated endocytosis did not affect HIV-1 integration, even though HIV-1 uptake was decreased, suggesting that clathrin-mediated endocytosis is not involved in HIV-1 restriction in LCs. CONCLUSIONS: Thus, our data strongly indicate that BGs belong to the caveolar endocytosis pathway and that caveolin-1 mediated HIV-1 uptake is an intrinsic restriction mechanism present in human LCs that prevents HIV-1 infection. Harnessing this particular internalization pathway has the potential to facilitate strategies to combat HIV-1 transmission.


Assuntos
Antígenos CD/metabolismo , Caveolina 1/metabolismo , Endocitose , HIV-1/fisiologia , Células de Langerhans/virologia , Lectinas Tipo C/metabolismo , Lectinas de Ligação a Manose/metabolismo , Internalização do Vírus , Vesículas Citoplasmáticas/virologia , Humanos
20.
Dev Comp Immunol ; 36(4): 741-50, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22182503

RESUMO

Numerous CXC chemokines have been identified in fish, however, their role in inflammation is not well established. Here, CXC chemokines of the CXCL8-like (CXCa_L1 and CXCL8_L2) and CXCL9/10/11-like (CXCb) subset were investigated in carp. Recombinant CXCa_L1, CXCL8_L2 and CXCb all stimulated chemotaxis of macrophages and granulocytes in vitro. CXCb also attracted lymphocytes. Distinct effects on phagocyte activation were observed: the CXCL8-like chemokines increase respiratory burst activity, but not nitrite production. The three chemokines differentially induced a moderate increase in IL-1ß, CXCa_L1 and CXCL8_L2 gene expression. Intracellular calcium mobilization in granulocytes upon CXCa_L1 stimulation implies signal transduction through G-protein coupled CXC receptors. Notably, upon intraperitoneal administration, carp CXCL8-like chemokines strongly induced in vivo leukocyte recruitment, including neutrophils and monocytes/macrophages, in contrast to CXCb, for which the number of recruited leukocytes was low. The results indicate functional homology for carp CXCL8-like and CXCb chemokines with mammalian CXCL8 and CXCL9-11, respectively.


Assuntos
Carpas/imunologia , Quimiocinas CXC/imunologia , Proteínas de Peixes/imunologia , Animais , Granulócitos/imunologia , Rim Cefálico/citologia , Macrófagos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...