Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 9(4)2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32326440

RESUMEN

Langerhans cells (LC) are the resident antigen presenting cells of the mucosal epithelium and play an essential role in initiating immune responses. LC are the only cells in the body to contain Birbeck granules (BG), which are unique cytoplasmic organelles comprised of c-type lectin langerin. Studies of BG have historically focused on morphological characterizations, but BG have also been implicated in viral antigen processing which suggests that they can serve a function in antiviral immunity. This study focused on investigating proteins that could be involved in BG formation to further characterize their structure using transmission electron microscopy (TEM). Here, we report a critical role for the protein annexin A2 (anxA2) in the proper formation of BG structures. When anxA2 expression is downregulated, langerin expression decreases, cytoplasmic BG are nearly ablated, and the presence of malformed BG-like structures increases. Furthermore, in the absence of anxA2, we found langerin was no longer localized to BG or BG-like structures. Taken together, these results indicate an essential role for anxA2 in facilitating the proper formation of BG.


Asunto(s)
Anexina A2/metabolismo , Gránulos Citoplasmáticos/metabolismo , Células de Langerhans/metabolismo , Antígenos CD , Línea Celular , Gránulos Citoplasmáticos/ultraestructura , Humanos , Células de Langerhans/ultraestructura , Lectinas Tipo C , Lectinas de Unión a Manosa , Subunidades de Proteína/metabolismo , Transporte de Proteínas
2.
Sci Adv ; 6(14): eaay2793, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32270032

RESUMEN

Activation-induced cytidine deaminase (AID) mediates immunoglobulin class switch DNA recombination (CSR) and somatic hypermutation (SHM), critical processes for maturation of the antibody response. Epigenetic factors, such as histone deacetylases (HDACs), would underpin B cell differentiation stage-specific AID expression. Here, we showed that NAD+-dependent class III HDAC sirtuin 1 (Sirt1) is highly expressed in resting B cells and down-regulated by stimuli inducing AID. B cell Sirt1 down-regulation, deprivation of NAD+ cofactor, or genetic Sirt1 deletion reduced deacetylation of Aicda promoter histones, Dnmt1, and nuclear factor-κB (NF-κB) p65 and increased AID expression. This promoted class-switched and hypermutated T-dependent and T-independent antibody responses or led to generation of autoantibodies. Genetic Sirt1 overexpression, Sirt1 boost by NAD+, or allosteric Sirt1 enhancement by SRT1720 repressed AID expression and CSR/SHM. By deacetylating histone and nonhistone proteins (Dnmt1 and NF-κB p65), Sirt1 transduces metabolic cues into epigenetic changes to play an important B cell-intrinsic role in modulating antibody and autoantibody responses.


Asunto(s)
Formación de Anticuerpos/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Citidina Desaminasa/genética , Epigénesis Genética , Sirtuina 1/metabolismo , Animales , Formación de Anticuerpos/inmunología , Biomarcadores , Citidina Desaminasa/metabolismo , Metilación de ADN , Humanos , Inmunofenotipificación , Ratones , Regiones Promotoras Genéticas , Linfocitos T/inmunología , Linfocitos T/metabolismo
4.
J Immunol ; 204(5): 1146-1157, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31932498

RESUMEN

Upon activation by CD40 or TLR signaling, B lymphocytes activate NF-κB to induce activation-induced cytidine deaminase and, therefore, Ig class switch DNA recombination, as central to the maturation of the Ab and autoantibody responses. In this study, we show that NF-κB activation is boosted by colocalization of engaged immune receptors, such as CD40, with RAB7 small GTPase on mature endosomes, in addition to signals emanating from the receptors localized on the plasma membrane, in mouse B cells. In mature endosomes, RAB7 directly interacts with TRAF6 E3 ubiquitin ligase, which catalyzes K63 polyubiquitination for NF-κB activation. RAB7 overexpression in Cd19+/creRosa26fl-STOP-fl-Rab7 mouse B cells upregulates K63 polyubiquitination activity of TRAF6, enhances NF-κB activation and activation-induced cytidine deaminase induction, and boosts IgG Ab and autoantibody levels. This, together with the extensive intracellular localization of CD40 and the strong correlation of RAB7 expression with NF-κB activation in mouse lupus B cells, shows that RAB7 is an integral component of the B cell NF-κB activation machinery, likely through interaction with TRAF6 for the assembly of "intracellular membrane signalosomes."


Asunto(s)
Linfocitos B/inmunología , Endosomas/inmunología , Cambio de Clase de Inmunoglobulina , FN-kappa B/inmunología , Factor 6 Asociado a Receptor de TNF/inmunología , Ubiquitinación/inmunología , Proteínas de Unión al GTP rab/inmunología , Animales , Antígenos CD19/genética , Antígenos CD19/inmunología , Linfocitos B/citología , Endosomas/genética , Ratones , Ratones Transgénicos , FN-kappa B/genética , Factor 6 Asociado a Receptor de TNF/genética , Ubiquitinación/genética , Proteínas de Unión al GTP rab/genética , Proteínas de Unión a GTP rab7
5.
Nat Commun ; 11(1): 60, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31896754

RESUMEN

Short-chain fatty acids (SCFAs) butyrate and propionate are metabolites from dietary fiber's fermentation by gut microbiota that can affect differentiation or functions of T cells, macrophages and dendritic cells. We show here that at low doses these SCFAs directly impact B cell intrinsic functions to moderately enhance class-switch DNA recombination (CSR), while decreasing at higher doses over a broad physiological range, AID and Blimp1 expression, CSR, somatic hypermutation and plasma cell differentiation. In human and mouse B cells, butyrate and propionate decrease B cell Aicda and Prdm1 by upregulating select miRNAs that target Aicda and Prdm1 mRNA-3'UTRs through inhibition of histone deacetylation (HDAC) of those miRNA host genes. By acting as HDAC inhibitors, not as energy substrates or through GPR-engagement signaling in these B cell-intrinsic processes, these SCFAs impair intestinal and systemic T-dependent and T-independent antibody responses. Their epigenetic impact on B cells extends to inhibition of autoantibody production and autoimmunity in mouse lupus models.


Asunto(s)
Anticuerpos/genética , Epigénesis Genética/efectos de los fármacos , Ácidos Grasos Volátiles/farmacología , Microbioma Gastrointestinal/inmunología , Animales , Anticuerpos/inmunología , Anticuerpos/metabolismo , Autoanticuerpos/genética , Autoanticuerpos/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Butiratos/farmacología , Citidina Desaminasa/antagonistas & inhibidores , Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , Citidina Desaminasa/metabolismo , Fibras de la Dieta , Ácidos Grasos Volátiles/aislamiento & purificación , Ácidos Grasos Volátiles/farmacocinética , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Inhibidores de Histona Desacetilasas/inmunología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Ratones Endogámicos C57BL , Ratones Mutantes , Factor 1 de Unión al Dominio 1 de Regulación Positiva/antagonistas & inhibidores , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/inmunología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Propionatos/farmacología , Distribución Tisular
6.
Front Microbiol ; 9: 2954, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30568638

RESUMEN

Viral life cycles consist of three main phases: (1) attachment and entry, (2) genome replication and expression, and (3) assembly, maturation, and egress. Each of these steps is intrinsically reliant on host cell factors and processes including cellular receptors, genetic replication machinery, endocytosis and exocytosis, and protein expression. Annexin A2 (AnxA2) is a membrane-associated protein with a wide range of intracellular functions and a recurrent host factor in a variety of viral infections. Spatially, AnxA2 is found in the nucleus and cytoplasm, vesicle-bound, and on the inner and outer leaflet of the plasma membrane. Structurally, AnxA2 exists as a monomer or in complex with S100A10 to form the AnxA2/S100A10 heterotetramer (A2t). Both AnxA2 and A2t have been implicated in a vast array of cellular functions such as endocytosis, exocytosis, membrane domain organization, and translational regulation through RNA binding. Accordingly, many discoveries have been made involving AnxA2 in viral pathogenesis, however, the reported work addressing AnxA2 in virology is highly compartmentalized. Therefore, the purpose of this mini review is to provide information regarding the role of AnxA2 in the lifecycle of multiple epithelial cell-targeting viruses to highlight recurrent themes, identify discrepancies, and reveal potential avenues for future research.

7.
Sci Rep ; 8(1): 11642, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-30076379

RESUMEN

Human papillomavirus (HPV) entry into epithelial cells is independent of canonical endocytic pathways. Upon interaction with host cells, HPV establishes infection by traversing through an endocytic pathway that is clathrin- and caveolin-independent, but dependent on the annexin A2/S100A10 heterotetramer (A2t). We examined the contribution of monomeric annexin A2 (AnxA2) vs. A2t in HPV infection and endocytosis, and further characterized the role of these molecules in protein trafficking. We specifically show that cell surface A2t is not required for HPV attachment, and in the absence of A2t virion internalization remains clathrin-independent. Without A2t, viral progression from early endosomes to multivesicular endosomes is significantly inhibited, capsid uncoating is dramatically reduced, and lysosomal degradation of HPV is accelerated. Furthermore, we present evidence that AnxA2 forms a complex with CD63, a known mediator of HPV trafficking. Overall, the observed reduction in infection is less significant in the absence of S100A10 alone compared to full A2t, supporting an independent role for monomeric AnxA2. More broadly, we show that successful infection by multiple oncogenic HPV types is dependent on A2t. These findings suggest that A2t is a central mediator of high-risk HPV intracellular trafficking post-entry and pre-viral uncoating.


Asunto(s)
Anexina A2/genética , Papillomaviridae/genética , Infecciones por Papillomavirus/genética , Proteínas S100/genética , Anexina A2/química , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Endocitosis/genética , Células Epiteliales/virología , Células HeLa , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/patogenicidad , Humanos , Lisosomas/genética , Papillomaviridae/patogenicidad , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Multimerización de Proteína/genética , Transporte de Proteínas/genética , Proteolisis , Proteínas S100/química , Virión/genética , Virión/patogenicidad
8.
Nat Commun ; 8: 14244, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28176781

RESUMEN

Antibody class-switch DNA recombination (CSR) is initiated by AID-introduced DSBs in the switch (S) regions targeted for recombination, as effected by Ku70/Ku86-mediated NHEJ. Ku-deficient B cells, however, undergo (reduced) CSR through an alternative(A)-NHEJ pathway, which introduces microhomologies in S-S junctions. As microhomology-mediated end-joining requires annealing of single-strand DNA ends, we addressed the contribution of single-strand annealing factors HR Rad52 and translesion DNA polymerase θ to CSR. Compared with their Rad52+/+ counterparts, which display normal CSR, Rad52-/- B cells show increased CSR, fewer intra-Sµ region recombinations, no/minimal microhomologies in S-S junctions, decreased c-Myc/IgH translocations and increased Ku70/Ku86 recruitment to S-region DSB ends. Rad52 competes with Ku70/Ku86 for binding to S-region DSB ends. It also facilitates a Ku-independent DSB repair, which favours intra-S region recombination and mediates, particularly in Ku absence, inter-S-S recombination, as emphasized by the significantly greater CSR reduction in Rad52-/- versus Rad52+/+ B cells on Ku86 knockdown.


Asunto(s)
Reparación del ADN por Unión de Extremidades/inmunología , Cambio de Clase de Inmunoglobulina/genética , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Reparación del ADN por Recombinación/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , Citidina Desaminasa/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/genética , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/inmunología , ADN Polimerasa Dirigida por ADN/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Cambio de Clase de Inmunoglobulina/inmunología , Región de Cambio de la Inmunoglobulina/genética , Autoantígeno Ku/genética , Autoantígeno Ku/inmunología , Autoantígeno Ku/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/genética , Proteína Recombinante y Reparadora de ADN Rad52/inmunología , Sulfonamidas , ADN Polimerasa theta
9.
Virol J ; 13(1): 187, 2016 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-27863502

RESUMEN

During sexual transmission of human immunodeficiency virus (HIV), macrophages are initial targets for HIV infection. Secretory leukocyte protease inhibitor (SLPI) has been shown to protect against HIV infection of macrophages through interactions with annexin A2 (A2), which is found on the macrophage cell surface as a heterotetramer (A2t) consisting of A2 and S100A10. Therefore, we investigated potential protein-protein interactions between A2 and HIV-1 gp120 through a series of co-immunoprecipitation assays and a single molecule pulldown (SiMPull) technique. Additionally, inhibitors of A2t (A2ti) that target the interaction between A2 and S100A10 were tested for their ability to impair productive HIV-1 infection of macrophages. Our data suggest that interactions between HIV-1 gp120 and A2 exist, though this interaction may be indirect. Furthermore, an anti-A2 antibody impaired HIV-1 particle production in macrophages in vitro, whereas A2ti did not indicating that annexin A2 may promote HIV-1 infection of macrophages in its monomeric rather than tetrameric form.


Asunto(s)
Anexina A2/antagonistas & inhibidores , VIH-1/inmunología , VIH-1/fisiología , Interacciones Huésped-Patógeno , Macrófagos/virología , Replicación Viral , Anexina A2/metabolismo , Anticuerpos/metabolismo , Centrifugación , Proteína gp120 de Envoltorio del VIH/metabolismo , Humanos , Inmunoprecipitación , Unión Proteica , Mapeo de Interacción de Proteínas
10.
AIDS Patient Care STDS ; 30(7): 291-306, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27410493

RESUMEN

In the last three decades, extensive research on human immunodeficiency virus (HIV) has highlighted its capability to exploit a variety of strategies to enter and infect immune cells. Although CD4(+) T cells are well known as the major HIV target, with infection occurring through the canonical combination of the cluster of differentiation 4 (CD4) receptor and either the C-C chemokine receptor type 5 (CCR5) or C-X-C chemokine receptor type 4 (CXCR4) coreceptors, HIV has also been found to enter other important immune cell types such as macrophages, dendritic cells, Langerhans cells, B cells, and granulocytes. Interestingly, the expression of distinct cellular cofactors partially regulates the rate in which HIV infects each distinct cell type. Furthermore, HIV can benefit from the acquisition of new proteins incorporated into its envelope during budding events. While several publications have investigated details of how HIV manipulates particular cell types or subtypes, an up-to-date comprehensive review on HIV tropism for different immune cells is lacking. Therefore, this review is meant to focus on the different receptors, coreceptors, and cofactors that HIV exploits to enter particular immune cells. Additionally, prophylactic approaches that have targeted particular molecules associated with HIV entry and infection of different immune cells will be discussed. Unveiling the underlying cellular receptors and cofactors that lead to HIV preference for specific immune cell populations is crucial in identifying novel preventative/therapeutic targets for comprehensive strategies to eliminate viral infection.


Asunto(s)
Antígenos CD4/metabolismo , Infecciones por VIH/virología , VIH-1/metabolismo , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , VIH-1/fisiología , Humanos , Receptores del VIH/metabolismo
11.
J Gen Virol ; 97(2): 422-434, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26555393

RESUMEN

Herpes simplex virus (HSV) was originally implicated in the aetiology of cervical cancer, and although high-risk human papillomavirus (HPV) is now the accepted causative agent, the epidemiological link between HSV and HPV-associated cancers persists. The annexin A2 heterotetramer (A2t) has been shown to mediate infectious HPV type 16 (HPV16) uptake by human keratinocytes, and secretory leukocyte protease inhibitor (SLPI), an endogenous A2t ligand, inhibits HPV16 uptake and infection. Interestingly, HSV infection induces a sustained downregulation of SLPI in epithelial cells, which we hypothesized promotes HPV16 infection through A2t. Here, we show that in vitro infection of human keratinocytes with HSV-1 or HSV-2, but not with an HSV-1 ICP4 deletion mutant that does not downregulate SLPI, leads to a >70% reduction of SLPI mRNA and a >60% decrease in secreted SLPI protein. Consequently, we observed a significant increase in the uptake of HPV16 virus-like particles and gene transduction by HPV16 pseudovirions (two- and 2.5-fold, respectively) in HSV-1- and HSV-2-infected human keratinocyte cell cultures compared with uninfected cells, whereas exogenously added SLPI reversed this effect. Using a SiMPull (single-molecule pulldown) assay, we demonstrated that endogenously secreted SLPI interacts with A2t on epithelial cells in an autocrine/paracrine manner. These results suggested that ongoing HSV infection and resultant downregulation of local levels of SLPI may impart a greater susceptibility for keratinocytes to HPV16 infection through the host cell receptor A2t, providing a mechanism that may, in part, provide an explanation for the aetiological link between HSV and HPV-associated cancers.


Asunto(s)
Interacciones Huésped-Patógeno , Papillomavirus Humano 16/fisiología , Queratinocitos/virología , Inhibidor Secretorio de Peptidasas Leucocitarias/metabolismo , Simplexvirus/fisiología , Internalización del Virus , Línea Celular , Regulación hacia Abajo , Humanos
12.
Papillomavirus Res ; 1: 12-21, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26665182

RESUMEN

Human papillomaviruses (HPV) establish persistent infections because of evolved immune evasion mechanisms, particularly HPV-mediated suppression of the immune functions of Langerhans cells (LC), the antigen presenting cells of the epithelium. Polyinosinic-polycytidilic acid (Poly-I:C) is broadly immunostimulatory with the ability to enhance APC expression of costimulatory molecules and inflammatory cytokines resulting in T cell activation. Here we investigated the activation of primary human LC derived from peripheral blood monocytes after exposure to HPV16 virus like particles followed by treatment with stabilized Poly-I:C compounds (s-Poly-I:C), and their subsequent induction of HPV16-specific T cells. Our results indicate that HPV16 particles alone were incapable of inducing LC activation as demonstrated by the lack of costimulatory molecules, inflammatory cytokines, chemokine-directed migration, and HPV16-specific CD8+ T cells in vitro. Conversely, s-Poly-I:C caused significant upregulation of costimulatory molecules and induction of chemokine-directed migration of LC that were pre-exposed to HPV16. In HLA-A*0201-positive donors, s-Poly-I:C treatment was able to induce CD8+ T cell immune responses against HPV16-derived peptides. Thus, s-Poly-I:C compounds are attractive for translation into therapeutics in which they could potentially mediate clearance of persistent HPV infection.

13.
Clin Immunol ; 161(2): 197-208, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26360252

RESUMEN

Human papillomavirus (HPV)-mediated suppression of Langerhans cell (LC) function can lead to persistent infection and development of cervical intraepithelial neoplasia (CIN). Women with HPV-induced high-grade CIN2/3 have not mounted an effective immune response against HPV, yet it is unknown if LC-mediated T cell activation from such women is functionally impaired against HPV. We investigated the functional activation of in vitro generated LC and their ability to induce HPV16-specific T cells from CIN2/3 patients after exposure to HPV16 followed by treatment with stabilized Poly-I:C (s-Poly-I:C). LC from patients exposed to HPV16 demonstrated a lack of costimulatory molecule expression, inflammatory cytokine secretion, and chemokine-directed migration. Conversely, s-Poly-I:C caused significant phenotypic and functional activation of HPV16-exposed LC, which resulted in de novo generation of HPV16-specific CD8(+) T cells. Our results highlight that LC of women with a history of persistent HPV infection can present HPV antigens and are capable of inducing an adaptive T cell immune response when given the proper stimulus, suggesting that s-Poly-I:C compounds may be attractive immunomodulators for LC-mediated clearance of persistent HPV infection.


Asunto(s)
Papillomavirus Humano 16/inmunología , Células de Langerhans/inmunología , Activación de Linfocitos/inmunología , Infecciones por Papillomavirus/inmunología , Poli I-C/inmunología , Displasia del Cuello del Útero/inmunología , Displasia del Cuello del Útero/virología , Adulto , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , ADN Viral/inmunología , Femenino , Humanos , Células de Langerhans/virología , Persona de Mediana Edad , Infecciones por Papillomavirus/virología , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/virología
14.
J Antimicrob Chemother ; 70(6): 1686-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25712315

RESUMEN

OBJECTIVES: High-risk human papillomavirus (HPV) infection leads to the development of several human cancers that cause significant morbidity and mortality worldwide. HPV type 16 (HPV16) is the most common of the cancer-causing genotypes and gains entry to the basal cells of the epithelium through a non-canonical endocytic pathway that involves the annexin A2/S100A10 heterotetramer (A2t). A2t is composed of two annexin A2 monomers bound to an S100A10 dimer and this interaction is a potential target to block HPV16 infection. Here, recently identified small molecule inhibitors of A2t (A2ti) were investigated for their ability to prevent HPV16 infection in vitro. METHODS: A2ti were added to HeLa cells in increasing concentrations prior to the addition of HPV16. Cytotoxicity was evaluated via trypan blue exclusion. HPV16 pseudovirion infection and fluorescently labelled HPV16 capsid internalization was measured with flow cytometry. RESULTS: A2ti blocked HPV16 infection by 100% without substantial cellular toxicity or reduction in cell growth. Furthermore, A2ti blocked HPV16 entry into epithelial cells by 65%, indicating that the observed inhibition of HPV16 infection is in part due to a block in entry and that non-infectious entry may occur in the absence of A2t binding. CONCLUSIONS: These results demonstrate that targeting A2t may be an effective strategy to prevent HPV16 infection.


Asunto(s)
Anexina A2/antagonistas & inhibidores , Antivirales/farmacología , Endocitosis/efectos de los fármacos , Papillomavirus Humano 16/fisiología , Internalización del Virus/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HeLa , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...