Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Immunity ; 41(4): 592-604, 2014 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-25308333

RESUMEN

Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is a pattern-recognition receptor for a variety of endogenous and exogenous ligands. However, LOX-1 function in the host immune response is not fully understood. Here, we report that LOX-1 expressed on dendritic cells (DCs) and B cells promotes humoral responses. On B cells LOX-1 signaling upregulated CCR7, promoting cellular migration toward lymphoid tissues. LOX-1 signaling on DCs licensed the cells to promote B cell differentiation into class-switched plasmablasts and led to downregulation of chemokine receptor CXCR5 and upregulation of chemokine receptor CCR10 on plasmablasts, enabling their exit from germinal centers and migration toward local mucosa and skin. Finally, we found that targeting influenza hemagglutinin 1 (HA1) subunit to LOX-1 elicited HA1-specific protective antibody responses in rhesus macaques. Thus, LOX-1 expressed on B cells and DC cells has complementary functions to promote humoral immune responses.


Asunto(s)
Linfocitos B/inmunología , Células Dendríticas/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Cambio de Clase de Inmunoglobulina/inmunología , Receptores Depuradores de Clase E/inmunología , Animales , Formación de Anticuerpos/inmunología , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Centro Germinal/citología , Humanos , Inmunoglobulina A/biosíntesis , Inmunoglobulina G/biosíntesis , Inmunoglobulina M/biosíntesis , Activación de Linfocitos/inmunología , Macaca mulatta , Masculino , Membrana Mucosa/citología , Receptores CCR10/biosíntesis , Receptores CCR7/biosíntesis , Receptores CXCR5/biosíntesis , Receptores Depuradores de Clase E/biosíntesis , Transducción de Señal/inmunología , Piel/citología
2.
Clin Immunol ; 232: 108874, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34740841

RESUMEN

Female sex hormones affect the immune response in the lower female genital tract. To understand their mechanisms of action, it is essential to define cell types expressing estrogen receptor (ER) and/or progesterone receptor (PR) in the human vaginal mucosa (VM). Here, we report that none of the dendritic cell (DC) subsets in the human VM expressed ERα or PR in situ. However, they were capable of expressing ERα, but not PR, after in vitro culture of the whole VM tissues. Similarly, ERα and/or PR expression by T cells in the VM tissues was also inducible rather than constitutive. In contrast, ERα and/or PR were constitutively expressed in HLA-DR- non-immune cell types (vimentin+, desmin+, or CD10+). These new findings will help us understand the mechanisms of action of female sex hormones in the modulation of immune response in the human VM and lower female genital tract.


Asunto(s)
Membrana Mucosa/metabolismo , Receptores de Estrógenos/biosíntesis , Receptores de Progesterona/biosíntesis , Vagina/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Células Dendríticas/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Linfocitos T/metabolismo
3.
Nat Immunol ; 9(5): 551-7, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18376401

RESUMEN

Although plasmacytoid dendritic cells (pDCs) respond to virus replication in a nonspecific way by producing large amounts of type I interferon, a rapid, direct function for pDCs in activating antiviral lymphocytes is less apparent. Here we show that pDCs were able to rapidly initiate antigen-specific antiviral CD8+ T cell responses. After being exposed to virus, pDCs efficiently and rapidly internalized exogenous viral antigens and then presented those antigens on major histocompatibility complex (MHC) class I to CD8+ T cells. Processing of exogenous antigen occurred in endocytic organelles and did not require transit of antigen to the cytosol. Intracellular stores of MHC class I partially localized together with the transferrin receptor and internalized transferrin in endosomes, which suggested that such recycling endosomes are sites for loading peptide onto MHC class I or for peptide transit. Our data demonstrate that pDCs use 'ready-made' stores of MHC class I to rapidly present exogenous antigen to CD8+ T cells.


Asunto(s)
Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Presentación de Antígeno , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Reactividad Cruzada , Endosomas/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Virus de la Influenza A/inmunología , Leucocitos Mononucleares , Activación de Linfocitos , Orgánulos/inmunología , Complejo de la Endopetidasa Proteasomal , Receptores de Transferrina/metabolismo
4.
Immunity ; 30(1): 120-9, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-19144318

RESUMEN

Mucosal immunoglobulin A (IgA) secreted by local plasma cells (PCs) is a critical component of mucosal immunity. Although IgA class switching can occur at mucosal sites, high-affinity PCs are optimally generated in germinal centers (GCs) in a T cell-dependent fashion. However, how CD4(+) helper T cells induce mucosal-homing IgA-PCs remains unclear. Here, we show that transforming growth factor beta1 (TGFbeta1) and interleukin 21 (IL-21), produced by follicular helper T cells (Tfh), synergized to generate abundant IgA-plasmablasts (PBs). In the presence of IL-21, TGFbeta1 promoted naive B cell proliferation and differentiation and overrode IL-21-induced IgG class switching in favor of IgA. Furthermore, TGFbeta1 and IL-21 downregulated CXCR5 while upregulating CCR10 on plasmablasts, enabling their exit from GCs and migration toward local mucosa. This was supported by the presence of CCR10(+)IgA(+)PBs in tonsil GCs. These findings show that Tfh contribute to mucosal IgA. Thus, mucosal vaccines should aim to induce robust Tfh responses.


Asunto(s)
Quimiotaxis de Leucocito , Inmunoglobulina A/metabolismo , Membrana Mucosa/inmunología , Células Plasmáticas/inmunología , Linfocitos T/inmunología , Linfocitos B/inmunología , Diferenciación Celular , Proliferación Celular , Niño , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoglobulina A/clasificación , Interleucina-2/genética , Interleucina-2/metabolismo , Interleucinas/fisiología , Reacción en Cadena de la Polimerasa , Receptores CCR10/metabolismo , Receptores CXCR5/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estándares de Referencia , Factor de Crecimiento Transformador beta1/fisiología , Regulación hacia Arriba
5.
J Immunol ; 195(4): 1723-31, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26123355

RESUMEN

Dendritic cells (DCs) can induce and control host immune responses. DC subset-dependent functional specialties and their ability to display functional plasticity, which is mainly driven by signals via pattern recognition receptors, identify DCs as immune orchestrators. A pattern recognition receptor, Dectin-1, is expressed on myeloid DCs and known to play important roles in Th17 induction and activation during fungal and certain bacterial infections. In this study, we first demonstrate that human plasmacytoid DCs express Dectin-1 in both mRNA and protein levels. More interestingly, Dectin-1-activated plasmacytoid DCs promote Th2-type T cell responses, whereas Dectin-1-activated myeloid DCs decrease Th2-type T cell responses. Such contrasting outcomes of Th2-type T cell responses by the two DC subsets are mainly due to their distinct abilities to control surface OX40L expression in response to ß-glucan. This study provides new insights for the regulation of host immune responses by Dectin-1 expressed on DCs.


Asunto(s)
Células Dendríticas/metabolismo , Expresión Génica , Lectinas Tipo C/genética , Células Mieloides/metabolismo , Células Th2/metabolismo , Citocinas/biosíntesis , Células Dendríticas/inmunología , Humanos , Memoria Inmunológica , Virus de la Influenza A/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lectinas Tipo C/química , Lectinas Tipo C/metabolismo , Datos de Secuencia Molecular , Células Mieloides/inmunología , Ligando OX40/metabolismo , Tonsila Palatina/citología , Tonsila Palatina/inmunología , Tonsila Palatina/metabolismo , Unión Proteica , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Quinasa Syk , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células Th2/inmunología , beta-Glucanos/metabolismo
6.
J Immunol ; 192(12): 5776-88, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24835401

RESUMEN

Recent compelling evidence indicates that Th17 confer host immunity against a variety of microbes, including extracellular and intracellular pathogens. Therefore, understanding mechanisms for the induction and activation of Ag-specific Th17 is important for the rational design of vaccines against pathogens. To study this, we employed an in vitro system in which influenza hemagglutinin (HA) 1 was delivered to dendritic cells (DCs) via Dectin-1 using anti-human Dectin-1 (hDectin-1)-HA1 recombinant fusion proteins. We found that healthy individuals maintained broad ranges of HA1-specific memory Th17 that were efficiently activated by DCs targeted with anti-hDectin-1-HA1. Nonetheless, these DCs were not able to induce a significant level of HA1-specific Th17 responses even in the presence of the Th17-promoting cytokines IL-1ß and IL-6. We further found that the induction of surface IL-1R1 expression by signals via TCRs and common γ-chain receptors was essential for naive CD4(+) T cell differentiation into HA1-specific Th17. This process was dependent on MyD88, but not IL-1R-associated kinase 1/4. Thus, interruptions in STAT3 or MyD88 signaling led to substantially diminished HA1-specific Th17 induction. Taken together, the de novo generation of pathogen-specific human Th17 requires complex, but complementary, actions of multiple signals. Data from this study will help us design a new and effective vaccine strategy that can promote Th17-mediated immunity against microbial pathogens.


Asunto(s)
Antígenos Virales/inmunología , Células Dendríticas/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Lectinas Tipo C/inmunología , Células Th17/inmunología , Células Dendríticas/citología , Femenino , Humanos , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Masculino , Factor 88 de Diferenciación Mieloide/inmunología , Células Th17/citología
7.
bioRxiv ; 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-39005456

RESUMEN

The interaction between antigens and antibodies (B cell receptors, BCRs) is the key step underlying the function of the humoral immune system in various biological contexts. The capability to profile the landscape of antigen-binding affinity of a vast number of BCRs will provide a powerful tool to reveal novel insights at unprecedented levels and will yield powerful tools for translational development. However, current experimental approaches for profiling antibody-antigen interactions are costly and time-consuming, and can only achieve low-to-mid throughput. On the other hand, bioinformatics tools in the field of antibody informatics mostly focus on optimization of antibodies given known binding antigens, which is a very different research question and of limited scope. In this work, we developed an innovative Artificial Intelligence tool, Cmai, to address the prediction of the binding between antibodies and antigens that can be scaled to high-throughput sequencing data. Cmai achieved an AUROC of 0.91 in our validation cohort. We devised a biomarker metric based on the output from Cmai applied to high-throughput BCR sequencing data. We found that, during immune-related adverse events (irAEs) caused by immune-checkpoint inhibitor (ICI) treatment, the humoral immunity is preferentially responsive to intracellular antigens from the organs affected by the irAEs. In contrast, extracellular antigens on malignant tumor cells are inducing B cell infiltrations, and the infiltrating B cells have a greater tendency to co-localize with tumor cells expressing these antigens. We further found that the abundance of tumor antigen-targeting antibodies is predictive of ICI treatment response. Overall, Cmai and our biomarker approach filled in a gap that is not addressed by current antibody optimization works nor works such as AlphaFold3 that predict the structures of complexes of proteins that are known to bind.

8.
bioRxiv ; 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38105939

RESUMEN

Profiling the binding of T cell receptors (TCRs) of T cells to antigenic peptides presented by MHC proteins is one of the most important unsolved problems in modern immunology. Experimental methods to probe TCR-antigen interactions are slow, labor-intensive, costly, and yield moderate throughput. To address this problem, we developed pMTnet-omni, an Artificial Intelligence (AI) system based on hybrid protein sequence and structure information, to predict the pairing of TCRs of αß T cells with peptide-MHC complexes (pMHCs). pMTnet-omni is capable of handling peptides presented by both class I and II pMHCs, and capable of handling both human and mouse TCR-pMHC pairs, through information sharing enabled this hybrid design. pMTnet-omni achieves a high overall Area Under the Curve of Receiver Operator Characteristics (AUROC) of 0.888, which surpasses competing tools by a large margin. We showed that pMTnet-omni can distinguish binding affinity of TCRs with similar sequences. Across a range of datasets from various biological contexts, pMTnet-omni characterized the longitudinal evolution and spatial heterogeneity of TCR-pMHC interactions and their functional impact. We successfully developed a biomarker based on pMTnet-omni for predicting immune-related adverse events of immune checkpoint inhibitor (ICI) treatment in a cohort of 57 ICI-treated patients. pMTnet-omni represents a major advance towards developing a clinically usable AI system for TCR-pMHC pairing prediction that can aid the design and implementation of TCR-based immunotherapeutics.

9.
Immunohorizons ; 3(3): 110-120, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-31240276

RESUMEN

Graft-versus-host disease (GVHD) is one of the major obstacles for the success of allogeneic hematopoietic stem cell transplantation. Here, we report that the interaction between OX40L and OX40 is of critical importance for both induction and progression of acute GVHD (aGVHD) driven by human T cells. Anti-human OX40L monoclonal antibody (hOX40L) treatment could thus effectively reduce the disease severity in a xenogeneic-aGVHD (x-aGVHD) model in both preventative and therapeutic modes. Mechanistically, blocking OX40L-OX40 interaction with an anti-hOX40L antibody reduces infiltration of human T cells in target organs, including liver, gut, lung, and skin. It also decreases IL-21- and TNF-producing T cell responses, while promoting regulatory T cell (Treg) responses without compromising the cytolytic activity of CD8+ T cells. Single blockade of hOX40L was thus more effective than dual blockade of IL-21 and TNF in reducing the severity of aGVHD as well as mortality. Data from this study indicate that OX40L-OX40 interactions play a central role in the pathogenesis of aGVHD induced by human T cells. Therapeutic strategies that can efficiently interrupt OX40L-OX40 interaction in patients might have potential to provide patients with an improved clinical benefit.


Asunto(s)
Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/metabolismo , Ligando OX40/metabolismo , Receptores OX40/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Enfermedad Aguda , Animales , Anticuerpos Monoclonales/farmacología , Citocinas/metabolismo , Citotoxicidad Inmunológica/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Progresión de la Enfermedad , Etanercept/farmacología , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/patología , Humanos , Interleucinas/antagonistas & inhibidores , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Leucocitos/metabolismo , Ratones , Ligando OX40/antagonistas & inhibidores , Unión Proteica , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-29507584

RESUMEN

BACKGROUND: T cells play a central role in chronic inflammation in asthma. However, the roles of individual subsets of T cells in the pathology of asthma in patients remain to be better understood. METHODS: We investigated the potential signatures of T cell subset phenotypes in asthma using fresh whole blood from adult atopic asthma patients (n = 43) and non-asthmatic control subjects (n = 22). We further assessed their potential clinical implications by correlating asthma severity. RESULTS: We report four major features of CD4+ T cells in the blood of atopic asthma patients. First, patients had a profound increase of CCR7+ memory CD4+ T cells, but not CCR7- memory CD4+ T cells. Second, an increase in CCR4+ CD4+ T cells in patients was mainly attributed to the increase of CCR7+ memory CD4+ T cells. Accordingly, the frequency of CCR4+CCR7+ memory CD4+ T cells correlated with asthma severity. Current common asthma therapeutics (including corticosteroids) were not able to affect the frequency of CCR4+CCR7+ memory CD4+ T cell subsets. Third, patients had an increase of Tregs, as assessed by measuring CD25, Foxp3, IL-10 and CTLA-4 expression. However, asthma severity was inversely correlated only with the frequency of CTLA-4+ CD4+ T cells. Lastly, patients and control subjects have similar frequencies of CD4+ T cells that express CCR5, CCR6, CXCR3, CXCR5, CD11a, or α4 integrin. However, the frequency of α4+ CD4+ T cells in patients correlated with asthma severity. CONCLUSIONS: CCR4+CCR7+ memory, but not CCR4+CCR7- memory, α4+, and CTLA4+ CD4+ T cells in patients show significant clinical implications in atopic asthma. Current common therapeutics cannot alter the frequency of such CD4+ T cell subsets in adult atopic asthma patients.

11.
EBioMedicine ; 5: 46-58, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27077111

RESUMEN

Dendritic cells (DCs) are major antigen-presenting cells that can efficiently prime and cross-prime antigen-specific T cells. Delivering antigen to DCs via surface receptors is thus an appealing strategy to evoke cellular immunity. Nonetheless, which DC surface receptor to target to yield the optimal CD8(+) and CD4(+) T cell responses remains elusive. Herein, we report the superiority of CD40 over 9 different lectins and scavenger receptors at evoking antigen-specific CD8(+) T cell responses. However, lectins (e.g., LOX-1 and Dectin-1) were more efficient than CD40 at eliciting CD4(+) T cell responses. Common and distinct patterns of subcellular and intracellular localization of receptor-bound αCD40, αLOX-1 and αDectin-1 further support their functional specialization at enhancing antigen presentation to either CD8(+) or CD4(+) T cells. Lastly, we demonstrate that antigen targeting to CD40 can evoke potent antigen-specific CD8(+) T cell responses in human CD40 transgenic mice. This study provides fundamental information for the rational design of vaccines against cancers and viral infections.


Asunto(s)
Presentación de Antígeno/inmunología , Ligando de CD40/inmunología , Células Dendríticas/inmunología , Inmunoterapia Activa , Activación de Linfocitos/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Humanos , Lectinas/inmunología , Lectinas Tipo C/inmunología , Ratones , Ratones Transgénicos , Proteínas Recombinantes de Fusión/inmunología , Receptores Depuradores de Clase E/inmunología
12.
Cancer Immunol Res ; 4(10): 823-834, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27485136

RESUMEN

Human papillomavirus (HPV), particularly HPV16 and HPV18, can cause cancers in diverse anatomical sites, including the anogenital and oropharyngeal (throat) regions. Therefore, development of safe and clinically effective therapeutic vaccines is an important goal. Herein, we show that a recombinant fusion protein of a humanized antibody to CD40 fused to HPV16.E6/7 (αCD40-HPV16.E6/7) can evoke HPV16.E6/7-specific CD8+ and CD4+ T-cell responses in head-and-neck cancer patients in vitro and in human CD40 transgenic (hCD40Tg) mice in vivo The combination of αCD40-HPV16.E6/7 and poly(I:C) efficiently primed HPV16.E6/7-specific T cells, particularly CD8+ T cells, in hCD40Tg mice. Inclusion of montanide enhanced HPV16.E6/7-specific CD4+, but not CD8+, T-cell responses. Poly(I:C) plus αCD40-HPV16.E6/7 was sufficient to mount both preventative and therapeutic immunity against TC-1 tumors in hCD40Tg mice, significantly increasing the frequency of HPV16-specific CD8+ CTLs in the tumors, but not in peripheral blood. In line with this, tumor volume inversely correlated with the frequency of HPV16.E6/7-specific CD8+ T cells in tumors, but not in blood. These data suggest that CD40-targeting vaccines for HPV-associated malignancies can provide a highly immunogenic platform with a strong likelihood of clinical benefit. Data from this study offer strong support for the development of CD40-targeting vaccines for other cancers in the future. Cancer Immunol Res; 4(10); 823-34. ©2016 AACR.


Asunto(s)
Antígenos CD40/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Neoplasias de Cabeza y Cuello/inmunología , Vacunas contra Papillomavirus/inmunología , Animales , Antivirales/inmunología , Linfocitos T CD4-Positivos/inmunología , Femenino , Papillomavirus Humano 16/inmunología , Humanos , Inmunidad Celular , Ratones Endogámicos C57BL , Ratones Transgénicos , Poli I-C/inmunología , Proteínas Recombinantes de Fusión/inmunología , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/prevención & control , Neoplasias del Cuello Uterino/virología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Immunol Lett ; 168(1): 89-97, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26381186

RESUMEN

Toll-like receptor 7 (TLR7) agonists are of interest as vaccine adjuvants and cancer therapeutics. Therefore, development of new TLR7 agonists that can efficiently promote host immune responses without evoking side effects is of great importance. Here, we describe two new compounds, J4 and F4, which elicit intracellular signaling exclusively via TLR7. Interestingly, both J4 and F4 induced less cytokine secretion (IL-1ß, IL-6, IL-10, IL-12p40, TNFα, and IL-12p70) from myeloid dendritic cells (mDCs) and monocytes than CL075 and R848; however, they all generated similar levels of phenotype maturation of antigen presenting cells (APCs), including plasmacytoid DCs. We further found that J4- and F4-induced APC activation was largely dependent on the activation of NF-κB and p38. Lastly, J4 and F4 could efficiently promote B cell proliferation and plasmablast differentiation as well as antigen-specific CD8(+) T cell responses in human in vitro. Therefore, these new TLR7 agonists could be employed to facilitate the development of new therapeutics and vaccine adjuvants against cancers and microbial infections.


Asunto(s)
Adenosina/análogos & derivados , Adyuvantes Inmunológicos/farmacología , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Receptor Toll-Like 7/agonistas , Adenosina/química , Adenosina/farmacología , Adyuvantes Inmunológicos/química , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Células HEK293 , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Estructura Molecular , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
14.
In Vivo ; 16(6): 451-7, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12494889

RESUMEN

A6 is an eight amino acid peptide derived from the non-receptor binding region of urokinase plasminogen activator (uPA), which interferes with the uPA/uPA receptor system. A6 has been synthesized as a potential anti-angiogenic, anti-cancer agent. The current study has investigated the potential therapeutic activity of A6 in the Lewis lung carcinoma (3LL) model of pulmonary metastasis. A6 was found to have direct anti-tumor activity against established 3LL pulmonary metastases at a low tumor burden (10-20 colonies per lung) and was therapeutic in combination with cyclophosphamide at high tumor burdens (> 100 colonies per lung). Mechanistic studies have revealed that A6 directly inhibits the invasion of 3LL cells through a Matrigel model basement membrane by 40-45%. Moreover, treatment with either A6 or doxorubicin resulted in thicker tubes in endothelial tube formation studies. Our results suggest that A6, by virtue of its anti-invasive and anti-angiogenic properties, might work additively or synergistically with chemotherapeutic agents and thereby contribute to enhanced therapy of established 3LL cancer metastases.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Activador de Plasminógeno de Tipo Uroquinasa/uso terapéutico , Animales , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/secundario , Ciclofosfamida/uso terapéutico , Doxorrubicina/uso terapéutico , Sinergismo Farmacológico , Quimioterapia Combinada , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Técnicas In Vitro , Ratones , Invasividad Neoplásica/prevención & control , Trasplante de Neoplasias , Neovascularización Patológica/prevención & control , ARN Mensajero/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
15.
Clin Vaccine Immunol ; 21(12): 1668-80, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25298110

RESUMEN

Despite the availability of annually formulated vaccines, influenza virus infection remains a worldwide public health burden. Therefore, it is important to develop preclinical challenge models that enable the evaluation of vaccine candidates while elucidating mechanisms of protection. Here, we report that naive rhesus macaques challenged with 2009 pandemic H1N1 (pH1N1) influenza virus do not develop observable clinical symptoms of disease but develop a subclinical biphasic fever on days 1 and 5 to 6 postchallenge. Whole blood microarray analysis further revealed that interferon activity was associated with fever. We then tested whether type I interferon activity in the blood is a correlate of vaccine efficacy. The animals immunized with candidate vaccines carrying hemagglutinin (HA) or nucleoprotein (NP) exhibited significantly reduced interferon activity on days 5 to 6 postchallenge. Supported by cellular and serological data, we conclude that blood interferon activity is a prominent marker that provides a convenient metric of influenza virus vaccine efficacy in the subclinical rhesus macaque model.


Asunto(s)
Anticuerpos Antivirales/sangre , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Animales , Humanos , Inmunización , Macaca mulatta , Vacunación
16.
Nat Commun ; 5: 5283, 2014 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-25335753

RESUMEN

The mechanisms by which microbial vaccines interact with human APCs remain elusive. Herein, we describe the transcriptional programs induced in human DCs by pathogens, innate receptor ligands and vaccines. Exposure of DCs to influenza, Salmonella enterica and Staphylococcus aureus allows us to build a modular framework containing 204 transcript clusters. We use this framework to characterize the responses of human monocytes, monocyte-derived DCs and blood DC subsets to 13 vaccines. Different vaccines induce distinct transcriptional programs based on pathogen type, adjuvant formulation and APC targeted. Fluzone, Pneumovax and Gardasil, respectively, activate monocyte-derived DCs, monocytes and CD1c+ blood DCs, highlighting APC specialization in response to vaccines. Finally, the blood signatures from individuals vaccinated with Fluzone or infected with influenza reveal a signature of adaptive immunity activation following vaccination and symptomatic infections, but not asymptomatic infections. These data, offered with a web interface, may guide the development of improved vaccines.


Asunto(s)
Células Dendríticas/citología , Células Dendríticas/microbiología , Transcripción Genética , Vacunas/química , Algoritmos , Animales , Antígenos CD1/metabolismo , Antígenos de Superficie/metabolismo , Análisis por Conglomerados , Citocinas/metabolismo , Células Dendríticas/metabolismo , Perros , Perfilación de la Expresión Génica , Glicoproteínas/metabolismo , Humanos , Subtipo H1N1 del Virus de la Influenza A , Interleucina-4/metabolismo , Células de Riñón Canino Madin Darby , Monocitos/citología , Monocitos/metabolismo , Análisis de Componente Principal , Salmonella enterica , Staphylococcus aureus , Trombomodulina , Transcriptoma
17.
AIDS ; 27(13): 2041-51, 2013 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-23615121

RESUMEN

OBJECTIVE: Targeting HIV antigens directly to dendritic cells using monoclonal antibodies against cell-surface receptors has been shown to evoke potent cellular immunity in animal models. The objective of this study was to configure an anti-human CD40 antibody fused to a string of five highly conserved CD4 and CD8 T-cell epitope-rich regions of HIV-1 Gag, Nef and Pol (αCD40.HIV5pep), and then to demonstrate the capacity of this candidate therapeutic vaccine to target these HIV peptide antigens to human dendritic cells to expand functional HIV-specific T cells. METHODS: Antigen-specific cytokine production using intracellular flow cytometry and multiplex bead-based assay, and suppression of viral inhibition, were used to characterize the T cells expanded by αCD40.HIV5pep from HIV-infected patient peripheral blood mononuclear cell (PBMC) and dendritic cell/T-cell co-cultures. RESULTS: This candidate vaccine expands memory CD4 and CD8 T cells specific to multiple epitopes within all five peptide regions across a wide range of major histocompatibility complex (MHC) haplotypes from HIV-infected patient PBMC and dendritic cell/T-cell co-cultures. These in vitro expanded HIV antigen-specific CD4 and CD8 T cells produce multiple cytokines and chemokines. αCD40.HIV5pep-expanded CD8 T cells have characteristics of cytotoxic effector cells and are able to kill autologous target cells and suppress HIV-1 replication in vitro. CONCLUSION: Our data demonstrate the therapeutic potential of this CD40-targeting HIV candidate vaccine in inducing a broad repertoire of multifunctional T cells in patients.


Asunto(s)
Vacunas contra el SIDA/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Antígenos VIH/inmunología , VIH-1/inmunología , Vacunas contra el SIDA/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Antígenos CD40/metabolismo , Proliferación Celular , Técnicas de Cocultivo , Antígenos VIH/genética , Antígenos VIH/metabolismo , VIH-1/genética , Humanos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo
18.
J Exp Med ; 209(7): 1335-48, 2012 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-22689824

RESUMEN

The development of autoantibodies is a hallmark of systemic lupus erythematosus (SLE). SLE serum can induce monocyte differentiation into dendritic cells (DCs) in a type I IFN-dependent manner. Such SLE-DCs activate T cells, but whether they promote B cell responses is not known. In this study, we demonstrate that SLE-DCs can efficiently stimulate naive and memory B cells to differentiate into IgG- and IgA-plasmablasts (PBs) resembling those found in the blood of SLE patients. SLE-DC-mediated IgG-PB differentiation is dependent on B cell-activating factor (BAFF) and IL-10, whereas IgA-PB differentiation is dependent on a proliferation-inducing ligand (APRIL). Importantly, SLE-DCs express CD138 and trans-present CD138-bound APRIL to B cells, leading to the induction of IgA switching and PB differentiation in an IFN-α-independent manner. We further found that this mechanism of providing B cell help is relevant in vivo, as CD138-bound APRIL is expressed on blood monocytes from active SLE patients. Collectively, our study suggests that a direct myeloid DC-B cell interplay might contribute to the pathogenesis of SLE.


Asunto(s)
Diferenciación Celular/inmunología , Lupus Eritematoso Sistémico/inmunología , Monocitos/inmunología , Células Plasmáticas/inmunología , Adolescente , Autoanticuerpos/inmunología , Autoanticuerpos/farmacología , Factor Activador de Células B/inmunología , Factor Activador de Células B/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Niño , Preescolar , Técnicas de Cocultivo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Citometría de Flujo , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Lactante , Interleucina-10/inmunología , Interleucina-10/metabolismo , Lupus Eritematoso Sistémico/sangre , Masculino , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/metabolismo , Suero/inmunología , Sindecano-1/inmunología , Sindecano-1/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Adulto Joven
19.
J Cell Biochem ; 88(3): 482-92, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12532325

RESUMEN

The proteasome is a multi-subunit protease complex that is involved in intracellular protein degradation in eukaryotes. Previously, we have reported that selective, synthetic chymotryptic proteasome inhibitors inhibit A-NK cell-mediated cytotoxicity by approximately 50%; however, the exact role of the proteasome in NK cell-mediated cytotoxicity remains unknown. Herein, we report that proteasome inhibitors, MG115 and MG132, decreased the proteasome chymotrypsin-like activity in the rat natural killer cell line RNK16 by 85% at a concentration of 5 microM. The viability of RNK16 cells was also reduced in the presence of these inhibitors. Both inhibitors induced the apoptosis of RNK16 cells, as shown by DNA fragmentation, caspase-3 activation and the appearance of sub-G-cell populations. An increase in the fraction of apoptotic cells was observed in a dose- and time-dependent manner in our studies. In addition, the activity of caspase-1, -2, -6, -7, -8, and -9, was increased following the treatment of RNK16 cells with these inhibitors. Further investigation revealed that the expression of Fas (CD95) protein on the RNK16 cell surface was increased after the treatment by MG115 or MG132, indicating that apoptosis induced by proteasome inhibitors in RNK16 cells might be mediated through the Fas (CD95)-mediated death pathway as well. Our studies indicate, for the first time, that proteasomal chymotryptic inhibitors can reduce natural killer cell viability and therefore indirectly inhibit cell-mediated cytotoxicity via the apoptosis-inducing properties of these agents.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Células Asesinas Naturales/metabolismo , Complejos Multienzimáticos/antagonistas & inhibidores , Receptor fas/metabolismo , Animales , Línea Celular , Cisteína Endopeptidasas/metabolismo , Activación Enzimática , Células Asesinas Naturales/citología , Leupeptinas/metabolismo , Complejos Multienzimáticos/metabolismo , Inhibidores de Proteasas/metabolismo , Complejo de la Endopetidasa Proteasomal , Ratas
20.
Biochem Biophys Res Commun ; 292(1): 184-9, 2002 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-11890690

RESUMEN

Urokinase plasminogen activator (uPA) and its receptor uPAR play a major role in immune cell-mediated, including natural killer (NK) cell-mediated, degradation of extracellular matrices. Herein, we investigate the effects of IL-2 on NK cell uPA and uPAR. RNA and protein analyses showed upregulation of uPA and uPAR following IL-2 stimulation. Gel-shift assays and Western blots detected uPA and uPAR mRNA binding proteins (mRNABPs), previously shown to destabilize uPA and uPAR mRNA. Following IL-2 stimulation, a downregulation of uPAR mRNABP and a reciprocal induction of uPAR mRNA were noted. The increase in uPA following IL-2 stimulation appeared to be more transcriptionally regulated. These data suggest that IL-2 upregulates both uPA and uPAR in NK cells through posttranscriptional as well as transcriptional mechanisms, partially explaining increases in NK cell invasiveness following IL-2 stimulation.


Asunto(s)
Interleucina-2/farmacología , Células Asesinas Naturales/enzimología , Receptores de Superficie Celular/genética , Regulación hacia Arriba , Activador de Plasminógeno de Tipo Uroquinasa/genética , Humanos , Células Asesinas Naturales/efectos de los fármacos , Cinética , Estabilidad del ARN , ARN Mensajero/biosíntesis , Proteínas de Unión al ARN/análisis , Receptores de Superficie Celular/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Activación Transcripcional , Células Tumorales Cultivadas , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA