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










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38370737

RESUMEN

Protein S (PS), the critical plasma cofactor for the anticoagulants tissue factor (TF) pathway inhibitor (TFPI) and activated protein C (APC), circulates in two functionally distinct pools: free (anticoagulant) or bound to complement component 4b-binding protein (C4BP) (anti-inflammatory). Acquired free PS deficiency is detected in several viral infections, but its cause is unclear. Here, we identified a shear-dependent interaction between PS and von Willebrand Factor (VWF) by mass spectrometry. Consistently, plasma PS and VWF comigrated in both native and agarose gel electrophoresis. The PS/VWF interaction was blocked by TFPI but not APC, suggesting an interaction with the C-terminal sex hormone binding globulin (SHBG) region of PS. Microfluidic systems, mimicking arterial laminar flow or disrupted turbulent flow, demonstrated that PS stably binds VWF as VWF unfolds under turbulent flow. PS/VWF complexes also localized to platelet thrombi under laminar arterial flow. In thrombin generation-based assays, shearing plasma decreased PS activity, an effect not seen in the absence of VWF. Finally, free PS deficiency in COVID-19 patients, measured using an antibody that binds near the C4BP binding site in SHBG, correlated with changes in VWF, but not C4BP, and with thrombin generation. Our data suggest that PS binds to a shear-exposed site on VWF, thus sequestering free PS and decreasing its anticoagulant activity, which would account for the increased thrombin generation potential. As many viral infections present with free PS deficiency, elevated circulating VWF, and increased vascular shear, we propose that the PS/VWF interaction reported here is a likely contributor to virus-associated thrombotic risk.

2.
Viruses ; 15(2)2023 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-36851646

RESUMEN

Adjuvants are essential components of subunit vaccines added to enhance immune responses to antigens through immunomodulation. Very few adjuvants have been approved for human use by regulatory agencies due to safety concerns. Current subunit vaccine adjuvants approved for human use are very effective in promoting humoral immune responses but are less effective at promoting T-cell immunity. In this study, we evaluated a novel pure enantio-specific cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (R-DOTAP) as an immunomodulator for subunit vaccines capable of inducing both humoral- and cellular-mediated immunity. Using recombinant protein antigens derived from SARS-CoV2 spike or novel computationally optimized broadly reactive influenza antigen (COBRA) proteins, we demonstrated that R-DOTAP nanoparticles promoted strong cellular- and antibody-mediated immune responses in both monovalent and bivalent vaccines. R-DOTAP-based vaccines induced antigen-specific and polyfunctional CD8+ and CD4+ effector T cells and memory T cells, respectively. Antibody responses induced by R-DOTAP showed a balanced Th1/Th2 type immunity, neutralizing activity and protection of mice from challenge with live SARS-CoV2 or influenza viruses. R-DOTAP also facilitated significant dose sparing of the vaccine antigens. These studies demonstrate that R-DOTAP is an excellent immune stimulator for the production of next-generation subunit vaccines containing multiple recombinant proteins.


Asunto(s)
COVID-19 , ARN Viral , Animales , Humanos , Ratones , Adyuvantes Inmunológicos , Cationes , COVID-19/prevención & control , Ácidos Grasos Monoinsaturados , Inmunidad , Lípidos , SARS-CoV-2 , Vacunas Sintéticas/genética , Anticuerpos Antivirales/inmunología
3.
Viruses ; 15(2)2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36851752

RESUMEN

It is clear that new approaches are needed to promote broadly protective immunity to viral pathogens, particularly those that are prone to mutation and escape from antibody-mediated immunity. Prototypic pathogens of this type are influenza and SARS-CoV-2, where the receptor-binding protein exhibits extremely high variability in its receptor-binding regions. T cells, known to target many viral proteins, and within these, highly conserved peptide epitopes, can contribute greatly to protective immunity through multiple mechanisms but are often poorly recruited by current vaccine strategies. Here, we have studied a promising novel pure enantio-specific cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (R-DOTAP), which was previously recognized for its ability to generate anti-tumor immunity through the induction of potent cytotoxic CD8 T cells. Using a preclinical mouse model, we have assessed an R-DOTAP nanoparticle adjuvant system for its ability to promote CD4 T cell responses to vaccination with recombinant influenza protein. Our studies revealed that R-DOTAP consistently outperformed a squalene-based adjuvant emulsion, even when it was introduced with a potent TLR agonist CpG, in the ability to elicit peptide epitope-specific CD4 T cells when quantified by IFN-γ and IL-2 ELISpot assays. Clinical testing of R-DOTAP containing vaccines in earlier work by others has demonstrated an acceptable safety profile. Hence, R-DOTAP can offer exciting opportunities as an immune stimulant for next-generation prophylactic recombinant protein-based vaccines.


Asunto(s)
COVID-19 , Vacunas contra la Influenza , Gripe Humana , Nanopartículas , Animales , Ratones , Humanos , Gripe Humana/prevención & control , Hemaglutininas , Escualeno , Linfocitos T CD4-Positivos , SARS-CoV-2 , Adyuvantes Inmunológicos , Vacunas Sintéticas , Vacunación , Cationes
4.
Sci Rep ; 10(1): 17090, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33051497

RESUMEN

The triterpene oil squalene is an essential component of nanoemulsion vaccine adjuvants. It is most notably in the MF59 adjuvant, a component in some seasonal influenza vaccines, in stockpiled, emulsion-based adjuvanted pandemic influenza vaccines, and with demonstrated efficacy for vaccines to other pandemic viruses, such as SARS-CoV-2. Squalene has historically been harvested from shark liver oil, which is undesirable for a variety of reasons. In this study, we have demonstrated the use of a Synthetic Biology (yeast) production platform to generate squalene and novel triterpene oils, all of which are equally as efficacious as vaccine adjuvants based on physiochemical properties and immunomodulating activities in a mouse model. These Synthetic Biology adjuvants also elicited similar IgG1, IgG2a, and total IgG levels compared to marine and commercial controls when formulated with common quadrivalent influenza antigens. Injection site morphology and serum cytokine levels did not suggest any reactogenic effects of the yeast-derived squalene or novel triterpenes, suggesting their safety in adjuvant formulations. These results support the advantages of yeast produced triterpene oils to include completely controlled growth conditions, just-in-time and scalable production, and the capacity to produce novel triterpenes beyond squalene.


Asunto(s)
Adyuvantes Inmunológicos/química , Vacunas contra la Influenza/inmunología , Triterpenos/química , Animales , Anticuerpos Antivirales/sangre , Betacoronavirus/aislamiento & purificación , COVID-19 , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/virología , Citocinas/sangre , Inmunoglobulina G/sangre , Vacunas contra la Influenza/química , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Pandemias/prevención & control , Neumonía Viral/prevención & control , Neumonía Viral/virología , SARS-CoV-2 , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Biología Sintética/métodos
5.
J Immunol ; 202(12): 3524-3536, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31053626

RESUMEN

Certain types of cationic lipids have shown promise in cancer immunotherapy, but their mechanism of action is poorly understood. In this study, we describe the properties of an immunotherapeutic consisting of the pure cationic lipid enantiomer R-1,2-dioleoyl-3-trimethyl-ammonium-propane (R-DOTAP) formulated with modified viral or self-peptide Ags. R-DOTAP formulations with peptide Ags stimulate strong cross-presentation and potent CD8 T cell responses associated with a high frequency of polyfunctional CD8 T cells. In a human papillomavirus tumor model system, a single s.c. injection of tumor-bearing mice with R-DOTAP plus human papillomavirus Ags induces complete regression of large tumors associated with an influx of Ag-specific CD8 T cells and a reduction of the ratio of regulatory/Ag-specific CD8 T cells. R-DOTAP also synergizes with an anti-PD1 checkpoint inhibitor, resulting in a significant inhibition of B16 melanoma tumor growth. We found that R-DOTAP stimulates type I IFN production by dendritic cells in vivo and in vitro. s.c. injection of R-DOTAP results in an IFN-dependent increase in draining lymph node size and a concomitant increase in CD69 expression. Using knockout mice, we show that type I IFN is required for the induction of CD8 T cell activity following administration of R-DOTAP plus Ag. This response requires Myd88 but not TRIF or STING. We also show that R-DOTAP stimulates both TLR7 and 9. Collectively, these studies reveal that R-DOTAP stimulates endosomal TLRs, resulting in a Myd88-dependent production of type I IFN. When administered with Ag, this results in potent Ag-specific CD8 T cell responses and antitumor activity.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Melanoma/terapia , Nanopartículas/metabolismo , Papillomaviridae/fisiología , Infecciones por Papillomavirus/terapia , Neoplasias Cutáneas/terapia , Linfocitos T Citotóxicos/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Ácidos Grasos Monoinsaturados/química , Humanos , Interferón Tipo I/metabolismo , Activación de Linfocitos , Melanoma/inmunología , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Nanopartículas/química , Infecciones por Papillomavirus/inmunología , Compuestos de Amonio Cuaternario/química , Neoplasias Cutáneas/inmunología , Linfocitos T Citotóxicos/trasplante
6.
J Immunol ; 200(2): 725-736, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29237778

RESUMEN

The integration of inflammatory signals is paramount in controlling the intensity and duration of immune responses. Eicosanoids, particularly PGE2, are critical molecules in the initiation and resolution of inflammation and in the transition from innate to acquired immune responses. Microsomal PGE synthase 1 (mPGES1) is an integral membrane enzyme whose regulated expression controls PGE2 levels and is highly expressed at sites of inflammation. PGE2 is also associated with modulation of autoimmunity through altering the IL-23/IL-17 axis and regulatory T cell (Treg) development. During a type II collagen-CFA immunization response, lack of mPGES1 impaired the numbers of CD4+ regulatory (Treg) and Th17 cells in the draining lymph nodes. Ag-experienced mPGES1-/- CD4+ cells showed impaired IL-17A, IFN-γ, and IL-6 production when rechallenged ex vivo with their cognate Ag compared with their wild-type counterparts. Additionally, production of PGE2 by cocultured APCs synergized with that of Ag-experienced CD4+ T cells, with mPGES1 competence in the APC compartment enhancing CD4+ IL-17A and IFN-γ responses. However, in contrast with CD4+ cells that were Ag primed in vivo, exogenous PGE2 inhibited proliferation and skewed IL-17A to IFN-γ production under Th17 polarization of naive T cells in vitro. We conclude that mPGES1 is necessary in vivo to mount optimal Treg and Th17 responses during an Ag-driven primary immune response. Furthermore, we uncover a coordination of autocrine and paracrine mPGES1-driven PGE2 production that impacts effector T cell IL-17A and IFN-γ responses.


Asunto(s)
Comunicación Autocrina , Dinoprostona/metabolismo , Comunicación Paracrina , Prostaglandina-E Sintasas/genética , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Animales , Epítopos de Linfocito T/inmunología , Regulación de la Expresión Génica , Inmunización , Inmunomodulación , Activación de Linfocitos/inmunología , Ratones , Fenotipo , Prostaglandina-E Sintasas/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E/genética
7.
J Trauma Acute Care Surg ; 77(2): 198-201, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25058241

RESUMEN

BACKGROUND: Transfusion-related immunomodulation consists of both proinflammatory and anti-inflammatory responses after transfusion of blood products. Stored red blood cells (RBCs) suppress human T-cell proliferation in vitro, but the mechanism remains unknown. We hypothesized that cytokine synthesis by T cells may be inhibited when stored RBCs are present and that suppression between fresh and stored RBCs would be different. METHODS: Purified human T cells were stimulated to proliferate with anti-CD3/anti-CD28 and then exposed to stored or fresh RBCs. Cells were placed in culture for 5 days. Cell culture supernatants were analyzed for the production of typical T-cell cytokines using multianalyte ELISArray kits. RESULTS: Stimulated T cells proliferated. RBC exposure markedly suppressed this proliferation. Interleukin 10, interleukin 17a, interferon γ, tumor necrosis factor α, and granulocyte macrophage colony-stimulating factor were increased in response to stimulation but depressed in the presence of stored RBCs. The use of fresh RBCs also resulted in depression of these cytokines when compared with stimulated T cells with no RBCs; however, this depression was less pronounced. CONCLUSION: T-cell activation is associated with both proinflammatory and anti-inflammatory cytokine release, comparable with patterns seen in trauma and acute injury. All of these responses are depressed by an exposure to stored RBCs. Decreased levels of these cytokines after RBC transfusion represents a potential contributor to the immunosuppressive complications seen in trauma patients after transfusion. This provides insight for future mechanistic studies to delineate the role of RBC transfusion in transfusion-related immunomodulation.


Asunto(s)
Citocinas/biosíntesis , Transfusión de Eritrocitos , Linfocitos T/metabolismo , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/inmunología , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Humanos , Técnicas In Vitro , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Interleucina-17/biosíntesis , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis
8.
Transfusion ; 54(5): 1340-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24188586

RESUMEN

BACKGROUND: Red blood cells (RBCs) suppress T-cell responsiveness through a mechanism requiring cell-cell contact. Questions remain as to whether this effect is an allogeneic response, related to cell death, or dependent on particular components of the RBCs. STUDY DESIGN AND METHODS: Peripheral T cells were isolated from healthy donors and exposed to stored allogeneic RBCs or autologous RBCs after processing. RBCs were lysed by hypotonic solvent to produce cellular ghosts. Tritiated thymidine proliferation assays were utilized. Cultures were saturated with interleukin (IL)-2 to determine whether impaired IL-2 synthesis played a role. RESULTS: T-cell proliferation was suppressed by both autologous and allogeneic RBCs. RBC membrane integrity does enhance T-cell suppression. T-cell death is not responsible for the suppressive changes. IL-2 synthesis is suppressed in RBC-exposed T cells but addition of exogenous IL-2 does not rescue proliferative capabilities. Proliferation of T cells was inhibited with RBC exposure but mitigated with the addition of fresh RBCs. CONCLUSIONS: T-cell suppression is enhanced by intact RBCs but this effect is unrelated solely to alloantigens. Neither apoptosis nor necrosis of T cells contributes to this phenomenon. IL-2 synthesis is suppressed after RBC exposure as a consequence of T-cell inhibition, but is not the primary cause of suppression. Fresh RBCs do not mediate T-cell suppression, indicating that changes in the RBC and development of the storage lesion may occur during initial blood bank processing.


Asunto(s)
Bancos de Sangre , Eritrocitos/fisiología , Linfocitos T/inmunología , Apoptosis , Células Cultivadas , Humanos , Tolerancia Inmunológica , Interleucina-2/farmacología , Activación de Linfocitos
9.
J Immunol ; 191(10): 4979-88, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24127557

RESUMEN

Microsomal PGE synthase-1 (mPGES-1) is an inducible enzyme that specifically catalyzes the conversion of PGH2 to PGE2. We showed that mPGES-1 null mice had a significantly reduced incidence and severity of collagen-induced arthritis compared with wild-type (WT) mice associated with a marked reduction in Abs to type II collagen. In this study, we further elucidated the role of mPGES-1 in the humoral immune response. Basal levels of serum IgM and IgG were significantly reduced in mPGES-1 null mice. Compared with WT mice, mPGES-1 null mice exhibited a significant reduction of hapten-specific serum Abs in response to immunization with the T cell-dependent (TD) Ag DNP-keyhole limpet hemocyanin. Immunization with the T cell-independent type 1 Ag trinitrophenyl-LPS or the T cell-independent type 2 Ag DNP-Ficoll revealed minimal differences between strains. Germinal center formation in the spleen of mPGES-1 null and WT mice were similar after immunization with DNP-keyhole limpet hemocyanin. To determine whether the effect of mPGES-1 and PGE2 was localized to hematopoietic or nonhematopoietic cells, we generated bone marrow chimeras. We demonstrated that mPGES-1 deficiency in nonhematopoietic cells was the critical factor for reduced TD Ab production. We conclude that mPGES-1 and PGE2-dependent phenotypic changes of nonhematopoietic/mesenchymal stromal cells play a key role in TD humoral immune responses in vivo. These findings may have relevance to the pathogenesis of rheumatoid arthritis and other autoimmune inflammatory diseases associated with autoantibody formation.


Asunto(s)
Artritis Experimental/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Oxidorreductasas Intramoleculares/deficiencia , Linfocitos T/inmunología , Animales , Artritis Experimental/genética , Trasplante de Médula Ósea , Células Cultivadas , Colágeno/inmunología , Femenino , Inmunidad Humoral , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Interleucina-2/metabolismo , Interleucina-4/metabolismo , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Prostaglandina-E Sintasas
10.
J Immunol ; 191(8): 4068-79, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24043900

RESUMEN

Heat shock transcription factor 1 (HSF1) is a major transcriptional regulator of the heat shock response in eukaryotic cells. HSF1 is evoked in response to a variety of cellular stressors, including elevated temperatures, oxidative stress, and other proteotoxic stressors. Previously, we demonstrated that HSF1 is activated in naive T cells at fever range temperatures (39.5°C) and is critical for in vitro T cell proliferation at fever temperatures. In this study, we demonstrated that murine HSF1 became activated to the DNA-binding form and transactivated a large number of genes in lymphoid cells strictly as a consequence of receptor activation in the absence of apparent cellular stress. Microarray analysis comparing HSF1(+/+) and HSF1(-/-) gene expression in T cells activated at 37°C revealed a diverse set of 323 genes significantly regulated by HSF1 in nonstressed T cells. In vivo proliferation studies revealed a significant impairment of HSF1(-/-) T cell expansion under conditions mimicking a robust immune response (staphylococcal enterotoxin B-induced T cell activation). This proliferation defect due to loss of HSF1 is observed even under nonfebrile temperatures. HSF1(-/-) T cells activated at fever temperatures show a dramatic reduction in cyclin E and cyclin A proteins during the cell cycle, although the transcription of these genes was modestly affected. Finally, B cell and hematopoietic stem cell proliferation from HSF1(-/-) mice, but not HSF1(+/+) mice, were also attenuated under stressful conditions, indicating that HSF1 is critical for the cell cycle progression of lymphoid cells activated under stressful conditions.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Activación de Linfocitos , Estrés Fisiológico , Linfocitos T/inmunología , Factores de Transcripción/metabolismo , Animales , Ciclo Celular , División Celular , Proliferación Celular , Células Cultivadas , Ciclina A/biosíntesis , Ciclina E/biosíntesis , Proteínas de Unión al ADN/genética , Enterotoxinas/inmunología , Fiebre/inmunología , Regulación de la Expresión Génica , Factores de Transcripción del Choque Térmico , Proteínas de Choque Térmico/metabolismo , Respuesta al Choque Térmico/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/genética
11.
J Surg Res ; 184(1): 567-71, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23764315

RESUMEN

BACKGROUND: Transfusion of packed red blood cells (RBCs) produces a myriad of immunologic derangements, from suppressive to stimulatory. Proliferation of human T cells is suppressed in vitro after exposure to processed red blood cells (PRBCs). We hypothesized that this effect would be mitigated by using fresh RBCs. We also hypothesized that this suppressive effect was a generalized effect on lymphocyte proliferation and would be observed in both CD4+ and CD8+ T-cell subpopulations as well as B cells. MATERIALS AND METHODS: We isolated human T cells from donor peripheral blood mononuclear cells and exposed them to either blood bank PRBCs or fresh RBCs from volunteer donors and stimulated them with anti-CD3/anti-CD28. Human B cells were stimulated with lipopolysaccharide and exposed to PRBCs or fresh RBCs. We measured proliferation of B cells by thymidine incorporation assays. We also treated RBCs with citrate-phosphate-dextrose (CPD) at different time points before culture them with stimulated T cells to determine the role of this common RBC storage solution in lymphocyte proliferation. RESULTS: In vitro proliferation of CD4+ and CD8+ T cells was suppressed by blood bank RBCs. This suppression is eliminated when fresh RBCs were used. The B cells showed inhibition of proliferation when exposed to similar conditions, which appeared to be consistent over serial dilutions. Fresh RBCs exposed to CPD did not appear suppressive in the first 6 h after exposure. CONCLUSIONS: T-cell and B-cell proliferation inhibition by blood banked RBCs suggests a generalized effect of RBCs on cellular proliferation. The lack of suppression by fresh RBCs further suggests that something involved in blood banking alters RBC properties such that they attain a suppressive phenotype. One such blood banking component, CPD, does not appear to affect this suppressive phenotype within the first 6 h.


Asunto(s)
Linfocitos B/citología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Transfusión de Eritrocitos , Eritrocitos/citología , Linfocitos B/inmunología , Almacenamiento de Sangre/métodos , Conservación de la Sangre/métodos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Células Cultivadas , Eritrocitos/inmunología , Citometría de Flujo , Humanos , Inmunomodulación/inmunología , Técnicas In Vitro
12.
J Immunol ; 190(5): 2199-207, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23359502

RESUMEN

Chronic infection with Toxoplasma gondii induces a potent resistance against reinfection, and IFN-γ production by CD8(+) T cells is crucial for the protective immunity. However, the molecular mechanisms that regulate the secondary response remain to be elucidated. In the current study, we examined the role of IL-2 in IFN-γ production by CD8(+) immune T cells in their secondary responses using T. gondii-specific CD8(+) T cell hybridomas and splenic CD8(+) immune T cells from chronically infected mice. The majority (92%) of CD8(+) T cell hybridomas produced large amounts of IFN-γ only when a low amount (0.5 ng/ml) of exogenous IL-2 was provided in combination with T. gondii Ags. Inhibition of cell proliferation by mitomycin C did not affect the enhancing effect of IL-2 on the IFN-γ production, and significant increases in transcription factor T-bet expression were associated with the IL-2-mediated IFN-γ amplification. Splenic CD8(+) immune T cells produced similar low levels of IL-2 in the secondary response to T. gondii, and a blocking of IL-2 signaling by anti-IL-2Rα Ab or inhibitors of JAK1 and JAK3 significantly reduced IFN-γ production of the T cells. This IL-2-mediated upregulation of IFN-γ production was observed in mitomycin C-treated CD8(+) immune T cells, thus independent from their cell division. Therefore, endogenous IL-2 produced by CD8(+) immune T cells can play an important autocrine-enhancing role on their IFN-γ production in the secondary responses to T. gondii, suggesting an importance of induction of CD8(+) immune T cells with an appropriate IL-2 production for vaccine development.


Asunto(s)
Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Interleucina-2/farmacología , Bazo/inmunología , Toxoplasmosis Animal/inmunología , Animales , Anticuerpos/farmacología , Linfocitos T CD8-positivos/parasitología , Proliferación Celular , Enfermedad Crónica , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Hibridomas/inmunología , Hibridomas/metabolismo , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-2/antagonistas & inhibidores , Interleucina-2/biosíntesis , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/genética , Janus Quinasa 1/inmunología , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/genética , Janus Quinasa 3/inmunología , Ratones , Ratones Endogámicos BALB C , Mitomicina/farmacología , Bazo/parasitología , Bazo/patología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología , Toxoplasma/inmunología , Toxoplasmosis Animal/parasitología , Toxoplasmosis Animal/patología
13.
J Surg Res ; 179(1): e183-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22482768

RESUMEN

BACKGROUND: Transfusion of packed red blood cells (PRBCs) is associated with morbidity and mortality. The mechanisms are not fully understood. Packed red blood cells deplete extracellular arginine and possess transporters for arginine, an amino acid essential for normal immunity. We hypothesize that the membrane y+ amino acid transporter contributes to arginine depletion in PRBCs. MATERIALS AND METHODS: We titrated PRBCs to a 10% hematocrit with phosphate-buffered saline, blocked PRBC y+ transporters using n-ethylmaleimide (0.2 mM), and measured arginine and ornithine levels using liquid chromatography-mass spectroscopy. We added radiolabeled L-arginine [4,5-(3)H] (10 µmol/L) added to similar culture conditions and measured arginine uptake in counts per minute (CPM). We examined storage periods of 6-9 d, 1-4 wk, and 6 wk, and correlated donor demographics with arginine uptake. RESULTS: n-Ethylmaleimide blockade of y+ transporters impaired PRBC arginine depletion from culture media (117.6 ± 8.6 µM versus 76.9 ± 5.8 µM; P < 0.001) and reduced intracellular L-arginine (7,574 ± 955 CPM versus 18,192 ± 1,376 CPM; P < 0.01). Arginine depletion increased with storage duration (1 wk versus 6 wk; P < 0.002). With n-ethylmaleimide treatment, 6-wk-old PRBCs preserved more culture arginine (P < 0.008) than at shorter durations. Nine-day storage duration increased L-arginine uptake compared with 6- to 8-day storage (n = 77, R = 0.225, P < 0.05). Extracellular arginine depletion and extracellular ornithine synthesis varied among donors and correlated inversely (R = -0.5, P = 0.01). CONCLUSIONS: Membrane y+ transporters are responsible for arginine depletion by PRBCs. Membrane y+ activity increases with storage duration. Arginine uptake varies among donors. Membrane biology of RBCs may have a role in the negative clinical effects associated with PRBC transfusion.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Arginina/metabolismo , Transportador de Aminoácidos Catiónicos 1/metabolismo , Transfusión de Eritrocitos/efectos adversos , Eritrocitos/metabolismo , Transporte Biológico , Transportador de Aminoácidos Catiónicos 1/antagonistas & inhibidores , Transportador de Aminoácidos Catiónicos 1/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Etilmaleimida/farmacología , Hematócrito , Humanos , Factores de Tiempo
14.
J Allergy Clin Immunol ; 131(2): 442-50, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23182172

RESUMEN

BACKGROUND: The prevalence of peanut allergies is increasing. Peanuts and many other allergen sources contain significant amounts of triglycerides, which affect absorption of antigens but have unknown effects on sensitization and anaphylaxis. We recently reported that dietary medium-chain triglycerides (MCTs), which bypass mesenteric lymph and directly enter portal blood, reduce intestinal antigen absorption into blood compared with long-chain triglycerides (LCTs), which stimulate mesenteric lymph flow and are absorbed in chylomicrons through mesenteric lymph. OBJECTIVE: We sought to test how dietary MCTs affect food allergy. METHODS: C3H/HeJ mice were fed peanut butter protein in MCT, LCT (peanut oil), or LCT plus an inhibitor of chylomicron formation (Pluronic L81). Peanut-specific antibodies in plasma, responses of the mice to antigen challenges, and intestinal epithelial cytokine expression were subsequently measured. RESULTS: MCT suppressed antigen absorption into blood but stimulated absorption into Peyer patches. A single gavage of peanut protein with MCT, as well as prolonged feeding in MCT-based diets, caused spontaneous allergic sensitization. MCT-sensitized mice experienced IgG-dependent anaphylaxis on systemic challenge and IgE-dependent anaphylaxis on oral challenge. MCT feeding stimulated jejunal-epithelial thymic stromal lymphopoietin, Il25, and Il33 expression compared with that seen after LCT feeding and promoted T(H)2 cytokine responses in splenocytes. Moreover, oral challenges of sensitized mice with antigen in MCT significantly aggravated anaphylaxis compared with challenges with the LCT. Importantly, the effects of MCTs could be mimicked by adding Pluronic L81 to LCTs, and in vitro assays indicated that chylomicrons prevent basophil activation. CONCLUSION: Dietary MCTs promote allergic sensitization and anaphylaxis by affecting antigen absorption and availability and by stimulating T(H)2 responses.


Asunto(s)
Anafilaxia/inmunología , Arachis/inmunología , Hipersensibilidad al Cacahuete/inmunología , Triglicéridos/inmunología , Alérgenos/inmunología , Anafilaxia/metabolismo , Animales , Anticuerpos/inmunología , Antígenos/inmunología , Arachis/química , Basófilos/inmunología , Basófilos/metabolismo , Quilomicrones/inmunología , Citocinas/inmunología , Dieta/métodos , Femenino , Inmunoglobulina E/inmunología , Inmunoglobulina G/inmunología , Interleucinas/inmunología , Absorción Intestinal/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Yeyuno/inmunología , Yeyuno/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratas , Ratas Sprague-Dawley , Células Th2/inmunología , Células Th2/metabolismo , Triglicéridos/administración & dosificación , Triglicéridos/metabolismo
15.
Eur J Pharm Biopharm ; 80(2): 315-22, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22068049

RESUMEN

The purpose of this study was to design novel nanocapsules (NCs) with surface-chelated nickel (Ni-NCs) as a vaccine delivery system for histidine (His)-tagged protein antigens. Ni-NCs were characterized for binding His-tagged model proteins through high-affinity non-covalent interactions. The mean diameter and zeta potential of the optimized Ni-NCs were 214.9 nm and -14.8 mV, respectively. The optimal binding ratio of His-tagged Green Fluorescent Protein (His-GFP) and His-tagged HIV-1 Gag p41 (His-Gag p41) to the Ni-NCs was 1:221 and 1:480 w/w, respectively. Treatment of DC2.4 cells with Ni-NCs did not result in significant loss in the cell viability up to 24h (<5%). We further evaluated the antibody response of the Ni-NCs using His-Gag p41 as a model antigen. Formulations were administered subcutaneously to BALB/c mice at day 0 (prime) and 14 (boost) followed by serum collection on day 28. Serum His-Gag p41-specific antibody levels were found to be significantly higher at 1 and 0.5 µg doses of Gag p41-His-Ni-NCs (His-Gag p41 equivalent) compared with His-Gag p41 (1 µg) adjuvanted with aluminum hydroxide (AH). The serum IgG2a levels induced by Gag p41-His-Ni-NCs (1 µg) were significantly higher than AH adjuvanted His-Gag p41. The Ni-NCs alone did not result in the elevation of systemic IL-12/p40 and CCL5/RANTES inflammatory cytokine levels upon subcutaneous administration in BALB/c mice. In conclusion, the proposed Ni-NCs can bind His-tagged proteins and have the potential to be used as antigen delivery system capable of generating strong antigen-specific antibodies at doses much lower than with aluminum-based adjuvant and causing no significant elevation of systemic pro-inflammatory IL-12/p40 and CCL5/RANTES cytokines.


Asunto(s)
Antígenos/inmunología , Histidina/química , Nanocápsulas , Vacunas/administración & dosificación , Animales , Antígenos/administración & dosificación , Citocinas/inmunología , Células Dendríticas/inmunología , Femenino , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/inmunología , Inflamación/inmunología , Inyecciones Subcutáneas , Lípidos/química , Ratones , Ratones Endogámicos BALB C , Níquel/química , Tamaño de la Partícula , Vacunas/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/administración & dosificación , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
16.
Infect Immun ; 79(1): 177-84, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20956571

RESUMEN

Heat shock factor 1 (HSF1) is a stress-induced transcription factor that promotes expression of genes that protect mammalian cells from the lethal effects of severely elevated temperatures (>42°C). However, we recently showed that HSF1 is activated at a lower temperature (39.5°C) in T cells, suggesting that HSF1 may be important for preserving T cell function during pathogen-induced fever responses. To test this, we examined the role of HSF1 in clearance of Listeria monocytogenes, an intracellular bacterial pathogen that elicits a strong CD8(+) T cell response in mice. Using temperature transponder microchips, we showed that the core body temperature increased approximately 2°C in L. monocytogenes-infected mice and that the fever response was maintained for at least 24 h. HSF1-deficient mice cleared a low-dose infection with slightly slower kinetics than did HSF1(+/+) littermate controls but were significantly more susceptible to challenges with higher doses of bacteria. Surprisingly, HSF1-deficient mice did not show a defect in CD8(+) T cell responses following sublethal infection. However, when HSF1-deficient mice were challenged with high doses of L. monocytogenes, increased levels of serum tumor necrosis factor alpha (TNF-α) and gamma interferon (IFN-γ) compared to those of littermate control mice were observed, and rapid death of the animals occurred within 48 to 60 h of infection. Neutralization of TNF-α enhanced the survival of HSF1-deficient mice. These results suggest that HSF1 is needed to prevent the overproduction of proinflammatory cytokines and subsequent death due to septic shock that can result following high-dose challenge with bacterial pathogens.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Fiebre/metabolismo , Listeria monocytogenes , Listeriosis/metabolismo , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Linfocitos T CD8-positivos/fisiología , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/fisiología , Genotipo , Factores de Transcripción del Choque Térmico , Interferón gamma/sangre , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/genética
17.
Int J Pharm ; 401(1-2): 87-92, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-20837122

RESUMEN

The present studies were aimed at investigating the engineering of NPs with protein-conjugated-surfactant at their surface. In order to increase the immunogenicity of a protein antigen, Brij 78 was functionalized by tresyl chloride and then further reacted with the primary amine of the model proteins ovalbumin (OVA) or horseradish peroxide (HRP). The reaction yielded Brij 78-OVA and Brij 78-HRP conjugates which were then used directly to form NP-OVA or NP-HRP using a one-step warm oil-in-water microemulsion precursor method with emulsifying wax as the oil phase, and Brij 78 and the Brij 78-OVA or Brij 78-HRP conjugate as surfactants. Similarly, Brij 700 was conjugated to HIV p24 antigen to yield Brij 700-p24 conjugate. The utility of these NPs for enhancing the immune responses to protein-based vaccines was evaluated in vivo using ovalbumin (OVA) as model protein and p24 as a relevant HIV antigen. In separate in vivo studies, female BALB/c mice were immunized by subcutaneous (s.c.) injection with NP-OVA and NP-p24 formulations along with several control formulations. These results suggested that with multiple antigens, covalent attachment of the antigen to the NP significantly enhanced antigen-specific immune responses. This facile covalent conjugation and incorporation method may be utilized to further incorporate other protein antigens, even multiple antigens, into an enhanced vaccine delivery system.


Asunto(s)
Adyuvantes Inmunológicos , Lípidos/química , Nanopartículas , Polietilenglicoles/química , Proteínas/química , Sulfonas/química , Adyuvantes Inmunológicos/química , Animales , Formación de Anticuerpos , Sistemas de Liberación de Medicamentos , Femenino , Proteína p24 del Núcleo del VIH/química , Proteína p24 del Núcleo del VIH/inmunología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/química , Ovalbúmina/inmunología , Proteínas/inmunología , Vacunas/química , Vacunas/inmunología
18.
J Trauma ; 69(2): 320-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20699740

RESUMEN

BACKGROUND: Packed red blood cell (PRBC) transfusion suppresses immunity and increases morbidity and mortality. Leukocyte reduction has failed to abrogate these effects, thus implicating red blood cells themselves or their components. PRBC impair proliferation of immortal (Jurkat) T cells by depleting arginine from the extracellular environment. The effect of PRBC on isolated ex vivo T-cell proliferation has not been reported. We hypothesize that PRBCs depress mitogen-stimulated proliferation in isolated human and mouse T cells. METHODS: Human peripheral T cells were isolated by Ficoll-Hypaque gradient, purified by magnetic separation, and stimulated with anti-CD3 or anti-CD28. DO11.10 transgenic mouse splenic T cells were stimulated with ovalbumin. Cells were cultured at 1 x 10(6)/mL in 96-well plates or in 24-transwell plates in the presence of PRBC (0.015-5% by volume, stored for 4-6 weeks). In culture media, arginine and citrulline were varied. Proliferation was measured at 72 hours by thymidine incorporation. T-cell viability, apoptosis, and receptor zeta chain were measured by flow cytometry. RESULTS: PRBC significantly depressed human peripheral and mouse splenic T-cell proliferation in a dose-dependent manner. PRBC arginase blockade by N-omega-hydroxy-nor-l-arginine only partly restored proliferation. Cell contact was required in both cell types for maximal effect. Depressed zeta chain in human peripheral T cells was partly restored by arginase blockade. Salvage by high-dose arginine and citrulline was unsuccessful. Decreased proliferation was not related to cell death. CONCLUSION: PRBC suppresses mitogen-stimulated human and antigen-stimulated mouse T-cell proliferation by mechanisms independent of arginine depletion. This is a novel mechanism for transfusion-associated immune suppression.


Asunto(s)
Arginina/metabolismo , Comunicación Celular/inmunología , Proliferación Celular , Transfusión de Eritrocitos/efectos adversos , Tolerancia Inmunológica , Linfocitos T/citología , Aminoácidos/análisis , Aminoácidos/metabolismo , Animales , Apoptosis/inmunología , Apoptosis/fisiología , Arginina/análisis , Técnicas de Cultivo de Célula , Células Cultivadas , Intervalos de Confianza , Regulación hacia Abajo/inmunología , Regulación hacia Abajo/fisiología , Eritrocitos/citología , Eritrocitos/inmunología , Citometría de Flujo , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/fisiología , Activación de Linfocitos/inmunología , Ratones , Linfocitos T/inmunología
19.
PLoS One ; 4(12): e8442, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20041190

RESUMEN

BACKGROUND: A small fraction of dietary protein survives enzymatic degradation and is absorbed in potentially antigenic form. This can trigger inflammatory responses in patients with celiac disease or food allergies, but typically induces systemic immunological tolerance (oral tolerance). At present it is not clear how dietary antigens are absorbed. Most food staples, including those with common antigens such as peanuts, eggs, and milk, contain long-chain triglycerides (LCT), which stimulate mesenteric lymph flux and postprandial transport of chylomicrons through mesenteric lymph nodes (MLN) and blood. Most dietary antigens, like ovalbumin (OVA), are emulsifiers, predicting affinity for chylomicrons. We hypothesized that chylomicron formation promotes intestinal absorption and systemic dissemination of dietary antigens. METHODOLOGY/PRINCIPAL FINDINGS: Absorption of OVA into MLN and blood was significantly enhanced when OVA was gavaged into fasted mice together with LCT compared with medium-chain triglycerides (MCT), which do not stimulate chylomicron formation. The effect of LCT was blocked by the addition of an inhibitor of chylomicron secretion, Pluronic L-81. Adoptively transferred OVA-specific DO11.10 T-cells proliferated more extensively in peripheral lymph nodes when OVA was gavaged with LCT than with MCT or LCT plus Pluronic L-81, suggesting that dietary OVA is systemically disseminated. Most dietary OVA in plasma was associated with chylomicrons, suggesting that these particles mediate systemic antigen dissemination. Intestinal-epithelial CaCo-2 cells secreted more cell-associated, exogenous OVA when stimulated with oleic-acid than with butyric acid, and the secreted OVA appeared to be associated with chylomicrons. CONCLUSIONS/SIGNIFICANCE: Postprandial chylomicron formation profoundly affects absorption and systemic dissemination of dietary antigens. The fat content of a meal may affect immune responses to dietary antigens by modulating antigen absorption and transport.


Asunto(s)
Antígenos/metabolismo , Quilomicrones/metabolismo , Dieta , Absorción Intestinal/fisiología , Ovalbúmina/metabolismo , Animales , Presentación de Antígeno/inmunología , Antígenos/administración & dosificación , Células CACO-2 , Quilomicrones/sangre , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/administración & dosificación , Transporte de Proteínas , Triglicéridos/metabolismo
20.
HIV AIDS (Auckl) ; 2009(1): 1-11, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21966230

RESUMEN

Lipid-based nanoparticles (NPs) with a small amount of surface-chelated nickel (Ni-NPs) were developed to easily formulate the HIV his-tagged Tat protein, as well as to formulate and co-deliver two HIV antigens (his-p24 and his-Nef) on one particle. Female BALB/c mice were immunized by s.c. injection with his-Tat/Ni-NP formulation (1.5 µg Tat-his/mouse) and control formulations on day 0 and 14. The day 28 anti-Tat specific IgG titer with his-Tat/Ni-NP was significantly greater than that with Alum/his-Tat. Furthermore, splenocytes from his-Tat/Ni-NP immunized mice secreted significantly higher IFN-γ than those from mice immunized with Alum/his-Tat. Although Ni-NPs did not show better adjuvant activity than Tat-coated anionic NPs made with sodium dodecyl sulfate (SDS/NPs), they were less toxic than SDS/NPs. The initial results indicated that co-immunization of mice using his-p24/his-Nef/Ni-NP induced greater antibody response compared to using Alum/his-p24/his-Nef. Co-delivery of two antigens using Ni-NPs also increased the immunogenicity of individual antigens compared to delivery of a single antigen by Ni-NPs. In conclusion, Ni-NPs are an efficient delivery system for HIV vaccines including both single antigen delivery and multiple antigen co-delivery.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...