Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Immunol ; 12: 718359, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867947

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with widespread inflammation, immune dysregulation, and is associated with the generation of destructive anti-DNA autoantibodies. We have shown previously the immune modulatory properties of pCons peptide in the induction of both CD4+ and CD8+ regulatory T cells which can in turn suppress development of the autoimmune disease in (NZB/NZW) F1 (BWF1) mice, an established model of lupus. In the present study, we add novel protein information and further demonstrate the molecular and cellular phenotypes of pCons-induced CD4+ and CD8+ Treg subsets. Flow cytometry analyses revealed that pCons induced CD8+ Treg cells with the following cell surface molecules: CD25highCD28high and low subsets (shown earlier), CD62Lhigh, CD122low, PD1low, CTLA4low, CCR7low and 41BBhigh. Quantitative real-time PCR (qRT-PCR) gene expression analyses revealed that pCons-induced CD8+ Treg cells downregulated the following several genes: Regulator of G protein signaling (RGS2), RGS16, RGS17, BAX, GPT2, PDE3b, GADD45ß and programmed cell death 1 (PD1). Further, we confirmed the down regulation of these genes by Western blot analyses at the protein level. To our translational significance, we showed herein that pCons significantly increased the percentage of CD8+FoxP3+ T cells and further increased the mean fluorescence intensity (MFI) of FoxP3 when healthy peripheral blood mononuclear cells (PBMCs) are treated with pCons (10 µg/ml, for 24-48 hours). In addition, we found that pCons reduced apoptosis in CD4+ and CD8+ T cells and B220+ B cells of BWF1 lupus mice. These data suggest that pCons stimulates cellular, immunological, and molecular changes in regulatory T cells which in turn protect against SLE autoimmunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Péptidos/genética , Péptidos/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/metabolismo , Apoptosis/genética , Apoptosis/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Femenino , Expresión Génica , Voluntarios Sanos , Humanos , Tolerancia Inmunológica/genética , Ratones , Ratones Endogámicos NZB , Persona de Mediana Edad , Péptidos/administración & dosificación , Proteínas RGS/genética , Proteínas RGS/inmunología , Adulto Joven
2.
Front Immunol ; 12: 725325, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34733276

RESUMEN

Recent evidence suggests the existence of a nexus between inflammatory pathways and the female sex hormone 17ß-estradiol, resulting in increased interferon-stimulated genes (ISGs), autoantibodies, and dysregulation of immune cells in SLE. However, the molecular mechanisms and the effect of estradiol on candidate target genes and their pathways remains poorly understood. Our previous work suggests that female SLE patients have increased estradiol levels compared to healthy controls. In the present study, we explored the effects of 17ß-estradiol treatment on expression of IFN (interferons)-stimulated genes and pro-inflammatory cytokines/chemokines. We found significantly increased (5-10-fold) expression of IFN-regulated genes in healthy females. Furthermore, we found significantly increased plasma levels of IL-6, IL-12, IL-17, IL-18, stem cell factor (SCF), and IL-21/IL-23 in SLE patients compared to healthy controls, and those levels positively correlated with the plasma levels of 17ß-estradiol. In addition, levels of IL-21 positively correlated with the SLE disease activity index (SLEDAI) score of SLE patients. In vitro treatment of PBMCs from either SLE patients or healthy controls with 17ß-estradiol at physiological concentration (~50 pg/ml) also significantly increased secretion of many pro-inflammatory cytokines and chemokines (IL-6, IL-12, IL-17, IL-8, IFN-γ; MIP1α, and MIP1ß) in both groups. Further our data revealed that 17ß-estradiol significantly increased the percentage of CD3+CD69+ and CD3+IFNγ+ T cells; whereas, simultaneous addition of 17ß-estradiol and an ERα inhibitor prevented this effect. Collectively, our findings indicate that 17ß-estradiol participates in the induction of pro-inflammatory cytokines and chemokines and further influences interferon genes and pathways.


Asunto(s)
Estradiol/metabolismo , Interferones/metabolismo , Leucocitos Mononucleares/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Adulto , Animales , Estudios de Casos y Controles , Quimiocinas/metabolismo , Citocinas/metabolismo , Estradiol/farmacología , Femenino , Humanos , Interferones/genética , Leucocitos Mononucleares/efectos de los fármacos , Modelos Lineales , Masculino , Ratones , Persona de Mediana Edad , Adulto Joven
3.
Front Immunol ; 12: 662901, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093553

RESUMEN

The regulation of autoimmunity and the molecular mechanisms by which different immune cells, including T cells, polymorphonuclear leukocytes (PMN-granulocytes), and B cells suppress autoimmune diseases is complex. We have shown previously that BWF1 lupus mice are protected from autoimmunity after i.v. injection or oral administration of tolerogenic doses of pCons, an artificial synthetic peptide based on sequences containing MHC class I and MHC class II determinants in the VH region of a J558-encoded BWF1 anti-DNA Ab. Several T cell subsets can transfer this tolerance. In this study, we determined the potential roles of granulocytes, B cells and regulatory T cells altered by pCons treatment in the BWF1 (NZB/NZW) mouse model of lupus. Immunophenotyping studies indicated that pCons treatment of BWF1 mice significantly increased CD4+FoxP3+ T cells, reduced the percent of B cells expressing CD19+CD5+ but increased the percent of CD19+CD1d+ regulatory B cells and increased the ability of the whole B cell population to suppress IgG anti-DNA production in vitro. pCons treatment significantly decreased the expression of CTLA-4 (cytotoxic T-lymphocyte-associated protein-4) in CD8+ T cells. In addition, peptide administration modified granulocytes so they became suppressive. We co-cultured sorted naïve B cells from mice making anti-DNA Ab (supported by addition of sorted naive CD4+ and CD8+ T cells from young auto-antibody-negative BWF1 mice) with sorted B cells or granulocytes from tolerized mice. Both tolerized granulocytes and tolerized B cells significantly suppressed the production of anti-DNA in vitro. In granulocytes from tolerized mice compared to saline-treated littermate controls, real-time PCR analysis indicated that expression of interferon-induced TNFAIP2 increased more than 2-fold while Ptdss2 and GATA1 mRNA were up-regulated more than 10-fold. In contrast, expression of these genes was significantly down-regulated in tolerized B cells. Further, another IFN-induced protein, Bcl2, was reduced in tolerized B cells as determined by Western blot analyses. In contrast, expression of FoxP3 was significantly increased in tolerized B cells. Together, these data suggest that B cells and granulocytes are altered toward suppressive functions by in vivo tolerization of BWF1 mice with pCons and it is possible these cell types participate in the clinical benefits seen in vivo.


Asunto(s)
Lupus Eritematoso Sistémico/inmunología , Animales , Anticuerpos Antinucleares/inmunología , Autoinmunidad , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Expresión Génica , Granulocitos/inmunología , Granulocitos/metabolismo , Tolerancia Inmunológica/inmunología , Inmunofenotipificación , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos NZB , Péptidos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
4.
Vaccines (Basel) ; 8(4)2020 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-33167311

RESUMEN

BACKGROUND: Immune checkpoint blockade that downregulates T cell evasion for effective immunity has provided a renewed interest in therapeutic cancer vaccines. METHODS: Utilizing murine lung cancer models, we determined: tumor burden, TIL cytolysis, immunohistochemistry, flow cytometry, RNA Sequencing, CD4 T cells, CD8 T cells, CXCL9 chemokine, and CXCL10 chemokine neutralization to evaluate the efficacy of Programmed cell death protein 1 (PD-1) blockade combined with chemokine (C-C motif) ligand 21-dendritic cell tumor antigen (CCL21-DC tumor Ag) vaccine. RESULTS: Anti-PD1 combined with CCL21-DC tumor Ag vaccine eradicated 75% of 12-day established tumors (150 mm3) that was enhanced to 90% by administering CCL21-DC tumor Ag vaccine prior to combined therapy. The effect of combined therapy was blocked by CD4, CD8, CXCL9, and CXCL10 neutralizing antibodies. CONCLUSION: PD-1 blockade therapy plus CCL21-DC tumor Ag vaccine could be beneficial to lung cancer patients.

5.
FASEB J ; 31(3): 998-1010, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27895106

RESUMEN

Tumor exosomes are emerging as antitumor immunity regulators; however, their effects on secondary exosome secretion by distal organs have not been explored. We have previously demonstrated that suppression of exosomes at the distal tumor site of pancreatic ductal adenocarcinoma (PDAC) ablated the development of salivary biomarker profile. Here, we explore the function of salivary exosomes from tumor-bearing mice in immune surveillance. We provide evidence that salivary exosomes from mice with PDAC exhibit a suppressive effect that results in reduced tumor-killing capacity by NK cells. Salivary exosomes from mice with PDAC where pancreatic tumors were engineered to suppress exosome biogenesis failed to suppress NK cell cytotoxic potential against tumor cells, as opposed to salivary exosomes from mice with PDAC with normal tumor exosome biogenesis. These results reveal an important and previously unknown mechanism of antitumor immune regulation and provide new insights into our understanding of the alterations of this biofluid during tumor development.-Katsiougiannis, S., Chia, D., Kim, Y., Singh, R. P., Wong, D. T. W. Saliva exosomes from pancreatic tumor-bearing mice modulate NK cell phenotype and antitumor cytotoxicity.


Asunto(s)
Carcinoma Ductal Pancreático/inmunología , Citotoxicidad Inmunológica , Exosomas/inmunología , Factores Inmunológicos/metabolismo , Células Asesinas Naturales/inmunología , Neoplasias Pancreáticas/inmunología , Saliva/inmunología , Animales , Línea Celular Tumoral , Femenino , Ratones , Ratones Endogámicos C57BL , Fenotipo
6.
Stem Cells Dev ; 23(14): 1611-24, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24650034

RESUMEN

Mesenchymal stem cells (MSCs) are being tested in a wide range of human diseases; however, loss of potency and inconsistent quality severely limit their use. To overcome these issues, we have utilized a developmental precursor called the hemangioblast as an intermediate cell type in the derivation of a highly potent and replenishable population of MSCs from human embryonic stem cells (hESCs). This method circumvents the need for labor-intensive hand-picking, scraping, and sorting that other hESC-MSC derivation methods require. Moreover, unlike previous reports on hESC-MSCs, we have systematically evaluated their immunomodulatory properties and in vivo potency. As expected, they dynamically secrete a range of bioactive factors, display enzymatic activity, and suppress T-cell proliferation that is induced by either allogeneic cells or mitogenic stimuli. However, they also display unique immunophenotypic properties, as well as a smaller size and >30,000-fold proliferative capacity than bone marrow-derived MSCs. In addition, this is the first report which demonstrates that hESC-MSCs can inhibit CD83 up-regulation and IL-12p70 secretion from dendritic cells and enhance regulatory T-cell populations induced by interleukin 2 (IL-2). This is also the first report which shows that hESC-MSCs have therapeutic efficacy in two different autoimmune disorder models, including a marked increase in survival of lupus-prone mice and a reduction of symptoms in an autoimmune model of uveitis. Our data suggest that this novel and therapeutically active population of MSCs could overcome many of the obstacles that plague the use of MSCs in regenerative medicine and serve as a scalable alternative to current MSC sources.


Asunto(s)
Diferenciación Celular/genética , Inmunomodulación , Células Madre Mesenquimatosas/citología , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular/inmunología , Linaje de la Célula , Proliferación Celular/genética , Células Dendríticas/citología , Células Dendríticas/inmunología , Humanos , Activación de Linfocitos/inmunología , Células Madre Mesenquimatosas/inmunología , Ratones , Células Madre Pluripotentes/inmunología , Linfocitos T Reguladores/inmunología
7.
Autoimmun Rev ; 11(9): 611-4, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22001419

RESUMEN

Regulatory T cells play an important role in the maintenance and regulation of immune tolerance and in the prevention of autoimmunity. Recent studies have demonstrated a deficiency in number and function of regulatory T cells in lupus and other autoimmune diseases. This may contribute to immune dysregulations and a defect in self-tolerance mechanisms. How to balance and "reset" the immune response from harmful pro-inflammatory to beneficial anti-inflammatory is the current strategy of the research. In this regard, several studies have been performed with various peptides, drugs, steroids and epigenetic agents to induce or modify regulatory cells and some measure of success has been achieved in the animal model of SLE and with lupus patient cells. Challenges ahead include the heterogeneous nature, phenotype and function of regulatory cells and the difficulties in manipulation of regulatory function in healthy versus diseased states. In this review, we will provide some recent findings indicating challenges and potential benefits of targeting of regulatory T cells in lupus.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Factores Inmunológicos/uso terapéutico , Lupus Eritematoso Sistémico/inmunología , Péptidos/uso terapéutico , Linfocitos T Reguladores/inmunología , Corticoesteroides/administración & dosificación , Corticoesteroides/uso terapéutico , Animales , Autoinmunidad/efectos de los fármacos , Recuento de Linfocito CD4 , Factores de Transcripción Forkhead/genética , Humanos , Tolerancia Inmunológica , Factores Inmunológicos/administración & dosificación , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/patología , Ratones , Péptidos/administración & dosificación , Linfocitos T Reguladores/efectos de los fármacos
8.
J Immunol ; 182(12): 7415-21, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494264

RESUMEN

Treatment of (NZB x NZW)F(1) (NZB/W) lupus-prone mice with the anti-DNA Ig-based peptide pConsensus prolongs the survival of treated animals and effectively delays the appearance of autoantibodies and glomerulonephritis. We have previously shown that part of these protective effects associated with the induction of CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) that suppressed autoantibody responses. Because the effects of pConsensus appeared secondary to qualitative rather than quantitative changes in Tregs, we investigated the molecular events induced by tolerance in Tregs and found that signaling pathways including ZAP70, p27, STAT1, STAT3, STAT6, SAPK, ERK, and JNK were not significantly affected. However, peptide tolerization affected in Tregs the activity of the MAPK p38, whose phosphorylation was reduced by tolerance. The pharmacologic inhibition of p38 with the pyridinyl imidazole inhibitor SB203580 in naive NZB/W mice reproduced in vivo the effects of peptide-induced tolerance and protected mice from lupus-like disease. Transfer experiments confirmed the role of p38 in Tregs on disease activity in the NZB/W mice. These data indicate that the modulation of p38 activity in lupus Tregs can significantly influence the disease activity.


Asunto(s)
ADN/inmunología , Tolerancia Inmunológica/inmunología , Inmunoglobulinas/inmunología , Lupus Eritematoso Sistémico/inmunología , Péptidos/inmunología , Linfocitos T Reguladores/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Recuento de Linfocito CD4 , ADN/genética , Regulación hacia Abajo/inmunología , Activación Enzimática/efectos de los fármacos , Femenino , Lupus Eritematoso Sistémico/enzimología , Lupus Eritematoso Sistémico/genética , Ratones , Péptidos/farmacología , Tasa de Supervivencia , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA