Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Immunol ; 195: 139-148, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29894743

RESUMEN

The major metabolic feature of diabetes is hyperglycemia which has been linked to the diabetes inflammatory processes, and diabetes-related vulnerability to infection. In the present study, we assessed how glucose affected PBMCs in type I interferon (IFN) production and subsequent signaling. We found that the moderately elevated glucose promoted, and high glucose suppressed type I IFN production, respectively. Pre-exposure to high glucose rendered monocytes more sensitive to IFN-α stimulation with heightened signaling, whereas, instantaneous addition of high glucose did not exhibit such effect. Consistent with this finding, the mRNA levels of IFN-α-induced IRF-7 in PBMCs were positively correlated with HbA1c levels of diabetes patients. Additionally, we found that high glucose promoted the production of other proinflammatory cytokines/chemokines. This study suggests that hyperglycemia may affect the inflammatory process in diabetes via promoting proinflammatory cytokines, as well as the host defense against microbial infections through impeding type I IFN production and signaling.


Asunto(s)
Complicaciones de la Diabetes/inmunología , Glucosa/metabolismo , Hiperglucemia/inmunología , Infecciones/inmunología , Inflamación/inmunología , Interferón Tipo I/metabolismo , Monocitos/inmunología , Adolescente , Adulto , Niño , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Inmunidad , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Masculino , Lectina 1 Similar a Ig de Unión al Ácido Siálico/metabolismo , Transducción de Señal , Células THP-1 , Adulto Joven
2.
Mol Ther ; 25(10): 2299-2308, 2017 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-28865999

RESUMEN

Hepatocellular carcinoma (HCC) has a high morbidity and mortality rate worldwide, with limited treatment options. Glypican-3 (GPC3) is a glycosylphosphatidylinositol-anchored glycoprotein that is overexpressed in most HCC tissues but not in normal tissues. GPC3-targeting antibody therapy shows limited response in a clinical trial due to the lack of a tumor-specific cytotoxic T lymphocyte (CTL) response. Here, in C57/B6 mice, we demonstrated that intravenous infusion of GPC3-coupled lymphocytes (LC/GPC3+) elicited robust GPC3-specific antibody and CTL responses, which effectively restricted proliferation and lysed cultured-HCC cells. Treatment with LC/GPC3+ induced durable tumor regression in HCC-bearing C57/B6 mice. Administration of LC/GPC3+ induced elevated levels of the cytotoxic T cell bioactive factors tumor necrosis factor alpha (TNF-α), interferon-γ (IFN-γ), granzyme B, and perforin, and substantially increased the number of infiltrating CD8+ T cells in tumor tissues. Moreover, immune responses elicited by LC/GPC3+ selectively suppressed GPC3+ tumors, but didn't affect the GPC3- tumors in BALB/c mice. Our findings provide the first preclinical evidence that intravenous infusion of the LC/GPC3+ complex can induce a strong anti-HCC effect through regulating systemic and local immune responses. These results indicate that the LC/GPC3+ complex could be developed as precision therapeutics for HCC patients in the future.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/prevención & control , Glipicanos/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/metabolismo , Interferón gamma/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo
3.
Mediators Inflamm ; 2018: 4316584, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30670926

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are identified as a heterogeneous population of cells with the function to suppress innate as well as adaptive immune responses. The initial studies of MDSCs were primarily focused on the field of animal tumor models or cancer patients. In cancer, MDSCs play the deleterious role to inhibit tumor immunity and to promote tumor development. Over the past few years, an increasing number of studies have investigated the role of MDSCs in autoimmune diseases. The beneficial effects of MDSCs in autoimmunity have been reported by some studies, and thus, immunosuppressive MDSCs may be a novel therapeutic target in autoimmune diseases. There are some controversial findings as well. Many questions such as the activation, differentiation, and suppressive functions of MDSCs and their roles in autoimmune diseases remain unclear. In this review, we have discussed the current understanding of MDSCs in autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Células Supresoras de Origen Mieloide/inmunología , Animales , Autoinmunidad/fisiología , Humanos
4.
J Immunol ; 193(3): 1024-34, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24973447

RESUMEN

Increasing evidence suggests that type 1 IFN (IFN-αß) is associated with pathogenesis of Th1-mediated type 1 diabetes (T1D). A major source of IFN-αß is plasmacytoid dendritic cells (pDCs). In this study, we analyzed peripheral blood pDC numbers and functions in at-risk, new-onset, and established T1D patients and controls. We found that subjects at risk for T1D and new-onset and established T1D subjects possessed significantly increased pDCs but similar number of myeloid DCs when compared with controls. pDC numbers were not affected by age in T1D subjects but declined with increasing age in control subjects. It was demonstrated that IFN-α production by PBMCs stimulated with influenza viruses was significantly higher in T1D subjects than in controls, and IFN-α production was correlated with pDC numbers in PBMCs. Of interest, only T1D-associated Coxsackievirus serotype B4 but not B3 induced majority of T1D PBMCs to produce IFN-α, which was confirmed to be secreted by pDCs. Finally, in vitro studies demonstrated IFN-α produced by pDCs augmented Th1 responses, with significantly greater IFN-γ-producing CD4(+) T cells from T1D subjects. These findings indicate that increased pDCs and their IFN-αß production may be associated with this Th1-mediated autoimmune disease, especially under certain viral infections linked to T1D pathogenesis.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/patología , Diabetes Mellitus Tipo 1/inmunología , Interferón-alfa/biosíntesis , Células TH1/inmunología , Regulación hacia Arriba/inmunología , Adolescente , Adulto , Recuento de Células Sanguíneas , Diferenciación Celular/inmunología , Células Cultivadas , Niño , Preescolar , Células Dendríticas/virología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/virología , Femenino , Humanos , Lactante , Interferón-alfa/fisiología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , Leucocitos Mononucleares/virología , Masculino , Orthomyxoviridae/inmunología , Células TH1/patología , Células TH1/virología , Adulto Joven
5.
Clin Immunol ; 160(1): 90-102, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25842187

RESUMEN

We developed a novel poly(lactic-co-glycolic acid)-based, microparticle (MP) system providing concurrent delivery of multiple encapsulated immuno-suppressive factors and antigen, for in vivo conditioning of dendritic cells (DCs) toward a tolerance promoting pathway. Subcutaneous administration prevents onset of type 1 diabetes (T1D) in NOD mice. Two MP sizes were made: phagocytosable MPs were fabricated encapsulating vitamin D3 or insulin B(9-23) peptide, while unphagocytosable MPs were fabricated encapsulating TGF-ß1 or GM-CSF. The combination of Vit D3/TGF-ß1 MPs confers an immature and LPS activation-resistant phenotype to DCs, and MP-delivered antigen is efficiently and functionally presented. Notably, two subcutaneous injections into 4week old NOD mice using the combination of MPs encapsulating Vit D3, Ins B, TGF-ß1 and GM-CSF protected 40% of mice from T1D development, significant in comparison to the control. This work represents one of the first applications of a biomaterial-based, MP vaccine system to successfully prevent autoimmune diabetes.


Asunto(s)
Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Portadores de Fármacos , Ácido Láctico , Ácido Poliglicólico , Vacunas/administración & dosificación , Animales , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Colecalciferol/farmacología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Tolerancia Inmunológica/inmunología , Insulina/farmacología , Lipopolisacáridos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Tamaño de la Partícula , Fragmentos de Péptidos/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Factor de Crecimiento Transformador beta1/farmacología , Vacunas/inmunología
6.
Biochem Biophys Res Commun ; 468(1-2): 46-52, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26545782

RESUMEN

It remains a top research priority to develop immunotherapeutic approaches to induce potent antigen-specific immune responses against tumors. However, in spite of some promising results, most strategies are ineffective because they generate low numbers of tumor-reactive cytotoxic T lymphocytes (CTLs). Here we designed a strategy to enhance antigen-specific immune response via administering sulfosuccinimidyl-4-[N-maleimidomethyl] cyclohexane-1-carboxylate (sulfo-SMCC)-conjugated melanoma tumor antigen GP10025-33 peptide-coupled syngeneic spleen cells in a mouse model of melanoma. We found that infusion of GP10025-33 peptide-coupled spleen cells significantly attenuated the growth of melanoma in prophylactic and therapeutic immunizations. Consistent with these findings, the adoptive transfer of spleen cells from immunized mice to naïve syngeneic mice was able to transfer anti-tumor effect, suggesting that GP10025-33 peptide-specific immune response was induced. Further studies showed that, CD8+ T cell proliferation and the frequency of interferon (IFN)-γ-producing CD8+ T cells upon ex vivo stimulation by GP10025-33 were significantly increased compared to control groups. Tumor antigen, GP10025-23 specific immune response was also confirmed by ELISpot and GP100-tetramer assays. This approach is simple, easy-handled, and efficiently delivering antigens to lymphoid tissues. Our study offers an opportunity for clinically translating this approach into tumor immunotherapy.


Asunto(s)
Inmunoconjugados/uso terapéutico , Inmunoterapia/métodos , Maleimidas/uso terapéutico , Melanoma/inmunología , Melanoma/prevención & control , Bazo/citología , Antígeno gp100 del Melanoma/uso terapéutico , Traslado Adoptivo/métodos , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Interferón gamma/inmunología , Maleimidas/administración & dosificación , Maleimidas/química , Melanoma/terapia , Ratones , Ratones Endogámicos C57BL , Péptidos/administración & dosificación , Péptidos/química , Péptidos/uso terapéutico , Bazo/inmunología , Antígeno gp100 del Melanoma/administración & dosificación , Antígeno gp100 del Melanoma/química
7.
Curr Diab Rep ; 15(11): 96, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26385483

RESUMEN

Recent evidence has highlighted the role of the innate immune system in type 1 diabetes (T1D) pathogenesis. Specifically, aberrant activation of the interferon response prior to seroconversion of T1D-associated autoantibodies supports a role for the interferon response as a precipitating event toward activation of autoimmunity. Melanoma differentiation-associated protein 5 (MDA5), encoded by IFIH1, mediates the innate immune system's interferon response to certain viral species that form double-stranded RNA (dsRNA), the MDA5 ligand, during their life cycle. Extensive research has associated single nucleotide polymorphisms (SNPs) within the coding region of IFIH1 with T1D. This review discusses the different risk and protective IFIH1 alleles in the context of recent structural and functional analysis that relate to MDA5 regulation of interferon responses. These studies have provided a functional hypothesis for IFIH1 T1D-associated SNPs' effects on MDA5-mediated interferon responses as well as supporting the genome-wide association (GWA) studies that first associated IFIH1 with T1D.


Asunto(s)
ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Diabetes Mellitus Tipo 1/genética , Polimorfismo de Nucleótido Simple , Animales , Diabetes Mellitus Tipo 1/inmunología , Predisposición Genética a la Enfermedad , Humanos , Helicasa Inducida por Interferón IFIH1 , Transducción de Señal
8.
Biochem Biophys Res Commun ; 446(4): 1035-41, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24661874

RESUMEN

Haploidentical hematopoietic stem cell transplantation (Haplo-HSCT) has been employed worldwide in recent years and led to favorable outcome in a group of patients who do not have human leukocyte antigen (HLA)-matched donors. However, the high incidence of severe graft-versus-host disease (GVHD) is a major problem for Haplo-HSCT. In the current study, we performed a proof of concept mouse study to test whether induction of allogeneic tolerance between two different parental strains was able to attenuate GVHD in Haplo-HSCT to the F1 mice. We induced alloantigen tolerance in C3H mice (H-2k) using ultraviolet B (UVB) irradiated immature dendritic cells (iDCs) derived from the cultures of Balb/c bone marrow cells. Then, we performed Haplo-HSCT using tolerant C3H mice as donors to F1 mice (C3H×Balb/c). The results demonstrated that this approach markedly reduced GVHD-associated death and significantly prolonged the survival of recipient mice in contrast to the groups with donors (C3H mice) that received infusion of non-UVB-irradiated DCs. Further studies showed that there were enhanced Tregs in the tolerant mice and alloantigen-specific T cell response was skewed to more IL-10-producing T cells, suggesting that these regulatory T cells might have contributed to the attenuation of GVHD. This study suggests that it is a feasible approach to preventing GVHD in Haplo-HSCT in children by pre-induction of alloantigen tolerance between the two parents. This concept may also lead to more opportunities in cell-based immunotherapy for GVHD post Haplo-HSCT.


Asunto(s)
Células Dendríticas/efectos de la radiación , Células Dendríticas/trasplante , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/métodos , Animales , Células Cultivadas , Células Dendríticas/inmunología , Enfermedad Injerto contra Huésped/inmunología , Humanos , Tolerancia Inmunológica , Interleucina-10/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Linfocitos T Reguladores/inmunología , Rayos Ultravioleta
9.
BMC Immunol ; 13: 70, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23237483

RESUMEN

BACKGROUND: ATG has been employed to deplete T cells in several immune-mediated conditions. However, whether ATG administration affects naïve and memory T cell differently is largely unknown. THE CONTEXT AND PURPOSE OF THE STUDY: In this study, we assessed how murine ATG therapy affected T cell subsets in NOD mice, based on their regulatory and naïve or memory phenotype, as well as its influence on antigen-specific immune responses. RESULTS: Peripheral blood CD4+ and CD8+ T cells post-ATG therapy declined to their lowest levels at day 3, while CD4+ T cells returned to normal levels more rapidly than CD8+ T cells. ATG therapy failed to eliminate antigen-primed T cells. CD4+ T cell responses post-ATG therapy skewed to T helper type 2 (Th2) and possibly IL-10-producing T regulatory type 1 (Tr1) cells. Intriguingly, Foxp3+ regulatory T cells (Tregs) were less sensitive to ATG depletion and remained at higher levels following in vivo recovery compared to controls. Of note, the frequency of Foxp3+ Tregs with memory T cell phenotype was significantly increased in ATG-treated animals. CONCLUSION: ATG therapy may modulate antigen-specific immune responses through inducing memory-like regulatory T cells as well as other protective T cells such as Th2 and IL-10-producing Tr1 cells.


Asunto(s)
Suero Antilinfocítico/farmacología , Memoria Inmunológica/efectos de los fármacos , Depleción Linfocítica , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos/farmacología , Suero Antilinfocítico/administración & dosificación , Autoantígenos/inmunología , Complejo CD3/inmunología , Proliferación Celular/efectos de los fármacos , Reactividad Cruzada/efectos de los fármacos , Reactividad Cruzada/inmunología , Femenino , Factores de Transcripción Forkhead/metabolismo , Inmunoglobulina G/inmunología , Interleucina-10/biosíntesis , Selectina L/metabolismo , Recuento de Linfocitos , Tejido Linfoide/efectos de los fármacos , Tejido Linfoide/inmunología , Ratones , Ratones Endogámicos NOD , Receptores de Antígenos de Linfocitos T/metabolismo , Bazo/efectos de los fármacos , Bazo/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th2/efectos de los fármacos , Células Th2/inmunología
10.
Diabetes Metab Res Rev ; 27(8): 809-12, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22069264

RESUMEN

BACKGROUND: Thioredoxin as a biological antioxidant plays an important role in regulating the redox system. The administration of recombinant thioredoxin has been demonstrated to be anti-inflammatory. In this study, the effect of recombinant human thioredoxin-1 (rhTrx-1) in preventing type 1 diabetes (T1D) in nonobese diabetic (NOD) mice was evaluated. METHODS: Eight-week-old NOD mice were treated with intravenous injection of rhTrx-1 (5 µg/mouse/day) for 5 weeks (5 days a week), followed by every other day for additional 5 weeks. Diabetes onset was monitored twice a week. Pancreatic histology and ß-cell mass were examined by hematoxylin and eosin (H&E) and insulin immunohistochemistry staining, respectively. Adoptive transfer experiments were executed to assess autoimmune T cells modulated by rhTrx treatment. RESULTS: The intravenous administration of rhTrx-1 significantly delayed and prevented T1D in NOD mice. The histology data showed that rhTrx-1 treatment markedly reduced insulitic lesions and significantly preserved insulin-producing ß cells. Adoptive transfer of spleen cells from rhTrx-1-treated mice into nonobese diabetic-severe combined immunodeficiency (NOD-SCID) mice significantly reduced the diabetes onset than transfer of those from phosphate-buffered saline-treated mice. Adoptive co-transfer experiments demonstrated that spleen cells from rhTrx-1-treated mice significantly delayed diabetes induced by the co-transferred diabetogenic spleen cells from the new-onset diabetic mice. CONCLUSIONS: Antioxidant rhTrx-1 effectively prevents T1D which may be attributed to its activity to modulate autoimmunity.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Tiorredoxinas/farmacología , Traslado Adoptivo , Animales , Autoinmunidad/efectos de los fármacos , Femenino , Humanos , Células Secretoras de Insulina/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Bazo/inmunología , Bazo/trasplante
11.
Clin Immunol ; 135(3): 374-83, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20223712

RESUMEN

In our previous studies, we demonstrated that infusion of apoptotic cells significantly prevented type 1 diabetes (T1D) in non-obese diabetic (NOD) mice. Extracorporeal photopheresis (ECP) is an apoptotic cell-based therapy used clinically for immune-mediated disorders. In this study, we examined the effect that intravenous delivery of apoptotic cells (ECP-treated) has in the prevention of T1D in NOD mice. We discovered that five weekly injections of ECP-treated NOD spleen cells, beginning at 8 weeks of age, significantly delayed diabetes onset. Furthermore, cell dose studies demonstrated that low dose ECP-treated spleen cells (2x10(5) cells/injection/mouse) had similar protective effects as compared to high dose (5x10(6) cells/injection). In contrast to ECP-treated cells alone, ECP-treated cells combined with beta cell antigens appeared to improve the protective effect as shown by the marked reduction in insulitis in the islets. Delivery of ECP-treated spleen cells or ECP-treated spleen cells plus beta cell antigen increased Foxp3(+) Tregs, and beta cell antigen-specific T cell proliferation was significantly suppressed in vivo in these two groups. In addition, we found that ECP-treated cells did not induce global immunosuppression or autoimmunity against nuclear antigens. In conclusion, ECP-treated cells provide a safe and effective approach in T1D prevention, suggesting that clinical ECP has great potential for managing human T1D.


Asunto(s)
Células Sanguíneas/trasplante , Diabetes Mellitus Tipo 1/terapia , Fotoféresis/métodos , Animales , Apoptosis/inmunología , Células Sanguíneas/inmunología , Separación Celular , Diabetes Mellitus Tipo 1/inmunología , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos NOD , Linfocitos T Reguladores/inmunología
12.
Curr Opin Organ Transplant ; 14(4): 338-43, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19444106

RESUMEN

PURPOSE OF REVIEW: This review is intended to introduce recent advances in the research surrounding extracorporeal photopheresis (ECP) with a focus on how apoptotic cells modulate antigen-presenting cells and induce regulatory T cells, given that ECP therapy induces apoptosis of leukocytes collected through leukapheresis. RECENT FINDINGS: It has been suggested that ECP therapy, unlike other immunosuppressive regimens, does not cause global immunosuppression, but induces immune tolerance. Recent clinical and animal studies demonstrate that ECP therapy induces antigen-specific regulatory T cells, including CD4+CD25+FoxP3+ T cells and IL-10-producing Tr1 cells, that may arise secondarily to the induction of tolerogenic antigen-presenting cells (APCs) by infusion of apoptotic cells. It has also been suggested that ECP therapy may induce IL-10-producing regulatory B cells and regulatory CD8+ T cells. Finally, several recent studies, which examined the cellular elements involved in the uptake of apoptotic cells, demonstrated that apoptotic cells modulate APCs through binding to specific receptors, particularly TAM receptors that provide inhibitory signals that block APC activation. SUMMARY: ECP therapy induces immune tolerance through modulation of antigen-presenting cells as well as induction of regulatory T cells. ECP therapy has great potential in the management of allogeneic transplantation and autoimmune diseases.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Apoptosis , Terapia de Inmunosupresión/métodos , Trasplante de Órganos , Fotoféresis , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante , Animales , Diferenciación Celular , Humanos , Interleucina-10/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal
13.
J Immunol Res ; 2018: 5463879, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29651443

RESUMEN

Dendritic cell (DC) immunotherapy has been effective for prevention of type 1 diabetes (T1D) in NOD mice but fails to protect if initiated after active autoimmunity. As autoreactivity expands inter- and intramolecularly during disease progression, we investigated whether DCs unpulsed or pulsed with ß cell antigenic dominant determinants (DD), subdominant determinants (SD), and ignored determinants (ID) could prevent T1D in mice with advanced insulitis. We found that diabetes was significantly delayed by DC therapy. Of interest, DCs pulsed with SD or ID appeared to provide better protection. T lymphocytes from DC-treated mice acquired spontaneous proliferating capability during in vitro culture, which could be largely eliminated by IL-2 neutralizing antibodies. This trend maintained even 29 weeks after discontinuing DC therapy and appeared antigen-independent. Furthermore, CD4+Foxp3+ T regulatory cells (Tregs) from DC-treated mice proliferated more actively in vitro compared to the controls, and Tregs from DC-treated mice showed significantly enhanced immunosuppressive activities in contrast to those from the controls. Our study demonstrates that DC therapy leads to long-lasting immunomodulatory effects in an antigen-dependent and antigen-independent manner and provides evidence for peptide-based intervention during a clinically relevant window to guide DC-based immunotherapy for autoimmune diabetes.


Asunto(s)
Células Dendríticas/fisiología , Diabetes Mellitus Tipo 1/terapia , Inmunoterapia Adoptiva/métodos , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Presentación de Antígeno , Autoantígenos/inmunología , Autoantígenos/metabolismo , Autoinmunidad , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Tolerancia Inmunológica , Epítopos Inmunodominantes/inmunología , Epítopos Inmunodominantes/metabolismo , Inmunomodulación , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD
14.
EBioMedicine ; 22: 10-17, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28663145

RESUMEN

Type 1 diabetes (T1D) has been associated with both genetic and environmental factors. Increasing incidence of T1D worldwide is prompting researchers to adopt different approaches to explain the biology of T1D, beyond the presence and activity of autoreactive lymphocytes. In this review, we propose inflammatory pathways as triggers for T1D. Within the scope of those inflammatory pathways and in understanding the pathogenesis of disease, we suggest that viruses, in particular Coxsackieviruses, act by causing a type 1 interferonopathy within the pancreas and the microenvironment of the islet. As such, this connection and common thread represents an exciting platform for the development of new diagnostic, treatment and/or prevention options.


Asunto(s)
Infecciones por Coxsackievirus/inmunología , Diabetes Mellitus Tipo 1/inmunología , Interferones/metabolismo , Enfermedades Pancreáticas/virología , Animales , Microambiente Celular , Inmunidad Innata , Islotes Pancreáticos/inmunología , Enfermedades Pancreáticas/inmunología , Transducción de Señal
15.
Ann N Y Acad Sci ; 1079: 153-6, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17130547

RESUMEN

Our previous work demonstrated peptide-pulsed mature myeloid dendritic cells (DC) presenting beta cell antigens induce tolerance. Here we determine whether immature DC (iDC) presenting dominant (insulin beta9-23 chain, proinsulin C19-A3) or ignored (glutamic acid decarboxylase 65(78-97)) antigen determinants promote tolerance. Nonobese diabetic (NOD) mice were given injections of either unpulsed or peptide-pulsed myeloid iDC beginning at 9 weeks of age for 3 consecutive weeks. Diabetes incidence in recipients of unpulsed iDC was comparable to unmanipulated animals ( approximately 80%), whereas GAD65(78-97) pulsed iDC recipients were protected from the disease (P = 0.05). We also analyzed splenic T cell proliferation responses to the panel of studied peptides in diabetic and nondiabetic recipients. When stimulated with insulin or proinsulin peptide, nondiabetic mice receiving the peptide-pulsed iDC had a 21- to 31-fold or 3.9- to 9.0-fold reduction in T cell response, respectively, as compared to the response of diabetic unpulsed recipients. However, only a 2.6- to 3.1-fold reduction in response to beta chain peptide, and a 1.5- to 3.4-fold reduction in proinsulin response were observed in diabetic mice receiving peptide-pulsed iDC. The reduction was not specific to the immunizing peptide, as reduced proliferation was observed to other diabetes-target peptides. We conclude that protective iDC-based therapies require target antigen presentation, and ignored determinants may be preferable perhaps due to an available naïve T cell repertoire. In addition, iDC presenting peptides induce a nonspecific reduction in T cell responses to beta cell antigens, possibly through the induction of regulatory T cells.


Asunto(s)
Traslado Adoptivo , Antígenos/inmunología , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Islotes Pancreáticos/inmunología , Animales , División Celular/efectos de los fármacos , División Celular/inmunología , Células Cultivadas , Células Dendríticas/trasplante , Femenino , Glutamato Descarboxilasa/inmunología , Tolerancia Inmunológica , Insulina/farmacología , Islotes Pancreáticos/patología , Ratones , Ratones Endogámicos NOD , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/farmacología , Bazo/citología , Bazo/metabolismo , Linfocitos T/inmunología , Linfocitos T/fisiología
17.
J Immunol Res ; 2016: 2419621, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27556047

RESUMEN

Our previous study demonstrated that transfusion of ultraviolet B-irradiated immature dendritic cells (UVB-iDCs) induced alloantigen-specific tolerance between two different strains of mice. Programmed death-1 (PD-1) and programmed death ligand-1 (PD-L1) have been suggested to play an important role in maintaining immune tolerance. In the present study, we seek to address whether PD-1/PD-L1 plays a role in the maintenance of UVB-iDC-induced tolerance. We first observe that the UVB-iDC-induced alloantigen-specific tolerance can be maintained for over 6 weeks. Supporting this, at 6 weeks after tolerance induction completion, alloantigen-specific tolerance is still able to be transferred to syngeneic naïve mice through adoptive transfer of CD4+ T cells. Furthermore, skin transplantation study shows that the survival of allogeneic grafts is prolonged in those tolerant recipients. Further studies show that PD-1/PD-L1 interaction is essential for maintaining the induced tolerance as blockade of PD-1/PD-L1 by anti-PD-L1 antibodies largely breaks the tolerance at both cellular and humoral immunological levels. Importantly, we show that PD-1/PD-L1 interaction in tolerant mice is also essential for controlling alloantigen-responding T cells, which have never experienced alloantigens. The above findings suggest that PD-1/PD-L1 plays a crucial role in maintaining immune tolerance induced by UVB-iDCs, as well as in actively controlling effector T cells specific to alloantigens.


Asunto(s)
Antígeno B7-H1/metabolismo , Tolerancia Inmunológica , Isoantígenos/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Rayos Ultravioleta , Traslado Adoptivo , Animales , Supervivencia de Injerto/inmunología , Tolerancia Inmunológica/efectos de la radiación , Activación de Linfocitos/inmunología , Ratones , Unión Proteica , Trasplante de Piel , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
18.
J Leukoc Biol ; 99(6): 1121-9, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26657791

RESUMEN

Myeloid-derived suppressor cells with immunosuppressive functions have been described to be associated with one of the mechanisms by which malignant tumors escape immune surveillance. However, little is known about the role of myeloid-derived suppressor cells in autoimmunity. In the current study, when we attempted to characterize the peritoneal cells in pristane-induced lupus model, as reported previously, we observed that there were markedly increased CD11b(+)Ly6C(hi) monocytes. Surprisingly, this type of monocytes was almost phenotypically identical to the reported monocytic myeloid-derived suppressor cells. Further analysis on how these CD11b(+)Ly6C(hi) cells affected T cell response showed that they strongly suppressed T cell proliferation in vitro in a manner dependent on cell-cell contact, NO, and PGE2. In addition, we found that CD11b(+)Ly6C(hi) monocytes inhibited Th1 differentiation but enhanced development of forkhead box p3(+)CD4(+) regulatory T cells. Consistent with the in vitro experimental results, the in vivo adoptive cell transfer study showed that infusion of pristane-treated syngeneic CD11b(+)Ly6C(hi) monocytes significantly suppressed the production of anti-keyhole limpet hemocyanin antibodies induced by keyhole limpet hemocyanin immunization. In addition, we found that CD11b(+)Ly6C(hi) monocytes were also increased significantly in spleen and peripheral blood and showed immunosuppressive characteristics similar to their peritoneal counterparts. Our findings indicate that CD11b(+)Ly6C(hi) monocytes in a pristane-induced lupus mouse model are monocytic myeloid-derived suppressor cells instead of inflammatory monocytes, as demonstrated previously. To our knowledge, this is the first to describe myeloid-derived suppressor cells in a pristane-induced lupus mouse model, which may lead to a better understanding of the role of CD11b(+)Ly6C(hi) monocytes in this specific pristane-induced lupus model.


Asunto(s)
Antígenos Ly/metabolismo , Antígeno CD11b/metabolismo , Terapia de Inmunosupresión , Lupus Eritematoso Sistémico/inducido químicamente , Lupus Eritematoso Sistémico/inmunología , Monocitos/patología , Traslado Adoptivo , Animales , Anticuerpos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Comunicación Celular , Proliferación Celular , Dinoprostona/biosíntesis , Modelos Animales de Enfermedad , Femenino , Hemocianinas/inmunología , Inmunización , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/patología , Ratones Endogámicos BALB C , Monocitos/metabolismo , Óxido Nítrico/biosíntesis , Bazo/patología , Terpenos
19.
Int Immunopharmacol ; 31: 158-68, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26735611

RESUMEN

In the present study, we report our recently developed new approach to inducing antigen-specific immune response. We use two nucleophilic substitution "click" chemistry processes to successfully couple protein antigens or peptides to mouse spleen cells or T cells by a heterobifunctional crosslinker, succinimidyl-4-(N-maleimidomethyl cyclohexane)-1-carboxylate (SMCC) or sulfo-SMCC. SMCC and its water-soluble analog sulfo-SMCC contain N-hydroxysuccinimide (NHS) ester and maleimide groups, which allow stable covalent conjugation of amine- and sulfhydryl-containing molecules in trans. Protein coupling to cells relies on the free sulfhydryls (thiols) on cell surfaces and the free amines on protein antigens. Although the amount of protein coupled to cells is limited due to the limited number of cell surface thiols, the injection of spleen cells coupled with antigenic proteins, such as keyhole limpet hemocyanin (KLH) or ovalbumin (OVA), induces a potent antigen-specific immune response in vivo, which is even stronger than that induced by the injection of a large dose of protein plus adjuvants. In addition, short peptides coupled to purified splenic T cells also potently elicit peptide-specific T cell proliferation in vivo after injection. Further studies show that antigen-coupled spleen cell treatment leads to augmented IFN-γ-producing T cells. Our study provides a unique antigen delivery method that efficiently distributes antigen to the entire immune system, subsequently eliciting a potent antigen-specific immune response with enhanced IFN-γ production. The findings in the present study suggest that this antigen-cell coupling strategy could be employed in immunotherapy for cancers, infectious diseases as well as immune-mediated disorders.


Asunto(s)
Antígenos/inmunología , Trasplante de Células , Reactivos de Enlaces Cruzados/química , Inmunoterapia/métodos , Maleimidas/química , Bazo/citología , Linfocitos T/inmunología , Animales , Antígenos/química , Células Cultivadas , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Bazo/química
20.
J Immunol Res ; 2015: 129682, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26258148

RESUMEN

In this study, we have evaluated our recently developed method for antigen-cell coupling using sulfosuccinimidyl-4-[N-maleimidomethyl]cyclohexane-1-carboxylate (sulfo-SMCC) heterobifunctional crosslinker in prevention and reversal of experimental autoimmune encephalomyelitis (EAE). We demonstrate that infusion of MOG35-55-coupled spleen cells (MOG-SP) significantly prevents and reverses EAE. Further studies show that the protected animals exhibit significantly delayed EAE upon EAE reinduction. Moreover, adoptive transfer of CD4+ T cells from the protected mice to naïve syngeneic mice renders the recipient mice resistant to EAE induction. Unexpectedly, CD4+ T cell proliferation is similar upon ex vivo stimulation by MOG35-55 amongst all groups. However, further analysis of those proliferating CD4+ T cells shows remarkable differences in Foxp3+ regulatory T cells (70% in MOG-SP groups versus 10-25% in control groups) and in IL-17+ cells (2-3% in MOG-SP groups versus 6-9% in control groups). In addition, we discover that MOG-SP treatment also significantly attenuates MOG35-55-responding IFN-γ-producing Th1 cells. These findings suggest that MOG-SP treatment induces EAE protective MOG35-55-specific regulatory T cells and suppresses EAE pathogenic Th17 and Th1 cells. Our study provides a novel approach for antigen-based EAE immunotherapy, which can potentially be translated into clinical application for immunotherapy of multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Inmunoconjugados/administración & dosificación , Maleimidas , Glicoproteína Mielina-Oligodendrócito , Bazo/citología , Bazo/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Encefalomielitis Autoinmune Experimental/diagnóstico , Encefalomielitis Autoinmune Experimental/prevención & control , Epítopos de Linfocito T/inmunología , Femenino , Inmunización , Activación de Linfocitos , Ratones , Fragmentos de Péptidos , Índice de Severidad de la Enfermedad , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA