Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Kidney Int ; 104(1): 28-31, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37349057

RESUMEN

CD4+ T cells that express forkhead box protein 3 are important in maintaining tolerance and restraining effector responses. Herrnstadt et al. use a model of rapidly progressive glomerulonephritis to examine the nature and role of forkhead box protein 3-positive and retinoid acid-related orphan receptor γt-positive regulatory T cells. These cells are prominent in experimental glomerulonephritis, both locally and systemically, and are present in kidneys of people with anti-neutrophil cytoplasmic antibody-associated vasculitis. Functionally, despite their expression of retinoid acid-related orphan receptor γt, associated with T-helper cell 17 cells, they regulate cellular immunity, both systemically and within the kidney.


Asunto(s)
Glomerulonefritis , Humanos , Riñón/metabolismo , Linfocitos T Reguladores , Factores de Transcripción Forkhead/metabolismo , Retinoides
2.
Vet Res ; 53(1): 61, 2022 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-35906635

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) is a prevalent and endemic swine pathogen which causes significant economic losses in the global swine industry. Multiple vaccines have been developed to prevent PRRSV infection. However, they provide limited protection. Moreover, no effective therapeutic drugs are yet available. Therefore, there is an urgent need to develop novel antiviral strategies to prevent PRRSV infection and transmission. Here we report that Toosendanin (TSN), a tetracyclic triterpene found in the bark or fruits of Melia toosendan Sieb. et Zucc., strongly suppressed type 2 PRRSV replication in vitro in Marc-145 cells and ex vivo in primary porcine alveolar macrophages (PAMs) at sub-micromolar concentrations. The results of transcriptomics revealed that TSN up-regulated the expression of IFI16 in Marc-145 cells. Furthermore, we found that IFI16 silencing enhanced the replication of PRRSV in Marc-145 cells and that the anti-PRRSV activity of TSN was dampened by IFI16 silencing, suggesting that the inhibition of TSN against PRRSV replication is IFI16-dependent. In addition, we showed that TSN activated caspase-1 and induced maturation of IL-1ß in an IFI16-dependent pathway. To verify the role of IL-1ß in PRRSV infection, we analyzed the effect of exogenous rmIL-1ß on PRRSV replication, and the results showed that exogenous IL-1ß significantly inhibited PRRSV replication in Marc-145 cells and PAMs in a dose-dependent manner. Altogether, our findings indicate that TSN significantly inhibits PRRSV replication at very low concentrations (EC50: 0.16-0.20 µM) and may provide opportunities for developing novel anti-PRRSV agents.


Asunto(s)
Síndrome Respiratorio y de la Reproducción Porcina , Virus del Síndrome Respiratorio y Reproductivo Porcino , Enfermedades de los Porcinos , Animales , Caspasa 1 , Línea Celular , Macrófagos Alveolares , Síndrome Respiratorio y de la Reproducción Porcina/metabolismo , Porcinos , Enfermedades de los Porcinos/metabolismo , Triterpenos , Replicación Viral
3.
Immunol Cell Biol ; 99(10): 1011-1025, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34251701

RESUMEN

Foxp3+ regulatory T cells (Tregs) represent a special lineage of CD4+ T cells. Analysis of Treg response during primary and secondary influenza virus infection clearly demonstrates a robust accumulation of Tregs into the infected lungs and the existence of a population of long-lived antigen-specific memory Tregs in the same tissues after resolution of the infection. However, it remains unknown whether these Tregs co-express Helios, a member of the Ikaros transcription factor family. In this study, Foxp3+ Helios+ and Foxp3+ Helios- Tregs in the lungs, mLNs and spleens of influenza virus-infected and uninfected control mice were tracked. The data show that while there is a co-existence of Foxp3+ Helios+ and Foxp3+ Helios- Tregs in the tissues, the accumulated Tregs in the lungs and lung-draining mediastinal lymph nodes (mLNs) of the infected mice are highly enriched for Foxp3+ Helios+ cells. It was further demonstrated that, after the clearance of primary infection, Foxp3+ Helios+ cells have the ability to persist in the tissues over their Helios- counterparts. More importantly, Foxp3+ Helios+ Tregs accumulated in an accelerated kinetics during recall response to reinfection. In vitro analysis of Treg suppressive function revealed that Foxp3+ Helios+ Tregs are more capable of suppressing influenza virus-specific CD8+ T cell activation, cytokine production and proliferation. Together, the data provide new insights into Treg responses during primary and secondary influenza virus infection and suggest that Foxp3+ Helios+ Tregs predominantly drive the Treg responses.


Asunto(s)
Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción Forkhead , Pulmón , Ratones
4.
Immunol Cell Biol ; 98(7): 563-576, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32330333

RESUMEN

The participation of dendritic cells (DCs) in CD8+ T-cell-mediated allograft rejection is a long-standing question of great clinical relevance for tissue transplantation. Here, we show that Batf3-/- mice demonstrate delayed allo-skin graft rejection and are deficient in priming allo-specific CD8+ T cells. Batf3-/- mouse priming is restored by transferring either purified CD8α+ or CD103+ DCs, demonstrating the critical role of these cells in alloreactivity. Using Db -restricted antiviral F5 transgenic T-cell receptor T cells, which we demonstrate are alloreactive with H-2Kd , we show that cross-dressing of CD8α+ and CD103+ primes CD8+ T-cell or allo-specific responses in vitro and in vivo. These findings suggest novel strategies for moderating tissue rejection based on interfering with DC cross-dressing or subsequent interaction with T cells.


Asunto(s)
Linfocitos T CD8-positivos , Células Dendríticas , Rechazo de Injerto/inmunología , Animales , Antígenos CD , Antígenos CD8 , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Cadenas alfa de Integrinas , Ratones , Ratones Endogámicos C57BL , Trasplante de Piel
5.
Immunol Cell Biol ; 97(9): 774-786, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31116465

RESUMEN

Memory regulatory T cells (mTregs) have been demonstrated to persist long-term in hosts after the resolution of primary influenza A virus (IAV) infection. However, whether such IAV infection-experienced (IAV-experienced) mTregs differentiate into a phenotypically and functionally distinct Treg subset and what function they play at the infection site remains poorly defined. In this study, we characterized the phenotype, examined the responsiveness and assessed the suppressive function of IAV-experienced memory Tregs (mTregs). In comparison with inexperienced naïve Tregs (nTregs), mTregs exhibited elevated expression of CD39, CD69, CD103, cytotoxic T lymphocyte-associated antigen-4, leukocyte function-associated antigen-1 and programmed cell death-1 and could be activated in an antigen-specific manner in vitro and in vivo. When mTregs and nTregs were adoptively cotransferred into recipient mice, mTregs had a competitive advantage in migrating to the IAV-infected lungs. mTregs were more capable of controlling in vitro proliferation of CD4+ and CD8+ T cells and suppressed CD40 and CD86 upregulation on bone marrow-derived dendritic cells. Adoptively transferred mTregs, but not adoptively transferred nTregs, significantly attenuated body weight loss, lung pathology and immune cell infiltration into the infected lungs after IAV infection. These results suggest that mTregs generated after IAV infection differentiate into a phenotypically distinct and functionally enhanced Treg subset that can be activated in an antigen-specific manner to exert immunosuppression. We propose vaccination to induce such mTregs as a potential novel strategy to protect against severe IAV infection.


Asunto(s)
Memoria Inmunológica , Virus de la Influenza A/inmunología , Pulmón/inmunología , Pulmón/virología , Infecciones por Orthomyxoviridae/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Movimiento Celular/inmunología , Proliferación Celular , Células Dendríticas/inmunología , Regulación hacia Abajo , Femenino , Pulmón/patología , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Monocitos/patología , Infiltración Neutrófila , Infecciones por Orthomyxoviridae/virología , Fenotipo , Pérdida de Peso
6.
Immunol Cell Biol ; 96(9): 981-993, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29738610

RESUMEN

The role of the immunoproteasome is perceived as confined to adaptive immune responses given its ability to produce peptides ideal for MHC Class-I binding. Here, we demonstrate that the immunoproteasome subunit, LMP2, has functions beyond its immunomodulatory role. Using LMP2-deficient mice, we demonstrate that LMP2 is crucial for lymphocyte development and survival in the periphery. Moreover, LMP2-deficient lymphocytes show impaired degradation of key BH3-only proteins, resulting in elevated levels of pro-apoptotic BIM and increased cell death. Interestingly, LMP2 is the sole immunoproteasome subunit required for BIM degradation. Together, our results suggest LMP2 has important housekeeping functions and represents a viable therapeutic target for cancer.


Asunto(s)
Proteína Proapoptótica que Interacciona Mediante Dominios BH3/inmunología , Cisteína Endopeptidasas/inmunología , Linfocitos/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Animales , Western Blotting , Línea Celular , Supervivencia Celular , Células Cultivadas , Cisteína Endopeptidasas/deficiencia , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal/deficiencia
7.
Antiviral Res ; 203: 105345, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35605699

RESUMEN

In addition to severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV-2 has become the third deadly coronavirus that infects humans and causes the new coronavirus disease (COVID-19). COVID-19 has already caused more than six million deaths worldwide and it is likely the biggest pandemic of this century faced by mankind. Although many studies on SARS-CoV-2 have been conducted, a detailed understanding of SARS-CoV-2 and COVID-19 is still lacking. Animal models are indispensable for studying its pathogenesis and developing vaccines and antivirals. In this review, we analyze animal models of coronavirus infections and explore their applications on antivirals and vaccines.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Coronavirus del Síndrome Respiratorio de Oriente Medio , Vacunas Virales , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/prevención & control , Modelos Animales , SARS-CoV-2
8.
FEBS J ; 288(10): 3164-3185, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33830641

RESUMEN

CD4+ T cells recognize peptides presented by major histocompatibility complex class II molecules (MHC-II). These peptides are generally derived from exogenous antigens. Macroautophagy has been reported to promote endogenous antigen presentation in viral infections. However, whether influenza A virus (IAV) infection-induced macroautophagy also leads to endogenous antigen presentation through MHC-II is still debated. In this study, we show that IAV infection leads to endogenous presentation of an immunodominant viral epitope NP311-325 by MHC-II to CD4+ T cells. Mechanistically, such MHC-II-restricted endogenous IAV antigen presentation requires de novo protein synthesis as it is inhibited by the protein synthesis inhibitor cycloheximide, and a functional ER-Golgi network as it is totally blocked by Brefeldin A. These results indicate that MHC-II-restricted endogenous IAV antigen presentation is dependent on de novo antigen and/or MHC-II synthesis, and transportation through the ER-Golgi network. Furthermore, such endogenous IAV antigen presentation by MHC-II is enhanced by TAP deficiency, indicating some antigenic peptides are of cytosolic origin. Most importantly, the bulk of such MHC-II-restricted endogenous IAV antigen presentation is blocked by autophagy inhibitors (3-MA and E64d) and deletion of autophagy-related genes, such as Beclin1 and Atg7. We have further demonstrated that in dendritic cells, IAV infection prevents autophagosome-lysosome fusion and promotes autophagosome fusion with MHC class II compartment (MIIC), which likely promotes endogenous IAV antigen presentation by MHC-II. Our results provide strong evidence that IAV infection-induced autophagosome formation facilitates endogenous IAV antigen presentation by MHC-II to CD4+ T cells. The implication for influenza vaccine design is discussed.


Asunto(s)
Presentación de Antígeno/genética , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Interacciones Huésped-Patógeno/genética , Subtipo H1N1 del Virus de la Influenza A/genética , Macroautofagia/genética , Animales , Antígenos Virales/química , Antígenos Virales/genética , Antígenos Virales/inmunología , Proteína 7 Relacionada con la Autofagia/deficiencia , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/inmunología , Beclina-1/deficiencia , Beclina-1/genética , Beclina-1/inmunología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/virología , Brefeldino A/farmacología , Linfocitos T CD4-Positivos/virología , Células Dendríticas/virología , Femenino , Expresión Génica , Células HEK293 , Antígenos de Histocompatibilidad Clase II/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Epítopos Inmunodominantes/química , Epítopos Inmunodominantes/genética , Epítopos Inmunodominantes/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Macroautofagia/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Plásmidos/química , Plásmidos/metabolismo , Transfección
9.
Expert Rev Respir Med ; 14(11): 1141-1147, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32762572

RESUMEN

INTRODUCTION: The innate immune response is the first line of defense and consists of physical, chemical and cellular defenses. The adaptive immune response is the second line of defense and is pathogen-specific. Innate immunity occurs immediately while adaptive immunity develops upon pathogen exposure, and is long-lasting, highly specific, and sustained by memory T cells. Respiratory virus infection typically induces effective immunity but over-exuberant responses are associated with pathophysiology. Cytokines expressed in response to viral infection can enhance biological responses, activate, and trigger signaling pathways leading to adaptive immunity Vaccines induce immunity, specifically B and T cell responses. Vaccination is generally efficacious, but for many viruses, our understanding of vaccination strategies and immunity is incomplete or in its infancy. Studies that examine innate and adaptive immune responses to respiratory virus infection will aid vaccine development and may reduce the burden of respiratory viral disease. AREAS COVERED: A literature search was performed using PubMed. The search covered: innate, adaptive, respiratory virus, vaccine development, B cell, and T cell. EXPERT OPINION: Immunity rests on two pillars, i.e. the innate and adaptive immune system, which function together on different tasks to maintain homeostasis. a better understanding of immunity is necessary for disease prevention and intervention.


Asunto(s)
Inmunidad Adaptativa/fisiología , Inmunidad Innata/fisiología , Infecciones del Sistema Respiratorio/inmunología , Virosis/inmunología , Animales , Humanos , Infecciones del Sistema Respiratorio/diagnóstico , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/terapia , Vacunación/métodos , Vacunación/tendencias , Vacunas Virales/uso terapéutico , Virosis/diagnóstico , Virosis/epidemiología , Virosis/terapia , Virus/inmunología
10.
Oncotarget ; 6(40): 42541-56, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26637810

RESUMEN

Fatal influenza outcomes result from a combination of rapid virus replication and collateral lung tissue damage caused by exaggerated pro-inflammatory host immune cell responses. There are few therapeutic agents that target both biological processes for the attenuation of influenza-induced lung pathology. We show that Saikosaponin A, a bioactive triterpene saponin with previouslyestablished anti-inflammatory effects, demonstrates both in vitro and in vivo anti-viral activity against influenza A virus infections. Saikosaponin A attenuated the replication of three different influenza A virus strains, including a highly pathogenic H5N1 strain, in human alveolar epithelial A549 cells. This anti-viral activity occurred through both downregulation of NF-κB signaling and caspase 3-dependent virus ribonucleoprotein nuclear export as demonstrated by NF-κB subunit p65 and influenza virus nucleoprotein nuclear translocation studies in influenza virus infected A549 cells. Critically, Saikosaponin A also attenuated viral replication, aberrant pro-inflammatory cytokine production and lung histopathology in the widely established H1N1 PR8 model of influenza A virus lethality in C57BL/6 mice. Flow cytometry studies of mouse bronchoalveolar lavage cells revealed that SSa exerted immunomodulatory effects through a selective attenuation of lung neutrophil and monocyte recruitment during the early peak of the innate immune response to PR8 infection. Altogether, our results indicate that Saikosaponin A possesses novel therapeutic potential for the treatment of pathological influenza virus infections.


Asunto(s)
Antivirales/farmacología , Pulmón/patología , Ácido Oleanólico/análogos & derivados , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones del Sistema Respiratorio/patología , Saponinas/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Virus de la Influenza A , Gripe Humana/patología , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ácido Oleanólico/farmacología , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/virología , Replicación Viral/efectos de los fármacos
11.
Int Immunopharmacol ; 14(1): 121-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22728095

RESUMEN

Saikosaponin a (SSa) and its epimer saikosaponin d (SSd) are major triterpenoid saponin derivatives from Radix bupleuri (RB), which has been long used in Chinese traditional medicine for treatment of various inflammation-related diseases. In the present study, the anti-inflammatory activity, as well as the underlying mechanism, of SSa and SSd was investigated in lipopolysaccharide (LPS)-induced RAW264.7 cells. Our results demonstrated that both SSa and SSd significantly inhibited the expression of inducible nitric-oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in LPS-induced RAW264.7 cells, and finally resulted in the reduction of nitric oxide (NO) and prostaglandin E(2) (PGE(2)). In addition, LPS-induced production of major pro-inflammatory cytokines: the tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), was suppressed in a dose-dependent manner by the treatment of SSa or SSd in RAW264.7 cells. Further analysis revealed that both SSa and SSd could inhibit translocation of nuclear factor-κB (NF-κB) from the cytoplasm to the nucleus in the LPS-induced RAW264.7 cells. Furthermore, SSa and SSd exhibited significant anti-inflammatory activity in two different murine models of acute inflammation, carrageenan-induced paw edema in rats and acetic acid-induced vascular permeability in mice. In conclusion, SSa and SSd showed potent anti-inflammatory activity through inhibitory effects on NF-κB activation and thereby on iNOS, COX-2 and pro-inflammatory cytokines.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Bupleurum/inmunología , Macrófagos/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Ácido Oleanólico/análogos & derivados , Saponinas/farmacología , Animales , Antiinflamatorios no Esteroideos/química , Permeabilidad Capilar/efectos de los fármacos , Línea Celular , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Terapia de Inmunosupresión , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Masculino , Medicina Tradicional China , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ácido Oleanólico/química , Ácido Oleanólico/farmacología , Saponinas/química , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Estereoisomerismo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/inmunología
12.
J Agric Food Chem ; 59(8): 3730-7, 2011 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-21401102

RESUMEN

This study was conducted to investigate the effects of apple polyphenol extract (APE) against influenza virus in mice loaded with restraint stress. The high-performance liquid chromatography (HPLC) fingerprint of APE was recorded, and the percentage composition of polyphenols was determined as 81.7%. Our results showed that restraint stress significantly promoted the mortality and duration of complications of mice infected with the H1N1 virus. However, oral administration of APE (100 and 200 mg/kg) improved the survival rates and prolonged living time of stressed mice infected with influenza virus in a dose-dependent manner. APE was further found to significantly improve the number of immunocytes, ratio of CD4 helper cells, secretion of IL-2, and capabilities of natural killer (NK) cytotoxicity (LU10/spleen) in spleens of restraint-stressed mice. In addition, APE also significantly decreased the level of lipid peroxidation and increased oxygen radical absorbance capacity (ORAC) in splenocytes. These results indicated that the protective effects of APE on mice infected with influenza virus were related to the alleviation of stress-induced impairment of immune functions and its antioxidant property might contribute to the immune recovery.


Asunto(s)
Flavonoides/farmacología , Inmovilización , Malus/química , Infecciones por Orthomyxoviridae/prevención & control , Fenoles/farmacología , Animales , Flavonoides/aislamiento & purificación , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Fenoles/aislamiento & purificación , Polifenoles , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA