Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 8: 889, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28824622

RESUMEN

Salmonella enterica serovar Typhimurium (S. Typhimurium) is a Gram-negative bacterium that produces disease in numerous hosts. In mice, oral inoculation is followed by intestinal colonization and subsequent systemic dissemination, which leads to severe pathogenesis without the activation of an efficient anti-Salmonella immune response. This feature suggests that the infection caused by S. Typhimurium may promote the production of anti-inflammatory molecules by the host that prevent efficient T cell activation and bacterial clearance. In this study, we describe the contribution of immune cells producing the anti-inflammatory cytokine interleukin-10 (IL-10) to the systemic infection caused by S. Typhimurium in mice. We observed that the production of IL-10 was required by S. Typhimurium to cause a systemic disease, since mice lacking IL-10 (IL-10-/-) were significantly more resistant to die after an infection as compared to wild-type (WT) mice. IL-10-/- mice had reduced bacterial loads in internal organs and increased levels of pro-inflammatory cytokines in serum at 5 days of infection. Importantly, WT mice showed high bacterial loads in tissues and no increase of cytokines in serum after 5 days of S. Typhimurium infection, except for IL-10. In WT mice, we observed a peak of il-10 messenger RNA production in ileum, spleen, and liver after 5 days of infection. Importantly, the adoptive transfer of T or B cells from WT mice restored the susceptibility of IL-10-/- mice to systemic S. Typhimurium infection, suggesting that the generation of regulatory cells in vivo is required to sustain a systemic infection by S. Typhimurium. These findings support the notion that IL-10 production from lymphoid cells is a key process in the infective cycle of S. Typhimurium in mice due to generation of a tolerogenic immune response that prevents bacterial clearance and supports systemic dissemination.

2.
J Immunol ; 199(1): 212-223, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28566367

RESUMEN

Human respiratory syncytial virus (hRSV) is the leading cause of severe lower respiratory tract infections in children. The development of novel prophylactic and therapeutic antiviral drugs against hRSV is imperative to control the burden of disease in the susceptible population. In this study, we examined the effects of inducing the activity of the host enzyme heme oxygenase-1 (HO-1) on hRSV replication and pathogenesis on lung inflammation induced by this virus. Our results show that after hRSV infection, HO-1 induction with metalloporphyrin cobalt protoporphyrin IX significantly reduces the loss of body weight due to hRSV-induced disease. Further, HO-1 induction also decreased viral replication and lung inflammation, as evidenced by a reduced neutrophil infiltration into the airways, with diminished cytokine and chemokine production and reduced T cell function. Concomitantly, upon cobalt protoporphyrin IX treatment, there is a significant upregulation in the production of IFN-α/ß mRNAs in the lungs. Furthermore, similar antiviral and protective effects occur by inducing the expression of human HO-1 in MHC class II+ cells in transgenic mice. Finally, in vitro data suggest that HO-1 induction can modulate the susceptibility of cells, especially the airway epithelial cells, to hRSV infection.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Pulmón/inmunología , Infecciones por Virus Sincitial Respiratorio/fisiopatología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/fisiología , Animales , Línea Celular , Citocinas/biosíntesis , Citocinas/inmunología , Replicación del ADN , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Hemo-Oxigenasa 1/genética , Humanos , Interferón-alfa/biosíntesis , Interferón-alfa/inmunología , Interferón beta/inmunología , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Ratones , Protoporfirinas/administración & dosificación , Protoporfirinas/farmacología , Infecciones por Virus Sincitial Respiratorio/inmunología , Linfocitos T/inmunología , Acoplamiento Viral , Internalización del Virus , Replicación Viral
3.
Virulence ; 8(6): 685-704, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27911218

RESUMEN

Human Respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV) are the two major etiological viral agents of lower respiratory tract diseases, affecting mainly infants, young children and the elderly. Although the infection of both viruses trigger an antiviral immune response that mediate viral clearance and disease resolution in immunocompetent individuals, the promotion of long-term immunity appears to be deficient and reinfection are common throughout life. A possible explanation for this phenomenon is that hRSV and hMPV, can induce aberrant T cell responses, which leads to exacerbated lung inflammation and poor T and B cell memory immunity. The modulation of immune response exerted by both viruses include different strategies such as, impairment of immunological synapse mediated by viral proteins or soluble factors, and the induction of pro-inflammatory cytokines by epithelial cells, among others. All these viral strategies contribute to the alteration of the adaptive immunity in order to increase the susceptibility to reinfections. In this review, we discuss current research related to the mechanisms underlying the impairment of T and B cell immune responses induced by hRSV and hMPV infection. In addition, we described the role each virulence factor involved in immune modulation caused by these viruses.


Asunto(s)
Metapneumovirus/patogenicidad , Infecciones por Paramyxoviridae/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/patogenicidad , Infecciones del Sistema Respiratorio/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa , Anciano , Animales , Linfocitos B/inmunología , Niño , Citocinas , Humanos , Evasión Inmune , Lactante , Metapneumovirus/inmunología , Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/virología , Neumonía/virología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/inmunología , Virus Sincitial Respiratorio Humano/fisiología , Infecciones del Sistema Respiratorio/virología , Replicación Viral
4.
Semin Respir Crit Care Med ; 37(4): 522-37, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27486734

RESUMEN

The human respiratory syncytial virus (hRSV) is by far the major cause of acute lower respiratory tract infections (ALRTIs) worldwide in infants and children younger than 2 years. The overwhelming number of hospitalizations due to hRSV-induced ALRTI each year is due, at least in part, to the lack of licensed vaccines against this virus. Thus, hRSV infection is considered a major public health problem and economic burden in most countries. The lung pathology developed in hRSV-infected individuals is characterized by an exacerbated proinflammatory and unbalanced Th2-type immune response. In addition to the adverse effects in airway tissues, hRSV infection can also cause neurologic manifestations in the host, such as seizures and encephalopathy. Although the origins of these extrapulmonary symptoms remain unclear, studies with patients suffering from neurological alterations suggest an involvement of the inflammatory response against hRSV. Furthermore, hRSV has evolved numerous mechanisms to modulate and evade the immune response in the host. Several studies have focused on elucidating the interactions between hRSV virulence factors and the host immune system, to rationally design new vaccines and therapies against this virus. Here, we discuss about the infection, pathology, and immune response triggered by hRSV in the host.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Infecciones del Sistema Respiratorio , Humanos , Evasión Inmune , Lactante , Recién Nacido , Pulmón/patología , Pulmón/virología , Infecciones por Virus Sincitial Respiratorio/complicaciones , Infecciones por Virus Sincitial Respiratorio/epidemiología , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitial Respiratorio Humano/inmunología , Infecciones del Sistema Respiratorio/complicaciones , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología
5.
Immunology ; 147(1): 55-72, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26451966

RESUMEN

Human respiratory syncytial virus (hRSV) is the leading cause of infant hospitalization related to respiratory disease. Infection with hRSV produces abundant infiltration of immune cells into the airways, which combined with an exacerbated pro-inflammatory immune response can lead to significant damage to the lungs. Human RSV re-infection is extremely frequent, suggesting that this virus may have evolved molecular mechanisms that interfere with host adaptive immunity. Infection with hRSV can be reduced by administering a humanized neutralizing antibody against the virus fusion protein in high-risk infants. Although neutralizing antibodies against hRSV effectively block the infection of airway epithelial cells, here we show that both, bone marrow-derived dendritic cells (DCs) and lung DCs undergo infection with IgG-coated virus (hRSV-IC), albeit abortive. Yet, this is enough to negatively modulate DC function. We observed that such a process is mediated by Fcγ receptors (FcγRs) expressed on the surface of DCs. Remarkably, we also observed that in the absence of hRSV-specific antibodies FcγRIII knockout mice displayed significantly less cellular infiltration in the lungs after hRSV infection, compared with wild-type mice, suggesting a potentially harmful, IgG-independent role for this receptor in hRSV disease. Our findings support the notion that FcγRs can contribute significantly to the modulation of DC function by hRSV and hRSV-IC. Further, we provide evidence for an involvement of FcγRIII in the development of hRSV pathogenesis.


Asunto(s)
Células Dendríticas/metabolismo , Pulmón/metabolismo , Activación de Linfocitos , Receptores de IgG/metabolismo , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitial Respiratorio Humano/patogenicidad , Linfocitos T/metabolismo , Inmunidad Adaptativa , Animales , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/metabolismo , Antivirales/farmacología , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/virología , Modelos Animales de Enfermedad , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/virología , Activación de Linfocitos/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Palivizumab/farmacología , Receptores de IgG/deficiencia , Receptores de IgG/genética , Receptores de IgG/inmunología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Virus Sincitial Respiratorio Humano/inmunología , Transducción de Señal , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/virología , Carga Viral , Replicación Viral
6.
Expert Opin Investig Drugs ; 24(12): 1613-30, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26457559

RESUMEN

INTRODUCTION: Human respiratory syncytial virus (hRSV) is the leading cause of acute lower respiratory tract infections worldwide in infants, as well as an important pathogen affecting the elderly and immunocompromised individuals. Despite more than a half a century of research, no licensed vaccines are available and only palivizumab has been approved to use in humans, mostly recommended or limited to high risk infants. Therefore, novel therapeutic and preventive drugs need to be developed to fight this major human pathogen. AREAS COVERED: This review discusses current therapeutic approaches in preclinical and clinical stages, aimed at controlling or preventing hRSV infection. These methods include passive immunization, experimental drugs, vaccine candidates and maternal immunization. EXPERT OPINION: Based on the results of various immunization strategies and therapeutic approaches, it is likely that the most effective strategy against hRSV will be a prophylactic tool aimed at developing a strong antiviral T-cell response capable of both, promoting the generation of hRSV-specific high affinity antibodies and leading the protective immunity required to prevent the disease caused by this virus. Alternatively, if prophylactic strategies fail, antiviral drugs and novel passive immunity strategies could significantly contribute to reducing hospitalization rates in susceptible individuals.


Asunto(s)
Antivirales/administración & dosificación , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Anciano , Animales , Antivirales/uso terapéutico , Diseño de Fármacos , Humanos , Huésped Inmunocomprometido , Lactante , Palivizumab/administración & dosificación , Palivizumab/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/epidemiología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/aislamiento & purificación , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/prevención & control , Infecciones del Sistema Respiratorio/virología
7.
Immunol Lett ; 162(1 Pt A): 237-47, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25268876

RESUMEN

Human Respiratory Syncytial Virus (hRSV) is the leading cause of lower respiratory tract diseases, affecting particularly newborns and young children. This virus is able to modulate the immune response, generating a pro-inflammatory environment in the airways that causes obstruction and pulmonary alterations in the infected host. To date, no vaccines are available for human use and the first vaccine that reached clinical trials produced an enhanced hRSV-associated pathology 50 years ago, resulting in the death of two children. Currently, only two therapeutic approaches have been used to treat hRSV infection in high risk children: 1. Palivizumab, a humanized antibody against the F glycoprotein that reduces to half the number of hospitalized cases and 2. Ribavirin, which fails to have a significant therapeutic effect. A major caveat for these approaches is their high economical cost, which highlights the need of new and affordable therapeutic or prophylactic tools to treat or prevents hRSV infection. Accordingly, several efforts are in progress to understand the hRSV-associated pathology and to characterize the immune response elicited by this virus. Currently, preclinical and clinical trials are being conducted to evaluate safety and efficacy of several drugs and vaccines, which have shown promising results. In this article, we discuss the most important advances in the development of drugs and vaccines, which could eventually lead to better strategies to treat or prevent the detrimental inflammation triggered by hRSV infection.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/fisiología , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Interacciones Huésped-Patógeno/inmunología , Humanos , Infecciones por Virus Sincitial Respiratorio/etiología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/inmunología
8.
Proc Natl Acad Sci U S A ; 111(31): E3214-23, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25056968

RESUMEN

Human respiratory syncytial virus (hRSV) is the leading cause of bronchiolitis and pneumonia in young children worldwide. The recurrent hRSV outbreaks and reinfections are the cause of a significant public health burden and associate with an inefficient antiviral immunity, even after disease resolution. Although several mouse- and human cell-based studies have shown that hRSV infection prevents naïve T-cell activation by antigen-presenting cells, the mechanism underlying such inhibition remains unknown. Here, we show that the hRSV nucleoprotein (N) could be at least partially responsible for inhibiting T-cell activation during infection by this virus. Early after infection, the N protein was expressed on the surface of epithelial and dendritic cells, after interacting with trans-Golgi and lysosomal compartments. Further, experiments on supported lipid bilayers loaded with peptide-MHC (pMHC) complexes showed that surface-anchored N protein prevented immunological synapse assembly by naive CD4(+) T cells and, to a lesser extent, by antigen-experienced T-cell blasts. Synapse assembly inhibition was in part due to reduced T-cell receptor (TCR) signaling and pMHC clustering at the T-cell-bilayer interface, suggesting that N protein interferes with pMHC-TCR interactions. Moreover, N protein colocalized with the TCR independently of pMHC, consistent with a possible interaction with TCR complex components. Based on these data, we conclude that hRSV N protein expression at the surface of infected cells inhibits T-cell activation. Our study defines this protein as a major virulence factor that contributes to impairing acquired immunity and enhances susceptibility to reinfection by hRSV.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Membrana Celular/metabolismo , Sinapsis Inmunológicas/inmunología , Nucleoproteínas/metabolismo , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales/metabolismo , Animales , Brefeldino A/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/patología , Comunicación Celular , Línea Celular , Membrana Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Antígenos de Histocompatibilidad/inmunología , Humanos , Sinapsis Inmunológicas/efectos de los fármacos , Membrana Dobles de Lípidos/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Péptidos/inmunología , Transporte de Proteínas/efectos de los fármacos , Receptores de Antígenos de Linfocitos T/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Replicación Viral/efectos de los fármacos
9.
J Immunol ; 192(1): 214-23, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24319265

RESUMEN

Along with the human respiratory syncytial virus (hRSV), the human metapneumovirus (hMPV) is one of the leading causes of childhood hospitalization and a major health burden worldwide. Unfortunately, owing to an inefficient immunological memory, hMPV infection provides limited immune protection against reinfection. Furthermore, hMPV can induce an inadequate Th2 type immune response that causes severe lung inflammation, leading to airway obstruction. Similar to hRSV, it is likely that an effective clearance of hMPV would require a balanced Th1 type immunity by the host, involving the activation of IFN-γ-secreting T cells. A recognized inducer of Th1 immunity is Mycobacterium bovis bacillus Calmette-Guérin (BCG), which has been used in newborns for many decades and in several countries as a tuberculosis vaccine. We have previously shown that immunization with BCG strains expressing hRSV Ags can induce an efficient immune response that protects against this virus. In this study, we show that immunization with rBCG strains expressing the phosphoprotein from hMPV also can induce protective Th1 immunity. Mice immunized with rBCG were protected against weight loss, airway inflammation, and viral replication in the lungs after hMPV infection. Our rBCG vaccine also induced the activation of hMPV-specific T cells producing IFN-γ and IL-2, which could protect from hMPV infection when transferred to recipient mice. These data strongly support the notion that rBCG induces protective Th1 immunity and could be considered as an efficient vaccine against hMPV.


Asunto(s)
Vacuna BCG/inmunología , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/inmunología , Células TH1/inmunología , Traslado Adoptivo , Animales , Anticuerpos Antibacterianos/inmunología , Línea Celular , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Humanos , Inmunidad Celular , Inmunoglobulina G/inmunología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Metapneumovirus/metabolismo , Ratones , Infecciones por Paramyxoviridae/patología , Infecciones por Paramyxoviridae/prevención & control , Fosfoproteínas/inmunología , Fosfoproteínas/metabolismo , Linfocitos T/inmunología , Linfocitos T/patología , Vacunas Sintéticas , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
10.
J Med Virol ; 86(7): 1256-66, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24150877

RESUMEN

Acute respiratory infections caused by the respiratory syncytial virus (RSV) are important health burdens that affect infants worldwide. RSV is also an important cause of morbidity and disease in adults, which causes enormous economic losses. At the present time, RSV infection is diagnosed by immunofluorescence, test pack and/or PCR, obtaining better results with PCR than with any other technique. The production of new monoclonal antibodies (mAbs) capable of detecting RSV in clinical samples is necessary to generate better and faster diagnosis tools for RSV. In this study, three new mAbs, directed against the RSV N and M2-1 proteins, were evaluated for the detection of RSV in clinical samples. Nasopharyngeal swabs were obtained from: 27 RSV-positive patients; 15 human metapneumovirus (hMPV)-positive patients; and 6 healthy controls. To evaluate RSV presence in these samples, clinical samples and RSV-infected cells were tested by Enzyme-Linked ImmunoSorbent Assay (ELISA), flow cytometry, immunofluorescence, and dot-blot assays. Specificity and sensitivity were determined for each mAb by using purified RSV antigens and antigens from different viruses. Infected cells and clinical samples tested with the three new mAbs resulted positive by immunofluorescence, ELISA, flow cytometry, and dot blot. No false positives were obtained in samples infected with other respiratory virus (hMPV) or from healthy controls. These results suggest that these new anti-RSV mAbs can be considered for the rapid and reliable detection of RSV on infected cells and clinical specimens by multiple immunological approaches.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antivirales , Pruebas Diagnósticas de Rutina/métodos , Infecciones por Virus Sincitial Respiratorio/diagnóstico , Virus Sincitial Respiratorio Humano/aislamiento & purificación , Adulto , Humanos , Inmunoensayo/métodos , Lactante , Nasofaringe/virología , Virus Sincitial Respiratorio Humano/inmunología , Sensibilidad y Especificidad
11.
Proc Natl Acad Sci U S A ; 110(22): 9112-7, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-23650398

RESUMEN

Respiratory syncytial virus (RSV) is the major cause of respiratory illness in infants worldwide. Neurologic alterations, such as seizures and ataxia, have been associated with RSV infection. We demonstrate the presence of RSV proteins and RNA in zones of the brain--such as the hippocampus, ventromedial hypothalamic nucleus, and brainstem--of infected mice. One month after disease resolution, rodents showed behavioral and cognitive impairment in marble burying (MB) and Morris water maze (MWM) tests. Our data indicate that the learning impairment caused by RSV is a result of a deficient induction of long-term potentiation in the hippocampus of infected animals. In addition, immunization with recombinant bacillus Calmette-Guérin (BCG) expressing RSV nucleoprotein prevented behavioral disorders, corroborating the specific effect of RSV infection over the central nervous system. Our findings provide evidence that RSV can spread from the airways to the central nervous system and cause functional alterations to the brain, both of which can be prevented by proper immunization against RSV.


Asunto(s)
Encéfalo/metabolismo , Discapacidades para el Aprendizaje/etiología , ARN Viral/metabolismo , Infecciones por Virus Sincitial Respiratorio/complicaciones , Infecciones por Virus Sincitial Respiratorio/inmunología , Proteínas Virales/metabolismo , Animales , Encéfalo/patología , Discapacidades para el Aprendizaje/prevención & control , Discapacidades para el Aprendizaje/virología , Potenciación a Largo Plazo/fisiología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos BALB C , Mycobacterium bovis/inmunología , Ratas , Ratas Sprague-Dawley , Infecciones por Virus Sincitial Respiratorio/metabolismo , Linfocitos T/inmunología , Vacunas Virales/inmunología
12.
Immunology ; 136(4): 414-24, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22587389

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multiple functional alterations affecting immune cells, such as B cells, T cells, dendritic cells (DCs) and monocytes. During SLE, the immunogenicity of monocytes and DCs is significantly up-regulated, promoting the activation of self-reactive T cells. Accordingly, it is important to understand the contribution of these cells to the pathogenesis of SLE and the mechanisms responsible for their altered functionality during disease. One of the key enzymes that control monocyte and DC function is haem oxygenase-1 (HO-1), which catalyses the degradation of the haem group into biliverdin, carbon monoxide and free iron. These products possess immunosuppressive and anti-inflammatory capacities. The main goal of this work was to determine HO-1 expression in monocytes and DCs from patients with SLE and healthy controls. Hence, peripheral blood mononuclear cells were obtained from 43 patients with SLE and 30 healthy controls. CD14(+) monocytes and CD4(+) T cells were sorted by FACS and HO-1 expression was measured by RT-PCR. In addition, HO-1 protein expression was determined by FACS. HO-1 levels in monocytes were significantly reduced in patients with SLE compared with healthy controls. These results were confirmed by flow cytometry. No differences were observed in other cell types, such as DCs or CD4(+) T cells, although decreased MHC-II levels were observed in DCs from patients with SLE. In conclusion, we found a significant decrease in HO-1 expression, specifically in monocytes from patients with SLE, suggesting that an imbalance of monocyte function could be partly the result of a decrease in HO-1 expression.


Asunto(s)
Células Dendríticas/enzimología , Hemo-Oxigenasa 1/metabolismo , Lupus Eritematoso Sistémico/enzimología , Lupus Eritematoso Sistémico/inmunología , Monocitos/enzimología , Adulto , Animales , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Femenino , Humanos , Receptores de Lipopolisacáridos/biosíntesis , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Monocitos/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de IgG/genética , Receptores de IgG/inmunología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...