Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 395
Filtrar
3.
Clin Immunol ; 235: 108933, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35074548

RESUMEN

We encountered two patients with partial interferon γ receptor 1 (IFN-γR1) deficiency in whom early diagnosis enhanced disease management. Patient 1 was a 44-year-old woman with enlarged lymph nodes diagnosed in a pre-pregnancy checkup, and pathological examination revealed a Mycobacterium avium infection. Based on her history of unknown multiple osteomyelitis during early childhood, mendelian susceptibility to mycobacterial disease was suspected. Genetic analysis revealed a novel heterozygous variant in IFNGR1. Genetic counseling was administered to the patient and her husband before they had their baby. Patient 2 was a 4-month-old boy whose father was previously diagnosed with autosomal dominant IFN-γR1 deficiency owing to Bacille de Calmette et Guérin (BCG) osteomyelitis. Genetic analysis showed that he had the same INFGR1 variant. He avoided BCG vaccination and has been disease-free since then. Early diagnosis is considered to be useful for genetic counseling and essential for preventing BCG osteomyelitis.


Asunto(s)
Vacuna BCG/efectos adversos , Vacuna BCG/inmunología , Osteomielitis/microbiología , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Adulto , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Humanos , Lactante , Masculino , Monocitos/metabolismo , Mycobacterium bovis , Linaje , Receptor de Interferón gamma
4.
J Allergy Clin Immunol ; 149(1): 252-261.e6, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34176646

RESUMEN

BACKGROUND: Patients with Mendelian susceptibility to mycobacterial disease (MSMD) experience recurrent and/or persistent infectious diseases associated with poorly virulent mycobacteria. Multifocal osteomyelitis is among the representative manifestations of MSMD. The frequency of multifocal osteomyelitis is especially high in patients with MSMD etiologies that impair cellular response to IFN-γ, such as IFN-γR1, IFN-γR2, or STAT1 deficiency. OBJECTIVES: This study sought to characterize the mechanism underlying multifocal osteomyelitis in MSMD. METHODS: GM colonies prepared from bone marrow mononuclear cells from patients with autosomal dominant (AD) IFN-γR1 deficiency, AD STAT1 deficiency, or STAT1 gain of function (GOF) and from healthy controls were differentiated into osteoclasts in the presence or absence of IFN-γ. The inhibitory effect of IFN-γ on osteoclastogenesis was investigated by quantitative PCR, immunoblotting, tartrate-resistant acid phosphatase staining, and pit formation assays. RESULTS: Increased osteoclast numbers were identified by examining the histopathology of osteomyelitis in patients with AD IFN-γR1 deficiency or AD STAT1 deficiency. In the presence of receptor activator of nuclear factor kappa-B ligand and M-CSF, GM colonies from patients with AD IFN-γR1 deficiency, AD STAT1 deficiency, or STAT1 GOF differentiated into osteoclasts, similar to GM colonies from healthy volunteers. IFN-γ concentration-dependent inhibition of osteoclast formation was impaired in GM colonies from patients with AD IFN-γR1 deficiency or AD STAT1 deficiency, whereas it was enhanced in GM colonies from patients with STAT1 GOF. CONCLUSIONS: Osteoclast differentiation is increased in AD IFN-γR1 deficiency and AD STAT1 deficiency due to an impaired response to IFN-γ, leading to excessive osteoclast proliferation and, by inference, increased bone resorption in infected foci, which may underlie multifocal osteomyelitis.


Asunto(s)
Infecciones por Mycobacterium , Osteogénesis/efectos de los fármacos , Osteomielitis , Receptores de Interferón/deficiencia , Factor de Transcripción STAT1/deficiencia , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Predisposición Genética a la Enfermedad , Humanos , Interferón gamma/farmacología , Mutación , Infecciones por Mycobacterium/genética , Mycobacterium avium , Osteoclastos/efectos de los fármacos , Osteomielitis/genética , Receptores de Interferón/genética , Factor de Transcripción STAT1/genética
5.
Int J Mol Sci ; 22(21)2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34769174

RESUMEN

A pivotal role of type I interferons in systemic lupus erythematosus (SLE) is widely accepted. Type III interferons (IFN-λ) however, the most recently discovered cytokines grouped within the interferon family, have not been extensively studied in lupus disease models yet. Growing evidence suggests a role for IFN-λ in regulating both innate and adaptive immune responses, and increased serum concentrations have been described in multiple autoimmune diseases including SLE. Using the pristane-induced lupus model, we found that mice with defective IFN-λ receptors (Ifnlr1-/-) showed increased survival rates, decreased lipogranuloma formation and reduced anti-dsDNA autoantibody titers in the early phase of autoimmunity development compared to pristane-treated wild-type mice. Moreover, Ifnlr1-/- mice treated with pristane had reduced numbers of inflammatory mononuclear phagocytes and cNK cells in their kidneys, resembling untreated control mice. Systemically, circulating B cells and monocytes (CD115+Ly6C+) were reduced in pristane-treated Ifnlr1-/- mice. The present study supports a significant role for type III interferons in the pathogenesis of pristane-induced murine autoimmunity as well as in systemic and renal inflammation. Although the absence of type III interferon receptors does not completely prevent the development of autoantibodies, type III interferon signaling accelerates the development of autoimmunity and promotes a pro-inflammatory environment in autoimmune-prone hosts.


Asunto(s)
Inmunidad Celular , Inmunidad Humoral , Interferones/inmunología , Leucocitos/inmunología , Lupus Eritematoso Sistémico , Terpenos/efectos adversos , Animales , Interferones/genética , Lupus Eritematoso Sistémico/inducido químicamente , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Noqueados , Receptores de Interferón/deficiencia , Receptores de Interferón/inmunología , Terpenos/farmacología , Interferón lambda
6.
PLoS One ; 16(10): e0258491, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34644348

RESUMEN

Mastitis is an inflammation of the mammary gland in the breast and is typically due to bacterial infection. In malaria-endemic areas, mastitis with accompanying fever can be challenging to differentiate from malaria. At the same time, it is unclear whether malaria infection is directly involved in the development of mastitis. In the present study, whether mastitis develops during infection with malaria parasites was investigated using a rodent malaria model with Plasmodium berghei (P. berghei; Pb) ANKA. The course of parasitemia in postpartum mice infected with Pb ANKA was similar to the course in infected virgin mice. However, infected postpartum mice died earlier than did infected virgin mice. In addition, the weight of pups from mice infected with Pb ANKA was significantly reduced compared with pups from uninfected mice. The macroscopic and histological analyses showed apparent changes, such as destruction of the alveolus wall and extensive presence of leukocytes, in mammary gland tissue in mice infected during the postpartum period. The findings suggest that women during the postpartum period are more vulnerable to complications when infected with malaria parasites, particularly women who do not acquire protective immunity against malaria parasites. Based on the proteomic analysis, IFN-γ signaling pathway-related proteins in mammary gland tissue of the infected postpartum mice were increased. Our results indicate that inflammation induced by IFN-γ, a proinflammatory cytokine, may contribute to negative histological changes in mammary gland tissue of postpartum mice infected with Pb ANKA. In IFN-γ receptor 1-deficient (IFNGR1-KO) mice, the histological changes in mammary gland tissue of the infected postpartum wild-type mice were improved to almost normal mammary gland structure. Furthermore, weight loss in pups delivered by infected IFNGR1-KO postpartum mice was not observed. Taken together, these findings indicate that inflammation induced by IFN-γ is associated with development of mastitis in postpartum mice infected with Pb ANKA. The present study results may increase our understanding of how disease aggravation occurs during postpartum malaria.


Asunto(s)
Malaria/patología , Glándulas Mamarias Animales/metabolismo , Animales , Modelos Animales de Enfermedad , Eritrocitos/parasitología , Eritrocitos/patología , Femenino , Interferón gamma/metabolismo , Malaria/fisiopatología , Glándulas Mamarias Animales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/análisis , Plasmodium berghei/patogenicidad , Periodo Posparto , Embarazo , Proteómica , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Transducción de Señal/genética , Regulación hacia Arriba , Receptor de Interferón gamma
7.
Elife ; 102021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34423779

RESUMEN

Arthropod-borne rickettsial pathogens cause mild and severe human disease worldwide. The tick-borne pathogen Rickettsia parkeri elicits skin lesions (eschars) and disseminated disease in humans; however, inbred mice are generally resistant to infection. We report that intradermal infection of mice lacking both interferon receptors (Ifnar1-/-;Ifngr1-/-) with as few as 10 R. parkeri elicits eschar formation and disseminated, lethal disease. Similar to human infection, eschars exhibited necrosis and inflammation, with bacteria primarily found in leukocytes. Using this model, we find that the actin-based motility factor Sca2 is required for dissemination from the skin to internal organs, and the outer membrane protein OmpB contributes to eschar formation. Immunizing Ifnar1-/-;Ifngr1-/- mice with sca2 and ompB mutant R. parkeri protects against rechallenge, revealing live-attenuated vaccine candidates. Thus, Ifnar1-/-;Ifngr1-/- mice are a tractable model to investigate rickettsiosis, virulence factors, and immunity. Our results further suggest that discrepancies between mouse and human susceptibility may be due to differences in interferon signaling.


Tick bites allow disease-causing microbes, including multiple species of Rickettsia bacteria, to pass from arthropods to humans. Being exposed to Rickettsia parkeri, for example, can cause a scab at the bite site, fever, headache and fatigue. To date, no vaccine is available against any of the severe diseases caused by Rickettsia species. Modelling human infections in animals could help to understand and combat these illnesses. R. parkeri is a good candidate for such studies, as it can give insight into more severe Rickettsia infections while being comparatively safer to handle. However, laboratory mice are resistant to this species of bacteria, limiting their use as models. To explore why this is the case, Burke et al. probed whether an immune mechanism known as interferon signalling protects laboratory rodents against R. parkeri. During infection, the immune system releases molecules called interferons that stick to 'receptors' at the surface of cells, triggering defense mechanisms that help to fight off an invader. Burke et al. injected R. parkeri into the skin of mice that had or lacked certain interferon receptors, showing that animals without two specific receptors developed scabs and saw the disease spread through their body. Further investigation showed that two R. parkeri proteins, known as OmpB or Sca2, were essential for the bacteria to cause skin lesions and damage internal organs. Burke et al. then used R. parkeri that lacked OmpB or Sca2 to test whether these modified, inoffensive microbes could act as 'vaccines'. And indeed, vulnerable laboratory mice which were first exposed to the mutant bacteria were then able to survive the 'normal' version of the microbe. Together, this work reveals that interferon signalling protects laboratory mice against R. parkeri infections. It also creates an animal model that can be used to study disease and vaccination.


Asunto(s)
Estudios de Asociación Genética , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Infecciones por Rickettsia/inmunología , Animales , Médula Ósea , Femenino , Inmunidad Innata , Inflamación , Listeria monocytogenes , Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Rickettsia , Infecciones por Rickettsia/patología , Garrapatas
8.
Int J Mol Sci ; 22(5)2021 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-33806448

RESUMEN

Lambda interferons mediate antiviral immunity by inducing interferon-stimulated genes (ISGs) in epithelial tissues. A common variant rs368234815TT/∆G creating functional gene from an IFNL4 pseudogene is associated with the expression of major ISGs in the liver but impaired clearance of hepatitis C. To explain this, we compared Halo-tagged and non-tagged IFNL3 and IFNL4 signaling in liver-derived cell lines. Transfection with non-tagged IFNL3, non-tagged IFNL4 and Halo-tagged IFNL4 led to a similar degree of JAK-STAT activation and ISG induction; however, the response to transfection with Halo-tagged IFNL3 was lower and delayed. Transfection with non-tagged IFNL3 or IFNL4 induced no transcriptome change in the cells lacking either IL10R2 or IFNLR1 receptor subunits. Cytosolic overexpression of signal peptide-lacking IFNL3 or IFNL4 in wild type cells did not interfere with JAK-STAT signaling triggered by interferons in the medium. Finally, expression profile changes induced by transfection with non-tagged IFNL3 and IFNL4 were highly similar. These data do not support the hypothesis about IFNL4-specific non-canonical signaling and point out that functional studies conducted with tagged interferons should be interpreted with caution.


Asunto(s)
Hepatocitos/inmunología , Hepatocitos/metabolismo , Interferones/genética , Interferones/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Línea Celular , Expresión Génica , Técnicas de Inactivación de Genes , Células Hep G2 , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferones/deficiencia , Subunidad beta del Receptor de Interleucina-10/deficiencia , Subunidad beta del Receptor de Interleucina-10/genética , Subunidad beta del Receptor de Interleucina-10/metabolismo , Interleucinas/deficiencia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Transfección
11.
Methods Mol Biol ; 2225: 107-123, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33108660

RESUMEN

Viruses have devised highly effective approaches that modulate the host immune response, blocking immune responses that are designed to eradicate viral infections. Over millions of years of evolution, virus-derived immune-modulating proteins have become extraordinarily potent, in some cases working at picomolar concentrations when expressed into surrounding tissues and effectively blocking host defenses against viral invasion and replication. The marked efficiency of these immune-modulating proteins is postulated to be due to viral engineering of host immune modulators as well as design and development of new strategies (i.e., some derived from host proteins and some entirely unique). Two key characteristics of viral immune modulators confer both adaptive advantages and desirable functions for therapeutic translation. First, many virus-derived immune modulators have evolved structures that are not readily recognized or regulated by mammalian immune pathways, ensuring little to no neutralizing antibody responses or proteasome-mediated degradation. Second, these immune modulators tend to target early steps in central immune responses, producing a powerful downstream inhibitory "domino effect" which may alter cell activation and gene expression.We have proposed that peptide metabolites of these immune-modulating proteins can enhance and extend protein function. Active immunomodulating peptides have been derived from both mammalian and viral proteins. We previously demonstrated that peptides derived from computationally predicted cleavage sites in the reactive center loop (RCL) of a viral serine proteinase inhibitor (serpin ) from myxoma virus, Serp-1 , can modify immune response activation. We have also demonstrated modulation of host gut microbiota produced by Serp-1 and RCL-derived peptide , S7, in a vascular inflammation model. Of interest, generation of derived peptides that maintain therapeutic function from a serpin can act by a different mechanism. Whereas Serp-1 has canonical serpin-like function to inhibit serine proteases, S7 instead targets mammalian serpins. Here we describe the derivation of active Serp- RCL peptides and their testing in inflammatory vasculitis models.


Asunto(s)
Factores Inmunológicos/inmunología , Myxoma virus/genética , Péptidos/inmunología , Serpinas/inmunología , Trasplante Homólogo/métodos , Vasculitis/terapia , Proteínas Virales/inmunología , Animales , Aorta Torácica , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Factores Inmunológicos/genética , Factores Inmunológicos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/genética , Péptidos/farmacología , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Serpinas/genética , Serpinas/farmacología , Vasculitis/inmunología , Vasculitis/patología , Proteínas Virales/genética , Proteínas Virales/farmacología , Receptor de Interferón gamma
12.
Virulence ; 11(1): 1656-1673, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33356838

RESUMEN

Summary: We characterized Mycobacterium bovis BCG isolates found in lung and brain samples from a previously vaccinated patient with IFNγR1 deficiency. The isolates collected displayed distinct genomic and phenotypic features consistent with host adaptation and associated changes in antibiotic susceptibility and virulence traits. Background: We report a case of a patient with partial recessive IFNγR1 deficiency who developed disseminated BCG infection after neonatal vaccination (BCG-vaccine). Distinct M. bovis BCG-vaccine derived clinical strains were recovered from the patient's lungs and brain. Methods: BCG strains were phenotypically (growth, antibiotic susceptibility, lipid) and genetically (whole genome sequencing) characterized. Mycobacteria cell infection models were used to assess apoptosis, necrosis, cytokine release, autophagy, and JAK-STAT signaling. Results: Clinical isolates BCG-brain and BCG-lung showed distinct Rv0667 rpoB mutations conferring high- and low-level rifampin resistance; the latter displayed clofazimine resistance through Rv0678 gene (MarR-like transcriptional regulator) mutations. BCG-brain and BCG-lung showed mutations in fadA2, fadE5, and mymA operon genes, respectively. Lipid profiles revealed reduced levels of PDIM in BCG-brain and BCG-lung and increased TAGs and Mycolic acid components in BCG-lung, compared to parent BCG-vaccine. In vitro infected cells showed that the BCG-lung induced a higher cytokine release, necrosis, and cell-associated bacterial load effect when compared to BCG-brain; conversely, both strains inhibited apoptosis and altered JAK-STAT signaling. Conclusions: During a chronic-disseminated BCG infection, BCG strains can evolve independently at different sites likely due to particular microenvironment features leading to differential antibiotic resistance, virulence traits resulting in dissimilar responses in different host tissues.


Asunto(s)
Vacuna BCG/efectos adversos , Vacuna BCG/inmunología , Mycobacterium bovis/inmunología , Mycobacterium bovis/patogenicidad , Receptores de Interferón/genética , Tuberculosis/sangre , Tuberculosis/diagnóstico , Animales , Antibacterianos/farmacología , Vacuna BCG/administración & dosificación , Encéfalo/microbiología , Bovinos , Preescolar , Farmacorresistencia Bacteriana , Humanos , Pulmón/microbiología , Masculino , Mutación , Mycobacterium bovis/efectos de los fármacos , Mycobacterium bovis/genética , Receptores de Interferón/deficiencia , Vacunación , Virulencia , Receptor de Interferón gamma
13.
Biochem Biophys Res Commun ; 533(4): 1519-1526, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33158480

RESUMEN

Cognitive impairment, acute or long-term, is a common complication in patients with severe bacterial infection. However, the underlying mechanisms are not fully verified and effective medicine is not available in clinics. Interferon gamma (IFNγ) is a pivotal cytokine against infection and is believed to be a tune in homeostasis of cognitive function. Here, we collected blood and cerebrospinal fluid (CF) from human subjects and mice, and found that plasma and CF levels of IFNγ were significantly increased in septic patients and endotoxin-challenged mice when compared with healthy controls. IFNγ signaling was boosted in the hippocampus of mice after a challenge of lipopolysaccharide (LPS), which was accompanied with cognitive impairment and decline of neurogenesis. Deficiency of IFNγ or its receptor (IFNγR) dramatically attenuated microglia-induced A1 astrocytes and consequently restored neurogenesis and cognitive function in endotoxemia mice model. Using primary microglia, astrocytes and neurons, we found that IFNγ remarkably increased LPS-mediated release of TNFα and IL-1α in microglia and consequently induced the transformation of astrocyte to A1 subtype, which ultimately resulted in neuron damage. Thus, IFNγ promotes cognitive impairment in endotoxemia by enhancing microglia-induced A1 astrocytes. Targeting IFNγ would be a novel strategy for preventing or treating cognitive dysfunction in patients with Gram-negative infection.


Asunto(s)
Astrocitos/fisiología , Disfunción Cognitiva/fisiopatología , Endotoxemia/fisiopatología , Interferón gamma/antagonistas & inhibidores , Neurogénesis/fisiología , Animales , Astrocitos/patología , Estudios de Casos y Controles , Células Cultivadas , Disfunción Cognitiva/genética , Disfunción Cognitiva/terapia , Modelos Animales de Enfermedad , Endotoxemia/genética , Endotoxemia/psicología , Silenciador del Gen , Humanos , Interferón gamma/deficiencia , Interferón gamma/genética , Interferón gamma/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Microglía/fisiología , Neurogénesis/genética , Tratamiento con ARN de Interferencia , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/fisiología , Receptor de Interferón gamma
14.
Front Immunol ; 11: 2189, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33042133

RESUMEN

Natural killer (NK) cells are important components of the innate immune defense against infections and cancers. Signal transducer and activator of transcription 1 (STAT1) is a transcription factor that is essential for NK cell maturation and NK cell-dependent tumor surveillance. Two alternatively spliced isoforms of STAT1 exist: a full-length STAT1α and a C-terminally truncated STAT1ß isoform. Aberrant splicing is frequently observed in cancer cells and several anti-cancer drugs interfere with the cellular splicing machinery. To investigate whether NK cell-mediated tumor surveillance is affected by a switch in STAT1 splicing, we made use of knock-in mice expressing either only the STAT1α (Stat1α/α) or the STAT1ß (Stat1ß/ß ) isoform. NK cells from Stat1α/α mice matured normally and controlled transplanted tumor cells as efficiently as NK cells from wild-type mice. In contrast, NK cells from Stat1ß/ß mice showed impaired maturation and effector functions, albeit less severe than NK cells from mice that completely lack STAT1 (Stat1-/- ). Mechanistically, we show that NK cell maturation requires the presence of STAT1α in the niche rather than in NK cells themselves and that NK cell maturation depends on IFNγ signaling under homeostatic conditions. The impaired NK cell maturation in Stat1ß/ß mice was paralleled by decreased IL-15 receptor alpha (IL-15Rα) surface levels on dendritic cells, macrophages and monocytes. Treatment of Stat1ß/ß mice with exogenous IL-15/IL-15Rα complexes rescued NK cell maturation but not their effector functions. Collectively, our findings provide evidence that STAT1 isoforms are not functionally redundant in regulating NK cell activity and that the absence of STAT1α severely impairs, but does not abolish, NK cell-dependent tumor surveillance.


Asunto(s)
Células Asesinas Naturales/citología , Linfopoyesis/fisiología , Factor de Transcripción STAT1/inmunología , Animales , Trasplante de Médula Ósea , Línea Celular Tumoral , Citotoxicidad Inmunológica , Vigilancia Inmunológica/efectos de los fármacos , Vigilancia Inmunológica/inmunología , Factor 3 de Genes Estimulados por el Interferón/deficiencia , Factor 3 de Genes Estimulados por el Interferón/genética , Factor 3 de Genes Estimulados por el Interferón/inmunología , Interleucina-15/farmacología , Subunidad alfa del Receptor de Interleucina-15 , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Depleción Linfocítica , Tejido Linfoide/citología , Linfoma/inmunología , Linfoma/patología , Linfopoyesis/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Receptores de Interferón/deficiencia , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética , Organismos Libres de Patógenos Específicos , Bazo/citología , Receptor de Interferón gamma
16.
PLoS One ; 15(9): e0239231, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32997686

RESUMEN

It is controversially discussed whether immune-deficient mice experience severity in the absence of infection. Because a comprehensive analysis of the well-being of immune-deficient mice under specific pathogen free conditions is missing, we used a multi-parametric test analyzing, corticosterone, weight, nest building and facial expression over a period of 9 month to determine the well-being of two immune-deficient mouse lines (recombination activating gene 2- and interferon gamma receptor-deficient mice). We do not find evidence for severity when comparing immune-deficient mice to their heterozygous immune-competent littermates. Our data challenge the assumption that immune-deficiency per se regardless of housing conditions causes severity. Based on our study we propose to use objective non-invasive parameters determined by laboratory animal science for decisions concerning severity of immune-deficient mice.


Asunto(s)
Corticosterona/genética , Proteínas de Unión al ADN/genética , Interferón gamma/genética , Ratones SCID/genética , Animales , Linfocitos B/inmunología , Corticosterona/inmunología , Humanos , Infecciones/genética , Infecciones/inmunología , Riñón/metabolismo , Riñón/patología , Ratones , Ratones SCID/inmunología , Dolor/genética , Dolor/patología , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Transducción de Señal/genética , Linfocitos T/inmunología , Testosterona/genética , Receptor de Interferón gamma
17.
J Clin Immunol ; 40(7): 1045-1047, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32696284

RESUMEN

This is a report of a successful bone marrow transplant in an IFN-γR1 patient with progressive mycobacterial infection. PURPOSE: Hematopoietic cell transplant in patients with interferon gamma receptor deficiencies has been fraught with challenges, not the least of which is failure of engraftment and infectious complications. METHODS: This is a report of a successful hematopoietic cell transplant in an actively infected patient of advanced age. RESULTS: This case report shows successful engraftment and resolution of infection posttransplant using a matched related donor in a single institution. CONCLUSION: A successful curative HCT despite persistent, disseminated, nontuberculous mycobacterial infection in a patient with AD-IFNγR1 suggests that this approach, while difficult, may be useful in other patients with otherwise refractory disease.


Asunto(s)
Trasplante de Médula Ósea , Genes Dominantes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Receptores de Interferón/deficiencia , Trasplante de Células Madre Hematopoyéticas , Humanos , Infecciones por Mycobacterium/diagnóstico , Infecciones por Mycobacterium/etiología , Infecciones por Mycobacterium/terapia , Infecciones por Mycobacterium no Tuberculosas/diagnóstico , Infecciones por Mycobacterium no Tuberculosas/etiología , Infecciones por Mycobacterium no Tuberculosas/terapia , Índice de Severidad de la Enfermedad , Trasplante Homólogo , Resultado del Tratamiento , Receptor de Interferón gamma
18.
Front Immunol ; 11: 1161, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32676075

RESUMEN

Interferon-γ receptor 1 (IFNγR1) deficiency is one of the inborn errors of IFN-γ immunity underlying Mendelian Susceptibility to Mycobacterial Disease (MSMD). This molecular circuit plays a crucial role in regulating the interaction between dendritic cells (DCs) and T lymphocytes, thus affecting DCs activation, maturation, and priming of T cells involved in the immune response against intracellular pathogens. We studied a girl who developed at the age of 2.5 years a Mycobacterium avium infection characterized by disseminated necrotizing granulomatous lymphadenitis, and we compared her findings with other patients with the same genetic condition. The patient carried a heterozygous 818del4 mutation in the IFNGR1 gene responsible of autosomal dominant (AD) partial IFNγR1 deficiency. During the acute infection blood cells immunophenotyping showed a marked reduction in DCs counts, including both myeloid (mDCs) and plasmacytoid (pDCs) subsets, that reversed after successful prolonged antimicrobial therapy. Histology of her abdomen lymph node revealed a profound depletion of tissue pDCs, as compared to other age-matched granulomatous lymphadenitis of mycobacterial origin. Circulating DCs depletion was also observed in another patient with AD partial IFNγR1 deficiency during mycobacterial infection. To conclude, AD partial IFNγR1 deficiency can be associated with a transient decrease in both circulating and tissular DCs during acute mycobacterial infection, suggesting that DCs counts monitoring might constitute a useful marker of treatment response.


Asunto(s)
Células Dendríticas/inmunología , Infección por Mycobacterium avium-intracellulare/inmunología , Enfermedades de Inmunodeficiencia Primaria/inmunología , Receptores de Interferón/deficiencia , Preescolar , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Mutación , Enfermedades de Inmunodeficiencia Primaria/genética , Receptores de Interferón/genética , Receptor de Interferón gamma
19.
J Immunol ; 204(8): 2192-2202, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32152071

RESUMEN

Type I and III IFNs play diverse roles in bacterial infections, being protective for some but deleterious for others. Using RNA-sequencing transcriptomics we investigated lung gene expression responses to Bordetella pertussis infection in adult mice, revealing that type I and III IFN pathways may play an important role in promoting inflammatory responses. In B. pertussis-infected mice, lung type I/III IFN responses correlated with increased proinflammatory cytokine expression and with lung inflammatory pathology. In mutant mice with increased type I IFN receptor (IFNAR) signaling, B. pertussis infection exacerbated lung inflammatory pathology, whereas knockout mice with defects in type I IFN signaling had lower levels of lung inflammation than wild-type mice. Curiously, B. pertussis-infected IFNAR1 knockout mice had wild-type levels of lung inflammatory pathology. However, in response to infection these mice had increased levels of type III IFN expression, neutralization of which reduced lung inflammation. In support of this finding, B. pertussis-infected mice with a knockout mutation in the type III IFN receptor (IFNLR1) and double IFNAR1/IFNLR1 knockout mutant mice had reduced lung inflammatory pathology compared with that in wild-type mice, indicating that type III IFN exacerbates lung inflammation. In marked contrast, infant mice did not upregulate type I or III IFNs in response to B. pertussis infection and were protected from lethal infection by increased type I IFN signaling. These results indicate age-dependent effects of type I/III IFN signaling during B. pertussis infection and suggest that these pathways represent targets for therapeutic intervention in pertussis.


Asunto(s)
Infecciones por Bordetella/inmunología , Bordetella pertussis/inmunología , Interferón Tipo I/inmunología , Interferones/inmunología , Infecciones del Sistema Respiratorio/inmunología , Factores de Edad , Animales , Infecciones por Bordetella/genética , Bordetella pertussis/patogenicidad , Femenino , Interferón Tipo I/genética , Interferones/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Infecciones del Sistema Respiratorio/genética , Análisis de Secuencia de ARN , Transducción de Señal/genética , Transducción de Señal/inmunología , Transcriptoma , Interferón lambda
20.
Cell Rep ; 30(12): 4041-4051.e4, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32209467

RESUMEN

During the 2013-2016 Ebola virus (EBOV) epidemic, a significant number of patients admitted to Ebola treatment units were co-infected with Plasmodium falciparum, a predominant agent of malaria. However, there is no consensus on how malaria impacts EBOV infection. The effect of acute Plasmodium infection on EBOV challenge was investigated using mouse-adapted EBOV and a biosafety level 2 (BSL-2) model virus. We demonstrate that acute Plasmodium infection protects from lethal viral challenge, dependent upon interferon gamma (IFN-γ) elicited as a result of parasite infection. Plasmodium-infected mice lacking the IFN-γ receptor are not protected. Ex vivo incubation of naive human or mouse macrophages with sera from acutely parasitemic rodents or macaques programs a proinflammatory phenotype dependent on IFN-γ and renders cells resistant to EBOV infection. We conclude that acute Plasmodium infection can safeguard against EBOV by the production of protective IFN-γ. These findings have implications for anti-malaria therapies administered during episodic EBOV outbreaks in Africa.


Asunto(s)
Resistencia a la Enfermedad/inmunología , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/complicaciones , Fiebre Hemorrágica Ebola/inmunología , Interferón gamma/metabolismo , Malaria/complicaciones , Plasmodium falciparum/fisiología , Animales , Femenino , Glicoproteínas/metabolismo , Fiebre Hemorrágica Ebola/prevención & control , Macrófagos Peritoneales/patología , Malaria/parasitología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptor de Interferón alfa y beta/metabolismo , Receptores de Interferón/deficiencia , Receptores de Interferón/metabolismo , Vesiculovirus/fisiología , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA