Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Nat Microbiol ; 8(8): 1468-1479, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37429907

RESUMEN

Although dengue virus (DENV) infection typically causes asymptomatic disease, DENV-infected patients can experience severe complications. A risk factor for symptomatic disease is pre-existing anti-DENV IgG antibodies. Cellular assays suggested that these antibodies can enhance viral infection of Fcγ receptor (FcγR)-expressing myeloid cells. Recent studies, however, revealed more complex interactions between anti-DENV antibodies and specific FcγRs by demonstrating that modulation of the IgG Fc glycan correlates with disease severity. To investigate the in vivo mechanisms of antibody-mediated dengue pathogenesis, we developed a mouse model for dengue disease that recapitulates the unique complexity of human FcγRs. In in vivo mouse models of dengue disease, we discovered that the pathogenic activity of anti-DENV antibodies is exclusively mediated through engagement of FcγRIIIa on splenic macrophages, resulting in inflammatory sequelae and mortality. These findings highlight the importance of IgG-FcγRIIIa interactions in dengue, with important implications for the design of safer vaccination approaches and effective therapeutic strategies.


Asunto(s)
Virus del Dengue , Dengue , Humanos , Animales , Ratones , Receptores de IgG , Macrófagos , Inmunoglobulina G
2.
Nat Commun ; 14(1): 2853, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-37202422

RESUMEN

Immunoglobulin G (IgG) antibodies contain a complex N-glycan embedded in the hydrophobic pocket between its heavy chain protomers. This glycan contributes to the structural organization of the Fc domain and determines its specificity for Fcγ receptors, thereby dictating distinct cellular responses. The variable construction of this glycan structure leads to highly-related, but non-equivalent glycoproteins known as glycoforms. We previously reported synthetic nanobodies that distinguish IgG glycoforms. Here, we present the structure of one such nanobody, X0, in complex with the Fc fragment of afucosylated IgG1. Upon binding, the elongated CDR3 loop of X0 undergoes a conformational shift to access the buried N-glycan and acts as a 'glycan sensor', forming hydrogen bonds with the afucosylated IgG N-glycan that would otherwise be sterically hindered by the presence of a core fucose residue. Based on this structure, we designed X0 fusion constructs that disrupt pathogenic afucosylated IgG1-FcγRIIIa interactions and rescue mice in a model of dengue virus infection.


Asunto(s)
Inmunoglobulina G , Receptores de IgG , Animales , Ratones , Glicosilación , Receptores de IgG/metabolismo , Fragmentos Fc de Inmunoglobulinas/metabolismo , Polisacáridos/química
3.
bioRxiv ; 2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36747840

RESUMEN

Immunoglobulin G (IgG) antibodies contain a single, complex N -glycan on each IgG heavy chain protomer embedded in the hydrophobic pocket between its Cγ2 domains. The presence of this glycan contributes to the structural organization of the Fc domain and determines its specificity for Fcγ receptors, thereby determining distinct cellular responses. On the Fc, the variable construction of this glycan structure leads to a family of highly-related, but non-equivalent glycoproteins known as glycoforms. We previously reported the development of synthetic nanobodies that distinguish IgG glycoforms without cross-reactivity to off-target glycoproteins or free glycans. Here, we present the X-ray crystal structure of one such nanobody, X0, in complex with its specific binding partner, the Fc fragment of afucosylated IgG1. Two X0 nanobodies bind a single afucosylated Fc homodimer at the upper Cγ2 domain, making both protein-protein and protein-carbohydrate contacts and overlapping the binding site for Fcγ receptors. Upon binding, the elongated CDR3 loop of X0 undergoes a conformational shift to access the buried N -glycan and acts as a 'glycan sensor', forming hydrogen bonds with the afucosylated IgG N -glycan that would otherwise be sterically hindered by the presence of a core fucose residue. Based on this structure, we designed X0 fusion constructs that disrupt pathogenic afucosylated IgG1-FcγRIIIa interactions and rescue mice in a model of dengue virus infection.

4.
Nature ; 599(7885): 465-470, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34547765

RESUMEN

Monoclonal antibodies with neutralizing activity against SARS-CoV-2 have demonstrated clinical benefits in cases of mild-to-moderate SARS-CoV-2 infection, substantially reducing the risk for hospitalization and severe disease1-4. Treatment generally requires the administration of high doses of these monoclonal antibodies and has limited efficacy in preventing disease complications or mortality among hospitalized patients with COVID-195. Here we report the development and evaluation of anti-SARS-CoV-2 monoclonal antibodies with optimized Fc domains that show superior potency for prevention or treatment of COVID-19. Using several animal disease models of COVID-196,7, we demonstrate that selective engagement of activating Fcγ receptors results in improved efficacy in both preventing and treating disease-induced weight loss and mortality, significantly reducing the dose required to confer full protection against SARS-CoV-2 challenge and for treatment of pre-infected animals. Our results highlight the importance of Fcγ receptor pathways in driving antibody-mediated antiviral immunity and exclude the possibility of pathogenic or disease-enhancing effects of Fcγ receptor engagement of anti-SARS-CoV-2 antibodies upon infection. These findings have important implications for the development of Fc-engineered monoclonal antibodies with optimal Fc-effector function and improved clinical efficacy against COVID-19 disease.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , COVID-19/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Cricetinae , Modelos Animales de Enfermedad , Femenino , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/farmacología , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Masculino , Ratones , Profilaxis Pre-Exposición , Receptores de IgG/química , Receptores de IgG/inmunología , Resultado del Tratamiento
5.
Harefuah ; 160(8): 488-492, 2021 Aug.
Artículo en Hebreo | MEDLINE | ID: mdl-34396721

RESUMEN

INTRODUCTION: The World Health Organization declared that the corona virus disease (COVID 19) is a global pandemic and public health emergency, following the outbreak in Wuhan, China, in December 2019. Fever, dry cough and fatigue were the main manifestations, but the primary concern is cases that deteriorated to severe pneumonia and acute respiratory distress syndrome (ARDS). The outbreak of COVID- 19 in Israel, in March 2020 caused major changes in the routine work in hospitals. Hasharon hospital, a small community hospital in Petah Tikvah was converted to corona patients' admissions only. In accordance with this new reality, the physiotherapy service prepared itself to provide treatments tailored to the clinical needs of the Covid 19 patients and the quarantine conditions of these patients' hospitalization. During a period of 2 months, we built a unique protocol adjusted to isolated patients and staff, including telemedicine and hands-on treatment. This article summarizes our experience of physiotherapy treatment in the acute and rehabilitation period.


Asunto(s)
COVID-19 , Telemedicina , Humanos , Pandemias , Modalidades de Fisioterapia , SARS-CoV-2
6.
Eur J Immunol ; 51(9): 2218-2224, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34268737

RESUMEN

Neutrophils play a crucial role in immune defense against and clearance of uropathogenic Escherichia coli (UPEC)-mediated urinary tract infection, the most common bacterial infection in healthy humans. CD300a is an inhibitory receptor that binds phosphatidylserine and phosphatidylethanolamine, presented on the membranes of apoptotic cells. CD300a binding to phosphatidylserine and phosphatidylethanolamine, also known as the "eat me" signal, mediates immune tolerance to dying cells. Here, we demonstrate for the first time that CD300a plays an important role in the neutrophil-mediated immune response to UPEC-induced urinary tract infection. We show that CD300a-deficient neutrophils have impaired phagocytic abilities and despite their increased accumulation at the site of infection, they are unable to reduce bacterial burden in the bladder, which results in significant exacerbation of infection and worse host outcome. Finally, we demonstrate that UPEC's pore forming toxin α-hemolysin induces upregulation of the CD300a ligand on infected bladder epithelial cells, signaling to neutrophils to be cleared.


Asunto(s)
Infecciones por Escherichia coli/prevención & control , Neutrófilos/inmunología , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/inmunología , Infecciones Urinarias/inmunología , Escherichia coli Uropatógena/inmunología , Animales , Apoptosis/inmunología , Infecciones por Escherichia coli/inmunología , Proteínas de Escherichia coli/metabolismo , Femenino , Técnicas de Inactivación de Genes , Proteínas Hemolisinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Fagocitosis/genética , Fagocitosis/inmunología , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Receptores Inmunológicos/genética , Vejiga Urinaria/inmunología , Vejiga Urinaria/microbiología , Vejiga Urinaria/patología , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/crecimiento & desarrollo
7.
Res Sq ; 2021 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-34075373

RESUMEN

Monoclonal antibodies (mAbs) with neutralizing activity against SARS-CoV-2 have demonstrated clinical benefit in cases of mild to moderate SARS-CoV-2 infection, substantially reducing the risk for hospitalization and severe disease1-4. Treatment generally requires the administration of high doses of these mAbs with limited efficacy in preventing disease complications or mortality among hospitalized COVID-19 patients5. Here we report the development and evaluation of Fc-optimized anti-SARS-CoV-2 mAbs with superior potency to prevent or treat COVID-19 disease. In several animal models of COVID-19 disease6,7, we demonstrate that selective engagement of activating FcγRs results in improved efficacy in both preventing and treating disease-induced weight loss and mortality, significantly reducing the dose required to confer full protection upon SARS-CoV-2 challenge and treatment of pre-infected animals. Our results highlight the importance of FcγR pathways in driving antibody-mediated antiviral immunity, while excluding any pathogenic or disease-enhancing effects of FcγR engagement of anti-SARS-CoV-2 antibodies upon infection. These findings have important implications for the development of Fc-engineered mAbs with optimal Fc effector function and improved clinical efficacy against COVID-19 disease.

8.
Proc Natl Acad Sci U S A ; 117(45): 28046-28055, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33093202

RESUMEN

An essential mechanism for severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection begins with the viral spike protein binding to the human receptor protein angiotensin-converting enzyme II (ACE2). Here, we describe a stepwise engineering approach to generate a set of affinity optimized, enzymatically inactivated ACE2 variants that potently block SARS-CoV-2 infection of cells. These optimized receptor traps tightly bind the receptor binding domain (RBD) of the viral spike protein and prevent entry into host cells. We first computationally designed the ACE2-RBD interface using a two-stage flexible protein backbone design process that improved affinity for the RBD by up to 12-fold. These designed receptor variants were affinity matured an additional 14-fold by random mutagenesis and selection using yeast surface display. The highest-affinity variant contained seven amino acid changes and bound to the RBD 170-fold more tightly than wild-type ACE2. With the addition of the natural ACE2 collectrin domain and fusion to a human immunoglobulin crystallizable fragment (Fc) domain for increased stabilization and avidity, the most optimal ACE2 receptor traps neutralized SARS-CoV-2-pseudotyped lentivirus and authentic SARS-CoV-2 virus with half-maximal inhibitory concentrations (IC50s) in the 10- to 100-ng/mL range. Engineered ACE2 receptor traps offer a promising route to fighting infections by SARS-CoV-2 and other ACE2-using coronaviruses, with the key advantage that viral resistance would also likely impair viral entry. Moreover, such traps can be predesigned for viruses with known entry receptors for faster therapeutic response without the need for neutralizing antibodies isolated from convalescent patients.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/química , Diseño de Fármacos , Ingeniería de Proteínas/métodos , Glicoproteína de la Espiga del Coronavirus/metabolismo , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/genética , Antivirales/metabolismo , Sitios de Unión , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Mutación , Biblioteca de Péptidos , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae , Glicoproteína de la Espiga del Coronavirus/química
9.
bioRxiv ; 2020 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-32766586

RESUMEN

An essential mechanism for SARS-CoV-1 and -2 infection begins with the viral spike protein binding to the human receptor protein angiotensin-converting enzyme II (ACE2). Here we describe a stepwise engineering approach to generate a set of affinity optimized, enzymatically inactivated ACE2 variants that potently block SARS-CoV-2 infection of cells. These optimized receptor traps tightly bind the receptor binding domain (RBD) of the viral spike protein and prevent entry into host cells. We first computationally designed the ACE2-RBD interface using a two-stage flexible protein backbone design process that improved affinity for the RBD by up to 12-fold. These designed receptor variants were affinity matured an additional 14-fold by random mutagenesis and selection using yeast surface display. The highest affinity variant contained seven amino acid changes and bound to the RBD 170-fold more tightly than wild-type ACE2. With the addition of the natural ACE2 collectrin domain and fusion to a human Fc domain for increased stabilization and avidity, the most optimal ACE2 receptor traps neutralized SARS-CoV-2 pseudotyped lentivirus and authentic SARS-CoV-2 virus with half-maximal inhibitory concentrations (IC50) in the 10-100 ng/ml range. Engineered ACE2 receptor traps offer a promising route to fighting infections by SARS-CoV-2 and other ACE2-utilizing coronaviruses, with the key advantage that viral resistance would also likely impair viral entry. Moreover, such traps can be predesigned for viruses with known entry receptors for faster therapeutic response without the need for neutralizing antibodies isolated or generated from convalescent patients.

10.
Sci Rep ; 9(1): 1351, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30718650

RESUMEN

Rheumatoid Arthritis (RA) causes chronic inflammation of joints. The cytokines TNFα and IFNγ are central players in RA, however their source has not been fully elucidated. Natural Killer (NK) cells are best known for their role in elimination of viral-infected and transformed cells, and they secrete pro-inflammatory cytokines. NK cells are present in the synovial fluids (SFs) of RA patients and are considered to be important in bone destruction. However, the phenotype and function of NK cells in the SFs of patients with erosive deformative RA (DRA) versus non-deformative RA (NDRA) is poorly characterized. Here we characterize the NK cell populations present in the blood and SFs of DRA and NDRA patients. We demonstrate that a distinct population of activated synovial fluid NK (sfNK) cells constitutes a large proportion of immune cells found in the SFs of DRA patients. We discovered that although sfNK cells in both DRA and NDRA patients have similar phenotypes, they function differently. The DRA sfNK secrete more TNFα and IFNγ upon exposure to IL-2 and IL-15. Consequently, we suggest that sfNK cells may be a marker for more severely destructive RA disease.


Asunto(s)
Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Células Asesinas Naturales/inmunología , Líquido Sinovial/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Artritis Reumatoide/sangre , Estudios de Casos y Controles , Femenino , Humanos , Interferón gamma/metabolismo , Masculino , Persona de Mediana Edad , Fenotipo , Receptores de Células Asesinas Naturales/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
iScience ; 11: 466-473, 2019 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-30661002

RESUMEN

Long, non-coding RNAs (lncRNAs) are involved in the regulation of many cellular processes. The lncRNA IFNG-AS1 was found to strongly influence the responses to several pathogens in mice by increasing interferon gamma (IFNγ) secretion. Studies have looked at IFNG-AS1 in T cells, yet IFNG-AS1 function in natural killer cells (NKs), an important source of IFNγ, remains unknown. Here, we show a previously undescribed sequence of IFNG-AS1 and report that it may be more abundant in cells than previously thought. Using primary human NKs and an NK line with IFNG-AS1 overexpression, we show that IFNG-AS1 is quickly induced upon NK cell activation, and that IFNG-AS1 overexpression leads to increased IFNγ secretion. Taken together, our work expands IFNG-AS1's activity to the innate arm of the type I immune response, helping to explain its notable effect in animal models of disease.

12.
Eur J Immunol ; 49(2): 228-241, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30536875

RESUMEN

Natural killer (NK) cells are innate lymphocytes that efficiently eliminate cancerous and infected cells. NKp46 is an important NK activating receptor shown to participate in recognition and activation of NK cells against pathogens, tumor cells, virally infected cells, and self-cells in autoimmune conditions, including type I and II diabetes. However, some of the NKp46 ligands are unknown and therefore investigating human NKp46 activity and its critical role in NK cell biology is problematic. We developed a unique anti-human NKp46 monocloncal antibody, denoted hNKp46.02 (02). The 02 mAb can induce receptor internalization and degradation. By binding to a unique epitope on a particular domain of NKp46, 02 lead NKp46 to lysosomal degradation. This downregulation therefore enables the investigation of all NKp46 activities. Indeed, using the 02 mAb we determined NK cell targets which are critically dependent on NKp46 activity, including certain tumor cells lines and human pancreatic beta cells. Most importantly, we showed that a toxin-conjugated 02 inhibits the growth of NKp46-positive cells; thus, exemplifying the potential of 02 in becoming an immunotherapeutic drug to treat NKp46-dependent diseases, such as, type I diabetes and NK and T cell related malignancies.


Asunto(s)
Anticuerpos Monoclonales/química , Antígenos Ly/metabolismo , Diabetes Mellitus Tipo 1 , Células Asesinas Naturales/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias , Animales , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Células Jurkat , Células K562 , Ratones , Neoplasias/diagnóstico , Neoplasias/metabolismo
13.
J Virol ; 92(17)2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29950412

RESUMEN

Natural killer (NK) cells are lymphocytes of the innate immune system capable of killing hazardous cells, including virally infected cells. NK cell-mediated killing is triggered by activating receptors. Prominent among these is the activating receptor NKG2D, which binds several stress-induced ligands, among them major histocompatibility complex (MHC) class I-related chain A (MICA). Most of the human population is persistently infected with human cytomegalovirus (HCMV), a virus which employs multiple immune evasion mechanisms, many of which target NK cell responses. HCMV infection is mostly asymptomatic, but in congenitally infected neonates and in immunosuppressed patients it can lead to serious complications and mortality. Here we discovered that an HCMV protein named UL148A whose role was hitherto unknown is required for evasion of NK cells. We demonstrate that UL148A-deficient HCMV strains are impaired in their ability to downregulate MICA expression. We further show that when expressed by itself, UL148A is not sufficient for MICA targeting, but rather acts in concert with an unknown viral factor. Using inhibitors of different cellular degradation pathways, we show that UL148A targets MICA for lysosomal degradation. Finally, we show that UL148A-mediated MICA downregulation hampers NK cell-mediated killing of HCMV-infected cells. Discovering the full repertoire of HCMV immune evasion mechanisms will lead to a better understanding of the ability of HCMV to persist in the host and may also promote the development of new vaccines and drugs against HCMV.IMPORTANCE Human cytomegalovirus (HCMV) is a ubiquitous pathogen which is usually asymptomatic but that can cause serious complications and mortality in congenital infections and in immunosuppressed patients. One of the difficulties in developing novel vaccines and treatments for HCMV is its remarkable ability to evade our immune system. In particular, HCMV directs significant efforts to thwarting cells of the innate immune system known as natural killer (NK) cells. These cells are crucial for successful control of HCMV infection, and yet our understanding of the mechanisms which HCMV utilizes to elude NK cells is partial at best. In the present study, we discovered that a protein encoded by HCMV which had no known function is important for preventing NK cells from killing HCMV-infected cells. This knowledge can be used in the future for designing more-efficient HCMV vaccines and for formulating novel therapies targeting this virus.


Asunto(s)
Citomegalovirus/fisiología , Antígenos de Histocompatibilidad Clase I/genética , Evasión Inmune , Células Asesinas Naturales/inmunología , Proteínas Virales de Fusión/fisiología , Línea Celular , Citomegalovirus/genética , Citomegalovirus/inmunología , Regulación hacia Abajo , Humanos , Activación de Linfocitos , Proteínas Virales de Fusión/genética
14.
Immunity ; 48(5): 951-962.e5, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29768178

RESUMEN

Natural killer cells (NKs) are abundant in the human decidua, regulating trophoblast invasion and angiogenesis. Several diseases of poor placental development are associated with first pregnancies, so we thus looked to characterize differences in decidual NKs (dNKs) in first versus repeated pregnancies. We discovered a population found in repeated pregnancies, which has a unique transcriptome and epigenetic signature, and is characterized by high expression of the receptors NKG2C and LILRB1. We named these cells Pregnancy Trained decidual NK cells (PTdNKs). PTdNKs have open chromatin around the enhancers of IFNG and VEGFA. Activation of PTdNKs led to increased production and secretion of IFN-γ and VEGFα, with the latter supporting vascular sprouting and tumor growth. The precursors of PTdNKs seem to be found in the endometrium. Because repeated pregnancies are associated with improved placentation, we propose that PTdNKs, which are present primarily in repeated pregnancies, might be involved in proper placentation.


Asunto(s)
Memoria Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Transcriptoma/inmunología , Útero/inmunología , Animales , Línea Celular Tumoral , Decidua/inmunología , Decidua/metabolismo , Femenino , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Embarazo , Útero/citología , Factor A de Crecimiento Endotelial Vascular/inmunología , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Sci Rep ; 7(1): 8661, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28819195

RESUMEN

Human cytomegalovirus (HCMV) is a major human pathogen, causing serious diseases in immunocompromised populations and congenially infected neonates. One of the main immune cells acting against the virus are Natural Killer (NK) cells. Killing by NK cells is mediated by a small family of activating receptors such as NKp30 that interact with the cellular ligand B7-H6. The outcome of B7-H6-NKp30 interaction was, so far, mainly studied with regard to NK recognition and killing of tumors. Here, we demonstrated that the expression of B7-H6 is upregulated following HCMV infection and that HCMV uses two of its genes: US18 and US20, to interfere with B7-H6 surface expression, in a mechanism involving endosomal degradation, in order to evade NK cell recognition.


Asunto(s)
Antígenos B7/genética , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Interacciones Huésped-Patógeno , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Proteínas Virales/genética , Antígenos B7/metabolismo , Línea Celular , Infecciones por Citomegalovirus/metabolismo , Citotoxicidad Inmunológica , Regulación de la Expresión Génica , Orden Génico , Genoma Viral , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Lisosomas/metabolismo , Receptor 3 Gatillante de la Citotoxidad Natural/genética , Receptor 3 Gatillante de la Citotoxidad Natural/metabolismo , Virulencia/inmunología
16.
J Immunol ; 198(9): 3662-3670, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28356383

RESUMEN

NK cells are part of the innate immune system, and are able to identify and kill hazardous cells. The discrimination between normal and hazardous cells is possible due to an array of inhibitory and activating receptors. NKG2D is one of the prominent activating receptors expressed by all human NK cells. This receptor binds stress-induced ligands, including human MICA, MICB, and UL16-binding proteins 1-6. The interaction between NKG2D and its ligands facilitates the elimination of cells under cellular stress, such as tumor transformation. However, the mechanisms regulating the expression of these ligands are still not well understood. Under normal conditions, the NKG2D ligands were shown to be posttranscriptionally regulated by cellular microRNAs and RNA-binding proteins (RBPs). Thus far, only the 3' untranslated regions (UTRs) of MICA, MICB, and UL16-binding protein 2 were shown to be regulated by RBPs and microRNAs, usually resulting in their downregulation. In this study we investigated whether MICB expression is controlled by RBPs through its 5'UTR. We used an RNA pull-down assay followed by mass spectrometry and identified vigilin, a ubiquitously expressed multifunctional RNA-binding protein. We demonstrated that vigilin binds and negatively regulates MICB expression through its 5'UTR. Additionally, vigilin downregulation in target cells led to a significant increase in NK cell activation against said target cells. Taken together, we have discovered a novel mode of MICB regulation.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Vigilancia Inmunológica , Células Asesinas Naturales/inmunología , Proteínas de Unión al ARN/metabolismo , Estrés Fisiológico/inmunología , Regiones no Traducidas 5'/genética , Línea Celular Tumoral , Regulación de la Expresión Génica/genética , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Ligandos , Activación de Linfocitos , Subfamilia K de Receptores Similares a Lectina de Células NK/agonistas , Unión Proteica , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética
17.
Eur J Immunol ; 47(4): 692-703, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28191644

RESUMEN

Natural killer (NK) cells are capable of killing various pathogens upon stimulation of activating receptors. Human metapneumovirus (HMPV) is a respiratory virus, which was discovered in 2001 and is responsible for acute respiratory tract infection in infants and children worldwide. HMPV infection is very common, infecting around 70% of all children under the age of five. Under immune suppressive conditions, HMPV infection can be fatal. Not much is known on how NK cells respond to HMPV. In this study, using reporter assays and NK-cell cytotoxicity assays performed with human and mouse NK cells, we demonstrated that the NKp46-activating receptor and its mouse orthologue Ncr1, both members of the natural cytotoxicity receptor (NCR) family, recognized an unknown ligand expressed by HMPV-infected human cells. We demonstrated that MHC class I is upregulated and MICA is downregulated upon HMPV infection. We also characterized mouse NK-cell phenotype in the blood and the lungs of HMPV-infected mice and found that lung NK cells are more activated and expressing NKG2D, CD43, CD27, KLRG1, and CD69 compared to blood NK cells regardless of HMPV infection. Finally, we demonstrated, using Ncr1-deficient mice, that NCR1 plays a critical role in controlling HMPV infection.


Asunto(s)
Antígenos Ly/metabolismo , Células Asesinas Naturales/inmunología , Pulmón/inmunología , Metapneumovirus/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Infecciones por Paramyxoviridae/inmunología , Animales , Antígenos Ly/genética , Niño , Citotoxicidad Inmunológica , Células HEK293 , Humanos , Lactante , Células Asesinas Naturales/virología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Carga Viral
18.
Cell Host Microbe ; 20(4): 527-534, 2016 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-27736647

RESUMEN

Natural killer (NK) cells form an important arm of the innate immune system and function to combat a wide range of invading pathogens, ranging from viruses to bacteria. However, the means by which NK cells accomplish recognition of pathogens with a limited repertoire of receptors remain largely unknown. In the current study, we describe the recognition of an emerging fungal pathogen, Candida glabrata, by the human NK cytotoxic receptor NKp46 and its mouse ortholog, NCR1. Using NCR1 knockout mice, we observed that this receptor-mediated recognition was crucial for controlling C. glabrata infection in vitro and in vivo. Finally, we delineated the fungal ligands to be the C. glabrata adhesins Epa1, Epa6, and Epa7 and demonstrated that clearance of systemic C. glabrata infections in vivo depends on their recognition by NCR1. As NKp46 and NCR1 have been previously shown to bind viral adhesion receptors, we speculate that NKp46/NCR1 may be a novel type of pattern recognition receptor.


Asunto(s)
Antígenos Ly/metabolismo , Candida glabrata/inmunología , Proteínas Fúngicas/metabolismo , Células Asesinas Naturales/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Animales , Antígenos Ly/genética , Candidiasis/inmunología , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos BALB C , Ratones Noqueados , Receptor 1 Gatillante de la Citotoxidad Natural/genética
19.
Cell Rep ; 17(4): 949-956, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27760325

RESUMEN

Herpes simplex virus 1 (HSV1) is a ubiquitous human pathogen that utilizes variable mechanisms to evade immune surveillance. The glycosylphosphatidylinositol (GPI) anchoring pathway is a multistep process in which a myriad of different proteins are covalently attached to a GPI moiety to be presented on the cell surface. Among the different GPI-anchored proteins there are many with immunological importance. We present evidence that the HSV1-encoded miR H8 directly targets PIGT, a member of the protein complex that covalently attaches proteins to GPI in the final step of GPI anchoring. This results in a membrane down-modulation of several different immune-related, GPI-anchored proteins, including ligands for natural killer-activating receptors and the prominent viral restriction factor tetherin. Thus, we suggest that by utilizing just one of dozens of miRNAs encoded by HSV1, the virus can counteract the host immune response at several key points.


Asunto(s)
Glicosilfosfatidilinositoles/metabolismo , Herpesvirus Humano 1/genética , Evasión Inmune , MicroARNs/metabolismo , Aciltransferasas/metabolismo , Antígenos CD/metabolismo , Citotoxicidad Inmunológica , Regulación hacia Abajo/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Células Asesinas Naturales/metabolismo , Ligandos , MicroARNs/genética
20.
Oncotarget ; 7(41): 66468-66479, 2016 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-27634893

RESUMEN

The innate sensing system is equipped with PRRs specialized in recognizing molecular structures (PAMPs) of various pathogens. This leads to the induction of anti-viral genes and inhibition of virus growth. Human Metapneumovirus (HMPV) is a major respiratory virus that causes an upper and lower respiratory tract infection in children. In this study we show that upon HMPV infection, the innate sensing system detects the viral RNA through the RIG-I sensor leading to induction of CEACAM1 expression. We further show that CEACAM1 is induced via binding of IRF3 to the CEACAM1 promoter. We demonstrate that induction of CEACAM1 suppresses the viral loads via inhibition of the translation machinery in the infected cells in an SHP2-dependent manner. In summary, we show here that HMPV-infected cells upregulates CEACAM1 to restrict HMPV infection.


Asunto(s)
Antígenos CD/inmunología , Moléculas de Adhesión Celular/inmunología , Inmunidad Innata/inmunología , Infecciones por Paramyxoviridae/inmunología , Animales , Antígenos CD/biosíntesis , Moléculas de Adhesión Celular/biosíntesis , Chlorocebus aethiops , Humanos , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/virología , Regulación hacia Arriba , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...