Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Transplant ; 24(5): 743-754, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38097018

RESUMEN

Antibody-mediated rejection (ABMR) is a leading cause of graft failure. Emerging evidence suggests a significant contribution of natural killer (NK) cells to microvascular inflammation (MVI). We investigated the influence of genetically determined NK cell functionality on ABMR development and activity. The study included 86 kidney transplant recipients subjected to systematic biopsies triggered by donor-specific antibody detection. We performed killer immunoglobulin-like receptor typing to predict missing self and genotyped polymorphisms determining NK cell functionality (FCGR3AV/F158 [rs396991], KLRC2wt/del, KLRK1HNK/LNK [rs1049174], rs9916629-C/T). Fifty patients had ABMR with considerable MVI and elevated NK cell transcripts. Missing self was not related to MVI. Only KLRC2wt/wt showed an association (MVI score: 2 [median; interquartile range: 0-3] vs 0 [0-1] in KLRC2wt/del recipients; P = .001) and remained significant in a proportional odds multivariable model (odds ratio, 7.84; 95% confidence interval, 2.37-30.47; P = .001). A sum score incorporating all polymorphisms and missing self did not outperform a score including only KLRC2 and FCGR3A variants, which were predictive in univariable analysis. NK cell genetics did not affect graft functional decline and survival. In conclusion, a functional KLRC2 polymorphism emerged as an independent determinant of ABMR activity, without a considerable contribution of missing self and other NK cell gene polymorphisms.


Asunto(s)
Rechazo de Injerto , Supervivencia de Injerto , Inflamación , Isoanticuerpos , Trasplante de Riñón , Células Asesinas Naturales , Donantes de Tejidos , Humanos , Células Asesinas Naturales/inmunología , Rechazo de Injerto/inmunología , Rechazo de Injerto/etiología , Rechazo de Injerto/patología , Trasplante de Riñón/efectos adversos , Masculino , Femenino , Persona de Mediana Edad , Donantes de Tejidos/provisión & distribución , Isoanticuerpos/inmunología , Pronóstico , Inflamación/inmunología , Estudios de Seguimiento , Supervivencia de Injerto/inmunología , Adulto , Factores de Riesgo , Microvasos/patología , Microvasos/inmunología , Genotipo , Fallo Renal Crónico/cirugía , Fallo Renal Crónico/inmunología , Fallo Renal Crónico/genética , Pruebas de Función Renal , Biomarcadores/análisis , Biomarcadores/metabolismo
2.
Kidney Int ; 101(4): 692-710, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34915041

RESUMEN

Detection of mismatched human leukocyte antigens by adaptive immune cells is considered as the main cause of transplant rejection, leading to either T-cell mediated rejection or antibody-mediated rejection. This canonical view guided the successful development of immunosuppressive therapies and shaped the diagnostic Banff classification for kidney transplant rejection that is used in clinics worldwide. However, several observations have recently emerged that question this dichotomization between T-cell mediated rejection and antibody-mediated rejection, related to heterogeneity in the serology, histology, and prognosis of the rejection phenotypes. In parallel, novel insights were obtained concerning the dynamics of donor-specific anti-human leukocyte antigen antibodies, the immunogenicity of donor-recipient non-human leukocyte antigen mismatches, and the autoreactivity against self-antigens. Moreover, the potential of innate allorecognition was uncovered, as exemplified by natural killer cell-mediated microvascular inflammation through missing self, and by the emerging evidence on monocyte-driven allorecognition. In this review, we highlight the gaps in the current classification of rejection, provide an overview of the expanding insights into the mechanisms of allorecognition, and critically appraise how these could improve our understanding and clinical approach to kidney transplant rejection. We argue that consideration of the complex interplay of various allorecognition mechanisms can foster a more integrated view of kidney transplant rejection and can lead to improved risk stratification, targeted therapies, and better outcome after kidney transplantation.


Asunto(s)
Trasplante de Riñón , Anticuerpos , Rechazo de Injerto/diagnóstico , Rechazo de Injerto/etiología , Rechazo de Injerto/prevención & control , Antígenos HLA , Humanos , Terapia de Inmunosupresión , Trasplante de Riñón/efectos adversos , Complicaciones Posoperatorias , Donantes de Tejidos
3.
J Am Soc Nephrol ; 32(8): 2070-2082, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34301794

RESUMEN

BACKGROUND: Circulating anti-HLA donor-specific antibodies (HLA-DSA) are often absent in kidney transplant recipients with microvascular inflammation (MVI). Missing self, the inability of donor endothelial cells to provide HLA I-mediated signals to inhibitory killer cell Ig-like receptors (KIRs) on recipient natural killer cells, can cause endothelial damage in vitro, and has been associated with HLA-DSA-negative MVI. However, missing self's clinical importance as a nonhumoral trigger of allograft rejection remains unclear. METHODS: In a population-based study of 924 consecutive kidney transplantations between March 2004 and February 2013, we performed high-resolution donor and recipient HLA typing and recipient KIR genotyping. Missing self was defined as the absence of A3/A11, Bw4, C1, or C2 donor genotype, with the presence of the corresponding educated recipient inhibitory KIR gene. RESULTS: We identified missing self in 399 of 924 transplantations. Co-occurrence of missing self types had an additive effect in increasing MVI risk, with a threshold at two concurrent types (hazard ratio [HR], 1.78; 95% confidence interval [95% CI], 1.26 to 2.53), independent of HLA-DSA (HR, 5.65; 95% CI, 4.01 to 7.96). Missing self and lesions of cellular rejection were not associated. No HLA-DSAs were detectable in 146 of 222 recipients with MVI; 28 of the 146 had at least two missing self types. Missing self associated with transplant glomerulopathy after MVI (HR, 2.51; 95% CI, 1.12 to 5.62), although allograft survival was better than with HLA-DSA-associated MVI. CONCLUSION: Missing self specifically and cumulatively increases MVI risk after kidney transplantation, independent of HLA-DSA. Systematic evaluation of missing self improves understanding of HLA-DSA-negative MVI and might be relevant for improved diagnostic classification and patient risk stratification.


Asunto(s)
Rechazo de Injerto/inmunología , Antígenos HLA/genética , Antígenos HLA/inmunología , Células Asesinas Naturales/inmunología , Receptores KIR/genética , Vasculitis/genética , Adulto , Anciano , Anticuerpos/sangre , Femenino , Genotipo , Supervivencia de Injerto , Antígeno HLA-A11/genética , Antígeno HLA-A11/inmunología , Antígeno HLA-A3/genética , Antígeno HLA-A3/inmunología , Antígenos HLA-B/genética , Antígenos HLA-B/inmunología , Antígenos HLA-C/genética , Antígenos HLA-C/inmunología , Prueba de Histocompatibilidad , Humanos , Trasplante de Riñón , Masculino , Microvasos , Persona de Mediana Edad , Receptores KIR2DL2/genética , Receptores KIR2DL3/genética , Donantes de Tejidos , Receptores de Trasplantes , Vasculitis/complicaciones
4.
J Am Soc Nephrol ; 32(2): 479-494, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33239394

RESUMEN

BACKGROUND: Binding of donor-specific antibodies (DSAs) to kidney allograft endothelial cells that does not activate the classic complement cascade can trigger the recruitment of innate immune effectors, including NK cells. Activated NK cells contribute to microvascular inflammation leading to chronic antibody-mediated rejection (AMR). Recipient NK cells can also trigger antibody-independent microvascular inflammation by sensing the absence of self HLA class I molecules ("missing self") on allograft endothelial cells. This translational study investigated whether the condition of missing self amplifies DSA-dependent NK cell activation to worsen chronic AMR. METHODS AND RESULTS: Among 1682 kidney transplant recipients who underwent an allograft biopsy at Lyon University Hospital between 2004 and 2017, 135 fulfilled the diagnostic criteria for AMR and were enrolled in the study. Patients with complement-fixing DSAs identified by a positive C3d binding assay (n=73, 54%) had a higher risk of transplant failure (P=0.002). Among the remaining patients with complement-independent chronic AMR (n=62, 46%), those in whom missing self was identified through donor and recipient genotyping exhibited worse allograft survival (P=0.02). In multivariable analysis, only proteinuria (HR: 7.24; P=0.01) and the presence of missing self (HR: 3.57; P=0.04) were independent predictors for transplant failure following diagnosis of chronic AMR. Cocultures of human NK cells and endothelial cells confirmed that addition of missing self to DSA-induced NK cell activation increased endothelial damage. CONCLUSIONS: The assessment of missing self at the time of diagnosis of chronic AMR identifies patients at higher risk for kidney transplant failure.


Asunto(s)
Aloinjertos/patología , Activación de Complemento/fisiología , Rechazo de Injerto/etiología , Antígenos de Histocompatibilidad Clase I/sangre , Trasplante de Riñón/efectos adversos , Células Asesinas Naturales/fisiología , Adulto , Aloinjertos/inmunología , Técnicas de Cultivo de Célula , Complemento C3d/metabolismo , Células Endoteliales/fisiología , Femenino , Rechazo de Injerto/sangre , Rechazo de Injerto/patología , Supervivencia de Injerto , Humanos , Células Asesinas Naturales/patología , Masculino , Persona de Mediana Edad , Adulto Joven
5.
Scand J Immunol ; 93(1): e12993, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33151595

RESUMEN

The discovery that NK cells are able to specifically recognize cells lacking the expression of self-MHC class I molecules provided the first insight into NK cell recognition of tumour cells. It started a flourishing field of NK cell research aimed at exploring the molecular nature of NK cell receptors involved in tumour cell recognition. While much of the important early work was conducted in murine experimental model systems, studies of human NK cells rapidly followed. Over the years, human NK cell research has swiftly progressed, aided by new detailed molecular information on human NK cell development, differentiation, molecular specificity, tissue heterogeneity and functional capacity. NK cells have also been studied in many different diseases aside from cancer, including viral diseases, autoimmunity, allergy and primary immunodeficiencies. These fields of research have all, indirectly or directly, provided further insights into NK cell-mediated recognition of target cells and paved the way for the development of NK cell-based immunotherapies for human cancer. Excitingly, NK cell-based immunotherapy now opens up for novel strategies aimed towards treating malignant diseases, either alone or in combination with other drugs. Reviewed here are some personal reflections of select contributions leading up to the current state-of-the-art in the field, with a particular emphasis on contributions from our own laboratory. This review is part of a series of articles on immunology in Scandinavia, published in conjunction with the 50th anniversary of the Scandinavian Society for Immunology.


Asunto(s)
Inmunoterapia , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Animales , Antígenos de Neoplasias/inmunología , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Interacciones Huésped-Patógeno , Humanos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Neoplasias/patología , Especificidad de Órganos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Resultado del Tratamiento
6.
Semin Immunol ; 31: 20-29, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28888619

RESUMEN

The contribution of natural killer (NK) cells to immunosurveillance of human cancer remains debatable. Here, we discuss advances in several areas of human NK cell research, many of which support the ability of NK cells to prevent cancer development and avoid relapse following adoptive immunotherapy. We describe the molecular basis for NK cell recognition of human tumor cells and provide evidence for NK cell-mediated killing of human primary tumor cells ex vivo. Subsequently, we highlight studies demonstrating the ability of NK cells to migrate to, and reside in, the human tumor microenvironment where selection of tumor escape variants from NK cells can occur. Indirect evidence for NK cell immunosurveillance against human malignancies is provided by the reduced incidence of cancer in individuals with high levels of NK cell cytotoxicity, and the significant clinical responses observed following infusion of human NK cells into cancer patients. Finally, we describe studies showing enhanced tumor progression, or increased cancer incidence, in patients with inherited and acquired defects in cellular cytotoxicity. All these observations have in common that they, either indirectly or directly, suggest a role for NK cells in mediating immunosurveillance against human cancer. This opens up for exciting possibilities with respect to further exploring NK cells in settings of adoptive immunotherapy in human cancer.


Asunto(s)
Citotoxicidad Inmunológica , Vigilancia Inmunológica , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Animales , Carcinogénesis/inmunología , Humanos , Escape del Tumor , Microambiente Tumoral
7.
J Virol ; 94(1)2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31597762

RESUMEN

Natural killer (NK) cells are a subset of innate lymphoid cells (ILC) capable of recognizing stressed and infected cells through multiple germ line-encoded receptor-ligand interactions. Missing-self recognition involves NK cell sensing of the loss of host-encoded inhibitory ligands on target cells, including MHC class I (MHC-I) molecules and other MHC-I-independent ligands. Mouse cytomegalovirus (MCMV) infection promotes a rapid host-mediated loss of the inhibitory NKR-P1B ligand Clr-b (encoded by Clec2d) on infected cells. Here we provide evidence that an MCMV m145 family member, m153, functions to stabilize cell surface Clr-b during MCMV infection. Ectopic expression of m153 in fibroblasts augments Clr-b cell surface levels. Moreover, infections using m153-deficient MCMV mutants (Δm144-m158 and Δm153) show an accelerated and exacerbated Clr-b downregulation. Importantly, enhanced loss of Clr-b during Δm153 mutant infection reverts to wild-type levels upon exogenous m153 complementation in fibroblasts. While the effects of m153 on Clr-b levels are independent of Clec2d transcription, imaging experiments revealed that the m153 and Clr-b proteins only minimally colocalize within the same subcellular compartments, and tagged versions of the proteins were refractory to coimmunoprecipitation under mild-detergent conditions. Surprisingly, the Δm153 mutant possesses enhanced virulence in vivo, independent of both Clr-b and NKR-P1B, suggesting that m153 potentially targets additional host factors. Nevertheless, the present data highlight a unique mechanism by which MCMV modulates NK ligand expression.IMPORTANCE Cytomegaloviruses are betaherpesviruses that in immunocompromised individuals can lead to severe pathologies. These viruses encode various gene products that serve to evade innate immune recognition. NK cells are among the first immune cells that respond to CMV infection and use germ line-encoded NK cell receptors (NKR) to distinguish healthy from virus-infected cells. One such axis that plays a critical role in NK recognition involves the inhibitory NKR-P1B receptor, which engages the host ligand Clr-b, a molecule commonly lost on stressed cells ("missing-self"). In this study, we discovered that mouse CMV utilizes the m153 glycoprotein to circumvent host-mediated Clr-b downregulation, in order to evade NK recognition. These results highlight a novel MCMV-mediated immune evasion strategy.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Células Asesinas Naturales/virología , Lectinas Tipo C/genética , Muromegalovirus/genética , Subfamilia B de Receptores Similares a Lectina de Células NK/genética , Receptores Inmunológicos/genética , Proteínas de la Matriz Viral/genética , Animales , Regulación de la Expresión Génica/inmunología , Prueba de Complementación Genética , Infecciones por Herpesviridae , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Células Asesinas Naturales/inmunología , Lectinas Tipo C/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Muromegalovirus/inmunología , Muromegalovirus/patogenicidad , Células 3T3 NIH , Subfamilia B de Receptores Similares a Lectina de Células NK/inmunología , Receptores Inmunológicos/inmunología , Transducción de Señal , Carga Viral , Proteínas de la Matriz Viral/deficiencia , Proteínas de la Matriz Viral/inmunología , Replicación Viral
8.
Cancer Immunol Immunother ; 67(4): 615-626, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29327110

RESUMEN

Although anti-disialoganglioside (GD2) antibodies are successfully used for neuroblastoma therapy, a third of patients with neuroblastoma experience treatment failure or serious toxicity. Various strategies have been employed in the clinic to improve antibody-dependent cell-mediated cytotoxicity (ADCC), such as the addition of interleukin (IL)-2 to enhance natural killer (NK) cell function, adoptive transfer of allogeneic NK cells to exploit immune surveillance, and retinoid-induced differentiation therapy. Nevertheless, these mechanisms are not fully understood. We developed a quantitative assay to test ADCC induced by the anti-GD2 antibody Hu14.18K322A in nine neuroblastoma cell lines and dissociated cells from orthotopic patient-derived xenografts (O-PDXs) in culture. IL-2 improved ADCC against neuroblastoma cells, and differentiation with all-trans retinoic acid stabilized GD2 expression on tumor cells and enhanced ADCC as well. Degranulation was highest in licensed NK cells that expressed CD158b (P < 0.001) and harbored a killer-cell immunoglobulin-like receptor (KIR) mismatch against the tumor-specific human leukocyte antigen (HLA; P = 0.016). In conclusion, IL-2 is an important component of immunotherapy because it can improve the cytolytic function of NK cells against neuroblastoma cells and could lower the antibody dose required for efficacy, thereby reducing toxicity. The effect of IL-2 may vary among individuals and a biomarker would be useful to predict ADCC following IL-2 activation. Sub-populations of NK cells may have different levels of activity dependent on their licensing status, KIR expression, and HLA-KIR interaction. Better understanding of HLA-KIR interactions and the molecular changes following retinoid-induced differentiation is necessary to delineate their role in ADCC.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Gangliósidos/inmunología , Células Asesinas Naturales/inmunología , Neuroblastoma/terapia , Terapia Combinada , Humanos , Interleucina-2/administración & dosificación , Neuroblastoma/inmunología , Neuroblastoma/patología , Tretinoina/administración & dosificación , Células Tumorales Cultivadas
9.
Semin Immunol ; 26(2): 138-44, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24629893

RESUMEN

Natural killer (NK) cells represent a first line of defense against pathogens and tumor cells. The activation of NK cells is regulated by the integration of signals deriving from activating and inhibitory receptors expressed on their surface. However, different NK cells respond differently to the same stimulus, be it target cells or agents that crosslink activating receptors. The processes that determine the level of NK cell responsiveness have been referred to collectively as NK cell education. NK cell education plays an important role in steady state conditions, where potentially auto-reactive NK cells are rendered tolerant to the surrounding environment. According to the "tuning" concept, the responsiveness of each NK cell is quantitatively adjusted to ensure self tolerance while at the same time ensuring useful reactivity against potential threats. MHC-specific inhibitory receptors displayed by NK cells play a major role in tuning NK cell responsiveness, but recent studies indicate that signaling from activating receptors is also important, suggesting that the critical determinant is an integrated signal from both types of receptors. An important and still unresolved question is whether NK cell education involves interactions with a specific cell population in the environment. Whether hematopoietic and/or non-hematopoietic cells play a role is still under debate. Recent results demonstrated that NK cell tuning exhibits plasticity in steady state conditions, meaning that it can be re-set if the MHC environment changes. Other evidence suggests, however, that inflammatory conditions accompanying infections may favor high responsiveness, indicating that inflammatory agents can over-ride the natural tendency of NK cells to adjust to the steady state environment. These findings raise many questions such as whether viruses and tumor cells manipulate NK cell responsiveness to evade immune-recognition. As knowledge of the underlying processes grows, the possibility of modulating NK cell responsiveness for therapeutic purposes is becoming increasingly attractive, and is now under serious investigation in clinical studies.


Asunto(s)
Células Asesinas Naturales/inmunología , Autotolerancia/inmunología , Animales , Citotoxicidad Inmunológica , Antígenos de Histocompatibilidad/inmunología , Antígenos de Histocompatibilidad/metabolismo , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo
10.
Immunogenetics ; 69(8-9): 547-556, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28699110

RESUMEN

The ability of NK cells to specifically recognize cells lacking expression of self-MHC class I molecules was discovered over 30 years ago. It provided the foundation for the "missing self" hypothesis. Research in the two past decades has contributed to a detailed understanding of the molecular mechanisms that determine the specificity and strength of NK cell-mediated "missing self" responses to tumor cells. However, in light of the recent remarkable breakthroughs in clinical cancer immunotherapy, the cytolytic potential of NK cells still remains largely untapped in clinical settings. There is abundant evidence demonstrating partial or complete loss of HLA class I expression in a wide spectrum of human tumor types. Such loss may result from immune selection of escape variants by tumor-specific CD8 T cells and has more recently also been linked to acquired resistance to checkpoint inhibition therapy. In the present review, we discuss the early predictions of the "missing self" hypothesis, its molecular basis and outline the potential for NK cell-based adoptive immunotherapy to convert checkpoint inhibitor therapy-resistant patients into clinical responders.


Asunto(s)
Antígeno CTLA-4/antagonistas & inhibidores , Células Asesinas Naturales/inmunología , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Neoplasias/inmunología , Receptores KIR/genética , Linfocitos T Citotóxicos/inmunología
11.
Cell Rep ; 38(10): 110503, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35235832

RESUMEN

Natural killer (NK) cells are innate immune cells that contribute to host defense against virus infections. NK cells respond to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in vitro and are activated in patients with acute coronavirus disease 2019 (COVID-19). However, by which mechanisms NK cells detect SARS-CoV-2-infected cells remains largely unknown. Here, we show that the Non-structural protein 13 of SARS-CoV-2 encodes for a peptide that is presented by human leukocyte antigen E (HLA-E). In contrast with self-peptides, the viral peptide prevents binding of HLA-E to the inhibitory receptor NKG2A, thereby rendering target cells susceptible to NK cell attack. In line with these observations, NKG2A-expressing NK cells are particularly activated in patients with COVID-19 and proficiently limit SARS-CoV-2 replication in infected lung epithelial cells in vitro. Thus, these data suggest that a viral peptide presented by HLA-E abrogates inhibition of NKG2A+ NK cells, resulting in missing self-recognition.


Asunto(s)
COVID-19 , Antígenos de Histocompatibilidad Clase I , Células Asesinas Naturales , Metiltransferasas , Subfamília C de Receptores Similares a Lectina de Células NK , ARN Helicasas , SARS-CoV-2 , Proteínas no Estructurales Virales , COVID-19/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Células Asesinas Naturales/inmunología , Metiltransferasas/inmunología , Subfamília C de Receptores Similares a Lectina de Células NK/inmunología , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Péptidos/metabolismo , ARN Helicasas/inmunología , Proteínas no Estructurales Virales/inmunología , Antígenos HLA-E
12.
Methods Mol Biol ; 2463: 291-310, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35344182

RESUMEN

This chapter is intended to serve as a practical guide for establishing a workflow using sequence-specific polymorphism PCR (SSP-PCR) for killer cell immunoglobulin-like receptor (KIR) genotyping in a clinical setting, especially in allogeneic hematopoietic stem cell transplantation (HSCT). As clinical evidence accumulates on the application of KIR and HLA genetics to guide donor selection in HSCT, there is an increasing need for KIR genotyping in clinical settings, and thus medical institutes may need to build this capability. Among the various KIR genotyping approaches now available, SSP-PCR methods are well-established and are the most cost-effective and will likely be the method of choice especially when expenses will be passed on to the patient. The protocol described in this chapter developed by Vilches et al. features small amplicon PCR and is suitable for KIR genotyping using FFPE-derived DNA as well as DNA extracted from blood samples. Setting up a laboratory workflow for in-house KIR genotyping is relatively straightforward; in this chapter, considerations for KIR genotyping to guide clinical decisions are discussed.In HSCT, a main objective of KIR genotyping is to apply the genetic analysis to predict donor and recipient combinations that have the most potential to produce NK cell alloresponses either through the missing-self mechanism or by action associated with activating KIR. The desired effects are reduction in acute GVHD and relapse rates and enhancement of overall survival. The information herein may also be useful to clinical laboratories considering the application of KIR genotyping in areas such as solid organ transplantation, NK cell-based treatment in other forms of cancer and autoimmune diseases, humanized antibody treatment, regenerative medicine, and reproductive medicine. Some background knowledge on KIR genetics will be necessary in managing a KIR genotyping platform. This chapter aims to address the main difficulties often encountered by physicians in understanding the KIR system, such as basic aspects of the nomenclature of KIR genes and haplotypes, genotypes, and determining presence/absence of KIR ligands in the patient and donor from the extensively diversified HLA class I allotypes. In describing the workflow, emphasis has been placed on the processes after genotype PCR and gel image acquisition: haplotype inference, generating B content scores, deduction of KIR ligands from HLA typing results, and the emerging algorithms for donor selection based on KIR and HLA genetics.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Genotipo , Enfermedad Injerto contra Huésped/genética , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Receptores KIR/genética , Flujo de Trabajo
13.
Pathogens ; 10(7)2021 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-34358058

RESUMEN

Immunoreceptor tyrosine-based inhibitory motif (ITIM)-bearing receptors (IRs) enable discrimination between self- and non-self molecules on the surface of host target cells. In this regard, they have a vital role in self-tolerance through binding and activating intracellular tyrosine phosphatases which can inhibit cellular activation. Yet, self-MHC class I (MHC I)-specific IRs are versatile in that they can also positively impact lymphocyte functionality, as exemplified by their role in natural killer (NK) cell education, often referred to as 'licensing'. Recent discoveries using defined mouse models of cytomegalovirus (CMV) infection have revealed that select self-MHC I IRs can increase NK cell antiviral defenses as well, whereas other licensing IRs cannot, or instead impede virus-specific NK responses for reasons that remain poorly understood. This review highlights a role for self-MHC I 'licensing' IRs in antiviral immunity, especially in the context of CMV infection, their impact on virus-specific NK cells during acute infection, and their potential to affect viral pathogenesis and disease.

14.
HLA ; 98(3): 191-199, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34050618

RESUMEN

The current transplant immunology dogma defends that allograft rejection is initiated by recipient's adaptive immune system. In this prevalent model, innate immune cells in general, and natural killer (NK) cells in particular, are merely considered as downstream effectors which participate in the destruction of the graft only upon recruitment by adaptive effectors: alloreactive T cells or donor-specific antibodies (DSA). Challenging this vision, recent data demonstrated that recipients' NK cells are capable of a form of allorecognition because they can sense the absence of self HLA class I molecules on the surface of graft endothelial cells. Missing-self triggers mTORC1-dependent activation of NK cells, which in turn promote the development of graft microvascular inflammation and detrimentally impact graft survival. The fact that some patients develop chronic vascular rejection in absence of DSA or genetically-predicted missing self suggests that other molecular mechanisms could underly NK cell allorecognition. This review provides an overview of these proven and putative molecular mechanisms and discusses future research directions in this emerging field in organ transplant immunology.


Asunto(s)
Células Endoteliales , Rechazo de Injerto , Alelos , Aloinjertos , Rechazo de Injerto/genética , Humanos , Células Asesinas Naturales
15.
Inflamm Regen ; 41(1): 2, 2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33407951

RESUMEN

In the regenerative medicine field, allogenic transplantation of regenerated tissues has been promoted because autologous transplantation setting is costly and time-consuming to prepare and therefore unsuitable for emergent treatment. To avoid a T cell-mediated immune rejection in the allogenic transplantation setting, induced pluripotent stem cells (iPSCs) derived from different HLA haplotype-homozygous (HLA-homo) donors have been prepared to be used as source of regenerated tissues. However, there still remain immunological issues, even when HLA-homo iPSCs are used. One issue is the immune response against minor histocompatibility antigens expressed on the regenerated tissues, and the other is the immune rejection mediated by NK cells. In this article, we introduce our research on NK cell reactivity against the regenerated tissues in the HLA homo-to-hetero transplantation setting. We further introduce several approaches taken by other groups that address the NK-mediated immune rejection issue.

16.
Cancers (Basel) ; 13(2)2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33467442

RESUMEN

Natural killer (NK) cells can kill target cells via the recognition of stress molecules and down-regulation of major histocompatibility complex class I (MHC-I). Some NK cells are educated to recognize and kill cells that have lost their MHC-I expression, e.g., tumor or virus-infected cells. A desired property of cancer immunotherapy is, therefore, to activate educated NK cells during anti-tumor responses in vivo. We here analyze NK cell responses to α-galactosylceramide (αGC), a potent activator of invariant NKT (iNKT) cells, or to exosomes loaded with αGC. In mouse strains which express different MHC-I alleles using an extended NK cell flow cytometry panel, we show that αGC induces a biased NK cell proliferation of educated NK cells. Importantly, iNKT cell-induced activation of NK cells selectively increased in vivo missing self-responses, leading to more effective rejection of tumor cells. Exosomes from antigen-presenting cells are attractive anti-cancer therapy tools as they may induce both innate and adaptive immune responses, thereby addressing the hurdle of tumor heterogeneity. Adding αGC to antigen-loaded dendritic-cell-derived exosomes also led to an increase in missing self-responses in addition to boosted T and B cell responses. This study manifests αGC as an attractive adjuvant in cancer immunotherapy, as it increases the functional capacity of educated NK cells and enhances the innate, missing self-based antitumor response.

17.
Cell Rep ; 32(4): 107969, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32726632

RESUMEN

Major histocompatibility complex class I (MHC-I)-restricted immune responses are largely attributed to cytotoxic T lymphocytes (CTLs). However, natural killer (NK) cells, as predicted by the missing-self hypothesis, have opposing requirements for MHC-I, suggesting that they may also demonstrate MHC-I-restricted effects. In mice, the Ly49 inhibitory receptors prevent NK cell killing of missing-self targets in effector responses, and they have a proposed second function in licensing or educating NK cells via self-MHC-I in vivo. Here we show MHC-I-restricted control of murine cytomegalovirus (MCMV) infection in vivo that is NK cell dependent. Using mice lacking specific Ly49 receptors, we show that control of MCMV requires inhibitory Ly49 receptors and an inhibitory signaling motif and the capacity for MCMV to downregulate MHC-I. Taken together, these data provide definitive evidence that the inhibitory receptors are required for missing-self rejection and are relevant to MHC-I-restricted NK cell control of a viral infection in vivo.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Células Asesinas Naturales/metabolismo , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Animales , Antígenos Ly , Infecciones por Citomegalovirus/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Muromegalovirus/inmunología , Muromegalovirus/patogenicidad , Subfamilia A de Receptores Similares a Lectina de Células NK/inmunología , Receptores de Células Asesinas Naturales , Virosis
18.
Methods Mol Biol ; 2097: 115-123, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31776923

RESUMEN

NK cells are innate lymphocytes that are vital to clearance of virally infected or malignantly transformed cells. Assessment of the cytotoxic response is an important component of NK cell research and investigation of human disease. Standard assays of NK cell-mediated cytotoxicity of CD107a degranulation or 51Cr release assay utilize cultured or freshly isolated NK cell populations in vitro. In addition to requirements to maintain multiple target cell lines and radioactivity precautions in the case of 51Cr, these are in vitro evaluations of a complex in vivo function. Here, we describe the in vivo assessment of NK cell-mediated cytotoxicity through the adoptive transfer of splenocytes and their subsequent rejection. This protocol offers rapid, quantitative, and concurrent assessment of NK cell-mediated cytotoxicity against the prototypic NK stimulations of "missing-self" and "nonself."


Asunto(s)
Traslado Adoptivo/métodos , Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Bazo/citología , Animales , Anticuerpos/metabolismo , Análisis de Datos , Depleción Linfocítica , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
19.
Elife ; 72018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30575523

RESUMEN

A recurrent theme in viral immune evasion is the sabotage of MHC-I antigen presentation, which brings virus the concomitant issue of 'missing-self' recognition by NK cells that use inhibitory receptors to detect surface MHC-I proteins. Here, we report that rodent herpesvirus Peru (RHVP) encodes a Qa-1 like protein (pQa-1) via RNA splicing to counteract NK activation. While pQa-1 surface expression is stabilized by the same canonical peptides presented by murine Qa-1, pQa-1 is GPI-anchored and resistant to the activity of RHVP pK3, a ubiquitin ligase that targets MHC-I for degradation. pQa-1 tetramer staining indicates that it recognizes CD94/NKG2A receptors. Consistently, pQa-1 selectively inhibits NKG2A+ NK cells and expression of pQa-1 can protect tumor cells from NK control in vivo. Collectively, these findings reveal an innovative NK evasion strategy wherein RHVP encodes a modified Qa-1 mimic refractory to MHC-I sabotage and capable of specifically engaging inhibitory receptors to circumvent NK activation.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Herpesviridae/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Células Asesinas Naturales/inmunología , Subfamília C de Receptores Similares a Lectina de Células NK/inmunología , Subfamília D de Receptores Similares a Lectina de las Células NK/inmunología , Secuencia de Aminoácidos , Animales , Presentación de Antígeno/inmunología , Secuencia de Bases , Citotoxicidad Inmunológica/genética , Células HEK293 , Herpesviridae/genética , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Células Asesinas Naturales/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Imitación Molecular/genética , Imitación Molecular/inmunología , Subfamília C de Receptores Similares a Lectina de Células NK/genética , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Subfamília D de Receptores Similares a Lectina de las Células NK/genética , Subfamília D de Receptores Similares a Lectina de las Células NK/metabolismo , Unión Proteica/inmunología , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico
20.
Immunol Lett ; 189: 40-47, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28414184

RESUMEN

Cytomegaloviruses (CMVs) have dedicated a large portion of their genome towards immune evasion targeting many aspects of the host immune system, particularly NK cells. However, the host managed to cope with the infection by developing multiple mechanisms to recognize viral threat and counterattack it, thus illustrating never-ending evolutionary interplay between CMV and its host. In this review, we will focus on several mechanisms of NK cell evasion by mouse CMV (MCMV), the role of host inhibitory and activating Ly49 receptors involved in the virus control and acquisition of adaptive features by NK cells as a consequence of MCMV infection.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Muromegalovirus/inmunología , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Animales , Autoantígenos/inmunología , Citotoxicidad Inmunológica , Evolución Molecular , Antígenos de Histocompatibilidad Clase I/metabolismo , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Ratones , Subfamilia A de Receptores Similares a Lectina de Células NK/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA