Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
1.
Nat Rev Cancer ; 21(3): 181-197, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33462501

RESUMEN

Aberrant signalling of ERBB family members plays an important role in tumorigenesis and in the escape from antitumour immunity in multiple malignancies. Molecular-targeted agents against these signalling pathways exhibit robust clinical efficacy, but patients inevitably experience acquired resistance to these molecular-targeted therapies. Although cancer immunotherapies, including immune checkpoint inhibitors (ICIs), have shown durable antitumour response in a subset of the treated patients in multiple cancer types, clinical efficacy is limited in cancers harbouring activating gene alterations of ERBB family members. In particular, ICI treatment of patients with non-small cell lung cancers with epidermal growth factor receptor (EGFR) alterations and breast cancers with HER2 alterations failed to show clinical benefits, suggesting that EGFR and HER2 signalling may have an essential role in inhibiting antitumour immune responses. Here, we discuss the mechanisms by which the signalling of ERBB family members affects not only autonomous cancer hallmarks, such as uncontrolled cell proliferation, but also antitumour immune responses in the tumour microenvironment and the potential application of immune-genome precision medicine into immunotherapy and molecular-targeted therapy focusing on the signalling of ERBB family members.


Asunto(s)
Neoplasias/inmunología , Receptores ErbB/genética , Receptores ErbB/fisiología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Complejo Mayor de Histocompatibilidad/fisiología , Terapia Molecular Dirigida , Mutación , Transducción de Señal/fisiología , Microambiente Tumoral
2.
Proc Natl Acad Sci U S A ; 117(45): 28251-28262, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33109721

RESUMEN

Toll-like receptor (TLR) recruitment to phagosomes in dendritic cells (DCs) and downstream TLR signaling are essential to initiate antimicrobial immune responses. However, the mechanisms underlying TLR localization to phagosomes are poorly characterized. We show herein that phosphatidylinositol-4-kinase IIα (PI4KIIα) plays a key role in initiating phagosomal TLR4 responses in murine DCs by generating a phosphatidylinositol-4-phosphate (PtdIns4P) platform conducive to the binding of the TLR sorting adaptor Toll-IL1 receptor (TIR) domain-containing adaptor protein (TIRAP). PI4KIIα is recruited to maturing lipopolysaccharide (LPS)-containing phagosomes in an adaptor protein-3 (AP-3)-dependent manner, and both PI4KIIα and PtdIns4P are detected on phagosomal membrane tubules. Knockdown of PI4KIIα-but not the related PI4KIIß-impairs TIRAP and TLR4 localization to phagosomes, reduces proinflammatory cytokine secretion, abolishes phagosomal tubule formation, and impairs major histocompatibility complex II (MHC-II) presentation. Phagosomal TLR responses in PI4KIIα-deficient DCs are restored by reexpression of wild-type PI4KIIα, but not of variants lacking kinase activity or AP-3 binding. Our data indicate that PI4KIIα is an essential regulator of phagosomal TLR signaling in DCs by ensuring optimal TIRAP recruitment to phagosomes.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/metabolismo , Células Dendríticas/inmunología , Complejo Mayor de Histocompatibilidad/fisiología , Fagosomas/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Células de la Médula Ósea , Citocinas/metabolismo , Lipopolisacáridos , Ratones , Transducción de Señal , Receptor Toll-Like 4/genética , Receptores Toll-Like/metabolismo
3.
J Immunol ; 204(12): 3351-3359, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32321756

RESUMEN

During normal T cell development in the thymus, αß TCRs signal immature thymocytes to differentiate into mature T cells by binding to peptide-MHC ligands together with CD4/CD8 coreceptors. Conversely, in MHC and CD4/CD8 coreceptor-deficient mice, the thymus generates mature T cells expressing MHC-independent TCRs that recognize native conformational epitopes rather than linear antigenic-peptides presented by MHC. To date, no structural information of MHC-independent TCRs is available, and their structural recognition of non-MHC ligand remains unknown. To our knowledge in this study, we determined the first structures of two murine MHC-independent TCRs (A11 and B12A) that bind with high nanomolar affinities to mouse adhesion receptor CD155. Solution binding demonstrated the Vαß-domain is responsible for MHC-independent B12A recognition of its ligand. Analysis of A11 and B12A sequences against various MHC-restricted and -independent TCR sequence repertoires showed that individual V-genes of A11 and B12A did not exhibit preference against MHC-restriction. Likewise, CDR3 alone did not discriminate against MHC binding, suggesting VDJ recombination together with Vα/Vß pairing determine their MHC-independent specificity for CD155. The structures of A11 and B12A TCR are nearly identical to those of MHC-restricted TCR, including the conformations of CDR1 and 2. Mutational analysis, together with negative-staining electron microscopy images, showed that the CDR regions of A11 and B12A recognized epitopes on D1 domain of CD155, a region also involved in CD155 binding to poliovirus and Tactile in human. Taken together, MHC-independent TCRs adopt canonical TCR structures to recognize native Ags, highlighting the importance of thymic selection in determining TCR ligand specificity.


Asunto(s)
Complejo Mayor de Histocompatibilidad/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores Virales/metabolismo , Animales , Células HEK293 , Humanos , Ligandos , Ratones , Péptidos/metabolismo , Poliovirus/metabolismo , Unión Proteica , Dominios Proteicos , Timocitos/metabolismo , Recombinación V(D)J/fisiología
4.
J Virol ; 94(3)2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31723020

RESUMEN

During the latent phase, Kaposi's sarcoma-associated herpes virus (KSHV) maintains itself inside the host by escaping the host immune surveillance mechanism through restricted protein expression. Latency-associated nuclear antigen (LANA), the most abundantly expressed protein, is essential for viral persistence, as it plays important roles in latent viral DNA replication and efficient segregation of the viral genome to the daughter cells following cell division. KSHV evades immune detection by maintaining the levels of LANA protein below a threshold required for detection by the host immune system but sufficient to maintain the viral genome. LANA achieves this by controlling its expression through regulation of its promoters and by inhibiting its presentation through interaction with the proteins of class I and class II major histocompatibility complex (MHC) pathways. In this study, we identified a mechanism of LANA expression and restricted immune recognition through formation of G-quadruplexes in LANA mRNA. We show that the formation of these stable structures in LANA mRNA inhibits its translation to control antigen presentation, which was supported by treatment of cells with TMPyP4, a G-quadruplex-stabilizing ligand. We identified heterogenous ribonucleoprotein A1 (hnRNP A1) as a G-quadruplex-unwinding helicase, which unfolds these stable secondary structures to regulate LANA translation.IMPORTANCE LANA, the most abundantly expressed protein during latency, is a multifunctional protein which is absolutely required for the persistence of KSHV in the host cell. Even though the functions of LANA in aiding pathogenesis of the virus have been extensively studied, the mechanism of how LANA escapes host's immune surveillance is not fully understood. This study sheds light on the autoregulatory role of LANA to modulate its expression and immune evasion through formation of G-quadruplexes in its mRNA. We used G-quadruplex-stabilizing ligand to define the inhibition in LANA expression and presentation on the cell surface through MHC class I. We defined the autoregulatory role of LANA and identified a cellular RNA helicase, hnRNP A1, regulating the translation of LANA mRNA. This interaction of hnRNP A1 with LANA mRNA could be exploited for controlling KSHV latency.


Asunto(s)
Antígenos Virales/metabolismo , G-Cuádruplex , Herpesvirus Humano 8/fisiología , Ribonucleoproteína Nuclear Heterogénea A1/metabolismo , Proteínas Nucleares/metabolismo , ARN Mensajero/metabolismo , Antígenos Virales/química , Antígenos Virales/genética , Secuencia de Bases , Línea Celular , ADN Viral , Genoma Viral , Herpesvirus Humano 8/genética , Ribonucleoproteína Nuclear Heterogénea A1/genética , Humanos , Complejo Mayor de Histocompatibilidad/fisiología , Proteínas Nucleares/química , Proteínas Nucleares/genética , Latencia del Virus/genética , Latencia del Virus/fisiología , Replicación Viral/genética
5.
Br J Hosp Med (Lond) ; 80(10): 568-573, 2019 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-31589501

RESUMEN

Malignancy is a micro-evolutionary phenomenon shaped by selection pressures. Chief among these is the adaptive immune system, which recognizes malignant cells as a threat and attempts to eradicate them. The task is not easily achieved - if it were, cancer would not be a part of our human experience. The field of immunotherapy has rapidly expanded over the last two decades. It has produced some of the most exciting results of 21st century medicine, and has deepened clinicians' understanding of the relationship between malignancy and the immune response. This review discusses this relationship and analyses key tools in the immunotherapy arsenal.


Asunto(s)
Sistema Inmunológico/fisiopatología , Inmunoterapia/métodos , Neoplasias/inmunología , Inmunidad Adaptativa/fisiología , Vacunas contra el Cáncer/farmacología , Puntos de Control del Ciclo Celular/fisiología , Humanos , Inmunoterapia Adoptiva/métodos , Complejo Mayor de Histocompatibilidad/fisiología , Linfocitos T/metabolismo
6.
Cell Syst ; 9(2): 159-166.e3, 2019 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-31176619

RESUMEN

The computational identification of peptides that can bind the major histocompatibility complex (MHC) with high affinity is an essential step in developing personal immunotherapies and vaccines. We introduce PUFFIN, a deep residual network-based computational approach that quantifies uncertainty in peptide-MHC affinity prediction that arises from observational noise and the lack of relevant training examples. With PUFFIN's uncertainty metrics, we define binding likelihood, the probability a peptide binds to a given MHC allele at a specified affinity threshold. Compared to affinity point estimates, we find that binding likelihood correlates better with the observed affinity and reduces false positives in high-affinity peptide design. When applied to examine an existing peptide vaccine, PUFFIN identifies an alternative vaccine formulation with higher binding likelihood. PUFFIN is freely available for download at http://github.com/gifford-lab/PUFFIN.


Asunto(s)
Biología Computacional/métodos , Complejo Mayor de Histocompatibilidad/fisiología , Unión Proteica/fisiología , Algoritmos , Bases de Datos de Proteínas , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Péptidos/metabolismo , Programas Informáticos , Incertidumbre
7.
Cytokine ; 119: 119-128, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30909148

RESUMEN

Neosporosis due to Neospora caninum causes abortions in farm animals such as cattle. No treatment and vaccine exist to fight this disease, responsible for considerable economic losses. It is thus important to better understand the immune responses occurring during the pathogenesis to control them in a global strategy against the parasite. In this context, we studied the roles of N. caninum glycosylphosphatidylinositols (GPIs), glycolipids defined as toxins in the related parasite Plasmodium falciparum. We demonstrated for the first time that GPIs could be excreted in the supernatant of N. caninum culture and trigger cell signalling through the Toll-like receptors 2 and 4. In addition, antibodies specific to N. caninum GPIs were detected in the serum of infected mice. As shown for other protozoan diseases, they could play a role in neutralizing GPIs. N. caninum GPIs were able to induce the production of tumour necrosis factor-α, interleukin(IL)-1ß and IL-12 cytokines by murine macrophages and dendritic cells. Furthermore, GPIs significantly reduced expression of major histocompatibility complex (MHC) molecules of class I on murine dendritic cells. In contrast to murine cells, bovine blood mononuclear cells produced increased levels of IFN-γ and IL-10, but reduced levels of IL-12p40 in response to GPIs. On these bovine cells, GPI had the tendency to up-regulate MHC class I, but to down-regulate MHC class II. Altogether, these results suggest that N. caninum GPIs might differentially participate in the responses of antigen presenting cells induced by the whole parasite in mouse models of neosporosis and in the natural cattle host.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Glicosilfosfatidilinositoles/metabolismo , Neospora/metabolismo , Animales , Bovinos , Células Cultivadas , Chlorocebus aethiops , Células Dendríticas/metabolismo , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Macrófagos/metabolismo , Complejo Mayor de Histocompatibilidad/fisiología , Ratones , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/metabolismo , Células Vero
8.
PLoS Biol ; 17(1): e3000131, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30703088

RESUMEN

Central players of the adaptive immune system are the groups of proteins encoded in the major histocompatibility complex (MHC), which shape the immune response against pathogens and tolerance to self-peptides. The corresponding genomic region is of particular interest, as it harbors more disease associations than any other region in the human genome, including associations with infectious diseases, autoimmune disorders, cancers, and neuropsychiatric diseases. Certain MHC molecules can bind to a much wider range of epitopes than others, but the functional implication of such an elevated epitope-binding repertoire has remained largely unclear. It has been suggested that by recognizing more peptide segments, such promiscuous MHC molecules promote immune response against a broader range of pathogens. If so, the geographical distribution of MHC promiscuity level should be shaped by pathogen diversity. Three lines of evidence support the hypothesis. First, we found that in pathogen-rich geographical regions, humans are more likely to carry highly promiscuous MHC class II DRB1 alleles. Second, the switch between specialist and generalist antigen presentation has occurred repeatedly and in a rapid manner during human evolution. Third, molecular positions that define promiscuity level of MHC class II molecules are especially diverse and are under positive selection in human populations. Taken together, our work indicates that pathogen load maintains generalist adaptive immune recognition, with implications for medical genetics and epidemiology.


Asunto(s)
Inmunidad Adaptativa/genética , Antígenos de Histocompatibilidad Clase II/genética , Complejo Mayor de Histocompatibilidad/genética , Secuencia de Aminoácidos/genética , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Evolución Biológica , Patógenos Transmitidos por la Sangre , Epítopos/genética , Epítopos/fisiología , Evolución Molecular , Variación Genética/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Complejo Mayor de Histocompatibilidad/fisiología , Péptidos/genética , Selección Genética/genética
9.
Mol Hum Reprod ; 25(2): 88-99, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30445548

RESUMEN

STUDY QUESTION: Does the genotype of the surrogate mother modulate the body composition and immunity of her offspring? SUMMARY ANSWER: C57BL/6J (B6) progenies carried by immunodeficient NOD SCID (NS) mothers had increased adaptive but decreased innate, immune responsiveness in comparison with the same genotype offspring carried by immunocompetent mothers, B6 and BALB/c (C); the B6 progenies carried by the same genotype mothers also showed higher body fat than the others. WHAT IS KNOWN ALREADY: Differences in the major histocompatibility complex (MHC) genes between mother and foetus is considered as an important factor in prenatal embryo development, whereas the impact of such dissimilarity on the phenotype of the mature progeny is unclear. STUDY DESIGN, SIZE, DURATION: Transplantation of two-cell mouse embryos into recipient females of the different MHC (H2) genotypes was used as an approach to simulate three variants of the immunogenic mother-foetus interaction: (i) bidirectional immunogenic dialogue between B6 (H2b haplotype) embryos and C (H2d haplotype) surrogate mother; (ii) one-way immunogenic interaction between B6 embryos and immunodeficient NS (H2g7 haplotype) surrogate mother and (iii) reduced immunogenetic dialogue between embryos and surrogate mother of the same H2b haplotype resulting in only a maternal response to HY antigens of male foetuses. Delivered by Caesarean section, pups were fostered by lactating B6 females and weighed after weaning (n = 171). Body mass and composition and innate and adaptive immunity were assessed in selected progeny groups at 9-11 weeks of age. PARTICIPANTS/MATERIALS, SETTING, METHODS: The study was performed on the specific pathogen-free mouse, inbred strains C57BL/6J, NOD SCID and BALB/c. Plasma progesterone in pregnant females was measured by enzyme-linked immunosorbent assay (ELISA). Body composition was determined by magnetic resonance spectroscopy using a low-field NMR spectrometer (EchoMRI, USA). To assess peritoneal macrophage responses (innate immunity) to anthrax, lactate dehydrogenase (LDH) and interleukin-1 (IL-1ß) were measured in a culture medium 24 h after the addition of both anthrax-lethal factor and anthrax-protective antigen. To assess adaptive immunity, 9-10 males in experimental groups were infected with Helicobacter hepaticus. Faeces collected 2 and 4 weeks after infection was used for quantitative assessment of the H. hepaticus DNA by real-time polymerase chain reaction. IgA, interferon (IFN-γ), tumour necrosis factor (TNFα), interleukin-17 (IL-17) and interleukin-10 (IL-10) in colon tissue and IgG in serum were determined in samples collected 4 weeks after gavage with H. hepaticus using ELISA. For statistical analyses, ANCOVA, post hoc least significant difference (LSD) test, Student's t-test, Spearman rank correlations and χ2 test were performed. P-value <0.05 was considered as a statistically significant difference. MAIN RESULTS AND THE ROLE OF CHANCE: ANCOVA with litter size and age as covariates revealed significant effects of the surrogate mother genotype on body mass and percent of fat in their adult progeny (F2149 = 15.60, P < 0.001 and F2149 = 5.02, P = 0.007, respectively). Adult B6 mice carried by B6 surrogate mothers were characterized by a higher percentage of body fat in comparison with offspring that were carried by NS and C females. In comparison with the male offspring carried by the B6 and C mothers, male B6 progenies carried by immunodeficient NS mothers had a higher humoral immune response (serum IgG) against oral infection with H. hepaticus, but lower in vitro macrophage IL-1ß reaction to the anthrax. Four weeks after the infection of offspring, concentrations of serum IgG and colon IL-10 correlated positively with maternal progesterone on Day 4 after embryo transfer and negatively with DNA of H. hepaticus. One-way ANOVA confirmed a statistically significant impact of surrogate mother genotype on adaptive (IgG) and innate (IL-1ß) immunity (F2.26 = 26.39, P < 0.001 and F2.27 = 5.89, P = 0.008, respectively). LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: The main limitation of our study is the number of combinations of mother and foetus interactions, in particular, transfer of only one embryo genotype was used. Also, it is a descriptive study, which requires further analysis of the epigenetic mechanisms of the observed phenotypic effects of surrogate mother genotype. WIDER IMPLICATIONS OF THE FINDINGS: Our experimental data demonstrate that the transfer of inbred embryos to surrogate mothers of the different genotypes is a prospective experimental model for the study of epigenetic effects of the immunogenetic interactions between mother and foetus. The experimental approach tested in our study will be in demand for the development of criteria for choosing surrogate mothers. In particular, immunocompetence of the surrogate mother along with genetic distance of her MHC alleles to the transferred embryos have a significant impact on offspring development. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the Russian FPI (6/099/2017), budget projects (0324-2016-0002 and 0324-2018-0016) and implemented using the equipment of the Centre for Genetic Resources of Laboratory Animals at ICG SB RAS, supported by the Ministry of Education and Science of Russia (Unique project identifier RFMEFI62117X0015). The authors report no conflicts of interest.


Asunto(s)
Transferencia de Embrión , Embrión de Mamíferos/metabolismo , Inmunidad Adaptativa/genética , Inmunidad Adaptativa/fisiología , Animales , Carbunco/inmunología , Composición Corporal/fisiología , Índice de Masa Corporal , Embrión de Mamíferos/inmunología , Femenino , Genotipo , Helicobacter hepaticus/inmunología , Helicobacter hepaticus/patogenicidad , Inmunidad Innata/genética , Inmunidad Innata/fisiología , Macrófagos/inmunología , Macrófagos/microbiología , Complejo Mayor de Histocompatibilidad/genética , Complejo Mayor de Histocompatibilidad/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Embarazo
10.
Bull Exp Biol Med ; 165(4): 584-588, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30121916

RESUMEN

Analysis of changes in lymphocyte subpopulations during co-culturing with multipotent mesenchymal stromal cells (MSC) revealed two distinct MSC groups: one group (A) increased HLA-DR expression on lymphocytes during co-culturing and the other (B) did not change it in comparison with lymphocyte monoculture. In stromal cells interacting with lymphocytes, expression of HLA-DR molecules was initiated, but only in samples that induced enhanced expression on lymphocytes and irrespectively of whether allogeneic or autologous lymphocytes were used for co-culturing with MSC. In group A, the relative expression of IDO1 significantly increased in comparison with group B. The revealed individual differences in MSC can explain why not all MSC samples are effective in the treatment of autoimmune diseases, acute "graft-versus-host" disease, and other pathologies.


Asunto(s)
Linfocitos/citología , Células Madre Mesenquimatosas/citología , Adolescente , Adulto , Niño , Femenino , Humanos , Activación de Linfocitos/fisiología , Complejo Mayor de Histocompatibilidad/fisiología , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Adulto Joven
11.
Cell ; 174(3): 672-687.e27, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30053426

RESUMEN

TCR-signaling strength generally correlates with peptide-MHC binding affinity; however, exceptions exist. We find high-affinity, yet non-stimulatory, interactions occur with high frequency in the human T cell repertoire. Here, we studied human TCRs that are refractory to activation by pMHC ligands despite robust binding. Analysis of 3D affinity, 2D dwell time, and crystal structures of stimulatory versus non-stimulatory TCR-pMHC interactions failed to account for their different signaling outcomes. Using yeast pMHC display, we identified peptide agonists of a formerly non-responsive TCR. Single-molecule force measurements demonstrated the emergence of catch bonds in the activating TCR-pMHC interactions, correlating with exclusion of CD45 from the TCR-APC contact site. Molecular dynamics simulations of TCR-pMHC disengagement distinguished agonist from non-agonist ligands based on the acquisition of catch bonds within the TCR-pMHC interface. The isolation of catch bonds as a parameter mediating the coupling of TCR binding and signaling has important implications for TCR and antigen engineering for immunotherapy.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/fisiología , Activación de Linfocitos/fisiología , Adulto , Femenino , Humanos , Cinética , Ligandos , Complejo Mayor de Histocompatibilidad/fisiología , Masculino , Persona de Mediana Edad , Simulación de Dinámica Molecular , Oligopéptidos , Péptidos , Unión Proteica/fisiología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal , Imagen Individual de Molécula , Linfocitos T/fisiología
12.
Eur Cytokine Netw ; 28(3): 102-110, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-29187337

RESUMEN

Toll-like receptor (TLR) agonists drive hematopoietic stem and progenitor cells (HSPCs) to differentiate along the myeloid lineage in vitro and also in vivo following infection. In this study, we used an in vitro model of HSPC differentiation to investigate the functional consequences (cytokine production) that exposing HSPCs to various pathogen-associated molecular patterns (PAMPs) and Candida albicans cells have on the subsequently derived macrophages. Mouse HSPCs (Lin- cells) were cultured with GM-CSF to induce macrophage differentiation in the presence or absence of the following pattern recognition receptor (PRR) agonists: Pam3CSK4 (TLR2 ligand), LPS (TLR4 ligand), depleted zymosan (which only activates Dectin-1), or inactivated C. albicans yeasts (which activate several PRRs, mainly TLR2 and Dectin-1). Our data show that only pure TLR2 ligand exposure (transient and continuous) impacts the inflammatory function of GM-CSF-derived macrophages, because Pam3CSK4-exposed HSPCs generate macrophages with a diminished ability to produce inflammatory cytokines. Interestingly, the Pam3CSK4-induced tolerance of macrophages (by transient exposure of HSPCs) is reinforced by subsequent exposure to C. albicans cells in GM-CSF-derived macrophages; however, the induced tolerance is partially reversed in M-CSF-derived macrophages. Therefore, the ability of macrophages to produce inflammatory cytokines is extremely dependent on how the HSPCs from which they are derived receive and integrate multiple microenvironmental signals (PRR ligands and/or CSFs).


Asunto(s)
Macrófagos/citología , Macrófagos/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Antígenos Ly/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Citocinas/metabolismo , Escherichia coli , Femenino , Citometría de Flujo , Complejo Mayor de Histocompatibilidad/fisiología , Ratones , Ratones Endogámicos C57BL , Receptores de Reconocimiento de Patrones/agonistas , Transducción de Señal/fisiología
13.
Bull Exp Biol Med ; 163(2): 230-234, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28726189

RESUMEN

We studied changes in the population of human multipotent mesenchymal stromal cells activated by IFNγ. The cells were cultured under standard conditions; IFNγ was added in various concentrations for 4 h or over 2 passages. It was shown that the total cell production significantly decreased after long-term culturing with IFNγ, but 4-h exposure did not affect this parameter. After 4-h culturing, the expression levels of IDO1, CSF1, and IL-6 increased by 300, 7, and 2.4 times, respectively, and this increase persisted 1 and 2 days after removal of IFNγ from the culture medium. The expression of class I and II MHC (HLA) on cell surface practically did not change immediately after exposure to IFNγ, but during further culturing, HLA-ABC (MHC I) and HLA-DR (MHC II) expression significantly increased, which abolished the immune privilege in these cells, the property allowing clinical use of allogenic multipotent mesenchymal stromal cells. Multipotent mesenchymal stromal cells can suppress proliferation of lymphocytes. The degree of this suppression depends on individual properties of multipotent mesenchymal stromal cell donor. Treatment with IFNγ did not significantly affect the intensity of inhibition of lymphocyte proliferation by these cells.


Asunto(s)
Interferón gamma/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Adolescente , Adulto , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Humanos , Interleucina-6/metabolismo , Activación de Linfocitos/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Complejo Mayor de Histocompatibilidad/fisiología , Masculino , Persona de Mediana Edad , Adulto Joven
14.
Mol Cancer Res ; 15(6): 635-650, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28356330

RESUMEN

The current concepts and practice of cancer immunotherapy evolved from classical experiments that distinguished "self" from "non-self" and the finding that humoral immunity is complemented by cellular immunity. Elucidation of the biology underlying immune checkpoints and interactions between ligands and ligand receptors that govern the immune system's ability to recognize tumor cells as foreign has led to the emergence of new strategies that mobilize the immune system to reverse this apparent tolerance. Some of these approaches have led to new therapies such as the use of mAbs to interfere with the immune checkpoint. Others have exploited molecular technologies to reengineer a subset of T cells to directly engage and kill tumor cells, particularly those of B-cell malignancies. However, before immunotherapy can become a more effective method of cancer care, there are many challenges that remain to be addressed and hurdles to overcome. Included are manipulation of tumor microenvironment (TME) to enhance T effector cell infiltration and access to the tumor, augmentation of tumor MHC expression for adequate presentation of tumor associated antigens, regulation of cytokines and their potential adverse effects, and reduced risk of secondary malignancies as a consequence of mutations generated by the various forms of genetic engineering of immune cells. Despite these challenges, the future of immunotherapy as a standard anticancer therapy is encouraging. Mol Cancer Res; 15(6); 635-50. ©2017 AACR.


Asunto(s)
Inmunoterapia/métodos , Complejo Mayor de Histocompatibilidad/fisiología , Neoplasias/inmunología , Neoplasias/terapia , Adenosina/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Tolerancia Inmunológica , Inmunoterapia/efectos adversos , Inmunoterapia/economía , Complejo Mayor de Histocompatibilidad/inmunología , Mutación , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Microambiente Tumoral
15.
Phys Rev E ; 95(1-1): 012403, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28208397

RESUMEN

The interactions between T-cell receptor (TCR) and peptide-major-histocompatibility complex (pMHC), which enable T-cell development and initiate adaptive immune responses, have been intensively studied. However, a central issue of how lipid rafts affect the TCR-pMHC interactions remains unclear. Here, by using a statistical-mechanical membrane model, we show that the binding affinity of TCR and pMHC anchored on two apposing cell membranes is significantly enhanced because of the lipid raft-induced signaling protein aggregation. This finding may provide an alternative insight into the mechanism of T-cell activation triggered by very low densities of pMHC. In the case of cell-substrate adhesion, our results indicate that the loss of lateral mobility of the proteins on the solid substrate leads to the inhibitory effect of lipid rafts on TCR-pMHC interactions. Our findings help to understand why different experimental methods for measuring the impact of lipid rafts on the receptor-ligand interactions have led to contradictory conclusions.


Asunto(s)
Complejo Mayor de Histocompatibilidad/fisiología , Microdominios de Membrana/metabolismo , Modelos Biológicos , Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Inmunidad Adaptativa , Animales , Adhesión Celular , Simulación por Computador , Difusión , Modelos Estadísticos , Método de Montecarlo , Agregado de Proteínas/fisiología
16.
Biochim Biophys Acta ; 1860(9): 1854-63, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27233453

RESUMEN

BACKGROUND: CD4 is a glycoprotein expressed on the surfaces of certain immune cells. On lymphocytes, an important function of CD4 is to co-engage Major Histocompatibility Complex (MHC) molecules with the T Cell Receptor (TCR), a process that is essential for antigen-specific activation of T cells. CD4 localizes dynamically into distinct membrane microdomains, an important feature of its immunoregulatory function that has also been shown to influence the efficiency of HIV replication. However, the mechanism by which CD4 localization is regulated and the biological significance of this is incompletely understood. METHODS: In this study, we used confocal microscopy, density-gradient centrifugation and flow cytometry to analyze dynamic redox-dependent effects on CD4 membrane domain localization. RESULTS: Blocking cell surface redox exchanges with both a membrane-impermeable sulfhydryl blocker (DTNB) and specific antibody inhibitors of Thioredoxin-1 (Trx1) induces translocation of CD4 into detergent-resistant membrane domains (DRM). In contrast, Trx1 inactivation does not change the localization of the chemokine receptor CCR5, suggesting that this effect is targeted. Moreover, DTNB treatment and Trx1 depletion coincide with strong inhibition of CD4-dependent HIV entry, but only moderate reductions in the infectivity of a CD4-independent HIV pseudovirion. CONCLUSIONS: Changes in the extracellular redox environment, potentially mediated by allosteric consequences of functional disulfide bond oxidoreduction, may represent a signal for translocation of CD4 into DRM clusters, and this sequestration, another potential mechanism by which the anti-HIV effects of cell surface oxidoreductase inhibition are exerted. GENERAL SIGNIFICANCE: Extracellular redox conditions may regulate CD4 function by potentiating changes in its membrane domain localization.


Asunto(s)
Antígenos CD4/metabolismo , Membrana Celular/metabolismo , Membrana Celular/virología , VIH-1/patogenicidad , Microdominios de Membrana/metabolismo , Tiorredoxinas/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Ácido Ditionitrobenzoico/farmacología , Células HeLa , Humanos , Complejo Mayor de Histocompatibilidad/fisiología , Oxidación-Reducción/efectos de los fármacos , Receptores CCR5/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T/virología , Internalización del Virus/efectos de los fármacos
17.
Immunogenetics ; 68(1): 3-18, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26392015

RESUMEN

Natural killer (NK) cells are immune cells that play a crucial role against viral infections and tumors. To be tolerant against healthy tissue and simultaneously attack infected cells, the activity of NK cells is tightly regulated by a sophisticated array of germline-encoded activating and inhibiting receptors. The best characterized mechanism of NK cell activation is "missing self" detection, i.e., the recognition of virally infected or transformed cells that reduce their MHC expression to evade cytotoxic T cells. To monitor the expression of MHC-I on target cells, NK cells have monomorphic inhibitory receptors which interact with conserved MHC molecules. However, there are other NK cell receptors (NKRs) encoded by gene families showing a remarkable genetic diversity. Thus, NKR haplotypes contain several genes encoding for receptors with activating and inhibiting signaling, and that vary in gene content and allelic polymorphism. But if missing-self detection can be achieved by a monomorphic NKR system why have these polygenic and polymorphic receptors evolved? Here, we review the expansion of NKR receptor families in different mammal species, and we discuss several hypotheses that possibly underlie the diversification of the NK cell receptor complex, including the evolution of viral decoys, peptide sensitivity, and selective MHC-downregulation.


Asunto(s)
Evolución Molecular , Células Asesinas Naturales , Receptores de Células Asesinas Naturales/fisiología , Animales , Evolución Biológica , Variación Genética , Haplotipos , Humanos , Células Asesinas Naturales/inmunología , Complejo Mayor de Histocompatibilidad/fisiología , Mamíferos/genética , Mamíferos/inmunología , Primates/genética , Primates/inmunología , Reproducción , Roedores/genética , Roedores/inmunología , Virosis/inmunología
18.
Pharmacotherapy ; 35(10): 963-76, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26497482

RESUMEN

The treatment of cancer has largely relied on killing tumor cells with nonspecific cytotoxic therapies and radiotherapy. This approach, however, has limitations including severe systemic toxicities, bystander effects on normal cells, recurrence of drug-resistant tumor cells, and the inability to target micrometastases or subclinical disease. An increased understanding of the critical role of the immune system in cancer development and progression has led to new treatment strategies using various immunotherapies. It is now recognized that established tumors have numerous mechanisms of suppressing the antitumor immune response including production of inhibitory cytokines, recruitment of immunosuppressive immune cells, and upregulation of coinhibitory receptors known as immune checkpoints. This review focuses on the immune checkpoint inhibitors, a novel class of immunotherapy first approved in 2011. Our objective is to highlight similarities and differences among the three immune checkpoint inhibitors approved by the U.S. Food and Drug Administration-ipilimumab, pembrolizumab, and nivolumab-to facilitate therapeutic decision making. We conducted a review of the published literature and conference proceedings and present a critical appraisal of the clinical evidence supporting their use in the treatment of metastatic melanoma and advanced squamous non-small cell lung cancer (NSCLC). We also compare and contrast their current place in cancer therapy and patterns of immune-related toxicities, and discuss the role of dual immune checkpoint inhibition and strategies for the management of immune-related adverse events. The immune checkpoint inhibitors have demonstrated a dramatic improvement in overall survival in patients with advanced melanoma and squamous NSCLC, along with acceptable toxicity profiles. These agents have a clear role in the first-line treatment of advanced melanoma and in the second-line treatment of advanced squamous NSCLC.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígeno CTLA-4/antagonistas & inhibidores , Melanoma/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Humanos , Inmunoterapia , Ipilimumab , Neoplasias Pulmonares/tratamiento farmacológico , Complejo Mayor de Histocompatibilidad/fisiología , Melanoma/genética , Melanoma/patología , Metástasis de la Neoplasia , Nivolumab , Proteínas Proto-Oncogénicas B-raf/genética , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptores de Antígenos de Linfocitos T/metabolismo , Estados Unidos
19.
Am J Transplant ; 15(6): 1580-90, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25824550

RESUMEN

Kidney allografts possess the ability to enable a short course of immunosuppression to induce tolerance of themselves and of cardiac allografts across a full-MHC barrier in miniature swine. However, the renal element(s) responsible for kidney-induced cardiac allograft tolerance (KICAT) are unknown. Here we investigated whether MHC disparities between parenchyma versus hematopoietic-derived "passenger" cells of the heart and kidney allografts affected KICAT. Heart and kidney allografts were co-transplanted into MHC-mismatched recipients treated with high-dose tacrolimus for 12 days. Group 1 animals (n = 3) received kidney and heart allografts fully MHC-mismatched to each other and to the recipient. Group 2 animals (n = 3) received kidney and heart allografts MHC-matched to each other but MHC-mismatched to the recipient. Group 3 animals (n = 3) received chimeric kidney allografts whose parenchyma was MHC-mismatched to the donor heart. Group 4 animals (n = 3) received chimeric kidney allografts whose passenger leukocytes were MHC-mismatched to the donor heart. Five of six heart allografts in Groups 1 and 3 rejected <40 days. In contrast, heart allografts in Groups 2 and 4 survived >150 days without rejection (p < 0.05). These data demonstrate that KICAT requires MHC-matching between kidney allograft parenchyma and heart allografts, suggesting that cells intrinsic to the kidney enable cardiac allograft tolerance.


Asunto(s)
Trasplante de Corazón , Corazón/fisiología , Histocompatibilidad/fisiología , Trasplante de Riñón , Riñón/fisiología , Complejo Mayor de Histocompatibilidad/fisiología , Tolerancia al Trasplante/fisiología , Aloinjertos , Animales , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Histocompatibilidad/inmunología , Terapia de Inmunosupresión , Inmunosupresores/uso terapéutico , Complejo Mayor de Histocompatibilidad/inmunología , Modelos Animales , Porcinos , Porcinos Enanos , Tacrolimus/uso terapéutico , Obtención de Tejidos y Órganos , Tolerancia al Trasplante/inmunología
20.
J Neuroinflammation ; 11: 57, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24666681

RESUMEN

BACKGROUND: Tissue-resident antigen-presenting cells (APC) exert a major influence on the local immune environment. Microglia are resident myeloid cells in the central nervous system (CNS), deriving from early post-embryonic precursors, distinct from adult hematopoietic lineages. Dendritic cells (DC) and macrophages infiltrate the CNS during experimental autoimmune encephalomyelitis (EAE). Microglia are not considered to be as effective APC as DC or macrophages. METHODS: In this work we compared the antigen presenting capacity of CD11c⁺ and CD11c⁻ microglia subsets with infiltrating CD11c⁺ APC, which include DC. The microglial subpopulations (CD11c⁻ CD45dim CD11b⁺ and CD11c⁺ CD45dim CD11b⁺) as well as infiltrating CD11c⁺ CD45high cells were sorted from CNS of C57BL/6 mice with EAE. Sorted cells were characterised by flow cytometry for surface phenotype and by quantitative real-time PCR for cytokine expression. They were co-cultured with primed T cells to measure induction of T cell proliferation and cytokine response. RESULTS: The number of CD11c⁺ microglia cells increased dramatically in EAE. They expressed equivalent levels of major histocompatibility complex and co-stimulatory ligands CD80 and CD86 as those expressed by CD11c⁺ cells infiltrating from blood. CD11c⁺ microglia differed significantly from blood-derived CD11c⁺ cells in their cytokine profile, expressing no detectable IL-6, IL-12 or IL-23, and low levels of IL-1ß. By contrast, CD11c⁻ microglia expressed low but detectable levels of all these cytokines. Transforming growth factor ß expression was similar in all three populations. Although CNS-resident and blood-derived CD11c⁺ cells showed equivalent ability to induce proliferation of myelin oligodendrocyte glycoprotein-immunised CD4⁺ T cells, CD11c⁺ microglia induced lower levels of T helper (Th)1 and Th17 cytokines, and did not induce Th2 cytokines. CONCLUSIONS: Our findings show distinct subtypes of APC in the inflamed CNS, with a hierarchy of functional competence for induction of CD4⁺ T cell responses.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Proliferación Celular , Sistema Nervioso Central/patología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Microglía/metabolismo , Linfocitos T/fisiología , Animales , Antígenos CD/metabolismo , Antígeno CD11c/metabolismo , Sistema Nervioso Central/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Citometría de Flujo , Complejo Mayor de Histocompatibilidad/fisiología , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA