Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nat Immunol ; 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956380

RESUMEN

Group 1 innate lymphoid cells (ILC1s) are cytotoxic and interferon gamma-producing lymphocytes lacking antigen-specific receptors, which include ILC1s and natural killer (NK) cells. In mice, ILC1s differ from NK cells, as they develop independently of the NK-specifying transcription factor EOMES, while requiring the repressor ZFP683 (ZNF683 in humans) for tissue residency. Here we identify highly variable ILC1 subtypes across tissues through investigation of human ILC1 diversity by single-cell RNA sequencing and flow cytometry. The intestinal epithelium contained abundant mature EOMES- ILC1s expressing PRDM1 rather than ZNF683, alongside a few immature TCF7+PRDM1- ILC1s. Other tissues harbored NK cells expressing ZNF683 and EOMES transcripts; however, EOMES protein content was variable. These ZNF683+ NK cells are tissue-imprinted NK cells phenotypically resembling ILC1s. The tissue ILC1-NK spectrum also encompassed conventional NK cells and NK cells distinguished by PTGDS expression. These findings establish a foundation for evaluating phenotypic and functional changes within the NK-ILC1 spectrum in diseases.

2.
Immunity ; 56(4): 797-812.e4, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-36801011

RESUMEN

The aryl-hydrocarbon receptor (AHR) is a ligand-activated transcription factor that buoys intestinal immune responses. AHR induces its own negative regulator, the AHR repressor (AHRR). Here, we show that AHRR is vital to sustaining intestinal intraepithelial lymphocytes (IELs). AHRR deficiency reduced IEL representation in a cell-intrinsic fashion. Single-cell RNA sequencing revealed an oxidative stress profile in Ahrr-/- IELs. AHRR deficiency unleashed AHR-induced expression of CYP1A1, a monooxygenase that generates reactive oxygen species, increasing redox imbalance, lipid peroxidation, and ferroptosis in Ahrr-/- IELs. Dietary supplementation with selenium or vitamin E to restore redox homeostasis rescued Ahrr-/- IELs. Loss of IELs in Ahrr-/- mice caused susceptibility to Clostridium difficile infection and dextran sodium-sulfate-induced colitis. Inflamed tissue of inflammatory bowel disease patients showed reduced Ahrr expression that may contribute to disease. We conclude that AHR signaling must be tightly regulated to prevent oxidative stress and ferroptosis of IELs and to preserve intestinal immune responses.


Asunto(s)
Ferroptosis , Linfocitos Intraepiteliales , Animales , Ratones , Linfocitos Intraepiteliales/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Estrés Oxidativo , Hidrocarburos
3.
Proc Natl Acad Sci U S A ; 120(52): e2318710120, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38109523

RESUMEN

Recent studies have characterized various mouse antigen-presenting cells (APCs) expressing the lymphoid-lineage transcription factor RORγt (Retinoid-related orphan receptor gamma t), which exhibit distinct phenotypic features and are implicated in the induction of peripheral regulatory T cells (Tregs) and immune tolerance to microbiota and self-antigens. These APCs encompass Janus cells and Thetis cell subsets, some of which express the AutoImmune REgulator (AIRE). RORγt+ MHCII+ type 3 innate lymphoid cells (ILC3) have also been implicated in the instruction of microbiota-specific Tregs. While RORγt+ APCs have been actively investigated in mice, the identity and function of these cell subsets in humans remain elusive. Herein, we identify a rare subset of RORγt+ cells with dendritic cell (DC) features through integrated single-cell RNA sequencing and single-cell ATAC sequencing. These cells, which we term RORγt+ DC-like cells (R-DC-like), exhibit DC morphology, express the MHC class II machinery, and are distinct from all previously reported DC and ILC3 subsets, but share transcriptional and epigenetic similarities with DC2 and ILC3. We have developed procedures to isolate and expand them in vitro, enabling their functional characterization. R-DC-like cells proliferate in vitro, continue to express RORγt, and differentiate into CD1c+ DC2-like cells. They stimulate the proliferation of allogeneic T cells. The identification of human R-DC-like cells with proliferative potential and plasticity toward CD1c+ DC2-like cells will prompt further investigation into their impact on immune homeostasis, inflammation, and autoimmunity.


Asunto(s)
Inmunidad Innata , Linfocitos , Humanos , Ratones , Animales , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Inflamación/metabolismo , Células Dendríticas
4.
Adv Exp Med Biol ; 1365: 97-112, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35567743

RESUMEN

The signature hallmark of adaptive immunity is the evolution of somatically rearranged antigen receptors, which confer both diversity and specificity to T and B lymphocytes. For decades, immunologists have observed cells which possess lymphoid characteristics yet lack such antigen-specific receptors. Collectively, these populations are referred to as innate lymphoid cells (ILCs) (Vivier et al. in Cell 174(5):1054-1066, 2018). Cytotoxic natural killer (NK) cells and lymphoid tissue-inducing cells (LTi), which contribute to the formation of lymphoid organs during embryogenesis, are the earliest described ILCs. Subsequently, diverse populations of ILCs have been described based on the signature cytokines they produce. Group 1 ILCs (ILC1) produce IFNγ, group 2 ILCs (ILC2) produce IL-5 and IL-13, and group 3 ILCs (ILC3) produce IL-22 and IL-17. In contrast to adaptive lymphocytes which take several days to undergo clonal expansion and acquire effector functions, ILCs secrete cytokines rapidly in response to activating signals in their tissue of residence. ILCs may also directly regulate adaptive lymphocytes and myeloid cells through co-stimulatory molecules and soluble factors. Thus, ILCs play important roles in both the initiation and amplification of the immune response. When properly regulated, ILCs maintain intestinal homeostasis and protect the host from infection by various pathogens. However, dysregulation of mucosal immunity drives intestinal inflammation and contributes to pathology, such as inflammatory bowel disease (IBD). In this review, we outline the roles that ILCs play in amplifying or regulating intestinal inflammation as well as ongoing efforts to target these disease mechanisms for IBD therapy.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Linfocitos , Citocinas , Humanos , Inmunidad Innata , Inflamación , Células Asesinas Naturales
5.
Eur J Immunol ; 47(4): 646-657, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28294319

RESUMEN

Drug-induced liver injury (DILI) is a major cause of acute liver failure (ALF), where hepatocyte necrotic products trigger liver inflammation, release of CXC chemokine receptor 2 (CXCR2) ligands (IL-8) and other neutrophil chemotactic molecules. Liver infiltration by neutrophils is a major cause of the life-threatening tissue damage that ensues. A GRPR (gastrin-releasing peptide receptor) antagonist impairs IL-8-induced neutrophil chemotaxis in vitro. We investigated its potential to reduce acetaminophen-induced ALF, neutrophil migration, and mechanisms underlying this phenomenon. We found that acetaminophen-overdosed mice treated with GRPR antagonist had reduced DILI and neutrophil infiltration in the liver. Intravital imaging and cell tracking analysis revealed reduced neutrophil mobility within the liver. Surprisingly, GRPR antagonist inhibited CXCL2-induced migration in vivo, decreasing neutrophil activation through CD11b and CD62L modulation. Additionally, this compound decreased CXCL8-driven neutrophil chemotaxis in vitro independently of CXCR2 internalization, induced activation of MAPKs (p38 and ERK1/2) and downregulation of neutrophil adhesion molecules CD11b and CD66b. In silico analysis revealed direct binding of GRPR antagonist and CXCL8 to the same binding spot in CXCR2. These findings indicate a new potential use for GRPR antagonist for treatment of DILI through a mechanism involving adhesion molecule modulation and possible direct binding to CXCR2.


Asunto(s)
Bombesina/análogos & derivados , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Neutrófilos/inmunología , Fragmentos de Péptidos/farmacología , Receptores de Bombesina/antagonistas & inhibidores , Receptores de Interleucina-8B/metabolismo , Animales , Bombesina/farmacología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Quimiotaxis/efectos de los fármacos , Humanos , Interleucina-8/metabolismo , Ratones , Ratones Endogámicos , Activación Neutrófila/efectos de los fármacos , Unión Proteica , Transducción de Señal/efectos de los fármacos
6.
Tumour Biol ; 39(3): 1010428317694321, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28351312

RESUMEN

Nerve fibers and neurotransmitters have increasingly been shown to have a role in tumor progression. Gastrin-releasing peptide is a neuropeptide linked to tumor aggressiveness, acting as an autocrine tumor growth factor by binding to its receptor, gastrin-releasing peptide receptor, expressed by many tumors. Although neuropeptides have been previously linked to tumor cell proliferation, more recent studies have uncovered roles for neuropeptides in chemotaxis and metastasis. Understanding the precise roles of such peptides in cancer is crucial to optimizing targeted therapy design. We have previously described that gastrin-releasing peptide acts directly as a chemotactic factor for neutrophils, dependent on PI3K, ERK, and p38. In this study, we investigated roles for gastrin-releasing peptide in lung adenocarcinoma. We asked if gastrin-releasing peptide would act as a proliferative and/or chemotactic stimulus for gastrin-releasing peptide receptor-expressing tumor cells. In A549 cells, a non-small cell lung carcinoma line, the treatment with gastrin-releasing peptide leads to activation of AKT and ERK1/2, and production of reactive oxygen species. Gastrin-releasing peptide induced migration of A549 cells, dependent on gastrin-releasing peptide receptor and PI3K, but not ERK. However, no proliferation was observed in these cells in response to gastrin-releasing peptide, and gastrin-releasing peptide did not promote resistance to treatment with a chemotherapy drug. Our results suggest that, similar to what happens in neutrophils, gastrin-releasing peptide is a migratory, rather than a proliferative, stimulus, for non-small cell lung carcinoma cells, indicating a putative role for gastrin-releasing peptide and gastrin-releasing peptide receptor in metastasis.


Asunto(s)
Adenocarcinoma/genética , Carcinogénesis/genética , Péptido Liberador de Gastrina/genética , Neoplasias Pulmonares/genética , Receptores de Bombesina/genética , Células A549 , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Antineoplásicos/administración & dosificación , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Péptido Liberador de Gastrina/administración & dosificación , Péptido Liberador de Gastrina/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Sistema de Señalización de MAP Quinasas/genética , Proteína Oncogénica v-akt/genética , Fosfatidilinositol 3-Quinasas/genética , Especies Reactivas de Oxígeno/metabolismo , Receptores de Bombesina/metabolismo
7.
Sci Immunol ; 9(95): eadi5374, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38758808

RESUMEN

The gut microbiota and tumor-associated macrophages (TAMs) affect tumor responses to anti-programmed cell death protein 1 (PD-1) immune checkpoint blockade. Reprogramming TAM by either blocking or deleting the macrophage receptor triggering receptor on myeloid cells 2 (TREM2) attenuates tumor growth, and lack of functional TREM2 enhances tumor elimination by anti-PD-1. Here, we found that anti-PD-1 treatment combined with TREM2 deficiency in mice induces proinflammatory programs in intestinal macrophages and a concomitant expansion of Ruminococcus gnavus in the gut microbiota. Gavage of wild-type mice with R. gnavus enhanced anti-PD-1-mediated tumor elimination, recapitulating the effect occurring in the absence of TREM2. A proinflammatory intestinal environment coincided with expansion, increased circulation, and migration of TNF-producing CD4+ T cells to the tumor bed. Thus, TREM2 remotely controls anti-PD-1 immune checkpoint blockade through modulation of the intestinal immune environment and microbiota, with R. gnavus emerging as a potential probiotic agent for increasing responsiveness to anti-PD-1.


Asunto(s)
Microbioma Gastrointestinal , Inmunoterapia , Macrófagos , Glicoproteínas de Membrana , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1 , Receptores Inmunológicos , Animales , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Ratones , Microbioma Gastrointestinal/inmunología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Inmunoterapia/métodos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Macrófagos/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones Noqueados , Femenino , Intestinos/inmunología
8.
Dev Cell ; 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38942017

RESUMEN

Recent advances in human genetics have shed light on the genetic factors contributing to inflammatory diseases, particularly Crohn's disease (CD), a prominent form of inflammatory bowel disease. Certain risk genes associated with CD directly influence cytokine biology and cell-specific communication networks. Current CD therapies primarily rely on anti-inflammatory drugs, which are inconsistently effective and lack strategies for promoting epithelial restoration and mucosal balance. To understand CD's underlying mechanisms, we investigated the link between CD and the FGFR1OP gene, which encodes a centrosome protein. FGFR1OP deletion in mouse intestinal epithelial cells disrupted crypt architecture, resulting in crypt loss, inflammation, and fatality. FGFR1OP insufficiency hindered epithelial resilience during colitis. FGFR1OP was crucial for preserving non-muscle myosin II activity, ensuring the integrity of the actomyosin cytoskeleton and crypt cell adhesion. This role of FGFR1OP suggests that its deficiency in genetically predisposed individuals may reduce epithelial renewal capacity, heightening susceptibility to inflammation and disease.

9.
iScience ; 26(2): 105991, 2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36824270

RESUMEN

The gut microbiota in early childhood is linked to asthma risk, but may continue to affect older patients with asthma. Here, we profile the gut microbiota of 38 children (19 asthma, median age 8) and 57 adults (17 asthma, median age 28) by 16S rRNA sequencing and find individuals with asthma harbored compositional differences from healthy controls in both adults and children. We develop a model to aid the design of mechanistic experiments in gnotobiotic mice and show enterotoxigenic Bacteroides fragilis (ETBF) is more prevalent in the gut microbiota of patients with asthma compared to healthy controls. In mice, ETBF, modulated by community context, can increase oxidative stress in the lungs during allergic airway inflammation (AAI). Our results provide evidence that ETBF affects the phenotype of airway inflammation in a subset of patients with asthma which suggests that therapies targeting the gut microbiota may be helpful tools for asthma control.

10.
J Clin Invest ; 133(13)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37279078

RESUMEN

Since the T-box transcription factors (TFs) T-BET and EOMES are necessary for initiation of NK cell development, their ongoing requirement for mature NK cell homeostasis, function, and molecular programming remains unclear. To address this, T-BET and EOMES were deleted in unexpanded primary human NK cells using CRISPR/Cas9. Deleting these TFs compromised in vivo antitumor response of human NK cells. Mechanistically, T-BET and EOMES were required for normal NK cell proliferation and persistence in vivo. NK cells lacking T-BET and EOMES also exhibited defective responses to cytokine stimulation. Single-cell RNA-Seq revealed a specific T-box transcriptional program in human NK cells, which was rapidly lost following T-BET and EOMES deletion. Further, T-BET- and EOMES-deleted CD56bright NK cells acquired an innate lymphoid cell precursor-like (ILCP-like) profile with increased expression of the ILC-3-associated TFs RORC and AHR, revealing a role for T-box TFs in maintaining mature NK cell phenotypes and an unexpected role of suppressing alternative ILC lineages. Our study reveals the critical importance of sustained EOMES and T-BET expression to orchestrate mature NK cell function and identity.


Asunto(s)
Inmunidad Innata , Proteínas de Dominio T Box , Humanos , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Células Asesinas Naturales/metabolismo , Factores de Transcripción/metabolismo , Citocinas/metabolismo
11.
Cancer Immunol Res ; 9(6): 600, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34365413

RESUMEN

In-depth immune profiling can guide the development and targeting of cancer immunotherapies. In this issue, Gherardin and colleagues find a strong signature of γδ T cells in the immune infiltrate of Merkel cell carcinoma that correlates with a more favorable prognosis and heightened responsiveness to checkpoint inhibitors. This may lead to future γδ T cell-targeted immunotherapies.See article by Gherardin et al., p. 612.


Asunto(s)
Carcinoma de Células de Merkel , Neoplasias Cutáneas , Humanos , Inmunoterapia , Pronóstico , Neoplasias Cutáneas/terapia , Linfocitos T
12.
Nat Commun ; 12(1): 1921, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33771991

RESUMEN

Crohn's disease (CD) is a chronic transmural inflammation of intestinal segments caused by dysregulated interaction between microbiome and gut immune system. Here, we profile, via multiple single-cell technologies, T cells purified from the intestinal epithelium and lamina propria (LP) from terminal ileum resections of adult severe CD cases. We find that intraepithelial lymphocytes (IEL) contain several unique T cell subsets, including NKp30+γδT cells expressing RORγt and producing IL-26 upon NKp30 engagement. Further analyses comparing tissues from non-inflamed and inflamed regions of patients with CD versus healthy controls show increased activated TH17 but decreased CD8+T, γδT, TFH and Treg cells in inflamed tissues. Similar analyses of LP find increased CD8+, as well as reduced CD4+T cells with an elevated TH17 over Treg/TFH ratio. Our analyses of CD tissues thus suggest a potential link, pending additional validations, between transmural inflammation, reduced IEL γδT cells and altered spatial distribution of IEL and LP T cell subsets.


Asunto(s)
Enfermedad de Crohn/inmunología , Linfocitos Intraepiteliales/inmunología , Análisis de la Célula Individual/métodos , Subgrupos de Linfocitos T/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Enfermedad de Crohn/patología , Perfilación de la Expresión Génica/métodos , Humanos , Linfocitos Intraepiteliales/metabolismo , Recuento de Linfocitos , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/inmunología , Células Th17/metabolismo
13.
Science ; 373(6553)2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34083450

RESUMEN

The meninges contain adaptive immune cells that provide immunosurveillance of the central nervous system (CNS). These cells are thought to derive from the systemic circulation. Through single-cell analyses, confocal imaging, bone marrow chimeras, and parabiosis experiments, we show that meningeal B cells derive locally from the calvaria, which harbors a bone marrow niche for hematopoiesis. B cells reach the meninges from the calvaria through specialized vascular connections. This calvarial-meningeal path of B cell development may provide the CNS with a constant supply of B cells educated by CNS antigens. Conversely, we show that a subset of antigen-experienced B cells that populate the meninges in aging mice are blood-borne. These results identify a private source for meningeal B cells, which may help maintain immune privilege within the CNS.


Asunto(s)
Subgrupos de Linfocitos B/fisiología , Linfocitos B/fisiología , Células de la Médula Ósea/fisiología , Sistema Nervioso Central/inmunología , Duramadre/citología , Linfopoyesis , Meninges/citología , Meninges/inmunología , Cráneo/anatomía & histología , Envejecimiento , Animales , Subgrupos de Linfocitos B/inmunología , Movimiento Celular , Sistema Nervioso Central/fisiología , Duramadre/inmunología , Fibroblastos/fisiología , Homeostasis , Privilegio Inmunológico , Ratones , Células Plasmáticas/fisiología , Análisis de la Célula Individual
14.
Cell Rep ; 33(5): 108331, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33147448

RESUMEN

Homeostatic mucosal immune responses are fine-tuned by naturally evolved interactions with native microbes, and integrating these relationships into experimental models can provide new insights into human diseases. Here, we leverage a murine-adapted airway microbe, Bordetella pseudohinzii (Bph), to investigate how chronic colonization impacts mucosal immunity and the development of allergic airway inflammation (AAI). Colonization with Bph induces the differentiation of interleukin-17A (IL-17A)-secreting T-helper cells that aid in controlling bacterial abundance. Bph colonization protects from AAI and is associated with increased production of secretory leukocyte protease inhibitor (SLPI), an antimicrobial peptide with anti-inflammatory properties. These findings are additionally supported by clinical data showing that higher levels of upper respiratory SLPI correlate both with greater asthma control and the presence of Haemophilus, a bacterial genus associated with AAI. We propose that SLPI could be used as a biomarker of beneficial host-commensal relationships in the airway.


Asunto(s)
Interacciones Microbiota-Huesped , Hipersensibilidad/microbiología , Hipersensibilidad/patología , Inflamación/patología , Pulmón/microbiología , Pulmón/patología , Microbiota , Inhibidor Secretorio de Peptidasas Leucocitarias/metabolismo , Células A549 , Adolescente , Adulto , Animales , Antígenos/metabolismo , Bordetella/fisiología , Niño , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Interacciones Microbiota-Huesped/genética , Humanos , Hipersensibilidad/complicaciones , Hipersensibilidad/inmunología , Inmunidad , Inflamación/complicaciones , Inflamación/inmunología , Inflamación/microbiología , Pulmón/inmunología , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Células Th17/inmunología , Transcriptoma/genética , Adulto Joven
15.
Cell Rep ; 31(9): 107720, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32492428

RESUMEN

Natural killer (NK) cells are cytotoxic innate lymphoid cells (ILCs) that mediate antiviral and antitumor responses and require the transcriptional regulator Eomesodermin (Eomes) for early development. However, the role of Eomes and its molecular program in mature NK cell biology is unclear. To address this, we develop a tamoxifen-inducible, type-1-ILC-specific (Ncr1-targeted) cre mouse and combine this with Eomes-floxed mice. Eomes deletion after normal NK cell ontogeny results in a rapid loss of NK cells (but not ILC1s), with a particularly profound effect on penultimately mature stage III NK cells. Mechanisms responsible for stage III reduction include increased apoptosis and impaired maturation from stage II precursors. Induced Eomes deletion also decreases NK cell cytotoxicity and abrogates in vivo rejection of major histocompatibility complex (MHC)-class-I-deficient cells. However, other NK cell functional responses, and stage IV NK cells, are largely preserved. These data indicate that mature NK cells have distinct Eomes-dependent and -independent stages.


Asunto(s)
Células Asesinas Naturales/inmunología , Proteínas de Dominio T Box/metabolismo , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Apoptosis , Puntos de Control del Ciclo Celular , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Receptores de Interleucina-15/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Bazo/citología , Bazo/inmunología , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética
16.
Cancer Discov ; 10(12): 1854-1871, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32826231

RESUMEN

Natural killer (NK) cells are an emerging cancer cellular therapy and potent mediators of antitumor immunity. Cytokine-induced memory-like (ML) NK cellular therapy is safe and induces remissions in patients with acute myeloid leukemia (AML). However, the dynamic changes in phenotype that occur after NK-cell transfer that affect patient outcomes remain unclear. Here, we report comprehensive multidimensional correlates from ML NK cell-treated patients with AML using mass cytometry. These data identify a unique in vivo differentiated ML NK-cell phenotype distinct from conventional NK cells. Moreover, the inhibitory receptor NKG2A is a dominant, transcriptionally induced checkpoint important for ML, but not conventional NK-cell responses to cancer. The frequency of CD8α+ donor NK cells is negatively associated with AML patient outcomes after ML NK therapy. Thus, elucidating the multidimensional dynamics of donor ML NK cells in vivo revealed critical factors important for clinical response, and new avenues to enhance NK-cell therapeutics. SIGNIFICANCE: Mass cytometry reveals an in vivo memory-like NK-cell phenotype, where NKG2A is a dominant checkpoint, and CD8α is associated with treatment failure after ML NK-cell therapy. These findings identify multiple avenues for optimizing ML NK-cell immunotherapy for cancer and define mechanisms important for ML NK-cell function.This article is highlighted in the In This Issue feature, p. 1775.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/metabolismo , Leucemia Mieloide Aguda/genética , Humanos , Leucemia Mieloide Aguda/patología
17.
Front Microbiol ; 8: 2447, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29312166

RESUMEN

Gastric infection by Helicobacter pylori is considered a risk factor for gastric and duodenal cancer, and extragastric diseases. Previous data have shown that, in a non-enzymatic way, H. pylori urease (HPU) activates neutrophils to produce ROS and also induces platelet aggregation, requiring ADP secretion modulated by the 12-lipoxygenase pathway, a signaling cascade also triggered by the physiological agonist collagen. Here we investigated further the effects on platelets of recombinant versions of the holoenzyme HPU, and of its two subunits (HpUreA and HpUreB). Although HpUreA had no aggregating activity on platelets, it partially inhibited collagen-induced aggregation. HpUreB induced platelet aggregation in the nanomolar range, and also interfered dose-dependently on both collagen- and ADP-induced platelet aggregation. HPU-induced platelet aggregation was inhibited by antibodies against glycoprotein VI (GPVI), the main collagen receptor in platelets. Flow cytometry analysis revealed exposure of P-selectin in HPU-activated platelets. Anti-glycoprotein IIbIIIa (GPIIbIIIa) antibodies increased the binding of FITC-labeled HPU to activated platelets, whereas anti-GPVI did not. Evaluation of post-transcriptional events in HPU-activated platelets revealed modifications in the pre-mRNA processing of pro-inflammatory proteins, with increased levels of mRNAs encoding IL-1ß and CD14. We concluded that HPU activates platelets probably through its HpUreB subunit. Activation of platelets by HPU turns these cells into a pro-inflammatory phenotype. Altogether, our data suggest that H. pylori urease, besides allowing bacterial survival within the gastric mucosa, may have an important, and so far overlooked, role in gastric inflammation mediated by urease-activated neutrophils and platelets.

18.
Vet Microbiol ; 201: 170-176, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28284605

RESUMEN

Mycoplasma hyopneumoniae is an economically significant swine pathogen that causes porcine enzootic pneumonia (PEP). Important processes for swine infection by M. hyopneumoniae depend on cell surface proteins, many of which are secreted by secretion pathways not completely elucidated so far. A putative type I signal peptidase (SPase I), a possible component of a putative Sec-dependent pathway, was annotated as a product of the sipS gene in the pathogenic M. hyopneumoniae 7448 genome. This M. hyopneumoniae putative SPase I (MhSPase I) displays only 14% and 23% of sequence identity/similarity to Escherichia coli bona fide SPase I, and, in complementation assays performed with a conditional E. coli SPase I mutant, only a partial restoration of growth was achieved with the heterologous expression of a recombinant MhSPase I (rMhSPase I). Considering the putative surface location of MhSPase I and its previously demonstrated capacity to induce a strong humoral response, we then assessed its potential to elicit a cellular and possible immunomodulatory response. In assays for immunogenicity assessment, rMhSPase I unexpectedly showed a cytotoxic effect on murine splenocytes. This cytotoxic effect was further confirmed using the swine epithelial PK(15) cell line in MTT and annexin V-flow cytometry assays, which showed that rMhSPase I induces apoptosis in a dose dependent-way. It was also demonstrated that this pro-apoptotic effect of rMhSPase I involves activation of a caspase-3 cascade. The potential relevance of the rMhSPase I pro-apoptotic effect for M. hyopneumoniae-host interactions in the context of PEP is discussed.


Asunto(s)
Apoptosis , Proteínas de la Membrana/metabolismo , Mycoplasma hyopneumoniae/enzimología , Neumonía Porcina por Mycoplasma/microbiología , Serina Endopeptidasas/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular , Proteínas de la Membrana/genética , Mycoplasma hyopneumoniae/genética , Serina Endopeptidasas/genética , Porcinos , Factores de Virulencia
20.
Vet Microbiol ; 190: 50-57, 2016 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-27283856

RESUMEN

Mycoplasma hyopneumoniae and Mycoplasma flocculare are two genetically close species found in the swine respiratory tract. Despite their similarities, while M. hyopneumoniae is the causative agent of porcine enzootic pneumonia, M. flocculare is a commensal bacterium. Genomic and transcriptional comparative analyses so far failed to explain the difference in pathogenicity between these two species. We then hypothesized that such difference might be, at least in part, explained by amino acid sequence and immunological or functional differences between ortholog surface proteins. In line with that, it was verified that approximately 85% of the ortholog surface proteins from M. hyopneumoniae 7448 and M. flocculare present one or more differential domains. To experimentally assess possible immunological implications of this kind of difference, the extracellular differential domains from one pair of orthologous surface proteins (MHP7448_0612, from M. hyopneumoniae, and MF_00357, from M. flocculare) were expressed in E. coli and used to immunize mice. The recombinant polypeptides (rMHP61267-169 and rMF35767-196, respectively) induced distinct cellular immune responses. While, rMHP61267-169 induced both Th1 and Th2 responses, rMF35767-196 induced just an early pro-inflammatory response. These results indicate that immunological properties determined by differential domains in orthologous surface protein might play a role in pathogenicity, contributing to elicit specific and differential immune responses against each species.


Asunto(s)
Inmunidad Celular/inmunología , Proteínas de la Membrana/inmunología , Mycoplasma hyopneumoniae/inmunología , Mycoplasma/inmunología , Dominios Proteicos/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Ratones , Proteínas Recombinantes/inmunología , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA