Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38870946

RESUMEN

Defective host defenses later in life are associated with changes in immune cell activities, suggesting that age-specific considerations are needed in immunotherapy approaches. In this study, we found that PD-1 and CTLA4-based cancer immunotherapies are unable to eradicate tumors in elderly mice. This defect in anti-tumor activity correlated with two known age-associated immune defects: diminished abundance of systemic naive CD8+ T cells and weak migratory activities of dendritic cells (DCs). We identified a vaccine adjuvant, referred to as a DC hyperactivator, which corrects DC migratory defects in the elderly. Vaccines containing tumor antigens and DC hyperactivators induced T helper type 1 (TH1) CD4+ T cells with cytolytic activity that drive anti-tumor immunity in elderly mice. When administered early in life, DC hyperactivators were the only adjuvant identified that elicited anti-tumor CD4+ T cells that persisted into old age. These results raise the possibility of correcting age-associated immune defects through DC manipulation.

2.
Annu Rev Immunol ; 33: 257-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25581309

RESUMEN

Receptors of the innate immune system detect conserved determinants of microbial and viral origin. Activation of these receptors initiates signaling events that culminate in an effective immune response. Recently, the view that innate immune signaling events rely on and operate within a complex cellular infrastructure has become an important framework for understanding the regulation of innate immunity. Compartmentalization within this infrastructure provides the cell with the ability to assign spatial information to microbial detection and regulate immune responses. Several cell biological processes play a role in the regulation of innate signaling responses; at the same time, innate signaling can engage cellular processes as a form of defense or to promote immunological memory. In this review, we highlight these aspects of cell biology in pattern-recognition receptor signaling by focusing on signals that originate from the cell surface, from endosomal compartments, and from within the cytosol.


Asunto(s)
Inmunidad Innata/fisiología , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Vías Biosintéticas , Membrana Celular/metabolismo , Endosomas/metabolismo , Humanos , Ligandos , Transducción de Señal
3.
Cell ; 185(4): 614-629.e21, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35148840

RESUMEN

Activation of the innate immune system via pattern recognition receptors (PRRs) is key to generate lasting adaptive immunity. PRRs detect unique chemical patterns associated with invading microorganisms, but whether and how the physical properties of PRR ligands influence the development of the immune response remains unknown. Through the study of fungal mannans, we show that the physical form of PRR ligands dictates the immune response. Soluble mannans are immunosilent in the periphery but elicit a potent pro-inflammatory response in the draining lymph node (dLN). By modulating the physical form of mannans, we developed a formulation that targets both the periphery and the dLN. When combined with viral glycoprotein antigens, this mannan formulation broadens epitope recognition, elicits potent antigen-specific neutralizing antibodies, and confers protection against viral infections of the lung. Thus, the physical properties of microbial ligands determine the outcome of the immune response and can be harnessed for vaccine development.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antígenos Virales/inmunología , Candida albicans/química , Mananos/inmunología , Hidróxido de Aluminio/química , Animales , Anticuerpos Neutralizantes/inmunología , Especificidad de Anticuerpos/inmunología , Linfocitos B/inmunología , COVID-19/inmunología , COVID-19/prevención & control , COVID-19/virología , Chlorocebus aethiops , Epítopos/inmunología , Inmunidad Innata , Inmunización , Inflamación/patología , Interferones/metabolismo , Lectinas Tipo C/metabolismo , Ligandos , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Senos Paranasales/metabolismo , Subunidades de Proteína/metabolismo , Lectina 1 Similar a Ig de Unión al Ácido Siálico/metabolismo , Solubilidad , Glicoproteína de la Espiga del Coronavirus/metabolismo , Linfocitos T/inmunología , Factor de Transcripción ReIB/metabolismo , Células Vero , beta-Glucanos/metabolismo
4.
Cell ; 184(19): 4953-4968.e16, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34492226

RESUMEN

Severe coronavirus disease 2019 (COVID-19) is characterized by overproduction of immune mediators, but the role of interferons (IFNs) of the type I (IFN-I) or type III (IFN-III) families remains debated. We scrutinized the production of IFNs along the respiratory tract of COVID-19 patients and found that high levels of IFN-III, and to a lesser extent IFN-I, characterize the upper airways of patients with high viral burden but reduced disease risk or severity. Production of specific IFN-III, but not IFN-I, members denotes patients with a mild pathology and efficiently drives the transcription of genes that protect against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In contrast, compared to subjects with other infectious or noninfectious lung pathologies, IFNs are overrepresented in the lower airways of patients with severe COVID-19 that exhibit gene pathways associated with increased apoptosis and decreased proliferation. Our data demonstrate a dynamic production of IFNs in SARS-CoV-2-infected patients and show IFNs play opposing roles at distinct anatomical sites.


Asunto(s)
COVID-19/patología , Interferones/metabolismo , Sistema Respiratorio/virología , Índice de Severidad de la Enfermedad , Factores de Edad , Envejecimiento/patología , COVID-19/genética , COVID-19/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Regulación de la Expresión Génica , Humanos , Interferones/genética , Leucocitos/patología , Leucocitos/virología , Pulmón/patología , Pulmón/virología , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/virología , Carga Viral
5.
Nat Immunol ; 21(1): 42-53, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31768073

RESUMEN

Pathogen-associated molecular patterns (PAMPs) have the capacity to couple inflammatory gene expression to changes in macrophage metabolism, both of which influence subsequent inflammatory activities. Similar to their microbial counterparts, several self-encoded damage-associated molecular patterns (DAMPs) induce inflammatory gene expression. However, whether this symmetry in host responses between PAMPs and DAMPs extends to metabolic shifts is unclear. Here, we report that the self-encoded oxidized phospholipid oxPAPC alters the metabolism of macrophages exposed to lipopolysaccharide. While cells activated by lipopolysaccharide rely exclusively on glycolysis, macrophages exposed to oxPAPC also use mitochondrial respiration, feed the Krebs cycle with glutamine, and favor the accumulation of oxaloacetate in the cytoplasm. This metabolite potentiates interleukin-1ß production, resulting in hyperinflammation. Similar metabolic adaptions occur in vivo in hypercholesterolemic mice and human subjects. Drugs that interfere with oxPAPC-driven metabolic changes reduce atherosclerotic plaque formation in mice, thereby underscoring the importance of DAMP-mediated activities in pathophysiological conditions.


Asunto(s)
Alarminas/inmunología , Lipopolisacáridos/inmunología , Macrófagos/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Fosfatidilcolinas/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Glucólisis/fisiología , Hipercolesterolemia/inmunología , Hipercolesterolemia/patología , Inflamación/prevención & control , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Fosforilación Oxidativa , Placa Aterosclerótica/patología , Placa Aterosclerótica/prevención & control
7.
Immunity ; 55(2): 224-236.e5, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34995475

RESUMEN

During gram-negative septicemia, interactions between platelets and neutrophils initiate a detrimental feedback loop that sustains neutrophil extracellular trap (NET) induction, disseminated intravascular coagulation, and inflammation. Understanding intracellular pathways that control platelet-neutrophil interactions is essential for identifying new therapeutic targets. Here, we found that thrombin signaling induced activation of the transcription factor NFAT in platelets. Using genetic and pharmacologic approaches, as well as iNFATuation, a newly developed mouse model in which NFAT activation can be abrogated in a cell-specific manner, we demonstrated that NFAT inhibition in activated murine and human platelets enhanced their activation and aggregation, as well as their interactions with neutrophils and NET induction. During gram-negative septicemia, NFAT inhibition in platelets promoted disease severity by increasing disseminated coagulation and NETosis. NFAT inhibition also partially restored coagulation ex vivo in patients with hypoactive platelets. Our results define non-transcriptional roles for NFAT that could be harnessed to address pressing clinical needs.


Asunto(s)
Plaquetas/efectos de los fármacos , Factores de Transcripción NFATC/antagonistas & inhibidores , Agregación Plaquetaria/efectos de los fármacos , Sepsis/patología , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Comunicación Celular/efectos de los fármacos , Gránulos Citoplasmáticos/metabolismo , Modelos Animales de Enfermedad , Trampas Extracelulares/metabolismo , Humanos , Inflamación , Ratones , Factores de Transcripción NFATC/metabolismo , Neutrófilos/metabolismo , Receptores de Trombina/metabolismo , Sepsis/metabolismo
8.
Immunity ; 54(7): 1369-1371, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260883

RESUMEN

Inflammaging drives age-related pathologies. In this issue of Immunity, Barkaway et al. illustrate how aged endothelial cells and mast cells promote reverse migration of neutrophils from inflamed tissue back into circulation, causing tissue damage at distal sites.


Asunto(s)
Células Endoteliales , Neutrófilos
9.
Immunity ; 54(6): 1186-1199.e7, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-33915108

RESUMEN

A cardinal feature of COVID-19 is lung inflammation and respiratory failure. In a prospective multi-country cohort of COVID-19 patients, we found that increased Notch4 expression on circulating regulatory T (Treg) cells was associated with disease severity, predicted mortality, and declined upon recovery. Deletion of Notch4 in Treg cells or therapy with anti-Notch4 antibodies in conventional and humanized mice normalized the dysregulated innate immunity and rescued disease morbidity and mortality induced by a synthetic analog of viral RNA or by influenza H1N1 virus. Mechanistically, Notch4 suppressed the induction by interleukin-18 of amphiregulin, a cytokine necessary for tissue repair. Protection by Notch4 inhibition was recapitulated by therapy with Amphiregulin and, reciprocally, abrogated by its antagonism. Amphiregulin declined in COVID-19 subjects as a function of disease severity and Notch4 expression. Thus, Notch4 expression on Treg cells dynamically restrains amphiregulin-dependent tissue repair to promote severe lung inflammation, with therapeutic implications for COVID-19 and related infections.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad Celular , Neumonía Viral/etiología , Neumonía Viral/metabolismo , Receptor Notch4/metabolismo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Anfirregulina/farmacología , Animales , Biomarcadores , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunohistoquímica , Inmunomodulación/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Virus de la Influenza A/fisiología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Ratones , Ratones Transgénicos , Neumonía Viral/patología , Receptor Notch4/antagonistas & inhibidores , Receptor Notch4/genética , Índice de Severidad de la Enfermedad
10.
Nat Immunol ; 18(10): 1084-1093, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28846084

RESUMEN

Interferon-λ (IFN-λ) is a central regulator of mucosal immunity; however, its signaling specificity relative to that of type I interferons is poorly defined. IFN-λ can induce antiviral interferon-stimulated genes (ISGs) in epithelia, while the effect of IFN-λ in non-epithelial cells remains unclear. Here we report that neutrophils responded to IFN-λ. We found that in addition to inducing ISG transcription, IFN-λ (but not IFN-ß) specifically activated a translation-independent signaling pathway that diminished the production of reactive oxygen species and degranulation in neutrophils. In mice, IFN-λ was elicited by enteric viruses and acted on neutrophils to decrease oxidative stress and intestinal damage. Thus, IFN-λ acted as a unique immunomodulatory agent by modifying transcriptional and non-translational neutrophil responses, which might permit a controlled development of the inflammatory process.


Asunto(s)
Gastroenteritis/etiología , Gastroenteritis/metabolismo , Interferón gamma/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Animales , Análisis por Conglomerados , Modelos Animales de Enfermedad , Gastroenteritis/patología , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Intestinos/patología , Ratones , Ratones Noqueados , Microbiota , Estrés Oxidativo , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo
11.
Nat Methods ; 20(5): 714-722, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37012480

RESUMEN

Major aims of single-cell proteomics include increasing the consistency, sensitivity and depth of protein quantification, especially for proteins and modifications of biological interest. Here, to simultaneously advance all these aims, we developed prioritized Single-Cell ProtEomics (pSCoPE). pSCoPE consistently analyzes thousands of prioritized peptides across all single cells (thus increasing data completeness) while maximizing instrument time spent analyzing identifiable peptides, thus increasing proteome depth. These strategies increased the sensitivity, data completeness and proteome coverage over twofold. The gains enabled quantifying protein variation in untreated and lipopolysaccharide-treated primary macrophages. Within each condition, proteins covaried within functional sets, including phagosome maturation and proton transport, similarly across both treatment conditions. This covariation is coupled to phenotypic variability in endocytic activity. pSCoPE also enabled quantifying proteolytic products, suggesting a gradient of cathepsin activities within a treatment condition. pSCoPE is freely available and widely applicable, especially for analyzing proteins of interest without sacrificing proteome coverage. Support for pSCoPE is available at http://scp.slavovlab.net/pSCoPE .


Asunto(s)
Proteoma , Proteómica , Proteoma/análisis , Proteómica/métodos , Espectrometría de Masas , Péptidos/química , Macrófagos
12.
Immunity ; 47(4): 697-709.e3, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29045901

RESUMEN

A heterogeneous mixture of lipids called oxPAPC, derived from dying cells, can hyperactivate dendritic cells (DCs) but not macrophages. Hyperactive DCs are defined by their ability to release interleukin-1 (IL-1) while maintaining cell viability, endowing these cells with potent aptitude to stimulate adaptive immunity. Herein, we found that the bacterial lipopolysaccharide receptor CD14 captured extracellular oxPAPC and delivered these lipids into the cell to promote inflammasome-dependent DC hyperactivation. Notably, we identified two specific components within the oxPAPC mixture that hyperactivated macrophages, allowing these cells to release IL-1 for several days, by a CD14-dependent process. In murine models of sepsis, conditions that promoted cell hyperactivation resulted in inflammation but not lethality. Thus, multiple phagocytes are capable of hyperactivation in response to oxPAPC, with CD14 acting as the earliest regulator in this process, serving to capture and transport these lipids to promote inflammatory cell fate decisions.


Asunto(s)
Células Dendríticas/inmunología , Inflamasomas/inmunología , Receptores de Lipopolisacáridos/inmunología , Fagocitos/inmunología , Fosfatidilcolinas/inmunología , Inmunidad Adaptativa/inmunología , Animales , Western Blotting , Línea Celular , Supervivencia Celular/inmunología , Células Dendríticas/metabolismo , Endocitosis/efectos de los fármacos , Endocitosis/inmunología , Femenino , Citometría de Flujo , Células HEK293 , Humanos , Inflamasomas/metabolismo , Interleucina-1/inmunología , Interleucina-1/metabolismo , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitos/metabolismo , Fosfatidilcolinas/metabolismo
13.
Cell ; 147(4): 868-80, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-22078883

RESUMEN

The transport of Toll-like Receptors (TLRs) to various organelles has emerged as an essential means by which innate immunity is regulated. While most of our knowledge is restricted to regulators that promote the transport of newly synthesized receptors, the regulators that control TLR transport after microbial detection remain unknown. Here, we report that the plasma membrane localized Pattern Recognition Receptor (PRR) CD14 is required for the microbe-induced endocytosis of TLR4. In dendritic cells, this CD14-dependent endocytosis pathway is upregulated upon exposure to inflammatory mediators. We identify the tyrosine kinase Syk and its downstream effector PLCγ2 as important regulators of TLR4 endocytosis and signaling. These data establish that upon microbial detection, an upstream PRR (CD14) controls the trafficking and signaling functions of a downstream PRR (TLR4). This innate immune trafficking cascade illustrates how pathogen detection systems operate to induce both membrane transport and signal transduction.


Asunto(s)
Endocitosis , Receptores de Lipopolisacáridos/metabolismo , Receptor Toll-Like 4/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Células Dendríticas/inmunología , Endosomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Fosfolipasa C gamma/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Quinasa Syk
14.
J Infect Dis ; 229(3): 876-887, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-37671668

RESUMEN

Mycobacterium tuberculosis (Mtb)-specific γ9δ2 T cells secrete granzyme A (GzmA) protective against intracellular Mtb growth. However, GzmA-enzymatic activity is unnecessary for pathogen inhibition, and the mechanisms of GzmA-mediated protection remain unknown. We show that GzmA homodimerization is essential for opsonization of mycobacteria, altered uptake into human monocytes, and subsequent pathogen clearance within the phagolysosome. Although monomeric and homodimeric GzmA bind mycobacteria, only homodimers also bind cluster of differentiation 14 (CD14) and Toll-like receptor 4 (TLR4). Without access to surface-expressed CD14 and TLR4, GzmA fails to inhibit intracellular Mtb. Upregulation of Rab11FIP1 was associated with inhibitory activity. Furthermore, GzmA colocalized with and was regulated by protein disulfide isomerase AI (PDIA1), which cleaves GzmA homodimers into monomers and prevents Mtb inhibitory activity. These studies identify a previously unrecognized role for homodimeric GzmA structure in opsonization, phagocytosis, and elimination of Mtb in human monocytes, and they highlight PDIA1 as a potential host-directed therapy for prevention and treatment of tuberculosis, a major human disease.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Granzimas/metabolismo , Monocitos/metabolismo , Receptor Toll-Like 4/metabolismo , Tuberculosis/microbiología
15.
J Med Virol ; 96(1): e29343, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38163281

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes COVID-19 and has brought a huge burden in terms of human lives. Strict social distance and influenza vaccination have been recommended to avoid co-infections between influenza viruses and SARS-CoV-2. Scattered reports suggested a protective effect of influenza vaccine on COVID-19 development and severity. We analyzed 51 studies on the capacity of influenza vaccination to affect infection with SARS-CoV-2, hospitalization, admission to Intensive Care Units (ICU), and mortality. All subjects taken into consideration did not receive any anti-SARS-CoV-2 vaccine, although their status with respect to previous infections with SARS-CoV-2 is not known. Comparison between vaccinated and not-vaccinated subjects for each of the four endpoints was expressed as odds ratio (OR), with 95% confidence intervals (CIs); all analyses were performed by DerSimonian and Laird model, and Hartung-Knapp model when studies were less than 10. In a total of 61 029 936 subjects from 33 studies, influenza vaccination reduced frequency of SARS-CoV-2 infection [OR plus 95% CI = 0.70 (0.65-0.77)]. The effect was significant in all studies together, in health care workers and in the general population; distance from influenza vaccination and the type of vaccine were also of importance. In 98 174 subjects from 11 studies, frequency of ICU admission was reduced with influenza vaccination [OR (95% CI) = 0.71 (0.54-0.94)]; the effect was significant in all studies together, in pregnant women and in hospitalized subjects. In contrast, in 4 737 328 subjects from 14 studies hospitalization was not modified [OR (95% CI) = 1.05 (0.82-1.35)], and in 4 139 660 subjects from 19 studies, mortality was not modified [OR (95% CI) = 0.76 (0.26-2.20)]. Our study emphasizes the importance of influenza vaccination in the protection against SARS-CoV-2 infection.


Asunto(s)
COVID-19 , Vacunas contra la Influenza , Femenino , Humanos , Embarazo , COVID-19/epidemiología , COVID-19/mortalidad , Hospitalización , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/epidemiología , Gripe Humana/prevención & control , SARS-CoV-2 , Vacunación
16.
Immunity ; 43(5): 909-22, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26546281

RESUMEN

Microbe-induced receptor trafficking has emerged as an essential means to promote innate immune signal transduction. Upon detection of bacterial lipopolysaccharides (LPS), CD14 induces an inflammatory endocytosis pathway that delivers Toll-like receptor 4 (TLR4) to endosomes. Although several regulators of CD14-dependent TLR4 endocytosis have been identified, the cargo-selection mechanism during this process remains unknown. We reveal that, in contrast to classic cytosolic interactions that promoted the endocytosis of transmembrane receptors, TLR4 was selected as cargo for inflammatory endocytosis entirely through extracellular interactions. Mechanistically, the extracellular protein MD-2 bound to and dimerized TLR4 in order to promote this endocytic event. Our analysis of LPS variants from human pathogens and gut commensals revealed a common mechanism by which bacteria prevent inflammatory endocytosis. We suggest that evasion of CD14-dependent endocytosis is an attribute that transcends the concept of pathogenesis and might be a fundamental feature of bacteria that inhabit eukaryotic hosts.


Asunto(s)
Bacterias/inmunología , Endocitosis/inmunología , Evasión Inmune/inmunología , Receptores de Lipopolisacáridos/metabolismo , Receptor Toll-Like 4/metabolismo , Células Cultivadas , Humanos , Inflamación/inmunología , Lipopolisacáridos/inmunología , Antígeno 96 de los Linfocitos/inmunología , Transporte de Proteínas/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología
17.
Am J Transplant ; 23(7): 935-945, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37080464

RESUMEN

Advances in immunosuppression have been relatively stagnant over the past 2 decades, and transplant recipients continue to experience long-term morbidity associated with immunosuppression regimens. Strategies to reduce or eliminate the dosage of immunosuppression medications are needed. We discovered a novel administration strategy using the classic adjuvant alum to condition murine islet transplant recipients, known as adjuvant conditioning (AC), to expand both polymorphonuclear and monocytic myeloid-derived suppressive cells (MDSCs) in vivo. These AC MDSCs potently suppress T cell proliferation when cultured together in vitro. AC MDSCs also facilitate naïve CD4+ T cells to differentiate into regulatory T cells. In addition, we were able to demonstrate a significant delay in alloislet rejection compared with that by saline-treated control following adjuvant treatment in a MDSC-dependent manner. Furthermore, AC MDSCs produce significantly more interleukin (IL)-10 than saline-treated controls, which we demonstrated to be critical for the increased T cell suppressor function of AC MDSCs as well as the observed protective effect of AC against alloislet rejection. Our data suggest that adjuvant-related therapeutics designed to expand MDSCs could be a useful strategy to prevent transplant rejection and curb the use of toxic immunosuppressive regimens currently used in transplant patients.


Asunto(s)
Células Supresoras de Origen Mieloide , Humanos , Animales , Ratones , Inmunosupresores/farmacología , Monocitos , Linfocitos T CD8-positivos , Terapia de Inmunosupresión
18.
Clin Microbiol Rev ; 34(3)2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-33789928

RESUMEN

Several viruses target the human respiratory tract, causing different clinical manifestations spanning from mild upper airway involvement to life-threatening acute respiratory distress syndrome (ARDS). As dramatically evident in the ongoing SARS-CoV-2 pandemic, the clinical picture is not always easily predictable due to the combined effect of direct viral and indirect patient-specific immune-mediated damage. In this review, we discuss the main RNA (orthomyxoviruses, paramyxoviruses, and coronaviruses) and DNA (adenoviruses, herpesviruses, and bocaviruses) viruses with respiratory tropism and their mechanisms of direct and indirect cell damage. We analyze the thin line existing between a protective immune response, capable of limiting viral replication, and an unbalanced, dysregulated immune activation often leading to the most severe complication. Our comprehension of the molecular mechanisms involved is increasing and this should pave the way for the development and clinical use of new tailored immune-based antiviral strategies.


Asunto(s)
Virus ADN , Lesión Pulmonar , Virus ARN , Infecciones del Sistema Respiratorio , Virosis , Adulto , Anciano , Antivirales/uso terapéutico , COVID-19 , Niño , Preescolar , Femenino , Humanos , Factores Inmunológicos/uso terapéutico , Lactante , Recién Nacido , Interferones/uso terapéutico , Pulmón/inmunología , Pulmón/virología , Lesión Pulmonar/diagnóstico , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/inmunología , Lesión Pulmonar/virología , Masculino , Persona de Mediana Edad , Pandemias , SARS-CoV-2
19.
Eur J Immunol ; 50(3): 317-325, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31986556

RESUMEN

Humans are constantly exposed to fungi, either in the form of commensals at epithelial barriers or as inhaled spores. Innate immune cells play a pivotal role in maintaining commensal relationships and preventing skin, mucosal, or systemic fungal infections due to the expression of pattern recognition receptors that recognize fungal cell wall components and modulate both their activation status and the ensuing adaptive immune response. Commensal fungi also play a critical role in the modulation of homeostasis and disease susceptibility at epithelial barriers. This review will outline cellular and molecular mechanisms of anti-fungal innate immunity focusing on C-type lectin receptors and their relevance in the context of host-fungi interactions at skin and mucosal surfaces in murine experimental models as well as patients susceptible to fungal infections.


Asunto(s)
Hongos/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Inmunidad Mucosa/inmunología , Lectinas Tipo C/inmunología , Micosis/inmunología , Animales , Humanos
20.
Semin Immunol ; 34: 61-67, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28855088

RESUMEN

Nanoparticles represent a new generation of drug delivery systems that can be engineered to harness optimal target selectivity for specific cells and tissues and high drug loading capacity, allowing for improved pharmacokinetics and enhanced bioavailability of therapeutics. The spontaneous propensity of both organic and colloidal nanoparticles to be captured by the cells of the reticuloendothelial system encouraged their utilization as passive targeting systems that can be preferentially directed to innate immune cells, such as macrophages, dendritic cells and neutrophils. The natural affinity for phagocytic cells suggests the possible implementation of nanoparticles as an immunotherapeutic platform for inflammatory diseases and autoimmune disorders. Here we discuss the recent advances in the application of nanotechnology to induce antigen-specific tolerance in autoimmunity and the use of nanoparticles for anti-inflammatory therapies, including treatment of inflammatory bowel diseases, psoriasis and rheumatoid arthritis.


Asunto(s)
Células Dendríticas/inmunología , Macrófagos/inmunología , Nanopartículas/metabolismo , Neutrófilos/inmunología , Autoinmunidad , Coloides/química , Sistemas de Liberación de Medicamentos , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Sistema Mononuclear Fagocítico , Nanomedicina , Nanopartículas/química , Nanotecnología , Fagocitosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA