Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 586(7829): 417-423, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32999463

RESUMEN

Microglia, the brain's resident macrophages, help to regulate brain function by removing dying neurons, pruning non-functional synapses, and producing ligands that support neuronal survival1. Here we show that microglia are also critical modulators of neuronal activity and associated behavioural responses in mice. Microglia respond to neuronal activation by suppressing neuronal activity, and ablation of microglia amplifies and synchronizes the activity of neurons, leading to seizures. Suppression of neuronal activation by microglia occurs in a highly region-specific fashion and depends on the ability of microglia to sense and catabolize extracellular ATP, which is released upon neuronal activation by neurons and astrocytes. ATP triggers the recruitment of microglial protrusions and is converted by the microglial ATP/ADP hydrolysing ectoenzyme CD39 into AMP; AMP is then converted into adenosine by CD73, which is expressed on microglia as well as other brain cells. Microglial sensing of ATP, the ensuing microglia-dependent production of adenosine, and the adenosine-mediated suppression of neuronal responses via the adenosine receptor A1R are essential for the regulation of neuronal activity and animal behaviour. Our findings suggest that this microglia-driven negative feedback mechanism operates similarly to inhibitory neurons and is essential for protecting the brain from excessive activation in health and disease.


Asunto(s)
Retroalimentación Fisiológica , Microglía/fisiología , Inhibición Neural , Neuronas/fisiología , 5'-Nucleotidasa/metabolismo , Potenciales de Acción , Adenosina/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antígenos CD/metabolismo , Apirasa/metabolismo , Calcio/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Inhibición Neural/genética , Receptor de Adenosina A1/metabolismo , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Factores de Tiempo
2.
J Infect Dis ; 230(1): 120-130, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052721

RESUMEN

BACKGROUND: Adenosine triphosphate (ATP) enhances neutrophil responses, but little is known about the role of ATP in influenza infections. METHODS: We used a mouse influenza model to study if ATP release is associated with neutrophil activation and disease progression. RESULTS: Influenza infection increased pulmonary ATP levels 5-fold and plasma ATP levels 3-fold vs healthy mice. Adding ATP at those concentrations to blood from healthy mice primed neutrophils and enhanced CD11b and CD63 expression, CD62L shedding, and reactive oxygen species production in response to formyl peptide receptor stimulation. Influenza infection also primed neutrophils in vivo, resulting in formyl peptide receptor-induced CD11b expression and CD62L shedding up to 3 times higher than that of uninfected mice. In infected mice, large numbers of neutrophils entered the lungs. These cells were significantly more activated than the peripheral neutrophils of infected mice and pulmonary neutrophils of healthy mice. Plasma ATP levels of infected mice and influenza disease progression corresponded with the numbers and activation level of their pulmonary neutrophils. CONCLUSIONS: Findings suggest that ATP release from the lungs of infected mice promotes influenza disease progression by priming peripheral neutrophils, which become strongly activated and cause pulmonary tissue damage after their recruitment to the lungs.


Asunto(s)
Adenosina Trifosfato , Progresión de la Enfermedad , Pulmón , Activación Neutrófila , Neutrófilos , Infecciones por Orthomyxoviridae , Animales , Adenosina Trifosfato/metabolismo , Pulmón/patología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Infecciones por Orthomyxoviridae/metabolismo , Ratones , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos C57BL , Ratones Endogámicos BALB C , Especies Reactivas de Oxígeno/metabolismo
3.
Brain Behav Immun ; 2024 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-39043348

RESUMEN

BACKGROUND: Sleep deficiencies, such as manifested in short sleep duration or insomnia symptoms, are known to increase the risk for multiple disease conditions involving immunopathology. Inflammation is hypothesized to be a mechanism through which deficient sleep acts as a risk factor for these conditions. Thus, one potential way to mitigate negative health consequences associated with deficient sleep is to target inflammation. Few interventional sleep studies investigated whether improving sleep affects inflammatory processes, but results suggest that complementary approaches may be necessary to target inflammation associated with sleep deficiencies. We investigated whether targeting inflammation through low-dose acetylsalicylic acid (ASA, i.e., aspirin) is able to blunt the inflammatory response to experimental sleep restriction. METHODS: 46 healthy participants (19F/27 M, age range 19-63 years) were studied in a double-blind randomized placebo-controlled crossover trial with three protocols each consisting of a 14-day at-home monitoring phase followed by an 11-day (10-night) in-laboratory stay (sleep restriction/ASA, sleep restriction/placebo, control sleep/placebo). In the sleep restriction/ASA condition, participants took low-dose ASA (81 mg/day) daily in the evening (22:00) during the at-home phase and the subsequent in-laboratory stay. In the sleep restriction/placebo and control sleep/placebo conditions, participants took placebo daily. Each in-laboratory stay started with 2 nights with a sleep opportunity of 8 h/night (23:00-07:00) for adaptation and baseline measurements. Under the two sleep restriction conditions, participants were exposed to 5 nights of sleep restricted to a sleep opportunity of 4 h/night (03:00-07:00) followed by 3 nights of recovery sleep with a sleep opportunity of 8 h/night. Under the control sleep condition, participants had a sleep opportunity of 8 h/night throughout the in-laboratory stay. During each in-laboratory stay, participants had 3 days of intensive monitoring (at baseline, 5th day of sleep restriction/control sleep, and 2nd day of recovery sleep). Variables, including pro-inflammatory immune cell function, C-reactive protein (CRP), and actigraphy-estimated measures of sleep, were analyzed using generalized linear mixed models. RESULTS: Low-dose ASA administration reduced the interleukin (IL)-6 expression in LPS-stimulated monocytes (p < 0.05 for condition*day) and reduced serum CRP levels (p < 0.01 for condition) after 5 nights of sleep restriction compared to placebo administration in the sleep restriction condition. Low-dose ASA also reduced the amount of cyclooxygenase (COX)-1/COX-2 double positive cells among LPS-stimulated monocytes after 2 nights of recovery sleep following 5 nights of sleep restriction compared to placebo (p < 0.05 for condition). Low-dose ASA further decreased wake after sleep onset (WASO) and increased sleep efficiency (SE) during the first 2 nights of recovery sleep (p < 0.001 for condition and condition*day). Baseline comparisons revealed no differences between conditions for all of the investigated variables (p > 0.05 for condition). CONCLUSION: This study shows that inflammatory responses to sleep restriction can be reduced by preemptive administration of low-dose ASA. This finding may open new therapeutic approaches to prevent or control inflammation and its consequences in those experiencing sleep deficiencies. TRIAL REGISTRATION: ClinicalTrials.gov NCT03377543.

4.
Immun Ageing ; 21(1): 45, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961477

RESUMEN

BACKGROUND: The function of polymorphonuclear neutrophils (PMNs) decreases with age, which results in infectious and inflammatory complications in older individuals. The underlying causes are not fully understood. ATP release and autocrine stimulation of purinergic receptors help PMNs combat microbial invaders. Excessive extracellular ATP interferes with these mechanisms and promotes inflammatory PMN responses. Here, we studied whether dysregulated purinergic signaling in PMNs contributes to their dysfunction in older individuals. RESULTS: Bacterial infection of C57BL/6 mice resulted in exaggerated PMN activation that was significantly greater in old mice (64 weeks) than in young animals (10 weeks). In contrast to young animals, old mice were unable to prevent the systemic spread of bacteria, resulting in lethal sepsis and significantly greater mortality in old mice than in their younger counterparts. We found that the ATP levels in the plasma of mice increased with age and that, along with the extracellular accumulation of ATP, the PMNs of old mice became increasingly primed. Stimulation of the formyl peptide receptors of those primed PMNs triggered inflammatory responses that were significantly more pronounced in old mice than in young animals. However, bacterial phagocytosis and killing by PMNs of old mice were significantly lower than that of young mice. These age-dependent PMN dysfunctions correlated with a decrease in the enzymatic activity of plasma ATPases that convert extracellular ATP to adenosine. ATPases depend on divalent metal ions, including Ca2+, Mg2+, and Zn2+, and we found that depletion of these ions blocked the hydrolysis of ATP and the formation of adenosine in human blood, resulting in ATP accumulation and dysregulation of PMN functions equivalent to those observed in response to aging. CONCLUSIONS: Our findings suggest that impaired hydrolysis of plasma ATP dysregulates PMN function in older individuals. We conclude that strategies aimed at restoring plasma ATPase activity may offer novel therapeutic opportunities to reduce immune dysfunction, inflammation, and infectious complications in older patients.

5.
J Infect Dis ; 2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37804120

RESUMEN

BACKGROUND: ATP enhances neutrophil responses, but little is known about the role of ATP in influenza infections. METHODS: We used a mouse influenza model to study if ATP release is associated with neutrophil activation and disease progression. RESULTS: Influenza infection increased pulmonary ATP levels 5-fold and plasma ATP levels 3-fold over the levels in healthy mice. Adding ATP at those concentrations to blood from healthy mice primed their neutrophils and enhanced CD11b and CD63 expression, CD62L shedding, and reactive oxygen species production in response to formyl peptide receptor (FPR) stimulation. Influenza infection also primed neutrophils in vivo, resulting in FPR-induced CD11b expression and CD62L shedding up to 3-times higher than that of uninfected mice. In infected mice, large numbers of neutrophils entered the lungs. These cells were significantly more activated than peripheral neutrophils of infected and pulmonary neutrophils of healthy mice. Plasma ATP levels of infected mice and influenza disease progression corresponded with the numbers and activation level of their pulmonary neutrophils. CONCLUSION: Our findings suggest that ATP release from the lungs of infected mice promotes influenza disease progression by priming peripheral neutrophils that become strongly activated and cause pulmonary tissue damage after their recruitment to the lungs.

6.
Thorax ; 78(2): 151-159, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35613855

RESUMEN

RATIONALE: The increased mortality and morbidity seen in critically injured patients appears associated with systemic inflammatory response syndrome (SIRS) and immune dysfunction, which ultimately predisposes to infection. Mitochondria released by injury could generate danger molecules, for example, ATP, which in turn would be rapidly scavenged by ectonucleotidases, expressed on regulatory immune cells. OBJECTIVE: To determine the association between circulating mitochondria, purinergic signalling and immune dysfunction after trauma. METHODS: We tested the impact of hepatocyte-derived free mitochondria on blood-derived and lung-derived CD8 T cells in vitro and in experimental mouse models in vivo. In parallel, immune phenotypic analyses were conducted on blood-derived CD8 T cells obtained from trauma patients. RESULTS: Isolated intact mitochondria are functional and generate ATP ex vivo. Extracellular mitochondria perturb CD8+ T cells in co-culture, inducing select features of immune exhaustion in vitro. These effects are modulated by scavenging ATP, modelled by addition of apyrase in vitro. Injection of intact mitochondria into recipient mice markedly upregulates the ectonucleotidase CD39, and other immune checkpoint markers in circulating CD8+ T cells. We note that mice injected with mitochondria, prior to instilling bacteria into the lung, exhibit more severe lung injury, characterised by elevated neutrophil influx and by changes in CD8+ T cell cytotoxic capacity. Importantly, the development of SIRS in injured humans, is likewise associated with disordered purinergic signalling and CD8 T cell dysfunction. CONCLUSION: These studies in experimental models and in a cohort of trauma patients reveal important associations between extracellular mitochondria, aberrant purinergic signalling and immune dysfunction. These pathogenic factors with immune exhaustion are linked to SIRS and could be targeted therapeutically.


Asunto(s)
Antígenos CD , Linfocitos T CD8-positivos , Animales , Humanos , Ratones , Adenosina Trifosfato/metabolismo , Biomarcadores/metabolismo , Linfocitos T CD8-positivos/metabolismo , Mitocondrias , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo
7.
Purinergic Signal ; 19(4): 651-662, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36596963

RESUMEN

Neutrophils (PMNs) require extracellular ATP and adenosine (ADO) to fight bacterial infections, which often have life-threatening consequences in pediatric patients. We wondered whether the ATP and ADO levels in the plasma of children change with age and if these changes influence the antimicrobial efficacy of the PMNs of these children. We measured plasma concentrations of ATP and ADO and the activities of the enzymes responsible for the breakdown of these mediators in plasma samples from healthy children and adolescents (n = 45) ranging in age from 0.2 to 15 years. In addition, using blood samples of these individuals, we compared how effective their PMNs were in the phagocytosis of bacteria. In an experimental sepsis model with young (10 days) and adolescent mice (10 weeks), we studied how age influenced the resilience of these animals to bacterial infections and whether addition of ATP could improve the antimicrobial capacity of their PMNs. We found that plasma ATP levels correlated with age and were significantly lower in infants (< 1 year) than in adolescents (12-15 years). In addition, we observed significantly higher plasma ATPase and adenosine deaminase activities in children (< 12 years) when compared to the adolescent population. The activities of these ATP and ADO breakdown processes correlated inversely with age and with the ability of PMNs to phagocytize bacteria. Similar to their human counterparts, young mice also had significantly lower plasma ATP levels when compared to adolescent animals. In addition, we found that mortality of young mice after bacterial infection was significantly higher than that of adolescent mice. Moreover, bacterial phagocytosis by PMNs of young mice was weaker when compared to that of older mice. Finally, we found that ATP supplementation could recover bacterial phagocytosis of young mice to levels similar to those of adolescent mice. Our findings suggest that rapid ATP hydrolysis in the plasma of young children lowers the antimicrobial functions of their PMNs and that this may contribute to the vulnerability of pediatric patients to bacterial infections.


Asunto(s)
Antiinfecciosos , Infecciones Bacterianas , Adolescente , Humanos , Ratones , Niño , Animales , Preescolar , Lactante , Neutrófilos/metabolismo , Adenosina/metabolismo , Adenosina Trifosfato/metabolismo , Infecciones Bacterianas/metabolismo , Antiinfecciosos/metabolismo , Fagocitosis
8.
Purinergic Signal ; 18(2): 223-239, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35132577

RESUMEN

ATP released into the bloodstream regulates immune responses and other physiological functions. Excessive accumulation of extracellular ATP interferes with these functions, and elevated plasma ATP levels could indicate infections and other pathological disorders. However, there is considerable disagreement about what constitutes normal plasma ATP levels. Therefore, we optimized a method to accurately assess ATP concentrations in blood. We found that rapid chilling of heparinized blood samples is essential to preserve in vivo ATP levels and that differential centrifugation minimizes inadvertent ATP release due to cell damage and mechanical stress. Plasma samples were stabilized with perchloric acid, etheno-derivatized, and delipidated for sensitive analysis of ATP and related compounds using high-performance liquid chromatography (HPLC) and fluorescence detection. We measured 33 ± 20 nM ATP, 90 ± 45 nM ADP, 100 ± 55 nM AMP, and 81 ± 51 nM adenosine in the blood of healthy human adults (n = 10). In critically ill patients, ATP levels were 6 times higher than in healthy subjects. The anticoagulant greatly affected results. ATP levels were nearly 8 times higher in EDTA plasma than in heparin plasma, while AMP levels were 3 times lower and adenosine was entirely absent in EDTA plasma. If EDTA blood was not immediately chilled, ATP, ADP, and AMP levels continued to rise, which indicates that EDTA interferes with the endogenous mechanisms that regulate plasma adenylate levels. Our optimized method eliminates artifacts that prevent accurate determination of plasma adenylates and will be useful for studying mechanisms that regulate adenylate levels and for monitoring of pathological processes in patients with infections and other diseases.


Asunto(s)
Adenosina Trifosfato , Adenosina , Adenosina Difosfato , Adenosina Monofosfato , Adenosina Trifosfato/análisis , Cromatografía Líquida de Alta Presión , Ácido Edético , Humanos
9.
Am J Otolaryngol ; 43(2): 103323, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34933164

RESUMEN

PURPOSE: Dysplasia and cancer of the upper aerodigestive tract are characterized by significant neoangiogenesis. This can be recognized by optical methods like the Storz Professional Image Enhancement System (SPIES). Up to now, there are no reports of using this novel technique for examining nasal diseases. The objective of this study was to evaluate the use of SPIES during sinus surgery to help differentiate various nasal pathologies and determine their extension. METHODS: Patients (n = 27) with different pathologies in the region of the paranasal sinuses were operated via functional endoscopic surgery using a 2D-HD-camera with white light and SPIES. In addition, 10 healthy individuals were examined. The system was evaluated using two different questionnaires. RESULTS: The handling and operation of SPIES was intuitive and easy. Use of SPIES did not prolong the procedure. There was no disturbing image distortion. SPIES seemed to improve the visualization, differentiation and evaluation of vascularization of paranasal pathologies and allowed for precise and accurate surgery. Compared to examination with the 2D-HD-camera and white light alone, SPIES appeared to facilitate the identification of mucosal pathologies. CONCLUSION: SPIES could be a promising adjunct tool to evaluate nasal pathologies intraoperatively. Especially in the case of vascularized tumors the enhanced image endoscopy seemed to be clearly superior to standard white light alone. In our study, the system facilitated the assessment of tumor extension and vascularization as well as the differentiation of healthy mucosa. Future randomized studies will be necessary to prove the potential of integrating this novel technique into the clinical routine for the differentiation of nasal pathologies and the improvement of resection margins during nasal tumor surgery.


Asunto(s)
Neoplasias Nasales , Enfermedades de los Senos Paranasales , Senos Paranasales , Endoscopía/métodos , Humanos , Aumento de la Imagen , Neoplasias Nasales/cirugía , Enfermedades de los Senos Paranasales/diagnóstico por imagen , Enfermedades de los Senos Paranasales/cirugía , Senos Paranasales/diagnóstico por imagen , Senos Paranasales/cirugía
10.
Am J Physiol Cell Physiol ; 320(1): C15-C29, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33052071

RESUMEN

Extracellular diphosphate and triphosphate nucleotides are released from activated or injured cells to trigger vascular and immune P2 purinergic receptors, provoking inflammation and vascular thrombosis. These metabokines are scavenged by ectonucleoside triphosphate diphosphohydrolase-1 (E-NTPDase1 or CD39). Further degradation of the monophosphate nucleoside end products occurs by surface ecto-5'-nucleotidase (NMPase) or CD73. These ectoenzymatic processes work in tandem to promote adenosinergic responses, which are immunosuppressive and antithrombotic. These homeostatic ectoenzymatic mechanisms are lost in the setting of oxidative stress, which exacerbates inflammatory processes. We have engineered bifunctional enzymes made up from ectodomains (ECDs) of CD39 and CD73 within a single polypeptide. Human alkaline phosphatase-ectodomain (ALP-ECD) and human acid phosphatase-ectodomain (HAP-ECD) fusion proteins were also generated, characterized, and compared with these CD39-ECD, CD73-ECD, and bifunctional fusion proteins. Through the application of colorimetrical functional assays and high-performance liquid chromatography kinetic assays, we demonstrate that the bifunctional ectoenzymes express high levels of CD39-like NTPDase activity and CD73-like NMPase activity. Chimeric CD39-CD73-ECD proteins were superior in converting triphosphate and diphosphate nucleotides into nucleosides when compared with ALP-ECD and HAP-ECD. We also note a pH sensitivity difference between the bifunctional fusion proteins and parental fusions, as well as ectoenzymatic property distinctions. Intriguingly, these innovative reagents decreased platelet activation to exogenous agonists in vitro. We propose that these chimeric fusion proteins could serve as therapeutic agents in inflammatory diseases, acting to scavenge proinflammatory ATP and also generate anti-inflammatory adenosine.


Asunto(s)
5'-Nucleotidasa/farmacología , Antiinflamatorios/farmacología , Apirasa/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Ingeniería de Proteínas , 5'-Nucleotidasa/química , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/metabolismo , Nucleótidos de Adenina/metabolismo , Antiinflamatorios/química , Antiinflamatorios/metabolismo , Apirasa/química , Apirasa/genética , Apirasa/metabolismo , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Proteínas Ligadas a GPI/farmacología , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Inhibidores de Agregación Plaquetaria/química , Inhibidores de Agregación Plaquetaria/metabolismo , Conformación Proteica , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Especificidad por Sustrato
11.
Eur J Pediatr ; 180(7): 2245-2251, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33709157

RESUMEN

Reported success rates of endoscopic choanal atresia (CA) surgery vary substantially due to a high heterogeneity in and between study groups. Comprehensive data on the unique patient cohort of newborns with bilateral CA are scarce. Our study aimed to close this gap by using narrow inclusion criteria and standardized surgical outcome parameters. A total of ten neonates who were diagnosed with bilateral complete CA and underwent endoscopic surgery at the Department of Otolaryngology, Head and Neck Surgery in the University Hospital of Munich between 2008 and 2017 were included. Preoperative findings, surgical procedures, outcome, and follow-up were analyzed. Standardized criteria were used to assess surgical outcome. Almost all patients (90%) required at least one revision procedure within the first 6 months after initial surgery because of symptomatic partial or complete restenosis. After that, all surviving patients remained asymptomatic until the end of the follow-up period.Conclusion: Endoscopic bilateral CA repair in neonates is a safe procedure with a high long-term success rate. However, compared to other patient groups with choanal obstruction, restenosis occurs frequently, and revision procedures are required in a large number of cases. This should be considered during preoperative planning and parent counseling. What is Known: • Bilateral complete choanal atresia (CA) is a neonatal emergency that requires surgical intervention. • Reported success rates of endoscopic choanal obstruction repair are highly variable and mostly derived from heterogenous study groups that do not reflect the situation in neonates adequately. What is New: • This study focuses exclusively on newborns with complete bilateral CA who underwent endoscopic surgery within the first 28 days of life and uses standardized criteria to assess outcome. • The long-term success rate of endoscopic bilateral CA repair in neonates is high; however, almost all patients require at least one revision procedure within the first 6 months.


Asunto(s)
Atresia de las Coanas , Atresia de las Coanas/cirugía , Endoscopía , Humanos , Recién Nacido , Nariz , Resultado del Tratamiento
12.
J Biol Chem ; 294(16): 6283-6293, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30787105

RESUMEN

T cell suppression contributes to immune dysfunction in sepsis. However, the underlying mechanisms are not well-defined. Here, we show that exposure of human peripheral blood mononuclear cells to bacterial lipopolysaccharide (LPS) can rapidly and dose-dependently suppress interleukin-2 (IL-2) production and T cell proliferation. We also report that these effects depend on monocytes. LPS did not prevent the interaction of monocytes with T cells, nor did it induce programmed cell death protein 1 (PD-1) signaling that causes T cell suppression. Instead, we found that LPS stimulation of monocytes led to the accumulation of extracellular ATP that impaired mitochondrial function, cell migration, IL-2 production, and T cell proliferation. Mechanistically, LPS-induced ATP accumulation exerted these suppressive effects on T cells by activating the purinergic receptor P2Y11 on the cell surface of T cells. T cell functions could be partially restored by enzymatic removal of extracellular ATP or pharmacological blocking of P2Y11 receptors. Plasma samples obtained from sepsis patients had similar suppressive effects on T cells from healthy subjects. Our findings suggest that LPS and ATP accumulation in the circulation of sepsis patients suppresses T cells by promoting inappropriate P2Y11 receptor stimulation that impairs T cell metabolism and functions. We conclude that inhibition of LPS-induced ATP release, removal of excessive extracellular ATP, or P2Y11 receptor antagonists may be potential therapeutic strategies to prevent T cell suppression and restore host immune function in sepsis.


Asunto(s)
Adenosina Trifosfato/metabolismo , Lipopolisacáridos/toxicidad , Mitocondrias/metabolismo , Receptores Purinérgicos P2/metabolismo , Sepsis/metabolismo , Linfocitos T/metabolismo , Adenosina Trifosfato/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Interleucina-2/inmunología , Interleucina-2/metabolismo , Células Jurkat , Masculino , Persona de Mediana Edad , Mitocondrias/inmunología , Mitocondrias/patología , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Antagonistas del Receptor Purinérgico P2/farmacología , Receptores Purinérgicos P2/inmunología , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Sepsis/patología , Linfocitos T/inmunología , Linfocitos T/patología
13.
Purinergic Signal ; 15(2): 127-137, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30919205

RESUMEN

Previous studies have shown that T cell receptor (TCR) and CD28 coreceptor stimulation involves rapid ATP release, autocrine purinergic feedback via P2X receptors, and mitochondrial ATP synthesis that promote T cell activation. Here, we show that ADP formation and autocrine stimulation of P2Y1 receptors are also involved in these purinergic signaling mechanisms. Primary human CD4 T cells and the human Jurkat CD4 T cell line express P2Y1 receptors. The expression of this receptor increases following T cell stimulation. Inhibition of P2Y1 receptors impairs the activation of mitochondria, as assessed by mitochondrial Ca2+ uptake, and reduces cytosolic Ca2+ signaling in response to TCR/CD28 stimulation. We found that the addition of exogenous ADP or overexpression of P2Y1 receptors significantly increased IL-2 mRNA transcription in response to TCR/CD28 stimulation. Conversely, antagonists or silencing of P2Y1 receptors reduced IL-2 mRNA transcription and attenuated T cell functions. We conclude that P2Y1 and P2X receptors have non-redundant, synergistic functions in the regulation of T cell activation. P2Y1 receptors may represent potential therapeutic targets to modulate T cell function in inflammation and host defense.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Activación de Linfocitos/fisiología , Receptores Purinérgicos P2X/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Transducción de Señal/inmunología , Comunicación Autocrina , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Humanos
14.
Crit Care Med ; 46(12): e1183-e1189, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30247270

RESUMEN

OBJECTIVES: Monocytes and macrophages produce interleukin-1ß by inflammasome activation which involves adenosine triphosphate release, pannexin-1 channels, and P2X7 receptors. However, interleukin-1ß can also be produced in an inflammasome-independent fashion. Here we studied if this mechanism also involves adenosine triphosphate signaling and how it contributes to inflammasome activation. DESIGN: In vitro studies with human cells and randomized animal experiments. SETTING: Preclinical academic research laboratory. SUBJECTS: Wild-type C57BL/6 and pannexin-1 knockout mice, healthy human subjects for cell isolation. INTERVENTIONS: Human monocytes and U937 macrophages were treated with different inhibitors to study how purinergic signaling contributes to toll-like receptor-induced cell activation and interleukin-1ß production. Wild-type and pannexin-1 knockout mice were subjected to cecal ligation and puncture to study the role of purinergic signaling in interleukin-1ß production and host immune defense. MEASUREMENTS AND MAIN RESULTS: Toll-like receptor agonists triggered mitochondrial adenosine triphosphate production and adenosine triphosphate release within seconds. Inhibition of mitochondria, adenosine triphosphate release, or P2 receptors blocked p38 mitogen-activated protein kinase and caspase-1 activation and interleukin-1ß secretion. Mice lacking pannexin-1 failed to activate monocytes, to produce interleukin-1ß, and to effectively clear bacteria following cecal ligation and puncture. CONCLUSIONS: Purinergic signaling has two separate roles in monocyte/macrophage activation, namely to facilitate the initial detection of danger signals via toll-like receptors and subsequently to regulate nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain containing 3 inflammasome activation. Further dissection of these mechanisms may reveal novel therapeutic targets for immunomodulation in critical care patients.


Asunto(s)
Adenosina Trifosfato/inmunología , Infecciones/inmunología , Inflamasomas/inmunología , Activación de Macrófagos/inmunología , Monocitos/inmunología , Animales , Técnicas de Cultivo de Célula , Conexinas/farmacología , Modelos Animales de Enfermedad , Compuestos Heterocíclicos con 3 Anillos , Humanos , Immunoblotting , Interleucina-1beta/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/farmacología , Transducción de Señal , Receptores Toll-Like/agonistas , Receptores Toll-Like/antagonistas & inhibidores
15.
Crit Care Med ; 45(1): e97-e104, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27548819

RESUMEN

OBJECTIVE: Sepsis remains an unresolved clinical problem. Therapeutic strategies focusing on inhibition of neutrophils (polymorphonuclear neutrophils) have failed, which indicates that a more detailed understanding of the underlying pathophysiology of sepsis is required. Polymorphonuclear neutrophil activation and chemotaxis require cellular adenosine triphosphate release via pannexin-1 channels that fuel autocrine feedback via purinergic receptors. In the current study, we examined the roles of endogenous and systemic adenosine triphosphate on polymorphonuclear neutrophil activation and host defense in sepsis. DESIGN: Prospective randomized animal investigation and in vitro studies. SETTING: Preclinical academic research laboratory. SUBJECTS: Wild-type C57BL/6 mice, pannexin-1 knockout mice, and healthy human subjects used to obtain polymorphonuclear neutrophils for in vitro studies. INTERVENTIONS: Wild-type and pannexin-1 knockout mice were treated with suramin or apyrase to block the endogenous or systemic effects of adenosine triphosphate. Mice were subjected to cecal ligation and puncture and polymorphonuclear neutrophil activation (CD11b integrin expression), organ (liver) injury (plasma aspartate aminotransferase), bacterial spread, and survival were monitored. Human polymorphonuclear neutrophils were used to study the effect of systemic adenosine triphosphate and apyrase on chemotaxis. MEASUREMENTS AND MAIN RESULTS: Inhibiting endogenous adenosine triphosphate reduced polymorphonuclear neutrophil activation and organ injury, but increased the spread of bacteria and mortality in sepsis. By contrast, removal of systemic adenosine triphosphate improved bacterial clearance and survival in sepsis by improving polymorphonuclear neutrophil chemotaxis. CONCLUSIONS: Systemic adenosine triphosphate impairs polymorphonuclear neutrophil functions by disrupting the endogenous purinergic signaling mechanisms that regulate cell activation and chemotaxis. Removal of systemic adenosine triphosphate improves polymorphonuclear neutrophil function and host defenses, making this a promising new treatment strategy for sepsis.


Asunto(s)
Adenosina Trifosfato/fisiología , Quimiotaxis de Leucocito/inmunología , Neutrófilos/fisiología , Sepsis/inmunología , Animales , Apirasa/farmacología , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Activación Neutrófila , Sepsis/mortalidad , Suramina/farmacología
16.
PLoS Pathog ; 11(10): e1005177, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26485519

RESUMEN

Exhausted T cells express multiple co-inhibitory molecules that impair their function and limit immunity to chronic viral infection. Defining novel markers of exhaustion is important both for identifying and potentially reversing T cell exhaustion. Herein, we show that the ectonucleotidse CD39 is a marker of exhausted CD8+ T cells. CD8+ T cells specific for HCV or HIV express high levels of CD39, but those specific for EBV and CMV do not. CD39 expressed by CD8+ T cells in chronic infection is enzymatically active, co-expressed with PD-1, marks cells with a transcriptional signature of T cell exhaustion and correlates with viral load in HIV and HCV. In the mouse model of chronic Lymphocytic Choriomeningitis Virus infection, virus-specific CD8+ T cells contain a population of CD39high CD8+ T cells that is absent in functional memory cells elicited by acute infection. This CD39high CD8+ T cell population is enriched for cells with the phenotypic and functional profile of terminal exhaustion. These findings provide a new marker of T cell exhaustion, and implicate the purinergic pathway in the regulation of T cell exhaustion.


Asunto(s)
Antígenos CD/inmunología , Apirasa/inmunología , Biomarcadores , Linfocitos T CD8-positivos/inmunología , Infecciones por Virus ARN/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Infecciones por Arenaviridae/inmunología , Cromatografía Líquida de Alta Presión , Enfermedad Crónica , Modelos Animales de Enfermedad , Citometría de Flujo , Infecciones por VIH/inmunología , Hepatitis C Crónica/inmunología , Humanos , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos
17.
J Infect Dis ; 213(3): 456-64, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26150546

RESUMEN

T cell suppression in sepsis is a well-known phenomenon; however, the underlying mechanisms are not fully understood. Previous studies have shown that T cell stimulation up-regulates mitochondrial adenosine triphosphate (ATP) production to fuel purinergic signaling mechanisms necessary for adequate T cell responses. Here we show that basal mitochondrial ATP production, ATP release, and stimulation of P2X1 receptors represent a standby purinergic signaling mechanism that is necessary for antigen recognition. Inhibition of this process impairs T cell vigilance and the ability of T cells to trigger T cell activation, up-regulate mitochondrial ATP production, and stimulate P2X4 and P2X7 receptors that elicit interleukin 2 production and T cell proliferation. T cells of patients with sepsis lack this standby purinergic signaling system owing to defects in mitochondrial function, ATP release, and calcium signaling. These defects impair antigen recognition and T cell function and are correlated with sepsis severity. Pharmacological targeting of these defects may improve T cell function and reduce the risk of sepsis.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Señalización del Calcio/fisiología , Mitocondrias/fisiología , Purinas/metabolismo , Receptores Purinérgicos/fisiología , Sepsis/inmunología , Adolescente , Adulto , Humanos , Células Jurkat , Suramina , Adulto Joven
18.
Purinergic Signal ; 12(3): 439-51, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27020575

RESUMEN

T cells respond to antigen stimulation with the rapid release of cellular ATP, which stimulates an autocrine feedback mechanism that regulates calcium influx through P2X receptors. This autocrine purinergic feedback mechanism plays an essential role in the activation of T cells resulting in cell proliferation and clonal expansion. We recently reported that increases in mitochondrial ATP production drive this stimulation-induced purinergic signaling mechanism but that low-level mitochondrial ATP production fuels basal T cell functions required to maintain vigilance of unstimulated T cells. Here we studied whether defects in these purinergic signaling mechanisms are involved in the unwanted proliferation of leukemia T cells. We found that acute leukemia T cells (Jurkat) possess a larger number and more active mitochondria than their healthy counterparts. Jurkat cells have higher intracellular ATP concentrations and generat more extracellular ATP than unstimulated T cells from healthy donors. As a result, increased purinergic signaling through P2X1 and P2X7 receptors elevates baseline levels of cytosolic Ca(2+) in Jurkat cells. We found that pharmacological inhibition of this basal purinergic signaling mechanism decreases mitochondrial activity, Ca(2+) signaling, and cell proliferation. Similar results were seen in the leukemic cell lines THP-1, U-937, and HL-60. Combined treatment with inhibitors of P2X1 or P2X7 receptors and the chemotherapeutic agent 6-mercaptopurine completely blocked Jurkat cell proliferation. Our results demonstrate that increased mitochondrial metabolism promotes autocrine purinergic signaling and uncontrolled proliferation of leukemia cells. These findings suggest that deranged purinergic signaling can result in T cell malignancy and that therapeutic targeting aimed at purinergic signaling is a potential strategy to combat T cell leukemia.


Asunto(s)
Adenosina Trifosfato/metabolismo , Señalización del Calcio/fisiología , Proliferación Celular/fisiología , Leucemia de Células T/metabolismo , Receptores Purinérgicos P2X/metabolismo , Línea Celular Tumoral , Citometría de Flujo , Humanos , Células Jurkat , Leucemia de Células T/patología , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
Purinergic Signal ; 12(4): 673-685, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27577957

RESUMEN

In neutrophils, adenosine triphosphate (ATP) release and autocrine purinergic signaling regulate coordinated cell motility during chemotaxis. Here, we studied whether similar mechanisms regulate the motility of breast cancer cells. While neutrophils and benign human mammary epithelial cells (HMEC) form a single leading edge, MDA-MB-231 breast cancer cells possess multiple leading edges enriched with A3 adenosine receptors. Compared to HMEC, MDA-MB-231 cells overexpress the ectonucleotidases ENPP1 and CD73, which convert extracellular ATP released by the cells to adenosine that stimulates A3 receptors and promotes cell migration with frequent directional changes. However, exogenous adenosine added to breast cancer cells or the A3 receptor agonist IB-MECA dose-dependently arrested cell motility by simultaneous stimulation of multiple leading edges, doubling cell surface areas and significantly reducing migration velocity by up to 75 %. We conclude that MDA-MB-231 cells, HMEC, and neutrophils differ in the purinergic signaling mechanisms that regulate their motility patterns and that the subcellular distribution of A3 adenosine receptors in MDA-MB-231 breast cancer cells contributes to dysfunctional cell motility. These findings imply that purinergic signaling mechanisms may be potential therapeutic targets to interfere with the motility of breast cancer cells in order to reduce the spread of cancer cells and the risk of metastasis.


Asunto(s)
Adenosina/análogos & derivados , Adenosina/farmacología , Neoplasias de la Mama/patología , Movimiento Celular/efectos de los fármacos , Agonistas del Receptor Purinérgico P1/farmacología , Receptor de Adenosina A3/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Transducción de Señal/efectos de los fármacos
20.
J Biol Chem ; 289(39): 26794-26803, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25104353

RESUMEN

Polymorphonuclear neutrophils (PMNs) form the first line of defense against invading microorganisms. We have shown previously that ATP release and autocrine purinergic signaling via P2Y2 receptors are essential for PMN activation. Here we show that mitochondria provide the ATP that initiates PMN activation. Stimulation of formyl peptide receptors increases the mitochondrial membrane potential (Δψm) and triggers a rapid burst of ATP release from PMNs. This burst of ATP release can be blocked by inhibitors of mitochondrial ATP production and requires an initial formyl peptide receptor-induced Ca(2+) signal that triggers mitochondrial activation. The burst of ATP release generated by the mitochondria fuels a first phase of purinergic signaling that boosts Ca(2+) signaling, amplifies mitochondrial ATP production, and initiates functional PMN responses. Cells then switch to glycolytic ATP production, which fuels a second round of purinergic signaling that sustains Ca(2+) signaling via P2X receptor-mediated Ca(2+) influx and maintains functional PMN responses such as oxidative burst, degranulation, and phagocytosis.


Asunto(s)
Adenosina Trifosfato/metabolismo , Comunicación Autocrina/fisiología , Señalización del Calcio/fisiología , Mitocondrias/metabolismo , Activación Neutrófila/fisiología , Neutrófilos/metabolismo , Receptores Purinérgicos P2X/metabolismo , Adenosina Trifosfato/inmunología , Degranulación de la Célula/fisiología , Femenino , Humanos , Masculino , Mitocondrias/inmunología , Neutrófilos/citología , Neutrófilos/inmunología , Fagocitosis/fisiología , Receptores Purinérgicos P2X/inmunología , Estallido Respiratorio/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA