Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ann Surg ; 278(6): e1277-e1288, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37154066

RESUMEN

OBJECTIVE: Injured tissue predisposes the subject to local and systemic infection. We studied injury-induced immune dysfunction seeking novel means to reverse such predisposition. BACKGROUND: Injury mobilizes primitive "DANGER signals" [danger-associated molecular patterns (DAMPs)] activating innate immunocyte (neutrophils, PMN) signaling and function. Mitochondrial formyl peptides activate G -protein coupled receptors (GPCR) like formyl peptide receptor-1. Mitochondrial DNA and heme activate toll-like receptors (TLR9 and TLR2/4). GPCR kinases (GRKs) can regulate GPCR activation. METHODS: We studied human and mouse PMN signaling elicited by mitochondrial DAMPs (GPCR surface expression; protein phosphorylation, or acetylation; Ca 2+ flux) and antimicrobial functions [cytoskeletal reorganization, chemotaxis (CTX), phagocytosis, bacterial killing] in cellular systems and clinical injury samples. Predicted rescue therapies were assessed in cell systems and mouse injury-dependent pneumonia models. RESULTS: Mitochondrial formyl peptides activate GRK2, internalizing GPCRs and suppressing CTX. Mitochondrial DNA suppresses CTX, phagocytosis, and killing through TLR9 through a novel noncanonical mechanism that lacks GPCR endocytosis. Heme also activates GRK2. GRK2 inhibitors like paroxetine restore functions. GRK2 activation through TLR9 prevented actin reorganization, implicating histone deacetylases (HDACs). Actin polymerization, CTX, bacterial phagocytosis, and killing were also rescued, therefore, by the HDAC inhibitor valproate. Trauma repository PMN showed GRK2 activation and cortactin deacetylation, which varied with severity and was most marked in patients developing infections. Either GRK2 or HDAC inhibition prevented loss of mouse lung bacterial clearance, but only the combination rescued clearance when given postinjury. CONCLUSIONS: Tissue injury-derived DAMPs suppress antimicrobial immunity through canonical GRK2 activation and a novel TLR-activated GRK2-pathway impairing cytoskeletal organization. Simultaneous GRK2/HDAC inhibition rescues susceptibility to infection after tissue injury.


Asunto(s)
Antiinfecciosos , Neutrófilos , Humanos , Ratones , Animales , Neutrófilos/metabolismo , Actinas/metabolismo , Receptor Toll-Like 9/metabolismo , ADN Mitocondrial/metabolismo , Péptidos/metabolismo , Hemo/metabolismo
2.
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
3.
Arterioscler Thromb Vasc Biol ; 41(6): 1915-1927, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33853347
4.
J Immunol ; 202(10): 2982-2990, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30952817

RESUMEN

Biliverdin reductase (BVR)-A is a pleotropic enzyme converting biliverdin to bilirubin and a signaling molecule that has cytoprotective and immunomodulatory effects. We recently showed that biliverdin inhibits the expression of complement activation fragment 5a receptor one (C5aR1) in RAW 264.7 macrophages. In this study, we investigated the role of BVR-A in determining macrophage inflammatory phenotype and function via regulation of C5aR1. We assessed expression of C5aR1, M1-like macrophage markers, including chemokines (RANTES, IP-10), as well as chemotaxis in response to LPS and C5a in bone marrow-derived macrophages from BVR fl/fl and LysM-Cre:BVR fl / fl mice (conditional deletion of BVR-A in myeloid cells). In response to LPS, macrophages isolated from LysM-Cre:BVR fl/fl showed significantly elevated levels of C5aR1 as well as chemokines (RANTES, IP10) but not proinflammatory markers, such as iNOS and TNF. An increase in C5aR1 expression was also observed in peritoneal macrophages and several tissues from LysM-Cre:BVR fl/fl mice in a model of endotoxemia. In addition, knockdown of BVR-A resulted in enhanced macrophage chemotaxis toward C5a. Part of the effects of BVR-A deletion on chemotaxis and RANTES expression were blocked in the presence of a C5aR1 neutralizing Ab, confirming the role of C5a-C5aR1 signaling in mediating the effects of BVR. In summary, BVR-A plays an important role in regulating macrophage chemotaxis in response to C5a via modulation of C5aR1 expression. In addition, macrophages lacking BVR-A are characterized by the expression of M1 polarization-associated chemokines, the levels of which depend in part on C5aR1 signaling.


Asunto(s)
Quimiocinas/inmunología , Quimiotaxis/inmunología , Complemento C5a/inmunología , Macrófagos/inmunología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/inmunología , Receptor de Anafilatoxina C5a/inmunología , Transducción de Señal/inmunología , Animales , Quimiocinas/genética , Quimiotaxis/genética , Complemento C5a/genética , Eliminación de Gen , Macrófagos/citología , Ratones , Ratones Transgénicos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Receptor de Anafilatoxina C5a/genética , Transducción de Señal/genética
5.
Proc Natl Acad Sci U S A ; 115(10): E2302-E2310, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29463714

RESUMEN

Ischemia reperfusion injury (IRI) is the predominant tissue insult associated with organ transplantation. Treatment with carbon monoxide (CO) modulates the innate immune response associated with IRI and accelerates tissue recovery. The mechanism has been primarily descriptive and ascribed to the ability of CO to influence inflammation, cell death, and repair. In a model of bilateral kidney IRI in mice, we elucidate an intricate relationship between CO and purinergic signaling involving increased CD39 ectonucleotidase expression, decreased expression of Adora1, with concomitant increased expression of Adora2a/2b. This response is linked to a >20-fold increase in expression of the circadian rhythm protein Period 2 (Per2) and a fivefold increase in serum erythropoietin (EPO), both of which contribute to abrogation of kidney IRI. CO is ineffective against IRI in Cd39-/- and Per2-/- mice or in the presence of a neutralizing antibody to EPO. Collectively, these data elucidate a cellular signaling mechanism whereby CO modulates purinergic responses and circadian rhythm to protect against injury. Moreover, these effects involve CD39- and adenosinergic-dependent stabilization of Per2. As CO also increases serum EPO levels in human volunteers, these findings continue to support therapeutic use of CO to treat IRI in association with organ transplantation, stroke, and myocardial infarction.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Monóxido de Carbono/administración & dosificación , Enfermedades Renales/tratamiento farmacológico , Riñón/efectos de los fármacos , Proteínas Circadianas Period/metabolismo , Daño por Reperfusión/prevención & control , Animales , Antígenos CD/genética , Apirasa/genética , Modelos Animales de Enfermedad , Humanos , Riñón/irrigación sanguínea , Riñón/metabolismo , Riñón/fisiopatología , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Circadianas Period/genética , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo
6.
Med Res Rev ; 40(4): 1147-1177, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31820474

RESUMEN

Treating acute kidney injury (AKI) represents an important unmet medical need both in terms of the seriousness of this medical problem and the number of patients. There is also a large untapped market opportunity in treating AKI. Over the years, there has been much effort in search of therapeutics with minimal success. However, over the same time period, new understanding of the underlying pathobiology and molecular mechanisms of kidney injury have undoubtedly helped the search for new therapeutics. Along this line, carbon monoxide (CO) has emerged as a promising therapeutic agent because of its demonstrated cytoprotective, and immunomodulatory effects. CO has also been shown to sensitize cancer, but not normal cells, to chemotherapy. This is particularly important in treating cisplatin-induced AKI, a common clinical problem that develops in patients receiving cisplatin therapies for a number of different solid organ malignancies. This review will examine and make the case that CO be developed into a therapeutic agent against AKI.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Monóxido de Carbono/uso terapéutico , Lesión Renal Aguda/etiología , Animales , Monóxido de Carbono/administración & dosificación , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Humanos
7.
Ann Surg ; 272(4): 604-610, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32932316

RESUMEN

OBJECTIVES: Sepsis and sterile both release "danger signals' that induce the systemic inflammatory response syndrome (SIRS). So differentiating infection from SIRS can be challenging. Precision diagnostic assays could limit unnecessary antibiotic use, improving outcomes. METHODS: After surveying human leukocyte cytokine production responses to sterile damage-associated molecular patterns (DAMPs), bacterial pathogen-associated molecular patterns, and bacteria we created a multiplex assay for 31 cytokines. We then studied plasma from patients with bacteremia, septic shock, "severe sepsis," or trauma (ISS ≥15 with circulating DAMPs) as well as controls. Infections were adjudicated based on post-hospitalization review. Plasma was studied in infection and injury using univariate and multivariate means to determine how such multiplex assays could best distinguish infective from noninfective SIRS. RESULTS: Infected patients had high plasma interleukin (IL)-6, IL-1α, and triggering receptor expressed on myeloid cells-1 (TREM-1) compared to controls [false discovery rates (FDR) <0.01, <0.01, <0.0001]. Conversely, injury suppressed many mediators including MDC (FDR <0.0001), TREM-1 (FDR <0.001), IP-10 (FDR <0.01), MCP-3 (FDR <0.05), FLT3L (FDR <0.05), Tweak, (FDR <0.05), GRO-α (FDR <0.05), and ENA-78 (FDR <0.05). In univariate studies, analyte overlap between clinical groups prevented clinical relevance. Multivariate models discriminated injury and infection much better, with the 2-group random-forest model classifying 11/11 injury and 28/29 infection patients correctly in out-of-bag validation. CONCLUSIONS: Circulating cytokines in traumatic SIRS differ markedly from those in health or sepsis. Variability limits the accuracy of single-mediator assays but machine learning based on multiplexed plasma assays revealed distinct patterns in sepsis- and injury-related SIRS. Defining biomarker release patterns that distinguish specific SIRS populations might allow decreased antibiotic use in those clinical situations. Large prospective studies are needed to validate and operationalize this approach.


Asunto(s)
Citocinas/sangre , Sepsis/sangre , Sepsis/diagnóstico , Síndrome de Respuesta Inflamatoria Sistémica/sangre , Síndrome de Respuesta Inflamatoria Sistémica/diagnóstico , Informes Anuales como Asunto , Diagnóstico Diferencial , Cirugía General , Pruebas Hematológicas/métodos , Humanos , Estudios Prospectivos , Sepsis/inmunología , Sociedades Médicas , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Estados Unidos
8.
Crit Care Med ; 48(2): e123-e132, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31939811

RESUMEN

OBJECTIVES: Trauma predisposes to systemic sterile inflammation (systemic inflammatory response syndrome) as well as infection, but the mechanisms linking injury to infection are poorly understood. Mitochondrial debris contains formyl peptides. These bind formyl peptide receptor-1, trafficking neutrophils to wounds, initiating systemic inflammatory response syndrome, and wound healing. Bacterial formyl peptides, however, also attract neutrophils via formyl peptide receptor-1. Thus, mitochondrial formyl peptides might suppress neutrophils antimicrobial function. Also, formyl peptide receptor-1 blockade used to mitigate systemic inflammatory response syndrome might predispose to sepsis. We examined how mitochondrial formyl peptides impact neutrophils functions contributing to antimicrobial responses and how formyl peptide receptor-1 antagonists affect those functions. DESIGN: Prospective study of human and murine neutrophils and clinical cohort analysis. SETTING: University research laboratory and level 1 trauma center. PATIENTS: Trauma patients, volunteer controls. ANIMAL SUBJECTS: C57Bl/6, formyl peptide receptor-1, and formyl peptide receptor-2 knockout mice. INTERVENTIONS: Human and murine neutrophils functions were activated with autologous mitochondrial debris, mitochondrial formyl peptides, or bacterial formyl peptides followed by chemokines or leukotrienes. The experiments were repeated using formyl peptide receptor-1 antagonist cyclosporin H, "designer" human formyl peptide receptor-1 antagonists (POL7178 and POL7200), or anti-formyl peptide receptor-1 antibodies. Mouse injury/lung infection model was used to evaluate effect of formyl peptide receptor-1 inhibition. MEASUREMENTS AND MAIN RESULTS: Human neutrophils cytosolic calcium, chemotaxis, reactive oxygen species production, and phagocytosis were studied before and after exposure to mitochondrial debris, mitochondrial formyl peptides, and bacterial formyl peptides. Mitochondrial formyl peptide and bacterial formyl peptides had similar effects on neutrophils. Responses to chemokines and leukotrienes were suppressed by prior exposure to formyl peptides. POL7200 and POL7178 were specific antagonists of human formyl peptide receptor-1 and more effective than cyclosporin H or anti-formyl peptide receptor-1 antibodies. Formyl peptides inhibited mouse neutrophils responses to chemokines only if formyl peptide receptor-1 was present. Formyl peptide receptor-1 blockade did not inhibit neutrophils bacterial phagocytosis or reactive oxygen species production. Cyclosporin H increased bacterial clearance in lungs after injury. CONCLUSIONS: Formyl peptides both activate and desensitize neutrophils. Formyl peptide receptor-1 blockade prevents desensitization, potentially both diminishing systemic inflammatory response syndrome and protecting the host against secondary infection after tissue trauma or primary infection.


Asunto(s)
Proteínas Mitocondriales/inmunología , Activación Neutrófila/inmunología , Receptores de Formil Péptido/antagonistas & inhibidores , Animales , Ciclosporina/farmacología , Humanos , Lesión Pulmonar/fisiopatología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Infecciones del Sistema Respiratorio/fisiopatología
10.
J Immunol ; 199(4): 1453-1464, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28674181

RESUMEN

Robust lung inflammation is one of the prominent features in the pathogenesis of acute lung injury (ALI). Macrophage migration and recruitment are often seen at the early stage of lung inflammatory responses to noxious stimuli. Using an acid inhalation-induced lung injury model, we explored the mechanisms by which acid exposure initiates macrophage recruitment and migration during development of ALI. The lung epithelium comprises a large surface area and functions as a first-line defense against noxious insults. We found that acid exposure induced a remarkable microvesicle (MV) release from lung epithelium as detected in bronchoalveolar lavage fluid. Significantly elevated RNA, rather than protein, was found in these epithelium-derived MVs after acid and included several highly elevated microRNAs, including microRNA (miR)-17 and miR-221. Acid-induced epithelial MV release promoted macrophage migration in vitro and recruitment into the lung in vivo and required, in part, MV shuttling of miR-17 and/or miR-221. Mechanistically, acid-induced epithelial MV miR-17/221 promoted ß1 integrin recycling and presentation back onto the surface of macrophages, in part via a Rab11-mediated pathway. Integrin ß1 is known to play an essential role in regulating macrophage migration. Taken together, acid-induced ALI results in epithelial MV shuttling of miR-17/221 that in turn modulates macrophage ß1 integrin recycling, promoting macrophage recruitment and ultimately contributing to lung inflammation.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Micropartículas Derivadas de Células , Integrina beta1/metabolismo , Pulmón/citología , Macrófagos/fisiología , MicroARNs/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Movimiento Celular , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/fisiología , Ácido Clorhídrico/administración & dosificación , Inflamación/inmunología , Integrina beta1/inmunología , Pulmón/patología , Pulmón/fisiología , Macrófagos/inmunología , Ratones , MicroARNs/aislamiento & purificación
11.
J Autoimmun ; 94: 122-133, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30098863

RESUMEN

In Crohn's disease, pathogenic Th17-cells express low levels of CD39 ectonucleotidase and are refractory to the immunosuppressive effects of unconjugated bilirubin (UCB), an endogenous ligand for aryl-hydrocarbon-receptor (AhR). This resistance to AhR ligation might be associated with alterations in responses to hypoxia. Limited exposure to hypoxia appears beneficial in acute tissue injury. However, in protracted inflammation, hypoxemia may paradoxically result in Th17-cell activation. We report here that in vitro exposure of Th17-cells from Crohn's disease patients to hypoxia limits responsiveness to AhR stimulation by UCB, as reflected by lower CD39 levels. Blockade of hypoxia-inducible-factor-1alpha (HIF-1α) upregulates CD39 and favors Th17-cell regulatory responses. Resistance of Th17-cells to AhR signaling results, in part, from HIF-1α-dependent induction of ATP-binding cassette (ABC) transporters: multidrug-resistance-protein-1 (MDR1) and multidrug-resistance-associated-protein-4 (MRP4). Increased ABC transporters promote efflux of suppressive AhR ligands, such as UCB, from Th17-cells. Inhibition of MDR1, MRP4 and/or HIF-1α with ritonavir (RTV) reconstitutes AhR function in Th17-cells, enhancing therapeutic effects of UCB in dextran-sulfate-sodium-induced experimental colitis. Deleterious effects of hypoxia on Th17-cells in Crohn's disease can be ameliorated either by inhibiting HIF-1α or by suppressing ABC transporters to increase UCB availability as an AhR substrate. Targeting HIF-1α-ABC transporters could provide innovative therapeutic pathways for IBD.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Colitis/inmunología , Enfermedad de Crohn/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/inmunología , Receptores de Hidrocarburo de Aril/inmunología , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/inmunología , Animales , Antiinflamatorios/inmunología , Antiinflamatorios/farmacología , Apirasa/genética , Apirasa/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Bilirrubina/inmunología , Bilirrubina/farmacología , Hipoxia de la Célula , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/genética , Enfermedad de Crohn/genética , Enfermedad de Crohn/patología , Sulfato de Dextran/administración & dosificación , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/inmunología , Membrana Mucosa/patología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Cultivo Primario de Células , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Receptores de Hidrocarburo de Aril/genética , Ritonavir/farmacología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/patología
12.
Circ Res ; 118(12): 1940-1959, 2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-27283533

RESUMEN

Understanding the processes governing the ability of the heart to repair and regenerate after injury is crucial for developing translational medical solutions. New avenues of exploration include cardiac cell therapy and cellular reprogramming targeting cell death and regeneration. An attractive possibility is the exploitation of cytoprotective genes that exist solely for self-preservation processes and serve to promote and support cell survival. Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. The remarkable cardioprotective effects ascribed to heme oxygenase-1 are best evidenced by its ability to regulate inflammatory processes, cellular signaling, and mitochondrial function ultimately mitigating myocardial tissue injury and the progression of vascular-proliferative disease. We discuss here new insights into the role of heme oxygenase-1 and heme on cardiovascular health, and importantly, how they might be leveraged to promote heart repair after injury.


Asunto(s)
Monóxido de Carbono/metabolismo , Hemo-Oxigenasa 1/metabolismo , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Animales , Hemo-Oxigenasa 1/genética , Humanos , Mitocondrias Cardíacas/metabolismo
13.
Heart Lung Circ ; 27(1): 122-129, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28487062

RESUMEN

BACKGROUND: Atrial fibrillation (AF) is the most frequent complication of surgery performed on cardiopulmonary bypass (CPB) and recent work associates CPB with postoperative inflammation. We have shown that all tissue injury releases mitochondrial damage associated molecular patterns (mtDAMPs) including mitochondrial DNA (mtDNA). This can act as a direct, early activator of neutrophils (PMN), eliciting a systemic inflammatory response syndrome (SIRS) while suppressing PMN function. Neutrophil Extracellular Traps (NETs) are crucial to host defence. They carry out NETosis wherein webs of granule proteins and chromatin trap and kill bacteria. We hypothesised that surgery performed on CPB releases mtDAMPs into the circulation. Molecular patterns thus mobilised during CPB might then participate in the pathogenesis of SIRS and predict postoperative complications like AF [1]. METHODS: We prospectively studied 16 patients undergoing elective operations on CPB. Blood was sampled preoperatively, at the end of CPB and on days 1-2 postoperatively. Plasma samples were analysed for mtDNA. Neutrophil IL-6 gene expression was studied to assess induction of SIRS. Neutrophils were also assayed for the presence of neutrophil extracellular traps (NETs/NETosis). These biologic findings were then correlated to clinical data and compared in patients with and without postoperative AF (POAF). RESULTS: Mitochondrial DNA was significantly elevated following CPB (six-fold increase post-CPB, p=0.008 and five-fold increase days 1-2, p=0.02). Patients with POAF showed greater increases in mtDNA post-CPB than those without. Postoperative AF was seen in all patients with a ≥2-fold increase of mtDNA (p=0.037 vs. <2-fold). Neutrophil IL-6 gene transcription increased postoperatively demonstrating SIRS that was greatest days 1-2 (p=0.039). Neutrophil extracellular trap (NET) formation was markedly suppressed in the post-CPB state. CONCLUSION: Mitochondrial DNA is released by CPB surgery and is associated with POAF. IL-6 gene expression increases after CPB, demonstrating the evolution of postoperative SIRS. Lastly, cardiac surgery on CPB also suppressed PMN NETosis. Taken together, our data suggest that mtDNA released during surgery on CPB, may be involved in the pathogenesis of SIRS and related postoperative inflammatory events like POAF and infections. Mitochondrial DNA may therefore prove to be an early biomarker for postoperative complications with the degree of association to be determined in appropriately sized studies. If mtDNA is directly involved in cardiac inflammation, mtDNA-induced toll-like receptor-9 (TLR9) signalling could also be targeted therapeutically.


Asunto(s)
Fibrilación Atrial/sangre , Puente Cardiopulmonar/efectos adversos , ADN Mitocondrial/sangre , Mitocondrias/genética , Complicaciones Posoperatorias , Anciano , Fibrilación Atrial/genética , Biomarcadores/sangre , ADN Mitocondrial/genética , Femenino , Cardiopatías/cirugía , Humanos , Masculino , Mitocondrias/metabolismo , Reacción en Cadena de la Polimerasa , Estudios Prospectivos
14.
Stroke ; 48(9): 2565-2573, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28747460

RESUMEN

BACKGROUND AND PURPOSE: Subarachnoid hemorrhage (SAH) is associated with a temporal pattern of stroke incidence. We hypothesized that natural oscillations in gene expression controlling circadian rhythm affect the severity of neuronal injury. We moreover predict that heme oxygenase-1 (HO-1/Hmox1) and its product carbon monoxide (CO) contribute to the restoration of rhythm and neuroprotection. METHODS: Murine SAH model was used where blood was injected at various time points of the circadian cycle. Readouts included circadian clock gene expression, locomotor activity, vasospasm, neuroinflammatory markers, and apoptosis. In addition, cerebrospinal fluid and peripheral blood leukocytes from SAH patients and controls were analyzed for clock gene expression. RESULTS: Significant elevations in the clock genes Per-1, Per-2, and NPAS-2 were observed in the hippocampus, cortex, and suprachiasmatic nucleus in mice subjected to SAH at zeitgeber time (ZT) 12 when compared with ZT2. Clock gene expression amplitude correlated with basal expression of HO-1, which was also significantly greater at ZT12. SAH animals showed a significant reduction in cerebral vasospasm, neuronal apoptosis, and microglial activation at ZT12 compared with ZT2. In animals with myeloid-specific HO-1 deletion (Lyz-Cre-Hmox1fl/fl ), Per-1, Per-2, and NPAS-2 expression was reduced in the suprachiasmatic nucleus, which correlated with increased injury. Treatment with low-dose CO rescued Lyz-Cre-Hmox1fl/fl mice, restored Per-1, Per-2, and NPAS-2 expression, and reduced neuronal apoptosis. CONCLUSIONS: Clock gene expression regulates, in part, the severity of SAH and requires myeloid HO-1 activity to clear the erythrocyte burden and inhibit neuronal apoptosis. Exposure to CO rescues the loss of HO-1 and thus merits further investigation in patients with SAH.


Asunto(s)
Monóxido de Carbono/metabolismo , Ritmo Circadiano/genética , Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Proteínas de la Membrana/genética , Hemorragia Subaracnoidea/genética , Factores de Transcripción ARNTL/genética , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas CLOCK/genética , Líquido Cefalorraquídeo/metabolismo , Hemo-Oxigenasa 1/metabolismo , Humanos , Inmunohistoquímica , Inflamación , Leucocitos/metabolismo , Locomoción , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas Circadianas Period/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Índice de Severidad de la Enfermedad , Núcleo Supraquiasmático/metabolismo , Vasoespasmo Intracraneal
15.
Am J Hematol ; 92(6): 569-582, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28378932

RESUMEN

Sickle Cell Disease (SCD) is a painful, lifelong hemoglobinopathy inherited as a missense point mutation in the hemoglobin (Hb) beta-globin gene. This disease has significant impact on quality of life and mortality, thus a substantial medical need exists to reduce the vaso-occlusive crises which underlie the pathophysiology of the disease. The concept that a gaseous molecule may exert biological function has been well known for over one hundred years. Carbon monoxide (CO), although studied in SCD for over 50 years, has recently emerged as a powerful cytoprotective biological response modifier capable of regulating a host of physiologic and therapeutic processes that, at low concentrations, exerts key physiological functions in various models of tissue inflammation and injury. CO is physiologically generated by the metabolism of heme by the heme oxygenase enzymes and is measurable in blood. A substantial amount of preclinical and clinical data with CO have been generated, which provide compelling support for CO as a potential therapeutic in a number of pathological conditions. Data underlying the therapeutic mechanisms of CO, including in SCD, have been generated by a plethora of in vitro and preclinical studies including multiple SCD mouse models. These data show CO to have key signaling impacts on a host of metallo-enzymes as well as key modulating genes that in sum, result in significant anti-inflammatory, anti-oxidant and anti-apoptotic effects as well as vasodilation and anti-adhesion of cells to the endothelium resulting in preservation of vascular flow. CO may also have a role as an anti-polymerization HbS agent. In addition, considerable scientific data in the non-SCD literature provide evidence for a beneficial impact of CO on cerebrovascular complications, suggesting that in SCD, CO could potentially limit these highly problematic neurologic outcomes. Research is needed and hopefully forthcoming, to carefully elucidate the safety and benefits of this potential therapy across the age spectrum of patients impacted by the host of pathophysiological complications of this devastating disease.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/metabolismo , Monóxido de Carbono/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Enfermedades Vasculares/etiología , Enfermedades Vasculares/prevención & control , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Animales , Monóxido de Carbono/administración & dosificación , Monóxido de Carbono/efectos adversos , Monóxido de Carbono/sangre , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Hemo Oxigenasa (Desciclizante)/sangre , Hemoglobinas/química , Hemoglobinas/genética , Hemoglobinas/metabolismo , Humanos , Transducción de Señal , Resultado del Tratamiento
16.
Microbiol Immunol ; 59(8): 452-65, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26146866

RESUMEN

Induction of mammalian heme oxygenase (HO)-1 and exposure of animals to carbon monoxide (CO) ameliorates experimental colitis. When enteric bacteria, including Escherichia coli, are exposed to low iron conditions, they express an HO-like enzyme, chuS, and metabolize heme into iron, biliverdin and CO. Given the abundance of enteric bacteria residing in the intestinal lumen, our postulate was that commensal intestinal bacteria may be a significant source of CO and those that express chuS and other Ho-like molecules suppress inflammatory immune responses through release of CO. According to real-time PCR, exposure of mice to CO results in changes in enteric bacterial composition and increases E. coli 16S and chuS DNA. Moreover, the severity of experimental colitis correlates positively with E. coli chuS expression in IL-10 deficient mice. To explore functional roles, E. coli were genetically modified to overexpress chuS or the chuS gene was deleted. Co-culture of chuS-overexpressing E. coli with bone marrow-derived macrophages resulted in less IL-12p40 and greater IL-10 secretion than in wild-type or chuS-deficient E. coli. Mice infected with chuS-overexpressing E. coli have more hepatic CO and less serum IL-12 p40 than mice infected with chuS-deficient E. coli. Thus, CO alters the composition of the commensal intestinal microbiota and expands populations of E. coli that harbor the chuS gene. These bacteria are capable of attenuating innate immune responses through expression of chuS. Bacterial HO-like molecules and bacteria-derived CO may represent novel targets for therapeutic intervention in inflammatory conditions.


Asunto(s)
Escherichia coli/enzimología , Escherichia coli/inmunología , Hemo Oxigenasa (Desciclizante)/inmunología , Hemo Oxigenasa (Desciclizante)/metabolismo , Evasión Inmune , Inmunidad Innata , Animales , Monóxido de Carbono/metabolismo , Células Cultivadas , Técnicas de Cocultivo , ADN Bacteriano/genética , ADN Ribosómico/genética , Escherichia coli/metabolismo , Eliminación de Gen , Expresión Génica , Hemo Oxigenasa (Desciclizante)/genética , Interleucina-10/metabolismo , Subunidad p40 de la Interleucina-12/metabolismo , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética
17.
J Immunol ; 191(5): 2089-95, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23872052

RESUMEN

We hypothesized B cells are involved in the pathogenesis of idiopathic pulmonary fibrosis (IPF), a progressive, restrictive lung disease that is refractory to glucocorticoids and other nonspecific therapies, and almost invariably lethal. Accordingly, we sought to identify clinically associated B cell-related abnormalities in these patients. Phenotypes of circulating B cells were characterized by flow cytometry. Intrapulmonary processes were evaluated by immunohistochemistry. Plasma B lymphocyte stimulating factor (BLyS) was assayed by ELISA. Circulating B cells of IPF subjects were more Ag differentiated, with greater plasmablast proportions (3.1 ± 0.8%) than in normal controls (1.3 ± 0.3%) (p < 0.03), and the extent of this differentiation correlated with IPF patient lung volumes (r = 0.44, p < 0.03). CD20(+) B cell aggregates, diffuse parenchymal and perivascular immune complexes, and complement depositions were all prevalent in IPF lungs, but much less prominent or absent in normal lungs. Plasma concentrations of BLyS, an obligate factor for B cell survival and differentiation, were significantly greater (p < 0.0001) in 110 IPF (2.05 ± 0.05 ng/ml) than among 53 normal (1.40 ± 0.04 ng/ml) and 90 chronic obstructive pulmonary disease subjects (1.59 ± 0.05 ng/ml). BLyS levels were uniquely correlated among IPF patients with pulmonary artery pressures (r = 0.58, p < 0.0001). The 25% of IPF subjects with the greatest BLyS values also had diminished 1-y survival (46 ± 11%), compared with those with lesser BLyS concentrations (81 ± 5%) (hazard ratio = 4.0, 95% confidence interval = 1.8-8.7, p = 0.0002). Abnormalities of B cells and BLyS are common in IPF patients, and highly associated with disease manifestations and patient outcomes. These findings have implications regarding IPF pathogenesis and illuminate the potential for novel treatment regimens that specifically target B cells in patients with this lung disease.


Asunto(s)
Factor Activador de Células B/sangre , Linfocitos B/citología , Linfocitos B/inmunología , Diferenciación Celular , Fibrosis Pulmonar Idiopática/inmunología , Anciano , Anciano de 80 o más Años , Diferenciación Celular/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Fibrosis Pulmonar Idiopática/sangre , Fibrosis Pulmonar Idiopática/patología , Inmunohistoquímica , Masculino , Persona de Mediana Edad
18.
Curr Opin Organ Transplant ; 20(1): 8-12, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25563986

RESUMEN

PURPOSE OF REVIEW: To update knowledge concerning the cause and consequences of the detrimental forms of innate immunity that inevitably occurs in peritransplant period tissue and cellular transplants. In addition, we review the information that a newly discovered, engraftment-promoting, and tolerance-inducing macrophage population is identified and characterized. RECENT FINDINGS: The allograft response mounted by adaptive immune cells is shaped by innate immunity. The early allograft response is uniquely intense as a result of activation of the innate immune response created by ischemia reperfusion injury in organ transplants, delayed revascularization of cell transplants, and hypoxia. Inflammation is created by both cellular 'debris' and cytokines. However, a newly discovered prominent, albeit fragile, tissue-resident, noninvasive, and immunoregulatory macrophage promotes engraftment and tolerance. The role of intracellular 'debris' as well as inflammation in evoking detrimental rejection-provoking peritransplant inflammation is emphasized as well as characterization of a prominent and highly immunoregulatory albeit fragile macrophage population that is tissue-resident and does not circulate is characterized. SUMMARY: Opportunity lies in the ability to rein in detrimental peri-transplant inflammation and in the ability to promote the longevity of a subpopulation of highly potent tissue-resident immunoregulatory macrophages.


Asunto(s)
Tolerancia Inmunológica/inmunología , Inmunidad Innata/inmunología , Trasplante de Órganos , Aloinjertos , Animales , Humanos
19.
Gastroenterology ; 144(4): 789-98, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23266559

RESUMEN

BACKGROUND & AIMS: Heme oxygenase-1 (HO-1) and its metabolic by-product, carbon monoxide (CO), protect against intestinal inflammation in experimental models of colitis, but little is known about their intestinal immune mechanisms. We investigated the interactions among CO, HO-1, and the enteric microbiota in mice and zebrafish. METHODS: Germ-free, wild-type, and interleukin (Il)10(-/-) mice and germ-free zebrafish embryos were colonized with specific pathogen-free (SPF) microbiota. Germ-free or SPF-raised wild-type and Il10(-/-) mice were given intraperitoneal injections of cobalt(III) protoporphyrin IX chloride (CoPP), which up-regulates HO-1, the CO-releasing molecule Alfama-186, or saline (control). Colitis was induced in wild-type mice housed in SPF conditions by infection with Salmonella typhimurium. RESULTS: In colons of germ-free, wild-type mice, SPF microbiota induced production of HO-1 via activation of nuclear factor erythroid 2-related factor 2-, IL-10-, and Toll-like receptor-dependent pathways; similar observations were made in zebrafish. SPF microbiota did not induce HO-1 in colons of germ-free Il10(-/-) mice. Administration of CoPP to Il10(-/-) mice before transition from germ-free to SPF conditions reduced their development of colitis. In Il10(-/-) mice, CO and CoPP reduced levels of enteric bacterial genomic DNA in mesenteric lymph nodes. In mice with S typhimurium-induced enterocolitis, CoPP reduced the numbers of live S typhimurium recovered from the lamina propria, mesenteric lymph nodes, spleen, and liver. Knockdown of HO-1 in mouse macrophages impaired their bactericidal activity against E coli, E faecalis, and S typhimurium, whereas exposure to CO or overexpression of HO-1 increased their bactericidal activity. HO-1 induction and CO increased acidification of phagolysosomes. CONCLUSIONS: Colonic HO-1 prevents colonic inflammation in mice. HO-1 is induced by the enteric microbiota and its homeostatic function is mediated, in part, by promoting bactericidal activities of macrophages.


Asunto(s)
Traslocación Bacteriana/fisiología , Monóxido de Carbono/farmacología , Colitis/prevención & control , Hemo-Oxigenasa 1/metabolismo , Salmonella typhimurium/fisiología , Animales , Traslocación Bacteriana/efectos de los fármacos , Western Blotting , Colitis/tratamiento farmacológico , Colitis/microbiología , Modelos Animales de Enfermedad , Escherichia coli/patogenicidad , Gentamicinas/farmacología , Hemo-Oxigenasa 1/biosíntesis , Macrófagos/citología , Macrófagos/fisiología , Metagenoma , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
Biochem Biophys Res Commun ; 449(1): 94-9, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24814708

RESUMEN

Macrophages play a crucial role in the maintenance and resolution of inflammation and express a number of pro- and anti-inflammatory molecules in response to stressors. Among them, the complement receptor 5a (C5aR) plays an integral role in the development of inflammatory disorders. Biliverdin and bilirubin, products of heme catabolism, exert anti-inflammatory effects and inhibit complement activation. Here, we define the effects of biliverdin on C5aR expression in macrophages and the roles of Akt and mammalian target of rapamycin (mTOR) in these responses. Biliverdin administration inhibited lipopolysaccharide (LPS)-induced C5aR expression (without altering basal expression), an effect partially blocked by rapamycin, an inhibitor of mTOR signaling. Biliverdin also reduced LPS-dependent expression of the pro-inflammatory cytokines TNF-α and IL-6. Collectively, these data indicate that biliverdin regulates LPS-mediated expression of C5aR via the mTOR pathway, revealing an additional mechanism underlying biliverdin's anti-inflammatory effects.


Asunto(s)
Biliverdina/farmacología , Lipopolisacáridos/farmacología , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Antioxidantes/farmacología , Línea Celular , Endotoxinas/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones , Receptor de Anafilatoxina C5a/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA