Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33649212

RESUMEN

The recently elucidated proresolving conjugates in tissue regeneration (CTR) maresin-CTR (MCTR), protectin-CTR (PCTR), and resolvin-CTR (RCTR), termed cysteinyl-specialized proresolving mediators (cys-SPMs) each promotes regeneration, controls infection, and accelerates resolution of inflammation. Here, we sought evidence for cys-SPM activation of primordial pathways in planaria (Dugesia japonica) regeneration that might link resolution of inflammation and regeneration. On surgical resection, planaria regeneration was enhanced with MCTR3, PCTR3, or RCTR3 (10 nM), each used for RNA sequencing. The three cys-SPMs shared up-regulation of 175 known transcripts with fold-change > 1.25 and combined false discovery rate (FDR) < 0.002, and 199 canonical pathways (FDR < 0.25), including NF-κB pathways and an ortholog of human TRAF3 (TNFR-associated factor 3). Three separate pathway analyses converged on TRAF3 up-regulation by cys-SPMs. With human macrophages, three cys-SPMs each dose-dependently increased TRAF3 expression in a cAMP-PKA-dependent manner. TRAF3 overexpression in macrophages enhanced Interleukin-10 (IL-10) and phagocytosis of Escherichia coli IL-10 also increased phagocytosis in a dose-dependent manner. Silencing of mouse TRAF3 in vivo significantly reduced IL-10 and macrophage phagocytosis. TRAF3 silencing in vivo also relieved cys-SPMs' actions in limiting polymorphonuclear neutrophil in E. coli exudates. These results identify cys-SPM-regulated pathways in planaria regeneration, uncovering a role for TRAF3/IL-10 in regulating mammalian phagocyte functions in resolution. Cys-SPM activation of TRAF3 signaling is a molecular component of both regeneration and resolution of infectious inflammation.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Planarias/inmunología , Regeneración/inmunología , Transducción de Señal/inmunología , Factor 3 Asociado a Receptor de TNF/inmunología , Animales , Infecciones por Escherichia coli/genética , Humanos , Inflamación/genética , Inflamación/inmunología , Neutrófilos/inmunología , Fagocitosis , Planarias/genética , Regeneración/genética , Transducción de Señal/genética , Factor 3 Asociado a Receptor de TNF/genética
2.
Prehosp Emerg Care ; 27(2): 269-274, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35820141

RESUMEN

We report the initial seven patients treated with nebulized ketamine for moderate to severe pain, via breath-actuated nebulizer, in an urban, ground-based emergency medical services (EMS) system. Ketamine for analgesia in the emergency setting has become widespread over the past decade. The addition of a non-parenteral, inexpensive, and well-tolerated ketamine delivery option is extremely desirable. We believe these initial data demonstrate promising pain reduction coupled with minimal side effects, indicating a potential role for nebulized ketamine in EMS.


Asunto(s)
Analgesia , Servicios Médicos de Urgencia , Ketamina , Humanos , Ketamina/uso terapéutico , Dolor/tratamiento farmacológico , Manejo del Dolor , Analgésicos/uso terapéutico
3.
Prehosp Emerg Care ; 27(7): 920-926, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37276174

RESUMEN

We report the initial six pediatric patients treated with ketamine for benzodiazepine-resistant status epilepticus in an urban, ground-based emergency medical services (EMS) system. Evidence for ketamine as a second-line agent for both adult and pediatric refractory seizure activity in the hospital setting has increased over the past decade. The availability of an inexpensive and familiar second-line prehospital anti-epileptic drug option is extremely desirable. We believe these initial data demonstrate promising seizure control effects without significant respiratory depression, indicating a potential role for ketamine in the EMS treatment of pediatric benzodiazepine-refractory seizures.


Asunto(s)
Servicios Médicos de Urgencia , Ketamina , Estado Epiléptico , Adulto , Humanos , Niño , Benzodiazepinas/uso terapéutico , Ketamina/uso terapéutico , Estado Epiléptico/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Anticonvulsivantes/uso terapéutico
4.
Int J Mol Sci ; 24(22)2023 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-38003333

RESUMEN

Omega-3 polyunsaturated fatty acids (PUFAs) play a vital role in human health, well-being, and the management of inflammatory diseases. Insufficient intake of omega-3 is linked to disease development. Specialized pro-resolving mediators (SPMs) are derived from omega-3 PUFAs and expedite the resolution of inflammation. They fall into categories known as resolvins, maresins, protectins, and lipoxins. The actions of SPMs in the resolution of inflammation involve restricting neutrophil infiltration, facilitating the removal of apoptotic cells and cellular debris, promoting efferocytosis and phagocytosis, counteracting the production of pro-inflammatory molecules like chemokines and cytokines, and encouraging a pro-resolving macrophage phenotype. This is an experimental pilot study in which ten healthy subjects were enrolled and received a single dose of 6 g of an oral SPM-enriched marine oil emulsion. Peripheral blood was collected at baseline, 3, 6, 9, 12, and 24 h post-administration. Temporal increases in plasma and serum SPM levels were found by using LC-MS/MS lipid profiling. Additionally, we characterized the temporal increases in omega-3 levels and established fundamental pharmacokinetics in both aforementioned matrices. These findings provide substantial evidence of the time-dependent elevation of SPMs, reinforcing the notion that oral supplementation with SPM-enriched products represents a valuable source of essential bioactive SPMs.


Asunto(s)
Ácidos Docosahexaenoicos , Ácidos Grasos Omega-3 , Humanos , Voluntarios Sanos , Cromatografía Liquida , Proyectos Piloto , Espectrometría de Masas en Tándem , Inflamación , Factor de Activación Plaquetaria , Mediadores de Inflamación
5.
Am J Physiol Heart Circ Physiol ; 322(6): H953-H970, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35333119

RESUMEN

All fats are not created equal, and despite the extensive literature, the effect of fat intake is the most debated question in obesity, cardiovascular, and cardiorenal research. Cellular and molecular mechanisms underlying cardiac dysfunction and consequent heart failure in the setting of obesity are not well understood. Our understanding of how fats are metabolically transformed after nonreperfused myocardial infarction (MI), in particular, is incomplete. Here, using male C57BL/6J mice (2 mo old), we determined the role of omega-6 fatty acids, provided as safflower oil (SO) for 12 wk, followed by supplementation with docosahexaenoic acid (DHA; n-3 fatty acids) for 8 wk before MI. With SO feeding, inflammation resolution was impaired. Specialized proresolving mediators (SPMs) increased in DHA-fed mice to reverse the effects of SO, whereas prostaglandins and thromboxane B2 were reduced in the spleen and amplified multiple resolving mechanisms in heart and kidney post-MI. DHA amplified the number of resolving macrophages and cardiac reparative pathways of the splenocardiac and cardiorenal networks in acute heart failure, with higher Treg cells in chronic heart failure and marked expression of Foxp3+ in the myocardium. Our findings indicate that surplus ingestion of SO intensified systemic, baseline, nonresolving inflammation, and DHA intake dominates splenocardiac resolving phase with the biosynthesis of SPMs and controlled cardiorenal inflammation in heart failure survivor mice.NEW & NOTEWORTHY Chronic and surplus dietary intake of safflower oil (SO) increased plasma creatinine dysregulated post-MI splenocardiac inflammation coincides with the dysfunctional cardiorenal network. In contrast, docosahexaenoic acid (DHA) increases post-MI survival in chronic heart failure. DHA transforms into specialized proresolving mediators (SPMs) and limited proinflammatory prostaglandins and thromboxanes following myocardial infarction (MI). DHA promotes Ly6Clow resolving macrophages and T regulatory cells (Foxp3+) in a splenocardiac manner post-MI.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Animales , Ácidos Docosahexaenoicos , Factores de Transcripción Forkhead , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/complicaciones , Prostaglandinas , Aceite de Cártamo
6.
Blood ; 134(17): 1458-1468, 2019 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-31300403

RESUMEN

Deep vein thrombosis (DVT) is a common cardiovascular disease with a major effect on quality of life, and safe and effective therapeutic measures to efficiently reduce existent thrombus burden are scarce. Using a comprehensive targeted liquid chromatography-tandem mass spectrometry-based metabololipidomics approach, we established temporal clusters of endogenously biosynthesized specialized proresolving mediators (SPMs) and proinflammatory and prothrombotic lipid mediators during DVT progression in mice. Administration of resolvin D4 (RvD4), an SPM that was enriched at the natural onset of thrombus resolution, significantly reduced thrombus burden, with significantly less neutrophil infiltration and more proresolving monocytes in the thrombus, as well as an increased number of cells in an early apoptosis state. Moreover, RvD4 promoted the biosynthesis of other D-series resolvins involved in facilitating resolution of inflammation. Neutrophils from RvD4-treated mice were less susceptible to an ionomycin-induced release of neutrophil extracellular traps (NETs), a meshwork of decondensed chromatin lined with histones and neutrophil proteins critical for DVT development. These results suggest that delivery of SPMs, specifically RvD4, modulates the severity of thrombo-inflammatory disease in vivo and improves thrombus resolution.


Asunto(s)
Ácidos Grasos Insaturados/uso terapéutico , Trombosis de la Vena/tratamiento farmacológico , Animales , Progresión de la Enfermedad , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Mediadores de Inflamación/inmunología , Lípidos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/efectos de los fármacos , Trombosis de la Vena/inmunología , Trombosis de la Vena/patología
7.
FASEB J ; 34(12): 15888-15906, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33047359

RESUMEN

A successful acute inflammatory response results in the elimination of infectious agents by neutrophils and monocytes, followed by resolution and repair through tissue-resident and recruited macrophages. Resolvins (D-series and E-series) are pro-resolving lipid mediators involved in resolution and tissue repair, whose intracellular signaling remains of interest. Here, we report that D-series resolvins (RvD1- RvD5) activate phospholipase D (PLD), a ubiquitously expressed membrane lipase enzyme activity in modulating phagocyte functions. The mechanism for PLD-mediated actions of Resolvin-D5 (RvD5) in polarizing macrophages (M1-like toward M2-like) was found to be two-pronged: (a) RvD5 inhibits post-transcriptional modifications, by miRs and 3'exonucleases that process PLD2 mRNA, thus increasing PLD2 expression and activity; and (b) RvD5 enhances PLD2-S6Kinase signaling required for membrane expansion and efferocytosis. In an in vivo model of second organ reflow injury, we found that RvD5 did not reduce lung neutrophil myeloperoxidase levels in PLD2-/- mice compared to WT and PLD1-/- mice, confirming a novel role of PLD2 as the isoform in RvD5-mediated resolution processes. These results demonstrate that RvD5-PLD2 are attractive targets for therapeutic interventions in vascular inflammation such as ischemia-reperfusion injury and cardiovascular diseases.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Inflamación/metabolismo , Fagocitos/efectos de los fármacos , Fosfolipasa D/metabolismo , Animales , Células Cultivadas , Femenino , Humanos , Inflamación/tratamiento farmacológico , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fagocitos/metabolismo , Fagocitosis/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Circ Res ; 124(2): 279-291, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30582456

RESUMEN

RATIONALE: CD69 is an immunomodulatory molecule induced during lymphocyte activation. Following stroke, T-lymphocytes upregulate CD69 but its function is unknown. OBJECTIVE: We investigated whether CD69 was involved in brain damage following an ischemic stroke. METHODS AND RESULTS: We used adult male mice on the C57BL/6 or BALB/c backgrounds, including wild-type mice and CD69-/- mice, and CD69+/+ and CD69-/- lymphocyte-deficient Rag2-/- mice, and generated chimeric mice. We induced ischemia by transient or permanent middle cerebral artery occlusion. We measured infarct volume, assessed neurological function, and studied CD69 expression, as well as platelet function, fibrin(ogen) deposition, and VWF (von Willebrand factor) expression in brain vessels and VWF content and activity in plasma, and performed the tail-vein bleeding test and the carotid artery ferric chloride-induced thrombosis model. We also performed primary glial cell cultures and sorted brain CD45-CD11b-CD31+ endothelial cells for mRNA expression studies. We blocked VWF by intravenous administration of anti-VWF antibodies. CD69-/- mice showed larger infarct volumes and worse neurological deficits than the wild-type mice after ischemia. This worsening effect was not attributable to lymphocytes or other hematopoietic cells. CD69 deficiency lowered the time to thrombosis in the carotid artery despite platelet function not being affected. Ischemia upregulated Cd69 mRNA expression in brain endothelial cells. CD69-deficiency increased fibrin(ogen) accumulation in the ischemic tissue, and plasma VWF content and activity, and VWF expression in brain vessels. Blocking VWF reduced infarct volume and reverted the detrimental effect of CD69-/- deficiency. CONCLUSIONS: CD69 deficiency promotes a prothrombotic phenotype characterized by increased VWF and worse brain damage after ischemic stroke. The results suggest that CD69 acts as a downregulator of endothelial activation.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Células Endoteliales/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Lectinas Tipo C/metabolismo , Activación de Linfocitos , Linfocitos T/metabolismo , Animales , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/genética , Coagulación Sanguínea , Plaquetas/metabolismo , Encéfalo/patología , Células Cultivadas , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Células Endoteliales/patología , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/patología , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción de Señal , Linfocitos T/patología , Factor de von Willebrand/metabolismo
9.
J Immunol ; 201(11): 3161-3165, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30355784

RESUMEN

Inflammatory resolution is a process that, when uncontrolled, impacts many organs and diseases. As an active, self-limited inflammatory process, resolution involves biosynthesis of specialized proresolving mediators (SPM) (e.g., lipoxins, resolvins [Rv], protectins, and maresins). Because vagal stimulation impacts inflammation, we examined human and mouse vagus ex vivo to determine if they produce lipid mediators. Using targeted lipid mediator metabololipidomics, we identified lipoxins, Rv, and protectins produced by both human and mouse vagus as well as PGs and leukotrienes. Human vagus produced SPM (e.g., RvE1, NPD1/PD1, MaR1, RvD5, and LXA4) on stimulation that differed from mouse (RvD3, RvD6, and RvE3), demonstrating species-selective SPM. Electrical vagus stimulation increased SPM in both human and mouse vagus as did incubations with Escherichia coli. Electrical vagus stimulation increased SPM and decreased PGs and leukotrienes. These results provide direct evidence for vagus SPM and eicosanoids. Moreover, they suggest that this vagus SPM circuit contributes to a new proresolving vagal reflex.


Asunto(s)
Eicosanoides/metabolismo , Infecciones por Escherichia coli/inmunología , Escherichia coli/fisiología , Mediadores de Inflamación/metabolismo , Nervio Vago/fisiología , Animales , Células Cultivadas , Estimulación Eléctrica , Humanos , Metabolismo de los Lípidos , Lipoxinas/metabolismo , Masculino , Ratones , Ratones Endogámicos , Técnicas de Cultivo de Órganos , Especificidad de la Especie , Estimulación del Nervio Vago
10.
Am J Pathol ; 188(4): 950-966, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29571326

RESUMEN

Resolvin conjugates in tissue regeneration (RCTRs) are new chemical signals that accelerate resolution of inflammation, infection, and tissue regeneration. Herein, using liquid chromatography-tandem mass spectrometry-based metabololipidomics, we identified RCTRs in human spleen, lymph node, bone marrow, and brain. In human spleen incubated with Staphylococcus aureus, endogenous RCTRs were increased along with conversion of deuterium-labeled docosahexaenoic acid, conferring pathway activation. Physical and biological properties of endogenous RCTRs were matched with those prepared by total organic synthesis. The complete stereochemical assignment of bioactive RCTR1 is 8R-glutathionyl-7S,17S-dihydroxy-4Z,9E,11E,13Z,15E,19Z-docosahexaenoic acid, RCTR2 is 8R-cysteinylglycinyl-7S,17S-dihydroxy-4Z,9E,11E,13Z,15E,19Z-docosahexaenoic acid, and RCTR3 is 8R-cysteinyl-7S,17S-dihydroxy-4Z,9E,11E,13Z,15E,19Z-docosahexaenoic acid. These stereochemically defined RCTRs stimulated human macrophage phagocytosis, efferocytosis, and planaria tissue generation. Proteome profiling demonstrated that RCTRs regulated both proinflammatory and anti-inflammatory cytokines with human macrophages. In microfluidic chambers, the three RCTRs limited human polymorphonuclear cell migration. In hind-limb ischemia-reperfusion-initiated organ injury, both RCTR2 and RCTR3 reduced polymorphonuclear cell infiltration into lungs. In infectious peritonitis, RCTR1 shortened the resolution intervals. Each RCTR (1 nmol/L) accelerated planaria tissue regeneration by approximately 0.5 days, with direct comparison to both maresin and protectin CTRs. Together, these results identify a new bioactive RCTR (ie, RCTR3) in human tissues and establish the complete stereochemistry and rank-order potencies of three RCTRs in vivo. Moreover, RCTR1, RCTR2, and RCTR3 each exert potent anti-inflammatory and proresolving actions with human leukocytes.


Asunto(s)
Ácidos Grasos Omega-3/química , Fagocitos/metabolismo , Regeneración/fisiología , Animales , Quimiotaxis , Infecciones por Escherichia coli/patología , Ácidos Grasos Omega-3/biosíntesis , Humanos , Inflamación/patología , Lesión Pulmonar/microbiología , Lesión Pulmonar/patología , Macrófagos/citología , Masculino , Metaboloma , Ratones , Fagocitos/citología , Fagocitosis , Planarias/fisiología , Proteoma/metabolismo , Daño por Reperfusión/microbiología , Daño por Reperfusión/patología , Bazo/metabolismo , Estereoisomerismo
11.
J Immunol ; 198(2): 842-851, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27994074

RESUMEN

Resolution of acute inflammation is an active process governed by specialized proresolving mediators, including resolvin (Rv)D2, that activates a cell surface G protein-coupled receptor, GPR18/DRV2. In this study, we investigated RvD2-DRV2-dependent resolution mechanisms using DRV2-deficient mice (DRV2-knockout [KO]). In polymicrobial sepsis initiated by cecal ligation and puncture, RvD2 (∼2.7 nmol/mouse) significantly increased survival (>50%) of wild-type mice and reduced hypothermia and bacterial titers compared with vehicle-treated cecal ligation and puncture mice that succumbed at 48 h. Protection by RvD2 was abolished in DRV2-KO mice. Mass spectrometry-based lipid mediator metabololipidomics demonstrated that DRV2-KO infectious exudates gave higher proinflammatory leukotriene B4 and procoagulating thromboxane B2, as well as lower specialized proresolving mediators, including RvD1 and RvD3, compared with wild-type. RvD2-DRV2-initiated intracellular signals were investigated using mass cytometry (cytometry by time-of-flight), which demonstrated that RvD2 enhanced phosphorylation of CREB, ERK1/2, and STAT3 in WT but not DRV2-KO macrophages. Monitored by real-time imaging, RvD2-DRV2 interaction significantly enhanced phagocytosis of live Escherichia coli, an action dependent on protein kinase A and STAT3 in macrophages. Taken together, we identified an RvD2/DRV2 axis that activates intracellular signaling pathways that increase phagocytosis-mediated bacterial clearance, survival, and organ protection. Moreover, these results provide evidence for RvD2-DRV2 and their downstream pathways in pathophysiology of infectious inflammation.


Asunto(s)
Ácidos Docosahexaenoicos/inmunología , Receptores Acoplados a Proteínas G/inmunología , Sepsis/inmunología , Animales , Cromatografía Liquida , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/inmunología , Masculino , Ratones , Ratones Noqueados , Fagocitosis/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Sepsis/metabolismo , Espectrometría de Masas en Tándem
12.
Stroke ; 45(5): 1453-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24676774

RESUMEN

BACKGROUND AND PURPOSE: Several lines of evidence support the involvement of mannose-binding lectin (MBL) in stroke brain damage. The lectin pathway of the complement system facilitates thrombin activation and clot formation under certain experimental conditions. In the present study, we examine whether MBL promotes thrombosis after ischemia/reperfusion and influences the course and prognosis of ischemic stroke. METHODS: Middle cerebral artery occlusion/reperfusion was performed in MBL-deficient (n=85) and wild-type (WT; n=83) mice, and the brain lesion was assessed by MRI at days 1 and 7. Relative cerebral blood flow was monitored up to 6 hours after middle cerebral artery occlusion with laser speckle contrast imaging. Fibrin(ogen) was analyzed in the brain vasculature and plasma, and the effects of thrombin inhibitor argatroban were evaluated to assess the role of MBL in thrombin activation. RESULTS: Infarct volumes and neurological deficits were smaller in MBL knockout mice than in WT mice. Relative cerebral blood flow values during middle cerebral artery occlusion and at reperfusion were similar in both groups, but decreased during the next 6 hours in the WT group only. Also, the WT mice showed more fibrin(ogen) in brain vessels and a better outcome after argatroban treatment. In contrast, argatroban did not improve the outcome in MBL knockout mice. CONCLUSIONS: MBL promotes brain damage and functional impairment after brain ischemia/reperfusion in mice. These effects are secondary to intravascular thrombosis and impaired relative cerebral blood flow during reperfusion. Argatroban protects WT mice, but not MBL knockout mice, emphasizing a role of MBL in local thrombus formation in acute ischemia/reperfusion.


Asunto(s)
Isquemia Encefálica/fisiopatología , Infarto de la Arteria Cerebral Media/fisiopatología , Lectina de Unión a Manosa/fisiología , Daño por Reperfusión/fisiopatología , Trombosis/etiología , Animales , Antitrombinas/administración & dosificación , Arginina/análogos & derivados , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Circulación Cerebrovascular/genética , Modelos Animales de Enfermedad , Fibrinógeno/antagonistas & inhibidores , Fibrinógeno/metabolismo , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Imagen por Resonancia Magnética , Masculino , Lectina de Unión a Manosa/deficiencia , Lectina de Unión a Manosa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación/genética , Ácidos Pipecólicos/administración & dosificación , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Sulfonamidas , Trombosis/genética
13.
Eur J Nucl Med Mol Imaging ; 40(3): 426-38, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23135322

RESUMEN

PURPOSE: Stroke induces strong expression of the 72-kDa heat-shock protein (HSP-70) in the ischaemic brain, and neuronal expression of HSP-70 is associated with the ischaemic penumbra. The aim of this study was to image induction of Hsp-70 gene expression in vivo after brain ischaemia using reporter mice. METHODS: A genomic DNA sequence of the Hspa1b promoter was used to generate an Hsp70-mPlum far-red fluorescence reporter vector. The construct was tested in cellular systems (NIH3T3 mouse fibroblast cell line) by transient transfection and examining mPlum and Hsp-70 induction under a challenge. After construct validation, mPlum transgenic mice were generated. Focal brain ischaemia was induced by transient intraluminal occlusion of the middle cerebral artery and the mice were imaged in vivo with fluorescence reflectance imaging (FRI) with an intact skull, and with confocal microscopy after opening a cranial window. RESULTS: Cells transfected with the Hsp70-mPlum construct showed mPlum fluorescence after stimulation. One day after induction of ischaemia, reporter mice showed a FRI signal located in the HSP-70-positive zone within the ipsilateral hemisphere, as validated by immunohistochemistry. Live confocal microscopy allowed brain tissue to be visualized at the cellular level. mPlum fluorescence was observed in vivo in the ipsilateral cortex 1 day after induction of ischaemia in neurons, where it is compatible with penumbra and neuronal viability, and in blood vessels in the core of the infarction. CONCLUSION: This study showed in vivo induction of Hsp-70 gene expression in ischaemic brain using reporter mice. The fluorescence signal showed in vivo the induction of Hsp-70 in penumbra neurons and in the vasculature within the ischaemic core.


Asunto(s)
Isquemia Encefálica/diagnóstico , Isquemia Encefálica/genética , Genes Reporteros/genética , Proteínas HSP70 de Choque Térmico/genética , Activación Transcripcional , Animales , Femenino , Proteínas Luminiscentes/genética , Ratones , Ratones Transgénicos , Microscopía Confocal , Células 3T3 NIH , Espectrometría de Fluorescencia , Proteína Fluorescente Roja
14.
Biomedicines ; 10(2)2022 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-35203665

RESUMEN

INTRODUCTION: Polycystic ovary syndrome (PCOS) is an endocrinological disorder that affects 5-15% of women of their reproductive age and is a frequent cause of infertility. Major symptoms include hyperandrogenism, ovulatory dysfunction, and often obesity and/or insulin resistance. PCOS also represents a state of chronic low-grade inflammation that is closely interlinked with the metabolic features. "Classical" pro-inflammatory lipid mediators such as prostaglandins (PG), leukotrienes (LT), or thromboxanes (TX) are derived from arachidonic acid (AA) and are crucial for the initial response. Resolution processes are driven by four families of so-called specialized pro-resolving mediators (SPMs): resolvins, maresins, lipoxins, and protectins. The study aimed to establish lipid mediator profiles of PCOS patients compared to healthy women to identify differences in their resolutive and pro-inflammatory lipid parameters. MATERIAL AND METHODS: Fifteen female patients (18-45 years) were diagnosed with PCOS according to Rotterdam criteria, and five healthy women, as a comparator group, were recruited for the study. The main outcome measures were: pro-inflammatory lipid mediators (PG, LT, TX) and their precursor AA, SPMs (resolvins, maresins, protectins, lipoxins), their precursors EPA, DHA, DPA, and their active biosynthesis pathway intermediates (18-HEPE, 17-HDHA, 14-HDHA). RESULTS: The level of pro-inflammatory parameters in serum was significantly higher in PCOS-affected women. The ratio (sum of pro-inflammatory molecules)/(sum of SPMs plus hydroxylated intermediates) reflecting the inflammatory state was significantly lower in the group of healthy women. CONCLUSION: There is a strong pro-inflammatory state in PCOS patients. Further research will clarify whether supplementation with SPMs or their precursors may improve this state.

15.
J Clin Invest ; 129(12): 5294-5311, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31657786

RESUMEN

Resolution of acute inflammation is an active process orchestrated by endogenous mediators and mechanisms pivotal in host defense and homeostasis. The macrophage mediator in resolving inflammation, maresin 1 (MaR1), is a potent immunoresolvent, stimulating resolution of acute inflammation and organ protection. Using an unbiased screening of greater than 200 GPCRs, we identified MaR1 as a stereoselective activator for human leucine-rich repeat containing G protein-coupled receptor 6 (LGR6), expressed in phagocytes. MaR1 specificity for recombinant human LGR6 activation was established using reporter cells expressing LGR6 and functional impedance sensing. MaR1-specific binding to LGR6 was confirmed using 3H-labeled MaR1. With human and mouse phagocytes, MaR1 (0.01-10 nM) enhanced phagocytosis, efferocytosis, and phosphorylation of a panel of proteins including the ERK and cAMP response element-binding protein. These MaR1 actions were significantly amplified with LGR6 overexpression and diminished by gene silencing in phagocytes. Thus, we provide evidence for MaR1 as an endogenous activator of human LGR6 and a novel role of LGR6 in stimulating MaR1's key proresolving functions of phagocytes.


Asunto(s)
Ácidos Docosahexaenoicos/metabolismo , Fagocitos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Ácidos Docosahexaenoicos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Silenciador del Gen , Células HEK293 , Humanos , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Fagocitosis , Fosforilación , ARN Interferente Pequeño/metabolismo , Células THP-1
17.
J Leukoc Biol ; 2018 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-29377345

RESUMEN

Local production and downstream metabolism of specialized proresolving lipid mediators (SPMs) are pivotal in regulating their biological actions during resolution of inflammation. Resolvin D4 (RvD4: 4S,5R,17S-trihydroxydocosa-6E,8E,10Z,13Z,15E,19Z hexaenoic acid) is one of the more recently elucidated SPMs with complete stereochemistry biosynthesized from docosahexaenoic acid . Here, we report a new multimilligram commercial synthesis that afforded enough material for matching, validation, and further evaluation of RvD4 functions. Using LC-MS-MS profiling, RvD4 was identified at bioactive amounts in human (1 pg/mL) and mouse bone marrow (12 pg/femur and tibia). In mouse bone marrow, ischemia increased the formation of RvD4 > 37-fold (455 pg/femur and tibia). Two separate mouse ischemic injury models were used, where RvD4 reduced second organ reperfusion lung injury > 50%, demonstrating organ protection. Structure-function relationships of RvD4 demonstrated > 40% increase in neutrophil and monocyte phagocytic function in human whole blood in comparison with 2 separate trans-containing double bond isomers that were inactive. These 2 isomers were prepared by organic synthesis: 4S,5R,17S-trihydroxydocosa-6E,8E,10E,13Z,15E,19Z-hexaenoic acid (10-trans-RvD4), a natural isomer, and 4S,5R,17S-trihydroxydocosa-6E,8E,10E,13E,15E,19Z-hexaenoic acid (10,13-trans-RvD4), a rogue isomer. Compared to leukotriene B4 , D-series resolvins (RvD1, RvD2, RvD3, RvD4, or RvD5) did not stimulate human neutrophil chemotaxis monitored via real-time microfluidics chambers. A novel 17-oxo-containing-RvD4 product of eicosanoid oxidoreductase was identified with human bone marrow cells. Comparison of 17-oxo-RvD4 to RvD4 demonstrated that with human leukocytes 17-oxo-RvD4 was inactive. Together, these provide commercial-scale synthesis that permitted a second independent validation of RvD4 complete stereochemical structure as well as evidence for RvD4 regulation in tissues and its stereoselective phagocyte responses.

18.
Nat Commun ; 9(1): 59, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29302056

RESUMEN

Proinflammatory eicosanoids (prostaglandins and leukotrienes) and specialized pro-resolving mediators (SPM) are temporally regulated during infections. Here we show that human macrophage phenotypes biosynthesize unique lipid mediator signatures when exposed to pathogenic bacteria. E. coli and S. aureus each stimulate predominantly proinflammatory 5-lipoxygenase (LOX) and cyclooxygenase pathways (i.e., leukotriene B4 and prostaglandin E2) in M1 macrophages. These pathogens stimulate M2 macrophages to produce SPMs including resolvin D2 (RvD2), RvD5, and maresin-1. E. coli activates M2 macrophages to translocate 5-LOX and 15-LOX-1 to different subcellular locales in a Ca2+-dependent manner. Neither attenuated nor non-pathogenic E. coli mobilize Ca2+ or activate LOXs, rather these bacteria stimulate prostaglandin production. RvD5 is more potent than leukotriene B4 at enhancing macrophage phagocytosis. These results indicate that M1 and M2 macrophages respond to pathogenic bacteria differently, producing either leukotrienes or resolvins that further distinguish inflammatory or pro-resolving phenotypes.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Leucotrieno B4/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Calcio/metabolismo , Humanos , Fagocitosis
20.
J Cereb Blood Flow Metab ; 30(3): 638-52, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19904289

RESUMEN

Oxidative and nitrosative stress are targets for intervention after ischemia/reperfusion. The aim of this study was to explore the effect of CR-6, a vitamin-E analogue that is antioxidant and scavenger of nitrogen-reactive species. Sprague-Dawley rats had the middle cerebral artery (MCA) occluded either for 90 mins or permanently. Cortical perfusion was continuously monitored by laser-Doppler flowmetry. CR-6 (100 mg/kg) was administered orally either at 2 and 8 h after MCA occlusion, or at 2 h only. Infarct volume, neurological deficit, and signs of reperfusion injury were evaluated. CR-6 was detected in plasma and brain by HPLC. CR-6 reduced glutathione consumption in the ischemic brain and superoxide generation in the isolated MCA. CR-6 decreased infarct volume and attenuated the neurological deficit at 1 and 7 days after ischemia/reperfusion, but not after permanent ischemia. Immediately after reperfusion, cortical blood flow values returned to their baseline (+/-20%) in several animals, whereas others showed hyper-perfusion (>20% of baseline). Reactive hyperemia was associated with adverse events such as increased cortical BBB leakage, edema, protein nitrotyrosination, COX-2 expression, and neutrophil accumulation; and with a poorer outcome, and CR-6 attenuated these effects. In conclusion, oral CR-6 administration after transient ischemia protects the brain from reperfusion injury.


Asunto(s)
Antioxidantes/farmacología , Benzopiranos/farmacología , Isquemia Encefálica/tratamiento farmacológico , Fármacos Neuroprotectores , Daño por Reperfusión/prevención & control , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiología , Western Blotting , Edema Encefálico/patología , Edema Encefálico/prevención & control , Isquemia Encefálica/patología , Infarto Cerebral/patología , Circulación Cerebrovascular/fisiología , Ciclooxigenasa 2/metabolismo , Hiperemia/sangre , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/patología , Inflamación/patología , Leucocitos/patología , Masculino , Infiltración Neutrófila , Estrés Oxidativo/efectos de los fármacos , Desempeño Psicomotor/fisiología , ARN/biosíntesis , ARN/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Especies de Nitrógeno Reactivo/metabolismo , Daño por Reperfusión/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA