Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Infect Immun ; 92(5): e0052223, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38629842

RESUMEN

Streptococcus pneumoniae (pneumococcus) remains a serious cause of pulmonary and systemic infections globally, and host-directed therapies are lacking. The aim of this study was to test the therapeutic efficacy of asapiprant, an inhibitor of prostaglandin D2 signaling, against pneumococcal infection. Treatment of young mice with asapiprant after pulmonary infection with invasive pneumococci significantly reduced systemic spread, disease severity, and host death. Protection was specific against bacterial dissemination from the lung to the blood but had no effect on pulmonary bacterial burden. Asapiprant-treated mice had enhanced antimicrobial activity in circulating neutrophils, elevated levels of reactive oxygen species (ROS) in lung macrophages/monocytes, and improved pulmonary barrier integrity indicated by significantly reduced diffusion of fluorescein isothiocyanate (FITC)-dextran from lungs into the circulation. These findings suggest that asapiprant protects the host against pneumococcal dissemination by enhancing the antimicrobial activity of immune cells and maintaining epithelial/endothelial barrier integrity in the lungs.


Asunto(s)
Infecciones Neumocócicas , Animales , Femenino , Ratones , Modelos Animales de Enfermedad , Pulmón/microbiología , Pulmón/patología , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Neutrófilos/efectos de los fármacos , Infecciones Neumocócicas/tratamiento farmacológico , Infecciones Neumocócicas/microbiología , Especies Reactivas de Oxígeno/metabolismo , Streptococcus pneumoniae/efectos de los fármacos
2.
Bioorg Med Chem Lett ; 102: 129675, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38417632

RESUMEN

NLRP3 is an intracellular sensor protein that detects a broad range of danger signals and environmental insults. Its activation results in a protective pro-inflammatory response designed to impair pathogens and repair tissue damage via the formation of the NLRP3 inflammasome. Assembly of the NLRP3 inflammasome leads to caspase 1-dependent secretory release of the pro-inflammatory cytokines IL-1ß and IL-18 as well as to gasdermin d-mediated pyroptotic cell death. Herein, we describe the discovery of a novel indazole series of high affinity, reversible inhibitors of NLRP3 activation through screening of DNA-encoded libraries and the potent lead compound 3 (BAL-0028, IC50 = 25 nM) that was identified directly from the screen. SPR studies showed that compound 3 binds tightly (KD range 104-123 nM) to the NACHT domain of NLRP3. A CADD analysis of the interaction of compound 3 with the NLRP3 NACHT domain proposes a binding site that is distinct from those of ADP and MCC950 and includes specific site interactions. We anticipate that compound 3 (BAL-0028) and other members of this novel indazole class of neutral inhibitors will demonstrate significantly different physical, biochemical, and biological properties compared to NLRP3 inhibitors previously identified.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamasomas/metabolismo , Sulfonamidas , Citocinas/metabolismo , Interleucina-1beta/metabolismo , Caspasa 1 , ADN
3.
J Neuroinflammation ; 19(1): 118, 2022 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-35610647

RESUMEN

BACKGROUND: The transient receptor potential vanilloid 1 (TRPV1) participates in thermosensation and inflammatory pain, but its immunomodulatory mechanisms remain enigmatic. N-Oleoyl dopamine (OLDA), an endovanilloid and endocannabinoid, is a TRPV1 agonist that is produced in the central nervous system and the peripheral nervous system. We studied the anti-inflammatory effects and TRPV1-dependent mechanisms of OLDA in models of inflammation and sepsis. METHODS: Mice were challenged intratracheally or intravenously with LPS, or intratracheally with S. aureus to induce pneumonia and sepsis, and then were treated intravenously with OLDA. Endpoints included plasma cytokines, leukocyte activation marker expression, mouse sepsis scores, lung histopathology, and bacterial counts. The role of TRPV1 in the effects of OLDA was determined using Trpv1-/- mice, and mice with TRPV1 knockdown pan-neuronally, in peripheral nervous system neurons, or in myeloid cells. Circulating monocytes/macrophages were depleted using clodronate to determine their role in the anti-inflammatory effects of OLDA in endotoxemic mice. Levels of exogenous OLDA, and of endovanilloids and endocannabinoids, at baseline and in endotoxemic mice, were determined by LC-MS/MS. RESULTS: OLDA administration caused an early anti-inflammatory response in endotoxemic and septic mice with high serum levels of IL-10 and decreased levels of pro-inflammatory cytokines. OLDA also reduced lung injury and improved mouse sepsis scores. Blood and lung bacterial counts were comparable between OLDA- and carrier-treated mice with S. aureus pneumonia. OLDA's effects were reversed in mice with pan-neuronal TRPV1 knockdown, but not with TRPV1 knockdown in peripheral nervous system neurons or myeloid cells. Depletion of monocytes/macrophages reversed the IL-10 upregulation by OLDA in endotoxemic mice. Brain and blood levels of endovanilloids and endocannabinoids were increased in endotoxemic mice. CONCLUSIONS: OLDA has strong anti-inflammatory actions in mice with endotoxemia or S. aureus pneumonia. Prior studies focused on the role of peripheral nervous system TRPV1 in modulating inflammation and pneumonia. Our results suggest that TRPV1-expressing central nervous system neurons also regulate inflammatory responses to endotoxemia and infection. Our study reveals a neuro-immune reflex that during acute inflammation is engaged proximally by OLDA acting on neuronal TRPV1, and through a multicellular network that requires circulating monocytes/macrophages, leads to the systemic production of IL-10.


Asunto(s)
Endotoxemia , Sepsis , Animales , Sistema Nervioso Central/metabolismo , Cromatografía Liquida , Citocinas/metabolismo , Dopamina/metabolismo , Endocannabinoides , Endotoxemia/inducido químicamente , Endotoxemia/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-10/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Sepsis/tratamiento farmacológico , Staphylococcus aureus , Canales Catiónicos TRPV/metabolismo , Espectrometría de Masas en Tándem
4.
Nature ; 605(7908): 146-151, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35314834

RESUMEN

Coronavirus disease 2019 (COVID-19) is especially severe in aged populations1. Vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are highly effective, but vaccine efficacy is partly compromised by the emergence of SARS-CoV-2 variants with enhanced transmissibility2. The emergence of these variants emphasizes the need for further development of anti-SARS-CoV-2 therapies, especially for aged populations. Here we describe the isolation of highly virulent mouse-adapted viruses and use them to test a new therapeutic drug in infected aged animals. Many of the alterations observed in SARS-CoV-2 during mouse adaptation (positions 417, 484, 493, 498 and 501 of the spike protein) also arise in humans in variants of concern2. Their appearance during mouse adaptation indicates that immune pressure is not required for selection. For murine SARS, for which severity is also age dependent, elevated levels of an eicosanoid (prostaglandin D2 (PGD2)) and a phospholipase (phospholipase A2 group 2D (PLA2G2D)) contributed to poor outcomes in aged mice3,4. mRNA expression of PLA2G2D and prostaglandin D2 receptor (PTGDR), and production of PGD2 also increase with ageing and after SARS-CoV-2 infection in dendritic cells derived from human peripheral blood mononuclear cells. Using our mouse-adapted SARS-CoV-2, we show that middle-aged mice lacking expression of PTGDR or PLA2G2D are protected from severe disease. Furthermore, treatment with a PTGDR antagonist, asapiprant, protected aged mice from lethal infection. PTGDR antagonism is one of the first interventions in SARS-CoV-2-infected animals that specifically protects aged animals, suggesting that the PLA2G2D-PGD2/PTGDR pathway is a useful target for therapeutic interventions.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Eicosanoides , Leucocitos Mononucleares , Ratones , Compuestos Orgánicos , Oxazoles , Piperazinas , Poliésteres , Prostaglandinas , Glicoproteína de la Espiga del Coronavirus , Sulfonamidas
5.
bioRxiv ; 2021 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-33907749

RESUMEN

Coronavirus disease 2019 (COVID-19) is especially severe in aged populations1. Resolution of the COVID-19 pandemic has been advanced by the recent development of SARS-CoV-2 vaccines, but vaccine efficacy is partly compromised by the recent emergence of SARS-CoV-2 variants with enhanced transmissibility2. The emergence of these variants emphasizes the need for further development of anti-SARS-CoV-2 therapies, especially in aged populations. Here, we describe the isolation of a new set of highly virulent mouse-adapted viruses and use them to test a novel therapeutic drug useful in infections of aged animals. Initially, we show that many of the mutations observed in SARS-CoV-2 during mouse adaptation (at positions 417, 484, 501 of the spike protein) also arise in humans in variants of concern (VOC)2. Their appearance during mouse adaptation indicates that immune pressure is not required for their selection. Similar to the human infection, aged mice infected with mouse-adapted SARS-CoV-2 develop more severe disease than young mice. In murine SARS, in which severity is also age-dependent, we showed that elevated levels of an eicosanoid, prostaglandin D2 (PGD2) and of a phospholipase, PLA2G2D, contributed to poor outcomes in aged mice3,4. Using our virulent mouse-adapted SARS-CoV-2, we show that infection of middle-aged mice lacking expression of DP1, a PGD2 receptor, or PLA2G2D are protected from severe disease. Further, treatment with a DP1 antagonist, asapiprant, protected aged mice from a lethal infection. DP1 antagonism is one of the first interventions in SARS-CoV-2-infected animals that specifically protects aged animals, and demonstrates that the PLA2G2D-PGD2/DP1 pathway is a useful target for therapeutic interventions.

6.
Sci Rep ; 11(1): 8675, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33883672

RESUMEN

In epithelial cancers, the epidermal growth factor receptor (EGFR) and integrin α6ß4 are frequently overexpressed and found to synergistically activate intracellular signaling pathways that promote cell proliferation and migration. In cancer cells, the ß4 subunit is phosphorylated at tyrosine residues not normally recognized as kinase substrates; however, the function of these phosphotyrosine residues in cancer cells is a subject of much debate. In EGFR-overexpressing carcinoma cells, we found that the Src family kinase (SFK) inhibitor PP2 reduces ß4 tyrosine phosphorylation following the activation of EGFR. However, siRNA mediated knockdown of the SFKs Src, Fyn, Yes and Lyn, individually or in combination, did not affect the EGF-induced phosphorylation of ß4. Using phospho-peptide affinity chromatography and mass spectrometry, we found that PLCγ1 binds ß4 at the phosphorylated residues Y1422/Y1440, but were unable to verify this interaction in A431 carcinoma cells that overexpress the EGFR. Furthermore, using A431 cells devoid of ß4 or reconstituted with phenylalanine specific mutants of ß4, the activation of several downstream signaling pathways, including PLCγ/PKC, MAPK and PI3K/Akt, were not substantially affected. We conclude that tyrosine-phosphorylated ß4 does not enhance EGFR-mediated signaling in EGFR-overexpressing cells, despite the fact that this integrin subunit is highly tyrosine phosphorylated in these cells.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Integrina beta4/metabolismo , Neoplasias Cutáneas/metabolismo , Tirosina/metabolismo , Animales , Línea Celular Tumoral , Humanos , Integrina beta4/fisiología , Espectrometría de Masas , Fosforilación , Fosfotirosina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Neoplasias Cutáneas/genética
7.
Shock ; 55(3): 349-356, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32826812

RESUMEN

ABSTRACT: Endothelial cells play a major role in inflammatory responses to infection and sterile injury. Endothelial cells express Toll-like receptor 4 (TLR4) and are activated by LPS to express inflammatory cytokines/chemokines, and to undergo functional changes, including increased permeability. The extracellular signal-regulated kinase 1/2 (ERK1/2) mediates pro-inflammatory signaling in monocytes and macrophages, but the role of ERK1/2 in LPS-induced activation of microvascular endothelial cells has not been defined. We therefore studied the role of ERK1/2 in LPS-induced inflammatory activation and permeability of primary human lung microvascular endothelial cells (HMVEC). Inhibition of ERK1/2 augmented LPS-induced IL-6 and vascular cell adhesion protein (VCAM-1) production by HMVEC. ERK1/2 siRNA knockdown also augmented IL-6 production by LPS-treated HMVEC. Conversely, ERK1/2 inhibition abrogated permeability and restored cell-cell junctions of LPS-treated HMVEC. Consistent with the previously described pro-inflammatory role for ERK1/2 in leukocytes, inhibition of ERK1/2 reduced LPS-induced cytokine/chemokine production by primary human monocytes. Our study identifies a complex role for ERK1/2 in TLR4-activation of HMVEC, independent of myeloid differentiation primary response gene (MyD88) and TIR domain-containing adaptor inducing IFN-ß (TRIF) signaling pathways. The activation of ERK1/2 limits LPS-induced IL-6 production by HMVEC, while at the same time promoting HMVEC permeability. Conversely, ERK1/2 activation promotes IL-6 production by human monocytes. Our results suggest that ERK1/2 may play an important role in the nuanced regulation of endothelial cell inflammation and vascular permeability in sepsis and injury.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Citocinas/biosíntesis , Células Endoteliales/fisiología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Células Cultivadas , Células Endoteliales/metabolismo , Femenino , Humanos , Lipopolisacáridos/administración & dosificación , Masculino
8.
J Immunol ; 199(4): 1465-1475, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28701511

RESUMEN

N-Arachidonoyl dopamine (NADA) is an endogenous lipid that potently activates the transient receptor potential vanilloid 1 (TRPV1), which mediates pain and thermosensation. NADA is also an agonist of cannabinoid receptors 1 and 2. We have reported that NADA reduces the activation of cultured human endothelial cells by LPS and TNF-α. Thus far, in vivo studies using NADA have focused on its neurologic and behavioral roles. In this article, we show that NADA potently decreases in vivo systemic inflammatory responses and levels of the coagulation intermediary plasminogen activator inhibitor 1 in three mouse models of inflammation: LPS, bacterial lipopeptide, and polymicrobial intra-abdominal sepsis. We also found that the administration of NADA increases survival in endotoxemic mice. Additionally, NADA reduces blood levels of the neuropeptide calcitonin gene-related peptide but increases the neuropeptide substance P in LPS-treated mice. We demonstrate that the anti-inflammatory effects of NADA are mediated by TRPV1 expressed by nonhematopoietic cells and provide data suggesting that neuronal TRPV1 may mediate NADA's anti-inflammatory effects. These results indicate that NADA has novel TRPV1-dependent anti-inflammatory properties and suggest that the endovanilloid system might be targeted therapeutically in acute inflammation.


Asunto(s)
Ácidos Araquidónicos/farmacología , Dopamina/análogos & derivados , Inflamación/metabolismo , Canales Catiónicos TRPV/metabolismo , Enfermedad Aguda , Animales , Ácidos Araquidónicos/metabolismo , Péptido Relacionado con Gen de Calcitonina/sangre , Modelos Animales de Enfermedad , Dopamina/metabolismo , Dopamina/farmacología , Inflamación/inmunología , Lipopéptidos/inmunología , Lipopolisacáridos/inmunología , Ratones , Inhibidor 1 de Activador Plasminogénico/metabolismo , Sepsis/metabolismo , Sustancia P/sangre
9.
Innate Immun ; 21(8): 827-46, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26403174

RESUMEN

The endothelium forms a vast network that dynamically regulates vascular barrier function, coagulation pathways and vasomotor tone. Microvascular endothelial cells are uniquely situated to play key roles during infection and injury, owing to their widespread distribution throughout the body and their constant interaction with circulating blood. While not viewed as classical immune cells, endothelial cells express innate immune receptors, including the Toll-like receptors (TLRs), which activate intracellular inflammatory pathways mediated through NF-κB and the MAP kinases. TLR agonists, including LPS and bacterial lipopeptides, directly upregulate microvascular endothelial cell expression of inflammatory mediators. Intriguingly, TLR activation also modulates microvascular endothelial cell permeability and the expression of coagulation pathway intermediaries. Microvascular thrombi have been hypothesized to trap microorganisms thereby limiting the spread of infection. However, dysregulated activation of endothelial inflammatory pathways is also believed to lead to coagulopathy and increased vascular permeability, which together promote sepsis-induced organ failure. This article reviews vascular endothelial cell innate immune pathways mediated through the TLRs as they pertain to sepsis, highlighting links between TLRs and coagulation and permeability pathways, and their role in healthy and pathologic responses to infection and sepsis.


Asunto(s)
Endotelio Vascular/inmunología , Inmunidad Innata/fisiología , Sepsis/inmunología , Receptores Toll-Like/inmunología , Coagulación Sanguínea , Permeabilidad Capilar , Endotelio Vascular/metabolismo , Humanos , Sepsis/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo
10.
Sci Signal ; 8(391): ra86, 2015 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-26307013

RESUMEN

Inflammatory critical illness is a syndrome that is characterized by acute inflammation and organ injury, and it is triggered by infections and noninfectious tissue injury, both of which activate innate immune receptors and pathways. Although reports suggest an anti-inflammatory role for the mitogen-activated protein kinase (MAPK) extracellular signal-regulated kinase 5 (ERK5), we previously found that ERK5 mediates proinflammatory responses in primary human cells in response to stimulation of Toll-like receptor 2 (TLR2). We inhibited the kinase activities and reduced the abundances of ERK5 and MEK5, a MAPK kinase directly upstream of ERK5, in primary human vascular endothelial cells and monocytes, and found that ERK5 promoted inflammation induced by a broad range of microbial TLR agonists and by the proinflammatory cytokines interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α). Furthermore, we found that inhibitors of MEK5 or ERK5 reduced the plasma concentrations of proinflammatory cytokines in mice challenged with TLR ligands or heat-killed Staphylococcus aureus, as well as in mice that underwent sterile lung ischemia-reperfusion injury. Finally, we found that inhibition of ERK5 protected endotoxemic mice from death. Together, our studies support a proinflammatory role for ERK5 in primary human endothelial cells and monocytes, and suggest that ERK5 is a potential therapeutic target in diverse disorders that cause inflammatory critical illness.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/inmunología , Proteína Quinasa 7 Activada por Mitógenos/inmunología , Monocitos/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Animales , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Interleucina-1beta/inmunología , Masculino , Ratones , Monocitos/patología , Síndrome de Respuesta Inflamatoria Sistémica/patología , Síndrome de Respuesta Inflamatoria Sistémica/terapia , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/inmunología , Factor de Necrosis Tumoral alfa/inmunología
11.
J Leukoc Biol ; 98(6): 983-94, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26216934

RESUMEN

Intravenous Ig is used to treat autoimmune or autoinflammatory disorders, but the mechanism by which it exerts its immunosuppressive activity is not understood completely. To examine the impact of intravenous Ig on macrophages, we compared cytokine production by LPS-activated macrophages in the presence and absence of intravenous Ig. Intravenous Ig treatment induced robust production of IL-10 in response to LPS, relative to LPS stimulation alone, and reduced production of proinflammatory cytokines. This anti-inflammatory, intravenous Ig-induced activation was sustained for 24 h but could only be induced if intravenous Ig were provided within 1 h of LPS stimulation. Intravenous Ig activation led to enhanced and prolonged activation of MAPKs, Erk1/2, p38, and Erk5, and inhibition of each reduced intravenous Ig-induced IL-10 production and suppression of IL-12/23p40. IL-10 production occurred rapidly in response to intravenous Ig + LPS and was sufficient to reduce proinflammatory IL-12/23p40 production in response to LPS. IL-10 induction and reduced IL-12/23p40 production were transcriptionally regulated. IL-10 played a direct role in reducing proinflammatory cytokine production by macrophages treated with intravenous Ig + LPS, as macrophages from mice deficient in the IL-10R ß chain or in IL-10 were compromised in their ability to reduce proinflammatory cytokine production. Finally, intraperitoneal injection of intravenous Ig or intravenous Ig + LPS into mice activated macrophages to produce high levels of IL-10 during subsequent or concurrent LPS challenge, respectively. These findings identify IL-10 as a key anti-inflammatory mediator produced by intravenous Ig-treated macrophages and provide insight into a novel mechanism by which intravenous Ig may dampen down inflammatory responses in patients with autoimmune or autoinflammatory diseases.


Asunto(s)
Inmunoglobulinas Intravenosas/farmacología , Interleucina-10/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Animales , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Humanos , Subunidad p40 de la Interleucina-12/inmunología , Lipopolisacáridos/toxicidad , Ratones , Ratones Noqueados , Receptores de Interleucina-10/inmunología
12.
J Biol Chem ; 289(19): 13079-100, 2014 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-24644287

RESUMEN

Although cannabinoids, such as Δ(9)-tetrahydrocannabinol, have been studied extensively for their psychoactive effects, it has become apparent that certain cannabinoids possess immunomodulatory activity. Endothelial cells (ECs) are centrally involved in the pathogenesis of organ injury in acute inflammatory disorders, such as sepsis, because they express cytokines and chemokines, which facilitate the trafficking of leukocytes to organs, and they modulate vascular barrier function. In this study, we find that primary human ECs from multiple organs express the cannabinoid receptors CB1R, GPR18, and GPR55, as well as the ion channel transient receptor potential cation channel vanilloid type 1. In contrast to leukocytes, CB2R is only minimally expressed in some EC populations. Furthermore, we show that ECs express all of the known endocannabinoid (eCB) metabolic enzymes. Examining a panel of cannabinoids, we demonstrate that the synthetic cannabinoid WIN55,212-2 and the eCB N-arachidonoyl dopamine (NADA), but neither anandamide nor 2-arachidonoylglycerol, reduce EC inflammatory responses induced by bacterial lipopeptide, LPS, and TNFα. We find that endothelial CB1R/CB2R are necessary for the effects of NADA, but not those of WIN55,212-2. Furthermore, transient receptor potential cation channel vanilloid type 1 appears to counter the anti-inflammatory properties of WIN55,212-2 and NADA, but conversely, in the absence of these cannabinoids, its inhibition exacerbates the inflammatory response in ECs activated with LPS. These data indicate that the eCB system can modulate inflammatory activation of the endothelium and may have important implications for a variety of acute inflammatory disorders that are characterized by EC activation.


Asunto(s)
Analgésicos/efectos adversos , Ácidos Araquidónicos/efectos adversos , Benzoxazinas/efectos adversos , Cannabinoides/efectos adversos , Dopamina/análogos & derivados , Morfolinas/efectos adversos , Naftalenos/efectos adversos , Analgésicos/farmacología , Ácidos Araquidónicos/farmacología , Proteínas Bacterianas/toxicidad , Benzoxazinas/farmacología , Cannabinoides/farmacología , Dopamina/efectos adversos , Dopamina/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Lipopéptidos/toxicidad , Morfolinas/farmacología , Naftalenos/farmacología , Receptores de Cannabinoides/metabolismo , Canales Catiónicos TRPV/metabolismo
13.
Neurosci Lett ; 562: 13-8, 2014 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-24406149

RESUMEN

This study aimed to investigate whether the regulation of 5-hydroxytryptamine-7 (5-HT7) receptors in the bilateral basolateral amygdala (BLA) could alter the amnesic effects of sevoflurane and change the hippocampal expression of Arc and neural apoptosis. Male Sprague-Dawley rats were randomized into ten groups. First, the animals received bilateral injection of SB269970 (20, 50, or 100 pmol/0.2 µl) or saline (0.2 µl) or AS-19 (2, 10, or 50 pmol/0.2 µl), followed by inhalation of 2% sevoflurane or air for 2h. Then, fear conditioning training was carried out, and the percentage of freezing was detected 24h later. Furthermore, hippocampal Arc protein level and neural apoptosis were measured. Pre-training inhalation of sevoflurane reduced the extent of freezing, and hippocampal Arc expression. The largest dose of SB269970 (100 pmol) could block sevoflurane-induced amnesia and reverse the inhibitive effect of sevoflurane on Arc expression, while the maximal dose of AS-19 could exacerbate the amnesic effect, and further inhibit Arc expression. Furthermore, pre-training inhalation of 2% sevoflurane for 6h could not induce neural apoptosis in the hippocampus. The amnesic effect of sevoflurane might partly attribute to its impairment of memory formation in the hippocampus via activation of 5-HT7 receptors in the BLA.


Asunto(s)
Amnesia/inducido químicamente , Amígdala del Cerebelo/efectos de los fármacos , Anestésicos por Inhalación/farmacología , Proteínas del Citoesqueleto/metabolismo , Hipocampo/efectos de los fármacos , Éteres Metílicos/farmacología , Proteínas del Tejido Nervioso/metabolismo , Receptores de Serotonina/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Hipocampo/metabolismo , Masculino , Memoria/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Sevoflurano , Factores de Tiempo
14.
J Vis Exp ; (78): e50677, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23995778

RESUMEN

The vascular endothelium plays an integral part in the inflammatory response. During the acute phase of inflammation, endothelial cells (ECs) are activated by host mediators or directly by conserved microbial components or host-derived danger molecules. Activated ECs express cytokines, chemokines and adhesion molecules that mobilize, activate and retain leukocytes at the site of infection or injury. Neutrophils are the first leukocytes to arrive, and adhere to the endothelium through a variety of adhesion molecules present on the surfaces of both cells. The main functions of neutrophils are to directly eliminate microbial threats, promote the recruitment of other leukocytes through the release of additional factors, and initiate wound repair. Therefore, their recruitment and attachment to the endothelium is a critical step in the initiation of the inflammatory response. In this report, we describe an in vitro neutrophil adhesion assay using calcein AM-labeled primary human neutrophils to quantitate the extent of microvascular endothelial cell activation under static conditions. This method has the additional advantage that the same samples quantitated by fluorescence spectrophotometry can also be visualized directly using fluorescence microscopy for a more qualitative assessment of neutrophil binding.


Asunto(s)
Adhesión Celular/fisiología , Comunicación Celular/fisiología , Células Endoteliales/citología , Microscopía Fluorescente/métodos , Neutrófilos/citología , Humanos
15.
Anesthesiology ; 117(4): 822-35, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22890118

RESUMEN

BACKGROUND: Ischemia-reperfusion (I-R) injury is a sterile inflammatory process that is commonly associated with diverse clinical situations such as hemorrhage followed by resuscitation, transient embolic events, and organ transplantation. I-R injury can induce lung dysfunction whether the I-R occurs in the lung or in a remote organ. Recently, evidence has emerged that receptors and pathways of the innate immune system are involved in recognizing sterile inflammation and overlap considerably with those involved in the recognition of and response to pathogens. METHODS: The authors used a mouse surgical model of transient unilateral left pulmonary artery occlusion without bronchial involvement to create ventilated lung I-R injury. In addition, they mimicked nutritional I-R injury in vitro by transiently depriving cells of all nutrients. RESULTS: Compared with sham-operated mice, mice subjected to ventilated lung I-R injury had up-regulated lung expression of inflammatory mediator messenger RNA for interleukin-1ß, interleukin-6, and chemokine (C-X-C motif) ligand-1 and -2, paralleled by histologic evidence of lung neutrophil recruitment and increased plasma concentrations of interleukin-1ß, interleukin-6, and high-mobility group protein B1 proteins. This inflammatory response to I-R required toll-like receptor-4 (TLR4). In addition, the authors demonstrated in vitro cooperativity and cross-talk between human macrophages and endothelial cells, resulting in augmented inflammatory responses to I-R. Remarkably, the authors found that selective depletion of alveolar macrophages rendered mice resistant to ventilated lung I-R injury. CONCLUSIONS: The data reveal that alveolar macrophages and the pattern recognition receptor toll-like receptor-4 are involved in the generation of the early inflammatory response to lung I-R injury.


Asunto(s)
Lesión Pulmonar Aguda/patología , Macrófagos Alveolares/fisiología , Daño por Reperfusión/patología , Receptor Toll-Like 4/fisiología , Lesión Pulmonar Aguda/etiología , Analgésicos no Narcóticos/administración & dosificación , Analgésicos no Narcóticos/farmacología , Animales , Antígenos CD11/genética , Antígenos CD11/fisiología , Línea Celular , Ácido Clodrónico/administración & dosificación , Ácido Clodrónico/farmacología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Liposomas , Pulmón/patología , Ratones , Ratones Noqueados , Infiltración Neutrófila , Estado Nutricional , Atelectasia Pulmonar/patología , Circulación Pulmonar/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Respiración Artificial , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/fisiología
16.
J Biol Chem ; 287(32): 26478-94, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22707717

RESUMEN

Endothelial cell (EC) Toll-like receptor 2 (TLR2) activation up-regulates the expression of inflammatory mediators and of TLR2 itself and modulates important endothelial functions, including coagulation and permeability. We defined TLR2 signaling pathways in EC and tested the hypothesis that TLR2 signaling differs in EC and monocytes. We found that ERK5, heretofore unrecognized as mediating TLR2 activation in any cell type, is a central mediator of TLR2-dependent inflammatory signaling in human umbilical vein endothelial cells, primary human lung microvascular EC, and human monocytes. Additionally, we observed that, although MEK1 negatively regulates TLR2 signaling in EC, MEK1 promotes TLR2 signaling in monocytes. We also noted that activation of TLR2 led to the up-regulation of intracellularly expressed TLR2 and inflammatory mediators via NF-κB, JNK, and p38-MAPK. Finally, we found that p38-MAPK, JNK, ERK5, and NF-κB promote the attachment of human neutrophils to lung microvascular EC that were pretreated with TLR2 agonists. This study newly identifies ERK5 as a key regulator of TLR2 signaling in EC and monocytes and indicates that there are fundamental differences in TLR signaling pathways between EC and monocytes.


Asunto(s)
Endotelio Vascular/citología , MAP Quinasa Quinasa 1/fisiología , Proteína Quinasa 7 Activada por Mitógenos/fisiología , Monocitos/citología , Receptor Toll-Like 2/fisiología , Adhesión Celular , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , FN-kappa B/metabolismo , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Receptor Toll-Like 2/metabolismo , Regulación hacia Arriba
17.
Int Immunopharmacol ; 13(4): 440-5, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22575870

RESUMEN

BACKGROUND: Many studies have shown that ghrelin can down-regulate inflammatory cytokine expression via the inhibition of NF-κB activity and therefore, its administration to septic patients is considered beneficial. However, our knowledge of ghrelin's effects on the upstream activators of the NF-κB pathway, such as NOD2, is still limited. This study aimed to investigate the possible involvement of the NOD2 signaling pathway in the anti-inflammatory effects of ghrelin. METHODS: Twenty-four male SD rats received cecal ligation and puncture (CLP) or sham operation, followed by infusion of saline or ghrelin. The lungs were harvested 6h after CLP or sham operation and analyzed for lung histopathology, neutrophil infiltration, inflammatory cytokines (TNF-α, and IL-6), NOD2 mRNA expression, and activation of NF-κB. Furthermore, survival was recorded for ten days in additional groups of rats. RESULTS: Compared with sham group, neutrophil infiltration, TNF-α and IL-6 levels, NOD2 mRNA expression, as well as NF-κB activation in lungs from rats undergoing CLP were significantly increased. After the administration of ghrelin, all inflammatory parameters analyzed were lower than those without ghrelin following CLP. In addition, ghrelin improved survival after CLP. CONCLUSION: Our results indicate that in a CLP model of sepsis, the beneficial effects that ghrelin has on inflammatory outcomes are mediated at least in part through inhibition of NOD2 expression upstream of NF-κB.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios no Esteroideos/administración & dosificación , Ghrelina/administración & dosificación , Proteína Adaptadora de Señalización NOD2/biosíntesis , Lesión Pulmonar Aguda/inmunología , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Ciego , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Ghrelina/efectos adversos , Humanos , Ligadura , Masculino , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD2/genética , Punciones , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Endogámicas , Activación Transcripcional/efectos de los fármacos
18.
Mol Biol Cell ; 23(8): 1475-85, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22357621

RESUMEN

During wound healing, hemidesmome disassembly enables keratinocyte migration and proliferation. Hemidesmosome dynamics are altered downstream of epidermal growth factor (EGF) receptor activation, following the phosphorylation of integrin ß4 residues S1356 and S1364, which reduces the interaction with plectin; however, this event is insufficient to drive complete hemidesmome disassembly. In the studies reported here, we used a fluorescence resonance energy transfer-based assay to demonstrate that the connecting segment and carboxy-terminal tail of the ß4 cytoplasmic domain interact, which facilitates the formation of a binding platform for plectin. In addition, analysis of a ß4 mutant containing a phosphomimicking aspartic acid residue at T1736 in the C-tail suggests that phosphorylation of this residue regulates the interaction with the plectin plakin domain. The aspartic acid mutation of ß4 T1736 impaired hemidesmosome formation in junctional epidermolysis associated with pyloric atresia/ß4 keratinocytes. Furthermore, we show that T1736 is phosphorylated downstream of protein kinase C and EGF receptor activation and is a substrate for protein kinase D1 in vitro and in cells, which requires its translocation to the plasma membrane and subsequent activation. In conclusion, we identify T1736 as a novel phosphorylation site that contributes to the regulation of hemidesmome disassembly, a dynamically regulated process involving the concerted phosphorylation of multiple ß4 residues.


Asunto(s)
Hemidesmosomas/metabolismo , Integrina beta4/metabolismo , Queratinocitos/metabolismo , Animales , Células COS , Línea Celular , Membrana Celular/metabolismo , Movimiento Celular , Proliferación Celular , Chlorocebus aethiops , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Hemidesmosomas/ultraestructura , Humanos , Integrina beta4/genética , Mutación , Fosforilación , Plectina/metabolismo , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Treonina/metabolismo
19.
Innate Immun ; 18(4): 602-16, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22186927

RESUMEN

The vascular endothelium is integrally involved in the host response to infection and in organ failure during acute inflammatory disorders such as sepsis. Gram-negative and Gram-positive bacterial lipoproteins circulate in sepsis and can directly activate the endothelium by binding to endothelial cell (EC) TLR2. In this report, we perform the most comprehensive analysis to date of the immune-related genes regulated after activation of endothelial TLR2 by bacterial di- and triacylated lipopeptides. We found that TLR2 activation specifically induces the expression of the genes IL-6, IL-8, CSF2, CSF3, ICAM1 and SELE by human umbilical vein ECs and human lung microvascular ECs. These proteins participate in neutrophil recruitment, adherence and activation at sites of inflammation. Significantly, our studies demonstrate that TLR2-mediated EC responses are specifically geared towards recruitment, activation, and survival of neutrophils and not mononuclear leukocytes, that ECs do not require priming by other inflammatory stimuli to respond to bacterial lipopeptides and, unlike mononuclear leukocytes, TLR2 agonists do not induce ECs to secrete TNF-α. This study suggests that endothelial TLR2 may be an important regulator of neutrophil trafficking to sites of infection in general, and that direct activation of lung endothelial TLR2 may contribute to acute lung injury during sepsis.


Asunto(s)
Endotelio Vascular/inmunología , Activación Neutrófila , Neutrófilos/inmunología , Sepsis/inmunología , Receptor Toll-Like 2/metabolismo , Proteínas Bacterianas/farmacología , Línea Celular , Movimiento Celular/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Selectina E/genética , Selectina E/metabolismo , Endotelio Vascular/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/genética , Factor Estimulante de Colonias de Granulocitos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Lipoproteínas/farmacología , Activación Neutrófila/efectos de los fármacos , ARN Interferente Pequeño/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Regulación hacia Arriba
20.
J Immunol ; 186(2): 1119-30, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21169547

RESUMEN

TLR2 activation induces cellular and organ inflammation and affects lung function. Because deranged endothelial function and coagulation pathways contribute to sepsis-induced organ failure, we studied the effects of bacterial lipoprotein TLR2 agonists, including peptidoglycan-associated lipoprotein, Pam3Cys, and murein lipoprotein, on endothelial function and coagulation pathways in vitro and in vivo. TLR2 agonist treatment induced diverse human endothelial cells to produce IL-6 and IL-8 and to express E-selectin on their surface, including HUVEC, human lung microvascular endothelial cells, and human coronary artery endothelial cells. Treatment of HUVEC with TLR2 agonists caused increased monolayer permeability and had multiple coagulation effects, including increased production of plasminogen activator inhibitor-1 (PAI-1) and tissue factor, as well as decreased production of tissue plasminogen activator and tissue factor pathway inhibitor. TLR2 agonist treatment also increased HUVEC expression of TLR2 itself. Peptidoglycan-associated lipoprotein induced IL-6 production by endothelial cells from wild-type mice but not from TLR2 knockout mice, indicating TLR2 specificity. Mice were challenged with TLR2 agonists, and lungs and plasmas were assessed for markers of leukocyte trafficking and coagulopathy. Wild-type mice, but not TLR2 mice, that were challenged i.v. with TLR2 agonists had increased lung levels of myeloperoxidase and mRNAs for E-selectin, P-selectin, and MCP-1, and they had increased plasma PAI-1 and E-selectin levels. Intratracheally administered TLR2 agonist caused increased lung fibrin levels. These studies show that TLR2 activation by bacterial lipoproteins broadly affects endothelial function and coagulation pathways, suggesting that TLR2 activation contributes in multiple ways to endothelial activation, coagulopathy, and vascular leakage in sepsis.


Asunto(s)
Anticoagulantes/fisiología , Coagulación Sanguínea/inmunología , Endotelio Vascular/fisiología , Proteínas de Escherichia coli/fisiología , Lipoproteínas/fisiología , Peptidoglicano/farmacología , Transducción de Señal/inmunología , Receptor Toll-Like 2/agonistas , Animales , Anticoagulantes/agonistas , Anticoagulantes/farmacología , Permeabilidad Capilar/inmunología , Línea Celular , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Proteínas de Escherichia coli/agonistas , Humanos , Inmunofenotipificación , Lipoproteínas/agonistas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/fisiología , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...