Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Mol Med ; 28(1): 148, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36494621

RESUMEN

BACKGROUND: Autoinflammatory diseases, a diverse group of inherited conditions characterized by excessive innate immune activation, have limited therapeutic options. Neuroimmune circuits of the inflammatory reflex control innate immune overactivation and can be stimulated to treat disease using the acetylcholinesterase inhibitor galantamine. METHODS: We tested the efficacy of galantamine in a rodent model of the prototypical autoinflammatory disease familial Mediterranean fever (FMF). Multiple chronic disease markers were evaluated in animals that received long-term galantamine treatment compared to vehicle. RESULTS: Long-term treatment with galantamine attenuated the associated splenomegaly and anemia which are characteristic features of this disease. Further, treatment reduced inflammatory cell infiltration into affected organs and a subcutaneous air pouch. CONCLUSIONS: These findings suggest that galantamine attenuates chronic inflammation in this mouse model of FMF. Further research is warranted to explore the therapeutic potential of galantamine in FMF and other autoinflammatory diseases.


Asunto(s)
Fiebre Mediterránea Familiar , Ratones , Animales , Fiebre Mediterránea Familiar/tratamiento farmacológico , Galantamina/farmacología , Galantamina/uso terapéutico , Acetilcolinesterasa/uso terapéutico , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico
2.
J Surg Res ; 268: 326-336, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34399355

RESUMEN

BACKGROUND: Acute mesenteric ischemia is a common surgical emergency. Restoration of blood flow is a critical objective of treating this pathology. However, many patients suffer from ischemia-reperfusion (I/R) injuries at the time of revascularization, requiring prolonged hospitalizations. B-1a cells are a subtype of B lymphocytes with roles in regulating inflammation and tissue injury by spontaneous release of natural IgM and IL-10. We hypothesized that treatment with B-1a cells protects mice from intestinal I/R. METHODS: Mesenteric ischemia was induced in mice by placing a vascular clip on the superior mesenteric artery for 60 minutes. At the time of reperfusion, B-1a cells or PBS control were instilled into the peritoneal cavity (PerC) of mice. PerC lavage, blood, intestine, and lungs were collected 4 h after reperfusion. Serum organ injury and inflammatory markers such as ALT, AST, LDH, lactate, IL-6, as well as lung and gut histology and myeloperoxidase (MPO) were assessed. RESULTS: In intestinal I/R, B-1a cell frequency and number in the PerC were significantly decreased compared to sham-operated mice. There was an increase in the serum levels of ALT, AST, LDH, lactate, and IL-6 when comparing the vehicle group with the sham group. These increases were significantly reduced in the B-1a cell treated group. B-1a cell treatment significantly decreased the intestine and lung injury scores as well as MPO content, compared to vehicle treated mice. B-1a cell treatment resulted in a reduction of apoptotic cells in these tissues. Serum IgM levels were decreased in intestinal I/R, while treatment with B-1a cells significantly increased their levels towards normal levels. CONCLUSIONS: B-1a cell treatment at the time of mesenteric reperfusion ameliorates end organ damage and reduces systemic inflammation through the improvement of serum IgM levels. Preserving B-1a cells pool could serve as a novel therapeutic avenue in intestinal I/R injury.


Asunto(s)
Lesión Pulmonar Aguda , Isquemia Mesentérica , Daño por Reperfusión , Lesión Pulmonar Aguda/patología , Animales , Linfocitos B/patología , Humanos , Intestinos/irrigación sanguínea , Isquemia Mesentérica/patología , Ratones , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/prevención & control
3.
J Leukoc Biol ; 109(6): 1019-1032, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33070370

RESUMEN

Extracellular cold-inducible RNA-binding protein (eCIRP) is a damage-associated molecular pattern. Neutrophils present in the mononuclear cell fraction of Ficoll gradient separation are called low-density neutrophils (LDNs). Here we report the novel role of eCIRP on LDNs' heterogeneity in sepsis. Sepsis was induced in male C57BL/6 wild-type (WT) and CIRP-/- mice by cecal ligation and puncture (CLP). At 20 h after CLP, LDNs in the blood were isolated by Ficoll gradient separation, followed by staining the cells with anti-Ly6G and anti-CD11b Abs and detection by flow cytometry. Sepsis or recombinant murine CIRP (rmCIRP) injection in mice resulted in significant increase in the frequency (%) and number of Ly6G+ CD11bhi and Ly6G+ CD11blo LDNs in the blood compared to sham- or vehicle-treated mice. At 20 h of CLP, CIRP-/- mice had significantly lower frequency and number of Ly6G+ CD11bhi and Ly6G+ CD11blo LDNs in the blood compared to WT mice. In sepsis mice or rmCIRP-injected mice, compared to Ly6G+ CD11blo LDNs, the expression of CXCR4, ICAM-1, and iNOS and formation of reactive oxygen species, and neutrophil extracellular traps in Ly6G+ CD11bhi LDNs in the blood were significantly increased. Treatment of WT bone marrow-derived neutrophils (BMDNs) with rmCIRP increased Ly6G+ CD11bhi LDN frequency, whereas treatment of TLR4-/- BMDNs with rmCIRP significantly decreased the frequency of Ly6G+ CD11bhi LDNs. BMDNs' stimulation with rmCIRP increased the expression of transcription factors in LDNs. eCIRP induces the formation of a proinflammatory phenotype Ly6G+ CD11bhi of LDNs through TLR4. Targeting eCIRP may provide beneficial outcomes in sepsis by decreasing proinflammatory Ly6G+ CD11bhi LDNs.


Asunto(s)
Antígenos Ly/metabolismo , Antígeno CD11b/metabolismo , Neutrófilos/metabolismo , Proteínas de Unión al ARN/metabolismo , Sepsis/etiología , Sepsis/metabolismo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Espacio Extracelular/metabolismo , Recuento de Leucocitos , Masculino , Ratones , Ratones Noqueados , Proteínas de Unión al ARN/genética , Sepsis/diagnóstico
4.
Mol Med ; 26(1): 71, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32677895

RESUMEN

BACKGROUND: Co-administration of human ghrelin and growth hormone (GH) reverse immunosuppression in septic aged animals, but the mechanism remains elusive. Here, we hypothesize that ghrelin and GH co-treatment restores the immune response in aged septic rats by inhibiting the production of transforming growth factor-ß (TGF-ß), an immunoregulatory cytokine, through the vagus nerve. METHODS: Male aged Fischer rats (22-23-month-old) were made septic by cecal ligation and puncture (CLP) with or without dissecting the vagus nerve (vagotomy). Human ghrelin and GH or vehicle (PBS) were administrated subcutaneously at 5 h post CLP. After 20 h of CLP, serum and spleens were harvested. RESULTS: Serum TGF-ß levels were increased in septic aged rats, while ghrelin and GH treatment significantly reduced its levels. Expression of TGF-ß in the spleen was upregulated after sepsis, while ghrelin and GH treatment significantly inhibited its expression. TNF-α and IL-6 levels were significantly reduced after ex vivo LPS stimulation of splenocytes from rats that underwent CLP compared to sham rats; while these levels were significantly higher in splenocytes from ghrelin and GH-treated CLP rats compared to vehicle-treated CLP rats. Ghrelin and GH treatment reduced program death receptor-1 (PD-1) expression, increased human leukocyte antigen-DR (HLA-DR) expression, attenuated lymphopenia, and cleaved caspase-3 levels in the spleen of septic aged rats. Vagotomy diminished the beneficial effects of ghrelin and GH treatment in septic rats. In vitro, the addition of ghrelin, GH, or ghrelin and GH together had no effect on restoring immune response in splenocytes from CLP rats following LPS stimulation, indicating the requirement of the vagus nerve for ghrelin and GH's effect. CONCLUSIONS: Ghrelin and GH attenuate immunosuppression in aged septic rats through the vagus nerve-dependent inhibition of TGF-ß production.


Asunto(s)
Ghrelina/farmacología , Hormona de Crecimiento Humana/farmacología , Inmunomodulación/efectos de los fármacos , Sepsis/etiología , Sepsis/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis , Nervio Vago/metabolismo , Animales , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Humanos , Terapia de Inmunosupresión , Masculino , Ratas , Sepsis/diagnóstico
5.
Surgery ; 168(3): 478-485, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32439208

RESUMEN

BACKGROUND: Intestinal ischemia-reperfusion injury results in morbidity and mortality from both local injury and systemic inflammation and acute lung injury. Extracellular cold-inducible RNA-binding protein is a damage associated molecular pattern that fuels systemic inflammation and potentiates acute lung injury. We recently discovered a triggering receptor expressed on myeloid cells-1 serves as a novel receptor for extracellular cold-inducible RNA-binding protein. We developed a 7-aa peptide, named M3, derived from the cold-inducible RNA-binding protein, which interferes with cold-inducible RNA-binding protein's binding to a triggering receptor expressed on myeloid cells-1. Here, we hypothesized that M3 protects mice against intestinal ischemia-reperfusion injury. METHODS: Intestinal ischemia was induced in C57BL/6 mice via clamping of the superior mesenteric artery for 60 minutes. At reperfusion, mice were treated intraperitoneally with M3 (10 mg/kg body weight) or normal saline vehicle. Mice were killed 4 hours after reperfusion and blood and lungs were collected for various analysis. A 24-hours survival after intestinal ischemia-reperfusion was assessed. RESULTS: Serum levels of organ injury markers aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, and lactate were increased with intestinal ischemia-reperfusion, while treatment with M3 significantly decreased their levels. Serum, intestinal, and lung levels of proinflammatory cytokines and chemokines were also increased by intestinal ischemia-reperfusion, and treatment with M3 significantly reduced these values. Intestinal ischemia-reperfusion caused significant histological intestinal and lung injuries, which were mitigated by M3. Treatment with M3 improved the survival from 40% to 80% after intestinal ischemia-reperfusion. CONCLUSION: Inhibition of triggering receptor expressed on myeloid cells-1 by an extracellular cold-inducible RNA-binding protein-derived small peptide (M3) decreased inflammation, reduced lung injury, and improved survival in intestinal ischemia-reperfusion injury. Thus, blocking the extracellular cold-inducible RNA-binding protein-triggering receptor expressed on myeloid cells-1 interaction is a promising therapeutic avenue for mitigating intestinal ischemia-reperfusion injury.


Asunto(s)
Intestinos/irrigación sanguínea , Fragmentos de Péptidos/uso terapéutico , Proteínas de Unión al ARN/uso terapéutico , Daño por Reperfusión/prevención & control , Receptor Activador Expresado en Células Mieloides 1/antagonistas & inhibidores , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/inmunología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Ratones , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/farmacología , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/farmacología , Daño por Reperfusión/complicaciones , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Receptor Activador Expresado en Células Mieloides 1/metabolismo
6.
JCI Insight ; 5(5)2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32027618

RESUMEN

Extracellular cold-inducible RNA-binding protein (eCIRP) is a recently discovered damage-associated molecular pattern. Understanding the precise mechanism by which it exacerbates inflammation is essential. Here we identified that eCIRP is a new biologically active endogenous ligand of triggering receptor expressed on myeloid cells-1 (TREM-1), fueling inflammation in sepsis. Surface plasmon resonance revealed a strong binding affinity between eCIRP and TREM-1, and fluorescence resonance energy transfer assay confirmed eCIRP's interaction with TREM-1 in macrophages. Targeting TREM-1 by its siRNA or a decoy peptide, LP17, or by using TREM-1-/- mice dramatically reduced eCIRP-induced inflammation. We developed a potentially novel 7-aa peptide derived from human eCIRP, M3, which blocked the interaction of TREM-1 and eCIRP. M3 suppressed inflammation induced by eCIRP or agonist TREM-1 antibody cross-linking in murine macrophages or human peripheral blood monocytes. M3 also inhibited eCIRP-induced systemic inflammation and tissue injury. Treatment with M3 further protected mice from sepsis, improved acute lung injury, and increased survival. Thus, we have discovered a potentially novel TREM-1 ligand and developed a new peptide, M3, to block eCIRP-TREM-1 interaction and improve outcomes in sepsis.


Asunto(s)
Inflamación/metabolismo , Proteínas de Unión al ARN/metabolismo , Sepsis/metabolismo , Receptor Activador Expresado en Células Mieloides 1/metabolismo , Lesión Pulmonar Aguda/prevención & control , Anciano , Animales , Humanos , Inflamación/complicaciones , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/farmacología , Células RAW 264.7 , Proteínas de Unión al ARN/química , Sepsis/complicaciones , Receptor Activador Expresado en Células Mieloides 1/genética
7.
Mol Med ; 25(1): 52, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31747882

RESUMEN

BACKGROUND: Alcohol intake predisposes to infections and sepsis. Alcohol and sepsis inhibit the expression of milk fat globule epidermal growth factor-factor VIII (MFG-E8), a glycoprotein essential for optimal efferocytosis, resulting in the release of proinflammatory molecules and increased sepsis severity. We previously reported that recombinant mouse (rm) MFG-E8 attenuates sepsis-induced organ injury in rats with acute alcohol intoxication. In order to develop a therapy that can be safely used in humans, we have produced recombinant human (rh) MFG-E8 and evaluated its efficacy to ameliorate sepsis after acute exposure to alcohol. METHODS: We induced acute alcohol intoxication with a bolus injection of alcohol (1.75 g/kg BW) followed by an intravenous infusion of 300 mg/kg/h alcohol for 10 h. Sepsis was then induced by cecal ligation and puncture (CLP). At -10, 0, and 10 h relative to CLP, rats received MFG-E8 or vehicle (albumin) intravenously. Animals were euthanized at 20 h after CLP for blood and tissue collection. Additional groups of animals were used for a survival study. RESULTS: Compared to vehicle, rhMFG-E8 treatment ameliorated blood levels of proinflammatory cytokines (% improvement: TNF-α 49.8%, IL-6 34.7%) and endotoxin (61.7%), as well as of transaminases (AST 36.2%, ALT 40.1%) and lactate (18.4%). Rats treated with rhMFG-E8 also had a significant histological attenuation of the acute lung injury, as well as a reduction in the number of apoptotic cells in the thymus (43.4%) and cleaved caspase 3 (38.7%) in the spleen. In addition, rhMFG-E8 improved the 10-day sepsis survival rate from 45 to 80% CONCLUSION: rhMFG-E8 significantly ameliorated sepsis in rats with acute alcohol exposure, demonstrating rhMFG-E8's potential to be developed as an effective therapy for sepsis in alcohol abusers.


Asunto(s)
Alcoholes/efectos adversos , Antígenos de Superficie/farmacología , Proteínas de la Leche/farmacología , Proteínas Recombinantes/farmacología , Sepsis , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratas , Ratas Sprague-Dawley , Sepsis/inducido químicamente , Sepsis/metabolismo , Sepsis/mortalidad
8.
Mol Med ; 25(1): 24, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31146675

RESUMEN

BACKGROUND: Alcohol abuse affects the brain regions responsible for memory, coordination and emotional processing. Binge alcohol drinking has shown reductions in brain activity, but the molecular targets have not been completely elucidated. We hypothesized that brain cells respond to excessive alcohol by releasing a novel inflammatory mediator, called cold inducible RNA-binding protein (CIRP), which is critical for the decreased brain metabolic activity and impaired cognition. METHODS: Male wild type (WT) mice and mice deficient in CIRP (CIRP-/-) were studied before and after exposure to binge alcohol level by assessment of relative brain glucose metabolism with fluorodeoxyglucose (18FDG) and positron emission tomography (PET). Mice were also examined for object-place memory (OPM) and open field (OF) tasks. RESULTS: Statistical Parametric Analysis (SPM) of 18FDG-PET uptake revealed marked decreases in relative glucose metabolism in distinct brain regions of WT mice after binge alcohol. Regional analysis (post hoc) revealed that while activity in the temporal (secondary visual) and limbic (entorhinal/perirhinal) cortices was decreased in WT mice, relative glucose metabolic activity was less suppressed in the CIRP-/- mice. Group and condition interaction analysis revealed differing responses in relative glucose metabolism (decrease in WT mice but increase in CIRP-/- mice) after alcohol in brain regions including the hippocampus and the cortical amygdala where the percent changes in metabolic activity correlated with changes in object discrimination performance. Behaviorally, alcohol-treated WT mice were impaired in exploring a repositioned object in the OPM task, and were more anxious in the OF task, whereas CIRP-/- mice were not impaired in these tasks. CONCLUSION: CIRP released from brain cells could be responsible for regional brain metabolic hypoactivity leading to cognitive impairment under binge alcohol conditions.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/complicaciones , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Proteínas de Unión al ARN/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Fluorodesoxiglucosa F18/análisis , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Tomografía de Emisión de Positrones , Proteínas de Unión al ARN/genética , Memoria Espacial/efectos de los fármacos
9.
Front Immunol ; 9: 2032, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30237803

RESUMEN

Patients surviving a septic episode exhibit persistent immune impairment and increased mortality due to enhanced vulnerability to infections. In the present study, using the cecal ligation and puncture (CLP) model of polymicrobial sepsis, we addressed the hypothesis that altered vagus nerve activity contributes to immune impairment in sepsis survivors. CLP-surviving mice exhibited less TNFα in serum following administration of LPS, a surrogate for an infectious challenge, than control-operated (control) mice. To evaluate the role of the vagus nerve in the diminished response to LPS, mice were subjected to bilateral subdiaphragmatic vagotomy at 2 weeks post-CLP. CLP-surviving vagotomized mice exhibited increased serum and tissue TNFα levels in response to LPS-challenge compared to CLP-surviving, non-vagotomized mice. Moreover, vagus nerve stimulation in control mice diminished the LPS-induced TNFα responses while having no effect in CLP mice, suggesting constitutive activation of vagus nerve signaling in CLP-survivors. The percentage of splenic CD4+ ChAT-EGFP+ T cells that relay vagus signals to macrophages was increased in CLP-survivors compared to control mice, and vagotomy in CLP-survivors resulted in a reduced percentage of ChAT-EGFP+ cells. Moreover, CD4 knockout CLP-surviving mice exhibited an enhanced LPS-induced TNFα response compared to wild-type mice, supporting a functional role for CD4+ ChAT+ T cells in mediating inhibition of LPS-induced TNFα responses in CLP-survivors. Blockade of the cholinergic anti-inflammatory pathway with methyllcaconitine, an α7 nicotinic acetylcholine receptor antagonist, restored LPS-induced TNFα responses in CLP-survivors. Our study demonstrates that the vagus nerve is constitutively active in CLP-survivors and contributes to the immune impairment.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Bacterias Gramnegativas/fisiología , Infecciones por Bacterias Grampositivas/inmunología , Sepsis/inmunología , Nervio Vago/fisiología , Animales , Ciego/cirugía , Modelos Animales de Enfermedad , Infecciones por Bacterias Grampositivas/metabolismo , Humanos , Tolerancia Inmunológica , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Sepsis/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Nervio Vago/cirugía , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
10.
J Leukoc Biol ; 104(6): 1159-1171, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30088666

RESUMEN

Sepsis is a severe inflammatory condition associated with high mortality. Transmigration of neutrophils into tissues increases their lifespan to promote deleterious function. Junctional adhesion molecule-C (JAM-C) plays a pivotal role in neutrophil transmigration into tissues. We aim to study the role of JAM-C on the aging of neutrophils to cause sepsis-induced acute lung injury (ALI). Sepsis was induced in C57BL/6J mice by cecal ligation and puncture (CLP) and JAM-C expression in serum was assessed. Bone marrow-derived neutrophils (BMDN) were treated with recombinant mouse JAM-C (rmJAM-C) ex vivo and their viability was assessed. CLP-operated animals were administrated with either isotype IgG or anti-JAM-C Ab at a concentration of 3 mg/kg and after 20 h, aged neutrophils (CXCR4+ ) were assessed in blood and lungs and correlated with systemic injury and inflammatory markers. Soluble JAM-C level in serum was up-regulated during sepsis. Treatment with rmJAM-C inhibited BMDN apoptosis, thereby increasing their lifespan. CLP increased the frequencies of CXCR4+ neutrophils in blood and lungs, while treatment with anti-JAM-C Ab significantly reduced the frequencies of CXCR4+ aged neutrophils. Treatment with anti-JAM-C Ab significantly reduced systemic injury markers (alanine aminotransferase, aspartate aminotransferase, and lactate dehydrogenase) as well as systemic and lung inflammatory cytokines (IL-6 and IL-1ß) and chemokine (macrophage inflammatory protein-2). The blockade of JAM-C improved lung histology and reduced neutrophil contents in lungs of septic mice. Thus, reduction of the pro-inflammatory aged neutrophils by blockade of JAM-C has a novel therapeutic potential in sepsis-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Moléculas de Adhesión Celular/antagonistas & inhibidores , Terapia Molecular Dirigida , Neutrófilos/inmunología , Sepsis/complicaciones , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/inmunología , Animales , Anticuerpos/uso terapéutico , Especificidad de Anticuerpos , Apoptosis , Moléculas de Adhesión Celular/sangre , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/fisiología , Senescencia Celular , Citocinas/sangre , Inmunoglobulina G/farmacología , Inmunoglobulinas/sangre , Inmunoglobulinas/inmunología , Inmunoglobulinas/fisiología , Pulmón/inmunología , Pulmón/patología , Antígeno de Macrófago-1/efectos de los fármacos , Antígeno de Macrófago-1/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/química , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CXCR4/análisis , Proteínas Recombinantes/farmacología , Migración Transendotelial y Transepitelial/efectos de los fármacos
11.
Mol Med ; 24(1): 26, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-30134811

RESUMEN

BACKGROUND: Sepsis morbidity and mortality are aggravated by acute lung injury (ALI) or acute respiratory distress syndrome (ARDS). Mouse B-1a cells are a phenotypically and functionally unique sub-population of B cells, providing immediate protection against infection by releasing natural antibodies and immunomodulatory molecules. We hypothesize that B-1a cells ameliorate sepsis-induced ALI. METHODS: Sepsis was induced in C57BL/6 mice by cecal ligation and puncture (CLP). PBS or B-1a cells were adoptively transferred into the septic mice intraperitoneally. After 20 h of CLP, lungs were harvested and assessed by PCR and ELISA for pro-inflammatory cytokines (IL-6, IL-1ß) and chemokine (MIP-2) expression, by histology for injury, by TUNEL and cleaved caspase-3 for apoptosis, and by myeloperoxidase (MPO) assay for neutrophil infiltration. RESULTS: We found that septic mice adoptively transferred with B-1a cells significantly decreased the mRNA and protein levels of IL-6, IL-1ß and MIP-2 in the lungs compared to PBS-treated mice. Mice treated with B-1a cells showed dramatic improvement in lung injury compared to PBS-treated mice after sepsis. We found apoptosis in the lungs was significantly inhibited in B-1a cell injected mice compared to PBS-treated mice after sepsis. B-1a cell treatment significantly down-regulated MPO levels in the lungs compared to PBS-treated mice in sepsis. The protective outcomes of B-1a cells in ALI was further confirmed by using B-1a cell deficient CD19-/- mice, which showed significant increase in the lung injury scores following sepsis as compared to WT mice. CONCLUSIONS: Our results demonstrate a novel therapeutic potential of B-1a cells to treat sepsis-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/terapia , Linfocitos B/trasplante , Sepsis/terapia , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Traslado Adoptivo , Animales , Citocinas/inmunología , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Peroxidasa/inmunología , Sepsis/complicaciones , Sepsis/inmunología , Sepsis/patología
12.
Sci Rep ; 8(1): 166, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29317708

RESUMEN

Cytoplasmic membrane-bound connexin 43 (Cx43) proteins oligomerize into hexameric channels (hemichannels) that can sometimes dock with hemichannels on adjacent cells to form gap junctional (GJ) channels. However, the possible role of Cx43 hemichannels in sterile and infectious inflammatory diseases has not been adequately defined due to the lack of selective interventions. Here we report that a proinflammatory mediator, the serum amyloid A (SAA), resembled bacterial endotoxin by stimulating macrophages to up-regulate Cx43 expression and double-stranded RNA-activated protein kinase R (PKR) phosphorylation in a TLR4-dependent fashion. Two well-known Cx43 mimetic peptides, the GAP26 and TAT-GAP19, divergently affected macrophage hemichannel activities in vitro, and differentially altered the outcome of lethal sepsis in vivo. By screening a panel of Cx43 mimetic peptides, we discovered that one cysteine-containing peptide, P5 (ENVCYD), effectively attenuated hemichannel activities, and significantly suppressed endotoxin-induced release of ATP and HMGB1 in vitro. In vivo, the P5 peptide conferred a significant protection against hepatic ischemia/reperfusion injury and lethal microbial infection. Collectively, these findings have suggested a pathogenic role of Cx43 hemichannels in sterile injurious as well as infectious inflammatory diseases possibly through facilitating extracellular ATP efflux to trigger PKR phosphorylation/activation.


Asunto(s)
Conexina 43/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Conexina 43/antagonistas & inhibidores , Conexina 43/química , Endotoxinas/metabolismo , Humanos , Inflamación/etiología , Inflamación/mortalidad , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Péptidos/metabolismo , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Sepsis/etiología , Sepsis/metabolismo , Sepsis/mortalidad , eIF-2 Quinasa/metabolismo
13.
Artículo en Inglés | MEDLINE | ID: mdl-29152323

RESUMEN

E. coli releases a 33 amino acid peptide melanocortin-like peptide of E. coli (MECO-1) that is identical to the C-terminus of the E. coli elongation factor-G (EF-G) and has interesting similarities to two prominent mammalian melanocortin hormones, alpha-melanocyte-stimulating hormone (alpha-MSH) and adrenocorticotropin (ACTH). Note that MECO-1 lacks HFRW, the common pharmacophore of the known mammalian melanocortin peptides. MECO-1 and the two hormones were equally effective in severely blunting release of cytokines (HMGB1 and TNF) from macrophage-like cells in response to (i) endotoxin (lipopolysaccharide) or (ii) pro-inflammatory cytokine HMGB-1. The in vitro anti-inflammatoty effects of MECO-1 and of alpha-MSH were abrogated by (i) antibody against melanocortin-1 receptor (MC1R) and by (ii) agouti, an endogenous inverse agonist of MC1R. In vivo MECO-1 was even more potent than alpha-MSH in rescuing mice from death due to (i) lethal doses of LPS endotoxin or (ii) cecal ligation and puncture, models of sterile and infectious sepsis, respectively.

14.
Sci Rep ; 7(1): 9235, 2017 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-28835626

RESUMEN

The Wnt/ß-catenin pathway has been involved in regulating inflammation in various infectious and inflammatory diseases. Sepsis is a life-threatening condition caused by dysregulated inflammatory response to infection with no effective therapy available. Recently elevated Wnt/ß-catenin signaling has been detected in sepsis. However, its contribution to sepsis-associated inflammatory response remains to be explored. In this study, we show that inhibition of Wnt/ß-catenin signaling reduces inflammation and mitigates sepsis-induced organ injury. Using in vitro LPS-stimulated RAW264.7 macrophages, we demonstrate that a small-molecule inhibitor of ß-catenin responsive transcription, iCRT3, significantly reduces the LPS-induced Wnt/ß-catenin activity and also inhibits TNF-α production and IκB degradation in a dose-dependent manner. Intraperitoneal administration of iCRT3 to C57BL/6 mice, subjected to cecal ligation and puncture-induced sepsis, decreases the plasma levels of proinflammatory cytokines and organ injury markers in a dose-dependent manner. The histological integrity of the lungs is improved with iCRT3 treatment, along with reduced lung collagen deposition and apoptosis. In addition, iCRT3 treatment also decreases the expression of the cytokines, neutrophil chemoattractants, as well as the MPO activity in the lungs of septic mice. Based on these findings we conclude that targeting the Wnt/ß-Catenin pathway may provide a potential therapeutic approach for treatment of sepsis.


Asunto(s)
Inflamación/metabolismo , Sepsis/etiología , Sepsis/metabolismo , Vía de Señalización Wnt , Animales , Apoptosis/efectos de los fármacos , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Inflamación/patología , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/inmunología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Oxazoles/farmacología , Células RAW 264.7 , Sepsis/tratamiento farmacológico , Sepsis/patología , Vía de Señalización Wnt/efectos de los fármacos
15.
Shock ; 46(4): 431-8, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26974422

RESUMEN

Intestinal ischemia-reperfusion (I/R) is associated with acute respiratory distress syndrome. Osteopontin (OPN), a glycoprotein secreted from immune-reactive cells, plays a deleterious role in various inflammatory diseases. Considering OPN as a pro-inflammatory molecule, we hypothesize that the treatment with its neutralizing antibody (anti-OPN Ab) protects mice against intestinal I/R-induced acute lung injury (ALI). Intestinal I/R was induced in mice by superior mesenteric artery occlusion with a vascular clip. After 45 min of occlusion, the clip was removed and anti-OPN Ab (25 µg/mouse) or normal IgG isotype control (25 µg/mouse) was immediately administrated intravenously. Blood, small intestine, and lung tissues were collected at 4 h after reperfusion for various analyses. After intestinal I/R, mRNA and protein levels of OPN were significantly induced in the small intestine, lungs, and blood relative to sham-operated animals. Compared with the IgG control group, treatment of anti-OPN Ab significantly reduced plasma levels of pro-inflammatory cytokine and chemokine (IL-6 and MIP-2) and organ injury markers (AST, ALT, and LDH). The histological architecture of the gut and lung tissues in anti-OPN Ab-treated intestinal I/R-induced mice showed significant improvement versus the IgG control mice. The lung inflammation measured by the levels of IL-6, IL-1ß, and MIP-2 was also significantly downregulated in the anti-OPN Ab-treated mice as compared with the IgG control mice. Besides, the lung MPO and neutrophil infiltration in anti-OPN Ab-treated mice showed significant reduction as compared with the IgG control animals. In conclusion, we have demonstrated beneficial outcomes of anti-OPN Ab treatment in protecting against ALI, implicating a novel therapeutic potential in intestinal I/R.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Osteopontina/metabolismo , Daño por Reperfusión/metabolismo , Lesión Pulmonar Aguda/etiología , Animales , Quimiocina CXCL2/metabolismo , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Intestinos/patología , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Osteopontina/genética , Daño por Reperfusión/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
PLoS One ; 11(1): e0144215, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26790027

RESUMEN

Systemic infection can initiate or exacerbate central nervous system (CNS) pathology, even in the absence of overt invasion of bacteria into the CNS. Recent epidemiological studies have demonstrated that human survivors of sepsis have an increased risk of long-term neurocognitive decline. There is thus a need for improved understanding of the physiological mechanisms whereby acute sepsis affects the CNS. In particular, MyD88-dependent activation of brain microvascular endothelial cells and a resulting loss of blood-brain barrier integrity have been proposed to play an important role in the effects of systemic inflammation on the CNS. Signaling through the vagus nerve has also been considered to be an important component of CNS responses to systemic infection. Here, we demonstrate that blood-brain barrier permeabilization and hippocampal transcriptional responses during polymicrobial sepsis occur even in the absence of MyD88-dependent signaling in cerebrovascular endothelial cells. We further demonstrate that these transcriptional responses can occur without vagus nerve input. These results suggest that redundant signals mediate CNS responses in sepsis. Either endothelial or vagus nerve activation may be individually sufficient to transmit systemic inflammation to the central nervous system. Transcriptional activation in the forebrain in sepsis may be mediated by MyD88-independent endothelial mechanisms or by non-vagal neuronal pathways.


Asunto(s)
Barrera Hematoencefálica/patología , Expresión Génica , Hipocampo/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Sepsis/genética , Nervio Vago/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Coinfección/genética , Coinfección/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio/citología , Endotelio/metabolismo , Femenino , Hipocampo/citología , Humanos , Masculino , Ratones , Factor 88 de Diferenciación Mieloide/genética , Sepsis/etiología , Sepsis/metabolismo , Transducción de Señal
17.
Bioelectron Med ; 3: 18-28, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-33145374

RESUMEN

When pathogens and toxins breech the epithelial barrier, antigens are transported by the lymphatic system to lymph nodes. In previously immunized animals, antigens become trapped in the draining lymph nodes, but the underlying mechanism that controls antigen restriction is poorly understood. Here we describe the role of neurons in sensing and restricting antigen flow in lymph nodes. The antigen keyhole-limpet hemocyanin (KLH) injected into the mouse hind paw flows from the popliteal lymph node to the sciatic lymph node, continuing through the upper lymphatics to reach the systemic circulation. Re-exposure to KLH in previously immunized mice leads to decreased flow from the popliteal to the sciatic lymph node as compared with naïve mice. Administering bupivacaine into the lymph node region restores antigen flow in immunized animals. In contrast, neural activation using magnetic stimulation significantly decreases antigen trafficking in naïve animals as compared with sham controls. Ablating NaV1.8 + sensory neurons significantly reduces antigen restriction in immunized mice. Genetic deletion of FcγRI/FcεRI also reverses the antigen restriction. Colocalization of PGP9.5-expressing neurons, FcγRI receptors and labeled antigen occurs at the antigen challenge site. Together, these studies reveal that neuronal circuits modulate antigen trafficking through a pathway that requires NaV1.8 and FcγR.

18.
Crit Care ; 19: 53, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25887405

RESUMEN

INTRODUCTION: Sepsis refers to severe systemic inflammation leading to acute lung injury (ALI) and death. Introducing novel therapies can reduce the mortality in ALI. Osteopontin (OPN), a secretory glycoprotein produced by immune reactive cells, plays a deleterious role in various inflammatory diseases. However, its role in ALI caused by sepsis remains unexplored. We hypothesize that treatment with an OPN-neutralizing antibody (anti-OPN Ab) protects mice against ALI during sepsis. METHODS: Sepsis was induced in 8-week-old male C57BL/6 mice by cecal ligation and puncture (CLP). Anti-OPN Ab or non-immunized IgG as control, at a dose of 50 µg/mouse, was intravenously injected at the time of CLP. After 20 hours, the expression of OPN and proinflammatory cytokines in tissues and plasma was examined by real-time PCR, Western blot, and ELISA. Plasma levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST) and lactate dehydrogenase (LDH) and the lung myeloperoxidase (MPO) levels were determined by colorimetric assays. Lung damage and neutrophil infiltrations were determined by histological H&E and Gr-1 staining, respectively. The effect of recombinant mouse OPN (rmOPN) on human neutrophil-like cell (HL-60) migration was performed by Boyden chamber assays and the involvement of intracellular signaling molecules in HL-60 cells was revealed by Western blot. RESULTS: After 20 hours of sepsis, mRNA and protein levels of OPN were significantly induced in lungs, spleen, and plasma. Treatment with an anti-OPN Ab in septic mice significantly reduced the plasma levels of ALT, AST, and LDH, and the proinflammatory cytokines IL-6, IL-1ß and the chemokine MIP-2, compared with the vehicle group. Similarly, the lung mRNA and protein expressions of proinflammatory cytokines and chemokine were greatly reduced in anti-OPN Ab-treated animals. The lung histological architecture, MPO and neutrophil infiltration were significantly improved in anti-OPN Ab-treated mice compared with the vehicle animals. Treatment of rmOPN in HL-60 cells significantly increased their migration, in vitro. The neutrophils treated with rmOPN remarkably increased the levels of phospho focal adhesion kinase (pFAK), phospho extracellular signal-regulated kinase (pERK) and phospho p38. CONCLUSIONS: Our findings clearly demonstrate the beneficial outcomes of anti-OPN Ab treatment in protecting against ALI, implicating a novel therapeutic strategy in sepsis.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Neutrófilos/fisiología , Osteopontina/inmunología , Sepsis/complicaciones , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/prevención & control , Animales , Movimiento Celular , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Osteopontina/antagonistas & inhibidores
19.
Mol Med ; 20: 601-11, 2015 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-25299421

RESUMEN

Inflammatory conditions characterized by excessive peripheral immune responses are associated with diverse alterations in brain function, and brain-derived neural pathways regulate peripheral inflammation. Important aspects of this bidirectional peripheral immune-brain communication, including the impact of peripheral inflammation on brain region-specific cytokine responses, and brain cholinergic signaling (which plays a role in controlling peripheral cytokine levels), remain unclear. To provide insight, we studied gene expression of cytokines, immune cell markers and brain cholinergic system components in the cortex, cerebellum, brainstem, hippocampus, hypothalamus, striatum and thalamus in mice after an intraperitoneal lipopolysaccharide injection. Endotoxemia was accompanied by elevated serum levels of interleukin (IL)-1ß, IL-6 and other cytokines and brain region-specific increases in Il1b (the highest increase, relative to basal level, was in cortex; the lowest increase was in cerebellum) and Il6 (highest increase in cerebellum; lowest increase in striatum) mRNA expression. Gene expression of brain Gfap (astrocyte marker) was also differentially increased. However, Iba1 (microglia marker) mRNA expression was decreased in the cortex, hippocampus and other brain regions in parallel with morphological changes, indicating microglia activation. Brain choline acetyltransferase (Chat ) mRNA expression was decreased in the striatum, acetylcholinesterase (Ache) mRNA expression was decreased in the cortex and increased in the hippocampus, and M1 muscarinic acetylcholine receptor (Chrm1) mRNA expression was decreased in the cortex and the brainstem. These results reveal a previously unrecognized regional specificity in brain immunoregulatory and cholinergic system gene expression in the context of peripheral inflammation and are of interest for designing future antiinflammatory approaches.


Asunto(s)
Encéfalo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Acetilcolinesterasa/genética , Animales , Encéfalo/metabolismo , Proteínas de Unión al Calcio/genética , Citocinas/sangre , Citocinas/genética , Proteínas Ligadas a GPI/genética , Proteína Ácida Fibrilar de la Glía , Inflamación/sangre , Masculino , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/genética , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/genética , ARN Mensajero/metabolismo , Receptores Muscarínicos/genética
20.
Brain Behav Immun ; 44: 19-27, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25063706

RESUMEN

Inflammatory conditions characterized by excessive immune cell activation and cytokine release, are associated with bidirectional immune system-brain communication, underlying sickness behavior and other physiological responses. The vagus nerve has an important role in this communication by conveying sensory information to the brain, and brain-derived immunoregulatory signals that suppress peripheral cytokine levels and inflammation. Brain muscarinic acetylcholine receptor (mAChR)-mediated cholinergic signaling has been implicated in this regulation. However, the possibility of controlling inflammation by peripheral administration of centrally-acting mAChR agonists is unexplored. To provide insight we used the centrally-acting M1 mAChR agonist xanomeline, previously developed in the context of Alzheimer's disease and schizophrenia. Intraperitoneal administration of xanomeline significantly suppressed serum and splenic TNF levels, alleviated sickness behavior, and increased survival during lethal murine endotoxemia. The anti-inflammatory effects of xanomeline were brain mAChR-mediated and required intact vagus nerve and splenic nerve signaling. The anti-inflammatory efficacy of xanomeline was retained for at least 20h, associated with alterations in splenic lymphocyte, and dendritic cell proportions, and decreased splenocyte responsiveness to endotoxin. These results highlight an important role of the M1 mAChR in a neural circuitry to spleen in which brain cholinergic activation lowers peripheral pro-inflammatory cytokines to levels favoring survival. The therapeutic efficacy of xanomeline was also manifested by significantly improved survival in preclinical settings of severe sepsis. These findings are of interest for strategizing novel therapeutic approaches in inflammatory diseases.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Agonistas Muscarínicos/farmacología , Piridinas/farmacología , Sepsis/prevención & control , Bazo/fisiología , Tiadiazoles/farmacología , Nervio Vago/fisiología , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/uso terapéutico , Citocinas/fisiología , Conducta de Enfermedad/efectos de los fármacos , Inyecciones Intraperitoneales , Lipopolisacáridos , Masculino , Ratones Endogámicos BALB C , Agonistas Muscarínicos/administración & dosificación , Agonistas Muscarínicos/uso terapéutico , Piridinas/administración & dosificación , Piridinas/uso terapéutico , Ratas Sprague-Dawley , Receptor Muscarínico M1/agonistas , Sepsis/mortalidad , Bazo/efectos de los fármacos , Bazo/inervación , Análisis de Supervivencia , Tiadiazoles/administración & dosificación , Tiadiazoles/uso terapéutico , Factor de Necrosis Tumoral alfa/sangre , Vagotomía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA