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











Base de datos
Intervalo de año de publicación
1.
Bosn J Basic Med Sci ; 22(2): 217-228, 2022 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-34813418

RESUMEN

Emerging evidence has shown that protocatechuic acid (PCA) has antioxidant and anti-inflammatory effects. It can alleviate the injury of sciatic nerve, while the mechanism of its therapeutic effect on neuralgia remains unknown . In vivo, chromium bowel ligation was used to establish a chronic constriction injury (CCI) rat model to induce sciatic nerve pain, then two doses of PCA were used to treat CCI rats. In vitro, 10 ng/mL TNF-α was used to stimulate glial satellite cells derived from the dorsal root ganglia (DRG) L4-L6 of the sciatic nerve to simulate sciatic nerve pain. PCA relieved mechanical allodynia and thermal hyperalgesia in CCI rats. CCK-8 assay revealed that PCA inhibited the proliferation of glial satellite cells induced by TNF-α. Moreover, ELISA demonstrated that PCA could improve the inflammatory response of rats caused by CCI and cells induced by TNF-α. Next, RT-qPCR and Western blot assays testified that PCA blocked the c-Jun N-terminal kinase/the chemokine ligand 1/CXC chemokine receptor 2 (JNK/CXCL1/CXCR2) pathway by inhibiting CXCL1 levels in cells induced by TNF-α and DRG of CCI rats. In conclusion, PCA can alleviate neuropathic pain of CCI rats, improve oxidative stress by inhibiting the JNK/CXCL1/CXCR2 signaling pathway, which provides a new perspective for the treatment of neuropathic pain caused by CCI.


Asunto(s)
Quimiocina CXCL1 , Hidroxibenzoatos , Sistema de Señalización de MAP Quinasas , Neuralgia , Receptores de Interleucina-8B , Animales , Quimiocina CXCL1/antagonistas & inhibidores , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuralgia/tratamiento farmacológico , Ratas , Receptores de Interleucina-8B/antagonistas & inhibidores
2.
J Neuroinflammation ; 18(1): 306, 2021 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-34963475

RESUMEN

BACKGROUND: Pathological interactions between ß-amyloid (Aß) and tau drive synapse loss and cognitive decline in Alzheimer's disease (AD). Reactive astrocytes, displaying altered functions, are also a prominent feature of AD brain. This large and heterogeneous population of cells are increasingly recognised as contributing to early phases of disease. However, the contribution of astrocytes to Aß-induced synaptotoxicity in AD is not well understood. METHODS: We stimulated mouse and human astrocytes with conditioned medium containing concentrations and species of human Aß that mimic those in human AD brain. Medium from stimulated astrocytes was collected and immunodepleted of Aß before being added to naïve rodent or human neuron cultures. A cytokine, identified in unbiased screens of stimulated astrocyte media and in postmortem human AD brain lysates was also applied to neurons, including those pre-treated with a chemokine receptor antagonist. Tau mislocalisation, synaptic markers and dendritic spine numbers were measured in cultured neurons and organotypic brain slice cultures. RESULTS: We found that conditioned medium from stimulated astrocytes induces exaggerated synaptotoxicity that is recapitulated following spiking of neuron culture medium with recombinant C-X-C motif chemokine ligand-1 (CXCL1), a chemokine upregulated in AD brain. Antagonism of neuronal C-X-C motif chemokine receptor 2 (CXCR2) prevented synaptotoxicity in response to CXCL1 and Aß-stimulated astrocyte secretions. CONCLUSIONS: Our data indicate that astrocytes exacerbate the synaptotoxic effects of Aß via interactions of astrocytic CXCL1 and neuronal CXCR2 receptors, highlighting this chemokine-receptor pair as a novel target for therapeutic intervention in AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/toxicidad , Astrocitos/patología , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/química , Sinapsis/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células Cultivadas , Medios de Cultivo Condicionados , Espinas Dendríticas/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neuronas/efectos de los fármacos , Receptores de Interleucina-8B/antagonistas & inhibidores , Proteínas tau/química , Proteínas tau/toxicidad
3.
J Dermatol Sci ; 104(1): 30-38, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34479772

RESUMEN

BACKGROUND: Organ transplant recipients show a high incidence for the formation of cutaneous squamous cell carcinoma (cSCC), while sirolimus appears to reduce the risk. GRO-α is a chemokine, which is overexpressed in many tumor entities and associated with malignant transformation. However, little is known about the expression and function of GRO-α in human cSCC. OBJECTIVE: Our aim was to investigate the relevance of the GRO-α (CXCL-1)/ CXCR2 axis in human cSCC and the potential impact of sirolimus. METHODS: We analyzed the GRO-α expression in human keratinocytes, different cSCC cell lines as well as cSCC tissue and investigated its effect on cell proliferation and migration. Additionally, we incubated cells with sirolimus and measured the expression of GRO-α and its receptor CXCR2. RESULTS: We showed that both constitutive as well as induced GRO-α expression is higher in in cSCC cell lines compared to keratinocytes and that GRO-α protein is detectable in human cSCC tissue. By GRO-α exposure and shRNA knock down, we identified GRO-α as a driving factor in proliferation and migration. Moreover, in a dermis equivalent GRO-α knocked down cSCC cell lines displayed a reduced capacity in tumor nest formation. Incubation with sirolimus significantly inhibited GRO-α expression in keratinocytes as well as tumor cell lines. Moreover, sirolimus decreased the expression of the corresponding receptor CXCR2. CONCLUSION: Taken together, our results suggest that the GRO-α/CXCR2 axis plays a role in human keratinocyte carcinogenesis and might represent a molecular mechanism for the preventive effect of mTOR inhibitors in cSCC development.


Asunto(s)
Carcinoma de Células Escamosas/prevención & control , Quimiocina CXCL1/metabolismo , Inhibidores mTOR/farmacología , Receptores de Interleucina-8B/metabolismo , Neoplasias Cutáneas/prevención & control , Carcinogénesis/efectos de los fármacos , Carcinogénesis/inmunología , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/genética , Técnicas de Silenciamiento del Gen , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Queratinocitos/metabolismo , Inhibidores mTOR/uso terapéutico , Receptores de Interleucina-8B/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Sirolimus/farmacología , Sirolimus/uso terapéutico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología
4.
Neurobiol Dis ; 158: 105468, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34358616

RESUMEN

CXCL1, a functional murine orthologue of the human chemokine CXCL8 (IL-8), and its CXCR1 and CXCR2 receptors were investigated in a murine model of acquired epilepsy developing following status epilepticus (SE) induced by intra-amygdala kainate. CXCL8 and its receptors were also studied in human temporal lobe epilepsy (TLE). The functional involvement of the chemokine in seizure generation and neuronal cell loss was assessed in mice using reparixin (formerly referred to as repertaxin), a non-competitive allosteric inhibitor of CXCR1/2 receptors. We found a significant increase in hippocampal CXCL1 level within 24 h of SE onset that lasted for at least 1 week. No changes were measured in blood. In analogy with human TLE, immunohistochemistry in epileptic mice showed that CXCL1 and its two receptors were increased in hippocampal neuronal cells. Additional expression of these molecules was found in glia in human TLE. Mice were treated with reparixin or vehicle during SE and for additional 6 days thereafter, using subcutaneous osmotic minipumps. Drug-treated mice showed a faster SE decay, a reduced incidence of acute symptomatic seizures during 48 h post-SE, and a delayed time to spontaneous seizures onset compared to vehicle controls. Upon reparixin discontinuation, mice developed spontaneous seizures similar to vehicle mice, as shown by EEG monitoring at 14 days and 2.5 months post-SE. In the same epileptic mice, reparixin reduced neuronal cell loss in the hippocampus vs vehicle-injected mice, as assessed by Nissl staining at completion of EEG monitoring. Reparixin administration for 2 weeks in mice with established chronic seizures, reduced by 2-fold on average seizure number vs pre-treatment baseline, and this effect was reversible upon drug discontinuation. No significant changes in seizure number were measured in vehicle-injected epileptic mice that were EEG monitored in parallel. Data show that CXCL1-IL-8 signaling is activated in experimental and human epilepsy and contributes to acute and chronic seizures in mice, therefore representing a potential new target to attain anti-ictogenic effects.


Asunto(s)
Quimiocina CXCL1/genética , Epilepsia del Lóbulo Temporal/genética , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8B/genética , Convulsiones/genética , Animales , Quimiocina CXCL1/antagonistas & inhibidores , Electroencefalografía , Epilepsia del Lóbulo Temporal/fisiopatología , Hipocampo/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Receptores de Interleucina-8A/antagonistas & inhibidores , Receptores de Interleucina-8B/antagonistas & inhibidores , Convulsiones/fisiopatología , Estado Epiléptico/genética , Estado Epiléptico/patología , Sulfonamidas/farmacología
5.
Phytomedicine ; 78: 153331, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32911383

RESUMEN

BACKGROUND: Breast cancer is the most common malignancy in women and metastasis is the leading cause of breast cancer-related deaths. Our previous studies have shown that XIAOPI formula, a newly approved drug by the State Food and Drug Administration of China (SFDA), can dramatically inhibit breast cancer metastasis by modulating the tumor-associated macrophages/C-X-C motif chemokine ligand 1 (TAMs/CXCL1) pathway. However, the bioactive compound accounting for the anti-metastatic effect of XIAOPI formula remains unclear. PURPOSE: This study was designed to separate the anti-metastatic bioactive compound from XIAOPI formula and to elucidate its action mechanisms. STUDY DESIGN/METHODS: TAMs/CXCL1 promoter activity-guided fractionation and multiple chemical structure identification approaches were conducted to screen the bioactive compound from XIAOPI formula. Breast cancer cells and TAMs were co-cultured in vitro or co-injected in vivo to simulate their coexistence. Multiple molecular biology experiments, zebrafish breast cancer xenotransplantation model and mouse breast cancer xenografts were applied to validate the anti-metastatic activity of the screened compound. RESULTS: Bioactivity-guided fractionation identified baohuoside I (BHS) as the key bioactive compound of XIAOPI formula in inhibiting TAMs/CXCL1 promoter activity. Functional studies revealed that BHS could significantly inhibit the migration and invasion as well as the expression of metastasis-related proteins in both human and mouse breast cancer cells, along with decreasing the proportion of breast cancer stem cells (CSCs). Furthermore, BHS could suppress the M2 phenotype polarization of TAMs and therefore attenuate their CXCL1 expression and secretion. Notably, mechanistic investigations validated TAMs/CXCL1 as the crucial target of BHS in suppressing breast cancer metastasis as exogenous addition of CXCL1 significantly abrogated the anti-metastatic effect of BHS on breast cancer cells. Moreover, BHS was highly safe in vivo as it exhibited no observable embryotoxicity or teratogenic effect on zebrafish embryos. More importantly, BHS remarkably suppressed breast cancer metastasis and TAMs/CXCL1 activity in both zebrafish breast cancer xenotransplantation model and mouse breast cancer xenografts. CONCLUSION: This study not only provides novel insights into TAMs/CXCL1 as a reliable screening target for anti-metastatic drug discovery, but also suggests BHS as a promising candidate drug for metastatic breast cancer treatment.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Quimiocina CXCL1/metabolismo , Flavonoides/farmacología , Macrófagos/efectos de los fármacos , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/genética , Técnicas de Cocultivo , Regulación hacia Abajo/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Embrión no Mamífero , Femenino , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Células Madre Neoplásicas/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/embriología
6.
Life Sci ; 248: 117456, 2020 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-32097666

RESUMEN

AIMS: In this study, we will investigate the therapeutic effects of berberine (BBR) in Helicobacter pylori (H. pylori) induced chronic atrophic gastritis (CAG). Furthermore, potential mechanisms of BBR in regulating IRF8-IFN-γ signaling axis will also be investigated. MATERIALS AND METHODS: H. pylori were utilized to establish CAG model of rats. Therapeutic effects of BBR on serum supernatant indices, and histopathology of stomach were analyzed in vivo. Moreover, GES-1 cells were infected by H. pylori, and intervened with BBR in vitro. Cell viability, morphology, proliferation, and quantitative analysis were detected by high-content screening (HCS) imaging assay. To further investigate the potential mechanisms of BBR, relative mRNA, immunohistochemistry and protein expression in IRF8-IFN-γ signaling axis were measured. KEY FINDINGS: Results showed serum supernatant indices including IL-17, CXCL1, and CXCL9 were downregulated by BBR intervention, while, G-17 increased significantly. Histological injuries of gastric mucosa induced by H. pylori also were alleviated. Moreover, cell viability and morphology changes of GES-1 cells were improved by BBR intervention. In addition, proinflammatory genes and IRF8-IFN-γ signaling axis related genes, including Ifit3, Upp1, USP18, Nlrc5, were suppressed by BBR administration in vitro and in vivo. The proteins expression related to IRF8-IFN-γ signaling axis, including Ifit3, IRF1 and Ifit1 were downregulated by BBR intervention.


Asunto(s)
Antiinflamatorios/farmacología , Berberina/farmacología , Gastritis Atrófica/tratamiento farmacológico , Infecciones por Helicobacter/tratamiento farmacológico , Factores Reguladores del Interferón/genética , Interferón gamma/genética , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/genética , Quimiocina CXCL1/inmunología , Quimiocina CXCL9/antagonistas & inhibidores , Quimiocina CXCL9/genética , Quimiocina CXCL9/inmunología , Enfermedad Crónica , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Gastritis Atrófica/genética , Gastritis Atrófica/inmunología , Gastritis Atrófica/microbiología , Regulación de la Expresión Génica , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/efectos de los fármacos , Helicobacter pylori/crecimiento & desarrollo , Helicobacter pylori/patogenicidad , Humanos , Factores Reguladores del Interferón/antagonistas & inhibidores , Factores Reguladores del Interferón/inmunología , Interferón gamma/antagonistas & inhibidores , Interferón gamma/inmunología , Interleucina-17/agonistas , Interleucina-17/genética , Interleucina-17/inmunología , Masculino , Proteínas NLR/antagonistas & inhibidores , Proteínas NLR/genética , Proteínas NLR/inmunología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Uridina Fosforilasa/antagonistas & inhibidores , Uridina Fosforilasa/genética , Uridina Fosforilasa/inmunología
7.
J Microbiol Immunol Infect ; 52(6): 872-879, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31759853

RESUMEN

BACKGROUND: Cytokines and chemokines play critical roles in the pathogenesis of asthma. Azithromycin, a macrolides, is frequently used in asthmatic children with lower respiratory tract infection and is reported having anti-inflammatory and immunomodulatory effects. However, the effects of azithromycin on the expression of TNF-α, Th1- and Th2-related chemokines, and neutrophil chemoattractant are unknown. We investigated the in vitro effects of azithromycin on the expression of TNF-α, Th1-related chemokine interferon-γ-inducible protein-10 (IP-10/CXCL10), Th2-related chemokine macrophage-derived chemokine (MDC/CCL22) and neutrophil chemoattractant growth-related oncogene-α (GRO-α/CXCL1) in THP-1 cells as a model for human monocytes. METHODS: THP-1 cells were pretreated with various concentrations of azithromycin before Toll-like receptor 4 (TLR4) agonist lipopolysaccharide (LPS) stimulation. TNF-α, IP-10, MDC and GRO-α were measured by ELISA. Intracellular signaling was investigated by pathway inhibitors and Western blot. RESULT: Azithromycin suppressed MDC and IP-10 expression in LPS-stimulated THP-1 cells. However, azithromycin had no effect LPS-induced TNF-α and GRO-α expression. Western blotting revealed that azithromycin suppressed LPS-induced phosphorylation of mitogen-activated protein kinase (MAPK)-JNK and ERK expression, and also suppressed LPS-induced phosphorylation of nuclear factor (NF) κB-p65 expression. CONCLUSION: Azithromycin suppressed LPS-induced MDC expression via the MAPK-JNK and the NFκB-p65 pathway. Azithromycin also suppressed LPS-induced IP-10 via the MAPK-JNK/ERK and the NFκB-p65 pathway. Azithromycin may benefit asthmatic patients by suppressing chemokines expression.


Asunto(s)
Antibacterianos/farmacología , Azitromicina/farmacología , Quimiocina CCL22/antagonistas & inhibidores , Quimiocina CXCL10/antagonistas & inhibidores , Monocitos/efectos de los fármacos , Monocitos/inmunología , Quimiocina CCL22/inmunología , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/inmunología , Quimiocina CXCL10/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Lipopolisacáridos , Sistema de Señalización de MAP Quinasas , Células THP-1 , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Factor de Transcripción ReIA , Factor de Necrosis Tumoral alfa/inmunología
8.
Neuropharmacology ; 151: 136-143, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30991054

RESUMEN

Painful peripheral neuropathy is the most dose-limiting side effect of paclitaxel (PTX), a widely used anti-cancer drug to treat solid tumours. The understanding of the mechanisms involved in this side effect is crucial to the development of new therapeutic approaches. CXCL1 chemokine and its receptor CXCR2 have been pointed as promising targets to treat chronic pain. Herein, we sought to evaluate the possible involvement of CXCL1 and CXCR2 in the pathogenesis of PTX-induced neuropathic pain in mice. PTX treatment led to increased levels of CXCL1 in both dorsal root ganglion and spinal cord samples. Systemic treatment with the anti-CXCL1 antibody (10 µg/kg, i.v.) or the selective CXCR2 antagonist (SB225002, 3 mg/kg, i.p.) had minor effect on PTX-induced mechanical hypersensitivity. On the other hand, the intrathecal (i.t.) treatment with anti-CXCL1 (1 ng/site) or SB225002 (10 µg/site) consistently inhibited the nociceptive responses of PTX-treated mice. Similar results were obtained by inhibiting the PI3Kγ enzyme a downstream pathway of CXCL1/CXCR2 signalling with either the selective AS605240 (5 µg/site, i.t.) or the non-selective wortmannin PI3K inhibitor (0.4 µg/site, i.t.). Overall, the data indicates that the up-regulation of CXCL1 is important for the development and maintenance of PTX-induced neuropathic pain in mice. Therefore, the spinal blockage of CXCL1/CXCR2 signalling might be a new innovative therapeutic approach to treat this clinical side effect of PTX.


Asunto(s)
Antineoplásicos Fitogénicos/efectos adversos , Quimiocina CXCL1/antagonistas & inhibidores , Paclitaxel/efectos adversos , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Receptores de Interleucina-8B/antagonistas & inhibidores , Médula Espinal/efectos de los fármacos , Animales , Quimiocina CXCL1/metabolismo , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Compuestos de Fenilurea/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Quinoxalinas/farmacología , Receptores de Interleucina-8B/metabolismo , Transducción de Señal/efectos de los fármacos , Médula Espinal/metabolismo , Tiazolidinedionas/farmacología
9.
Theranostics ; 9(3): 853-867, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30809313

RESUMEN

Rationale: The expression of the chemokine (C-X-C motif) ligand 1 (CXCL1), an inflammatory protein, has been reported to be up-regulated in many human cancers. The mechanisms through which aberrant cellular CXCL1 levels promote specific steps in tumor growth and progression are unknown. Methods: We described the anticancer effects and mechanism of action of HL2401, a monoclonal antibody directed at CXCL1 with in vitro and in vivo data on bladder and prostate cancers. Results: HL2401 inhibited proliferation and invasion of bladder and prostate cells along with disrupting endothelial sprouting in vitro. Furthermore, novel mechanistic investigations revealed that CXCL1 expression stimulated interleukin 6 (IL6) expression and repressed tissue inhibitor of metalloproteinase 4 (TIMP4). Systemic administration of HL2401 in mice bearing bladder and prostate xenograft tumors retarded tumor growth through the inhibition of cellular proliferation and angiogenesis along with an induction of apoptosis. Our findings reveal a previously undocumented relationship between CXCL1, IL6 and TIMP4 in solid tumor biology. Principal conclusions: Taken together, our results argue that CXCL1 plays an important role in sustaining the growth of bladder and prostate tumors via up-regulation of IL6 and down-regulation of TIMP4. Targeting these critical interactions with a CXCL1 monoclonal antibody offers a novel strategy to therapeutically manage bladder and prostate cancers.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antineoplásicos Inmunológicos/administración & dosificación , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL1/antagonistas & inhibidores , Interleucina-6/antagonistas & inhibidores , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Neoplasias de la Próstata/patología , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Trasplante Heterólogo , Resultado del Tratamiento , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/patología , Inhibidor Tisular de Metaloproteinasa-4
10.
Neuropsychopharmacol Rep ; 38(3): 145-148, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30175527

RESUMEN

AIM: We have previously demonstrated that upregulation of CC chemokines through dopamine receptor signaling in the prefrontal cortex (PFC) underlies methamphetamine (Meth)-induced reward. Given the common pharmacological property of Meth and cocaine (Coca), which are highly addictive psychostimulants, we hypothesized that chemokines may also contribute to Coca-induced reward. The aim of this study was to identify a key chemokine-mediating Coca-induced reward in mice. METHODS: The mRNA expression levels of chemokines were measured by reverse transcription-quantitative polymerase chain reaction. Coca-induced reward was evaluated by conditioned place preference test. RESULTS: We found that mRNA expression levels of CC chemokine ligand 2 (CCL2), CCL7, and CXC chemokine ligand 1 (CXCL1) were upregulated in the PFC after a single administration of Coca (20 mg/kg, s.c.). Upregulation of CXCL1, but not CCL2 and CCL7, mRNA in the PFC was also observed after repeated administration of Coca. A single administration of dopamine D1 receptor agonist SKF 81297 (10 mg/kg, s.c.), but not D2 receptor agonist sumanirole, upregulated CXCL1 mRNA in the PFC. Coca-induced reward was attenuated by the pretreatment of SB 225002 (5 mg/kg, s.c.), a selective antagonist of CXC chemokine receptor 2 (CXCR2, cognate receptor for CXCL1). CONCLUSIONS: Collectively, we identified CXCL1 as a key regulator in Coca-induced reward and propose that pharmacological approach targeting CXCL1 could be a novel pharmacotherapy for Coca-induced reward.


Asunto(s)
Quimiocina CXCL1/genética , Trastornos Relacionados con Cocaína/metabolismo , Recompensa , Animales , Benzazepinas/farmacología , Bencimidazoles/farmacología , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/metabolismo , Cocaína/farmacología , Trastornos Relacionados con Cocaína/fisiopatología , Agonistas de Dopamina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Compuestos de Fenilurea/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
Inflammation ; 41(6): 2003-2011, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30073566

RESUMEN

The purpose of this study was to investigate the inhibition neuroinflammation mechanisms of hyperbaric oxygen therapy (HBOT). Primary astrocytes were incubated with lipopolysaccharide (LPS) after which they underwent HBOT and separate administration of inflammatory cytokine inhibitors. The respective expression of inflammatory factors was then detected. Results showed that LPS significantly induced increases in the expression levels of chemokine (C-X-C motif) ligand 1 (CXCL1), chemokine C-C motif ligand 2 (CCL2), phospho-nuclear factor-kappa B (p-NF-κB), phospho-c-Jun N-terminal kinase (p-JNK), phospho-extracellular signal-regulated kinase (p-ERK), and phospho-p38 (p-p38) in cultured astrocytes and peaked at 3 h. HBOT downregulated the expression of some inflammation mediators including CXCL1 and CCL2. Furthermore, HBOT inhibited the expression of some up-stream regulators of inflammation mediators including p-NF-κB, p-JNK, p-p38 (at 3 and 6 h), and p-ERK (3 h). Inhibitors of NF-κB, ERK, and JNK (BAY117082, PD98059, and SP600125) significantly suppressed the expression of CXCL1 and CCL2 that were induced by LPS for 3 h. However, the p38 inhibitor, SB203580, had no obvious effect on expression levels of CXCL1 and CCL2. In conclusion, we found that HBOT inhibits neuroinflammation via regulation of the LPS-induced NF-κB/mitogen-activated protein kinases (MAPKs, JNK, and ERK) -CCL2/CXCL1 signaling pathways.


Asunto(s)
Astrocitos/metabolismo , Oxigenoterapia Hiperbárica , Inflamación/prevención & control , Lipopolisacáridos/farmacología , Transducción de Señal , Animales , Astrocitos/patología , Células Cultivadas , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CXCL1/antagonistas & inhibidores , Humanos , Sistema de Señalización de MAP Quinasas , FN-kappa B/antagonistas & inhibidores
12.
Proc Natl Acad Sci U S A ; 115(38): E8948-E8957, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30158168

RESUMEN

Angiogenesis is essential in the early stage of solid tumor recurrence, but how a suspensive tumor is reactivated before angiogenesis is mostly unknown. Herein, we stumble across an interesting phenomenon that s.c. xenografting human lung cancer tissues can awaken the s.c. suspensive tumor in nude mice. We further found that a high level of insulin-like growth factor 1 (IGF1) was mainly responsible for triggering the transition from suspensive tumor to progressive tumor in this model. The s.c. suspensive tumor is characterized with growth arrest, avascularity, and a steady-state level of proliferating and apoptotic cells. Intriguingly, CD133+ lung cancer stem cells (LCSCs) are highly enriched in suspensive tumor compared with progressive tumor. Mechanistically, high IGF1 initiates LCSCs self-renewal from asymmetry to symmetry via the activation of a PI3K/Akt/ß-catenin axis. Next, the expansion of LCSC pool promotes angiogenesis by increasing the production of CXCL1 and PlGF in CD133+ LCSCs, which results in lung cancer recurrence. Clinically, a high level of serum IGF1 in lung cancer patients after orthotopic lung cancer resection as an unfavorable factor is strongly correlated with the high rate of recurrence and indicates an adverse progression-free survival. Vice versa, blocking IGF1 or CXCL1/PlGF with neutralizing antibodies can prevent the reactivation of a suspensive tumor induced by IGF1 stimulation in the mouse model. Collectively, the expansion of LCSC pool before angiogenesis induced by IGF1 is a key checkpoint during the initiation of cancer relapse, and targeting serum IGF1 may be a promising treatment for preventing recurrence in lung cancer patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neoplasias Pulmonares/patología , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/patología , Neovascularización Patológica/patología , Antígeno AC133/metabolismo , Animales , Carcinoma de Pulmón de Células no Pequeñas/sangre , Línea Celular Tumoral , Proliferación Celular , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/metabolismo , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/análisis , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Neoplasias Pulmonares/sangre , Ratones , Ratones Desnudos , Recurrencia Local de Neoplasia/sangre , Neovascularización Patológica/sangre , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Crecimiento Placentario/antagonistas & inhibidores , Factor de Crecimiento Placentario/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo
13.
Drug Deliv ; 25(1): 797-806, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29542355

RESUMEN

Ovarian cancer is the leading cause of cancer death among gynecological malignancies. The high mortality rate has not been significantly reduced despite advances in surgery and chemotherapy. Gene therapy shows therapeutic potential, but several key issues must be resolved before clinical application. To minimize toxicity in noncancerous tissues, tumor-specific ligands are conjugated to vectors to increase the selectivity of drug delivery. The expression pattern of follicle-stimulating hormone (FSH) receptor in normal and cancer tissues provides an opportunity for highly selective drug delivery in ovarian cancer. Furthermore, tumor-specific promoters can conditionally regulate therapeutic gene expression in tumor or normal tissues. The mucin 16 (MUC16) promoter might be a potential tool to drive ovarian cancer-localized gene expression since MUC16/CA125 is overexpressed in most ovarian carcinomas. Here, we screened the possible MUC16 promoter sequences and constructed MUC16 promoter-driven gro-α shRNA plasmid vectors. The vectors were specifically delivered into ovarian cancer cells via FSH peptide-conjugated nanoparticles. The predicted promoter sequence with TAAA repeats showed high transcriptional activity. The nanoparticle complex containing MUC16 promoter-driven gro-α shRNA and FSH peptides had the ability to decrease gro-α protein secretion in ovarian cancer cells and block tumor growth without obvious toxic effects in a nude mouse model bearing ovarian cancer. Our study provides a novel gene delivery system using a MUC16 promoter trigger and FSH peptide-mediated active targeting in ovarian cancer, and this system may be a promising strategy for specific genetic therapeutic delivery.


Asunto(s)
Hormona Folículo Estimulante de Subunidad beta/administración & dosificación , Proteínas de la Membrana/antagonistas & inhibidores , Nanopartículas/química , Neoplasias Ováricas/terapia , Regiones Promotoras Genéticas , ARN Interferente Pequeño/administración & dosificación , Tratamiento con ARN de Interferencia/métodos , Animales , Antígeno Ca-125/genética , Antígeno Ca-125/metabolismo , Carcinoma/metabolismo , Carcinoma/patología , Carcinoma/terapia , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Biología Computacional , Femenino , Hormona Folículo Estimulante de Subunidad beta/química , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Hormona Folículo Estimulante de Subunidad beta/uso terapéutico , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Electrónica de Transmisión , Nanopartículas/ultraestructura , Invasividad Neoplásica/prevención & control , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Tamaño de la Partícula , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/uso terapéutico , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/uso terapéutico , Propiedades de Superficie , Carga Tumoral
14.
Eur Heart J ; 39(20): 1818-1831, 2018 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-29514257

RESUMEN

Aims: Chemokine-mediated monocyte infiltration into the damaged heart represents an initial step in inflammation during cardiac remodelling. Our recent study demonstrates a central role for chemokine receptor CXCR2 in monocyte recruitment and hypertension; however, the role of chemokine CXCL1 and its receptor CXCR2 in angiotensin II (Ang II)-induced cardiac remodelling remain unknown. Methods and results: Angiotensin II (1000 ng kg-1 min-1) was administrated to wild-type (WT) mice treated with CXCL1 neutralizing antibody or CXCR2 inhibitor SB265610, knockout (CXCR2 KO) or bone marrow (BM) reconstituted chimeric mice for 14 days. Microarray revealed that CXCL1 was the most highly upregulated chemokine in the WT heart at Day 1 after Ang II infusion. The CXCR2 expression and the CXCR2+ immune cells were time-dependently increased in Ang II-infused hearts. Moreover, administration of CXCL1 neutralizing antibody markedly prevented Ang II-induced hypertension, cardiac dysfunction, hypertrophy, fibrosis, and macrophage accumulation compared with Immunoglobulin G (IgG) control. Furthermore, Ang II-induced cardiac remodelling and inflammatory response were also significantly attenuated in CXCR2 KO mice and in WT mice treated with SB265610 or transplanted with CXCR2-deficienct BM cells. Co-culture experiments in vitro further confirmed that CXCR2 deficiency inhibited macrophage migration and activation, and attenuated Ang II-induced cardiomyocyte hypertrophy and fibroblast differentiation through multiple signalling pathways. Notably, circulating CXCL1 level and CXCR2+ monocytes were higher in patients with heart failure compared with normotensive individuals. Conclusions: Angiotensin II-induced infiltration of monocytes in the heart is largely mediated by CXCL1-CXCR2 signalling which initiates and aggravates cardiac remodelling. Inhibition of CXCL1 and/or CXCR2 may represent new therapeutic targets for treating hypertensive heart diseases.


Asunto(s)
Cardiomegalia/metabolismo , Quimiocina CXCL1/fisiología , Monocitos/fisiología , Receptores de Interleucina-8B/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Angiotensina II , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Cardiomegalia/prevención & control , Movimiento Celular/fisiología , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/sangre , Femenino , Fibrosis , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Miocardio/patología , Receptores de Interleucina-8B/sangre , Receptores de Interleucina-8B/deficiencia , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
15.
Cell Rep ; 22(13): 3574-3586, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29590624

RESUMEN

Microglia are a key immune-competent cell type that respond to environmental and physiological changes during ischemic stroke. However, the molecular mechanisms controlling post-ischemic microglia activity are unclear. Understanding these mechanisms may ultimately reduce disease burden and allow the manipulation of microglia responses to shape the outcomes of stroke. Here, we report that, after experimentally induced stroke, ZEB1 is highly expressed in ipsilateral cerebral hemisphere, where it is upregulated mainly in microglia. Using a conditional transgenic mouse, we found that ZEB1 upregulation in microglia regulates immune responses in the CNS and alleviates brain injury after ischemic stroke. Our data indicate that ZEB1 overexpression mediates microglia responses and, in turn, inhibits the production of astrocytic CXCL1 through the TGF-ß1-dependent pathway. Reduced CXCL1 leads to a decline in neutrophil infiltration into the brain, thereby reducing CNS inflammation. Our results demonstrate the importance of ZEB1 in microglia-orchestrated neuroinflammation and suggest a potential means for reducing stroke-induced neurological injury.


Asunto(s)
Isquemia Encefálica/metabolismo , Microglía/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Enfermedad Aguda , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/inmunología , Microglía/patología , Regulación hacia Arriba , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/inmunología
16.
Artículo en Inglés | MEDLINE | ID: mdl-29410948

RESUMEN

CXCL-1, also called keratinocyte-derived cytokine (KC), is a predominant chemokine produced in glial cells upon infection with Theiler's murine encephalomyelitis virus (TMEV). In this study, we assessed the role of KC in the development of TMEV-induced demyelinating disease by utilizing polyclonal anti-KC antibodies as well as KC-expressing recombinant TMEV. Our results indicate that the level of KC produced after infection with TMEV or stimulation with various TLRs is significantly higher in various cells from susceptible SJL mice compared to those in cells from resistant B6 mice. SJL mice treated with rabbit anti-KC antibodies displayed accelerated development of TMEV-induced demyelinating disease, elevated viral loads in the CNS and decreased antiviral T cell responses. In addition, infection of susceptible SJL mice with recombinant KC-TMEV produced biologically active KC, which resulted in the accelerated pathogenesis of demyelinating disease and elevated T cell responses to viral antigens compared to mice infected with control recombinant HEL-TMEV. These results strongly suggest that both the lack of KC during TMEV infection and the excessive presence of the chemokine promote the pathogenesis of demyelinating disease. Therefore, a balance in the level of KC during TMEV infection appears to be critically important in controlling the pathogenesis of demyelinating disease.


Asunto(s)
Infecciones por Cardiovirus/complicaciones , Infecciones por Cardiovirus/virología , Quimiocina CXCL1/metabolismo , Enfermedades Desmielinizantes/etiología , Enfermedades Desmielinizantes/metabolismo , Theilovirus/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Astrocitos/inmunología , Astrocitos/metabolismo , Astrocitos/virología , Quimiocina CXCL1/antagonistas & inhibidores , Enfermedades Desmielinizantes/diagnóstico , Enfermedades Desmielinizantes/tratamiento farmacológico , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/farmacología , Interferón gamma/biosíntesis , Queratinocitos/metabolismo , Queratinocitos/virología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/virología , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/patología , Médula Espinal/virología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
17.
J Neurointerv Surg ; 10(1): 93-97, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28196918

RESUMEN

INTRODUCTION: Macrophages and neutrophils have been separately implicated in cerebral aneurysm formation. The interactions between different myeloid subsets and the contributions of macrophage phenotypes in these lesions over time are not known. The purpose of the study was to examine macrophage phenotypic changes in cerebral aneurysms. METHODS: We induced aneurysm formation in C57BL/6 mice and quantified contributions of M1 and M2 macrophages in aneurysm specimens with or without neutrophil blockade. In our aneurysm model, the left common carotid and right renal arteries were ligated, and mice were placed on a hypertensive high fat diet. One week later, stereotactic injection with elastase solution into the basal cisterns was performed. An angiotensin II secreting osmotic pump was implanted. The mice were then treated with anti-CXCL1 antibody or IgG control antibody. Animals were euthanized at 3 days, or 1 or 2 weeks. The circle of Willis was analyzed using immunohistochemistry for M1 and M2 macrophage phenotype contributions. RESULTS: Proinflammatory M1/M2 ratio increased in cerebral aneurysm formation over time, from 0.56 at 3 days to 1.75 at 2 weeks (p<0.0001). In contrast, anti-CXCL1 antibody blockade led to polarization towards an anti-inflammatory phenotype with an M1/M2 ratio of 0.95 at 2 weeks compared with IgG treated mice (p=0.0007). CONCLUSIONS: CXCL1 dependent neutrophil inflammation appears to have an important role in macrophage polarization to M1 phenotype in cerebral aneurysm development.


Asunto(s)
Aneurisma Intracraneal/inmunología , Aneurisma Intracraneal/patología , Macrófagos/inmunología , Macrófagos/patología , Animales , Antiinflamatorios/administración & dosificación , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/inmunología , Femenino , Inmunoglobulina G/administración & dosificación , Inmunoglobulinas Intravenosas/administración & dosificación , Aneurisma Intracraneal/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fenotipo
18.
Biol Pharm Bull ; 40(5): 729-732, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28458362

RESUMEN

Chemokine receptors CXCR1 and CXCR2 are conserved between guinea pigs and humans, but the distinct role of each receptor in chemotactic responses of neutrophils against chemokine ligands has not been elucidated due in part to the lack of specific inhibitors against these receptors in guinea pigs. In this study, we investigated the roles of guinea pig CXCR1 and CXCR2 on neutrophils in chemotactic responses to guinea pig interleukin (IL)-8 and growth-regulated oncogene (GRO)α by using specific inhibitory antibodies against these receptors. Neutrophil migration induced by IL-8 was partially inhibited by either anti-CXCR1 antibody or anti-CXCR2 antibody. In addition, the migration was inhibited completely when both anti-CXCR1 and anti-CXCR2 antibodies were combined. On the other hand, neutrophil migration induced by GROα was not inhibited by anti-CXCR1 antibody while inhibited profoundly by anti-CXCR2 antibody. These results indicated that CXCR1 and CXCR2 mediated migration induced by the IL-8 synergistically and only CXCR2 mediated migration induced by GROα in guinea pig neutrophils. Our findings on ligand selectivity of CXCR1 and CXCR2 in guinea pigs are consistent with those in humans.


Asunto(s)
Quimiocina CXCL1/farmacología , Interleucina-8/farmacología , Neutrófilos/fisiología , Receptores de Interleucina-8A/fisiología , Receptores de Interleucina-8B/fisiología , Animales , Anticuerpos Bloqueadores/farmacología , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL1/antagonistas & inhibidores , Quimiotaxis/efectos de los fármacos , Femenino , Cobayas , Enfermedades del Sistema Inmune , Interleucina-8/antagonistas & inhibidores , Trastornos Leucocíticos , Neutrófilos/inmunología , Receptores de Interleucina-8A/inmunología , Receptores de Interleucina-8B/inmunología
19.
J Cell Mol Med ; 21(7): 1411-1419, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28176455

RESUMEN

Adiponectin (APN) is known to promote the osteogenic differentiation of human jaw bone marrow mesenchymal stem cells (h-JBMMSCs). However, the underlying mechanism has not been fully elucidated. Previously, we showed that APN could promote h-JBMMSC osteogenesis via APPL1-p38 by up-regulating osteogenesis-related genes. Here, we aimed to determine whether APN could promote h-JBMMSC chemotaxis through CXCL1/CXCL8. The CCK-8, wound healing and transwell assays were used to evaluate the proliferation, migration and chemotaxis of h-JBMMSCs with or without APN treatment. Chemotaxis-related genes were screened using RNA-seq, and the results were validated using real-time PCR and ELISA. We also performed Western blot using the AMPK inhibitor, WZ4003, and the p38 MAPK inhibitor, SB203580, to identify the signalling pathway involved. We found that APN could promote h-JBMMSC chemotaxis in the co-culture transwell system. CXCL1 and CXCL8 were screened and confirmed as the up-regulated target genes. The APN-induced CXCL1/8 up-regulation to promote chemotaxis could be blocked by CXCR2 inhibitor SB225002. Western blot revealed that the phosphorylation of AMPK and p38 MAPK increased in a time-dependent manner with APN treatment. Additionally, WZ4003 and SB203580 could suppress the APN-induced overexpression of CXCL1 and CXCL8. The results of the transwell chemotaxis assay also supported the above results. Our data suggest that APN can promote h-JBMMSC chemotaxis by up-regulating CXCL1 and CXCL8.


Asunto(s)
Adiponectina/administración & dosificación , Quimiocina CXCL1/genética , Interleucina-8/genética , Desarrollo Maxilofacial/genética , Osteogénesis/genética , Adiponectina/metabolismo , Anilidas/farmacología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL1/antagonistas & inhibidores , Quimiotaxis/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-8/antagonistas & inhibidores , Desarrollo Maxilofacial/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/genética
20.
J Leukoc Biol ; 100(5): 1125-1134, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27286792

RESUMEN

During intraocular bacterial infections, the primary innate responders are neutrophils, which may cause bystander damage to the retina or perturb the clarity of the visual axis. We hypothesized that cytokine IL-6 and chemokine CXCL1 contributed to rapid neutrophil recruitment during Bacillus cereus endophthalmitis, a severe form of intraocular infection that is characterized by explosive inflammation and retinal damage that often leads to rapid vision loss. To test this hypothesis, we compared endophthalmitis pathogenesis in C57BL/6J, IL-6-/-, and CXCL1-/- mice. Bacterial growth in eyes of CXCL1-/-, IL-6-/-, and C67BL/6J mice was similar. Retinal function retention was greater in eyes of IL-6-/- and CXCL1-/- mice compared with that of C57BL/6J, despite these eyes having similar bacterial burdens. Neutrophil influx into eyes of CXCL1-/- mice was reduced to a greater degree compared with that of eyes of IL6-/- mice. Histology confirmed significantly less inflammation in eyes of CXCL1-/- mice, but similar degrees of inflammation in IL6-/- and C57BL/6J eyes. Because inflammation was reduced in eyes of infected CXCL1-/- mice, we tested the efficacy of anti-CXCL1 in B. cereus endophthalmitis. Retinal function was retained to a greater degree and there was less overall inflammation in eyes treated with anti-CXCL1, which suggested that anti-CXCL1 may have therapeutic efficacy in limiting inflammation during B. cereus endophthalmitis. Taken together, our results indicate that absence of IL-6 did not affect overall pathogenesis of endophthalmitis. In contrast, absence of CXCL1, in CXCL1-/- mice or after anti-CXCL1 treatment, led to an improved clinical outcome. Our findings suggest a potential benefit in targeting CXCL1 to control inflammation during B. cereus and perhaps other types of intraocular infections.


Asunto(s)
Bacillus cereus , Quimiocina CXCL1/fisiología , Quimiotaxis de Leucocito/fisiología , Endoftalmitis/inmunología , Infecciones Bacterianas del Ojo/inmunología , Interleucina-6/fisiología , Neutrófilos/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Bacillus cereus/crecimiento & desarrollo , Bacillus cereus/aislamiento & purificación , Carga Bacteriana , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/deficiencia , Quimiocina CXCL1/genética , Electrorretinografía , Endoftalmitis/microbiología , Infecciones Bacterianas del Ojo/microbiología , Mediadores de Inflamación/análisis , Interleucina-6/deficiencia , Interleucina-6/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peroxidasa/análisis , Retina/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA