Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 195
Filtrar
1.
Int J Mol Sci ; 24(13)2023 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-37446007

RESUMEN

Some chemoattractants and leukocytes such as M1 and M2 macrophages are known to be involved in the development of glomerulosclerosis during diabetic nephropathy (DN). In the course of diabetes, an altered and defective cellular metabolism leads to the increase in adenosine levels, and thus to changes in the polarity (M1/M2) of macrophages. MRS1754, a selective antagonist of the A2B adenosine receptor (A2BAR), attenuated glomerulosclerosis and decreased macrophage-myofibroblast transition in DN rats. Therefore, we aimed to investigate the effect of MRS1754 on the glomerular expression/secretion of chemoattractants, the intraglomerular infiltration of leukocytes, and macrophage polarity in DN rats. Kidneys/glomeruli of non-diabetic, DN, and MRS1754-treated DN rats were processed for transcriptomic analysis, immunohistopathology, ELISA, and in vitro macrophage migration assays. The transcriptomic analysis identified an upregulation of transcripts and pathways related to the immune system in the glomeruli of DN rats, which was attenuated using MRS1754. The antagonism of the A2BAR decreased glomerular expression/secretion of chemoattractants (CCL2, CCL3, CCL6, and CCL21), the infiltration of macrophages, and their polarization to M2 in DN rats. The in vitro macrophages migration induced by conditioned-medium of DN glomeruli was significantly decreased using neutralizing antibodies against CCL2, CCL3, and CCL21. We concluded that the pharmacological blockade of the A2BAR decreases the transcriptional expression of genes/pathways related to the immune response, protein expression/secretion of chemoattractants, as well as the infiltration of macrophages and their polarization toward the M2 phenotype in the glomeruli of DN rats, suggesting a new mechanism implicated in the antifibrotic effect of MRS1754.


Asunto(s)
Acetamidas , Antagonistas del Receptor de Adenosina A2 , Polaridad Celular , Factores Quimiotácticos , Nefropatías Diabéticas , Glomérulos Renales , Macrófagos , Purinas , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/inmunología , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/metabolismo , Factores Quimiotácticos/antagonistas & inhibidores , Factores Quimiotácticos/genética , Factores Quimiotácticos/metabolismo , Polaridad Celular/efectos de los fármacos , Polaridad Celular/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Antagonistas del Receptor de Adenosina A2/farmacología , Receptor de Adenosina A2B , Acetamidas/farmacología , Purinas/farmacología , Animales , Ratas , Movimiento Celular/efectos de los fármacos , Masculino , Ratas Sprague-Dawley , Transcripción Genética/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Inmunidad/efectos de los fármacos , Inmunidad/genética
2.
ChemMedChem ; 16(18): 2851-2863, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34047071

RESUMEN

In silico approaches identified 1, N-(6-((4-bromo- benzyl)amino)hexyl)-3,5-bis(trifluoromethyl)benzene sulfonamide, as a potential inhibitor of the S100A2-p53 protein-protein interaction, a validated pancreatic cancer drug target. Subsequent cytotoxicity screening revealed it to be a 2.97 µM cell growth inhibitor of the MiaPaCa-2 pancreatic cell line. This is in keeping with our hypothesis that inhibiting this interaction would have an anti-pancreatic cancer effect with S100A2, the validated PC drug target. A combination of focused library synthesis (three libraries, 24 compounds total) and cytotoxicity screening identified a propyl alkyl diamine spacer as optimal; the nature of the terminal phenyl substituent had limited impact on observed cytotoxicity, whereas N-methylation was detrimental to activity. In total 15 human cancer cell lines were examined, with most analogues showing broad-spectrum activity. Near uniform activity was observed against a panel of six pancreatic cancer cell lines: MiaPaCa-2, BxPC-3, AsPC-1, Capan-2, HPAC and PANC-1. In all cases there was good to excellent correlation between the predicted docking pose in the S100A2-p53 binding groove and the observed cytotoxicity, especially in the pancreatic cancer cell line with high endogenous S100A2 expression. This supports S100A2 as a pancreatic cancer drug target.


Asunto(s)
Antineoplásicos/farmacología , Factores Quimiotácticos/antagonistas & inhibidores , Proteínas S100/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Factores Quimiotácticos/metabolismo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Proteínas S100/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
3.
ChemMedChem ; 16(18): 2864-2881, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34047450

RESUMEN

In silico screening predicted 1 (N-(4-((4-(3-(4-(3-methoxyphenyl)-1H-1,2,3-triazol-1-yl)propyl)piperazin-1-yl) sulfonyl)-phenyl)acetamide) as an inhibitor of the S100A2-p53 protein-protein interaction. S100A2 is a validated pancreatic cancer drug target. In the MiaPaCa-2 pancreatic cell line, 1 was a ∼50 µM growth inhibitor. Synthesis of five focused compound libraries and cytotoxicity screening revealed increased activity from the presence of electron withdrawing moieties on the sulfonamide aromatic ring, with the 3,5-bis-CF3 Library 3 analogues the most active, with GI50 values of 0.91 (3-ClPh; 13 i; BxPC-3, Pancreas) to 9.0 µM (4-CH3 ; 13 d; PANC-1, Pancreas). Activity was retained against an expanded pancreatic cancer cell line panel (MiaPaCa-2, BxPC-3, AsPC-1, Capan-2, PANC-1 and HPAC) and the normal cell line MCF10A (breast). Bulky 4-disposed substituents on the terminal phenyl ring enhanced broad spectrum activity with growth inhibition values spanning 1.1 to 3.1 µM (4-C(CH3 )3 ; 13 e; BxPC-3 and AsPC-1 (pancreas), respectively). Central alkyl spacer contraction from propyl to ethyl proved detrimental to activity with Library 4 and 5.5- to 10-fold less cytotoxic than the propyl linked Library 2 and Library 3. The data herein was consistent with the predicted binding poses of the compounds evaluated. The highest levels of cytotoxicity were observed with those analogues best capable of adopting a near identical pose to the p53-peptide in the S100A2-p53 binding groove.


Asunto(s)
Antineoplásicos/farmacología , Factores Quimiotácticos/antagonistas & inhibidores , Proteínas S100/antagonistas & inhibidores , Triazoles/farmacología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Factores Quimiotácticos/metabolismo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Proteínas S100/metabolismo , Relación Estructura-Actividad , Triazoles/síntesis química , Triazoles/química , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
4.
J Med Chem ; 61(14): 5934-5948, 2018 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-29972644

RESUMEN

5-Oxo-6,8,11,14-eicosatetraenoic acid (5-oxo-ETE) is a potent lipid mediator that induces tissue eosinophilia via the selective OXE receptor (OXE-R), which is an attractive therapeutic target in eosinophilic diseases. We previously identified indole OXE-R antagonists that block 5-oxo-ETE-induced primate eosinophil activation. Although these compounds possess good oral absorption, their plasma levels decline rapidly due to extensive oxidation of their hexyl side chain. We have now succeeded in dramatically increasing antagonist potency and resistance to metabolism by replacing the hexyl group with phenylpentyl or phenylhexyl side chains. Compared with our previous lead compound S-230, our most potent antagonist, S-C025, has an IC50 (120 pM) over 80 times lower and a substantially longer plasma half-life. A single major metabolite, which retains antagonist activity (IC50, 690 pM) and has a prolonged lifetime in plasma was observed. These new highly potent OXE-R antagonists may provide a novel strategy for the treatment of eosinophilic disorders like asthma.


Asunto(s)
Ácidos Araquidónicos/antagonistas & inhibidores , Factores Quimiotácticos/antagonistas & inhibidores , Granulocitos/citología , Granulocitos/efectos de los fármacos , Ácidos Pentanoicos/farmacología , Receptores Eicosanoides/antagonistas & inhibidores , Animales , Calcio/metabolismo , Femenino , Humanos , Concentración 50 Inhibidora , Macaca fascicularis , Ácidos Pentanoicos/química , Ácidos Pentanoicos/metabolismo , Ácidos Pentanoicos/farmacocinética , Estereoisomerismo , Distribución Tisular
5.
Biochem Pharmacol ; 138: 107-118, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28476332

RESUMEN

We have developed a selective indole antagonist (230) targeting the OXE receptor for the potent eosinophil chemoattractant 5-oxo-ETE (5-oxo-6,8,11,14-eicosatetraenoic acid), that may be useful for the treatment of eosinophilic diseases such as asthma. In previous studies we identified ω2-oxidation of the hexyl side chain of racemic 230 as a major metabolic route in monkeys, but also obtained evidence for another pathway that appeared to involve hydroxylation of the hexyl side chain close to the indole. The present study was designed to investigate the metabolism of the active S-enantiomer of 230 (S230) and to identify the novel hydroxy metabolite and its chirality. Following oral administration, S230 rapidly appeared in the blood along with metabolites formed by a novel and highly stereospecific α-hydroxylation pathway, resulting in the formation of αS-hydroxy-S230. The chirality of α-hydroxy-S230 was determined by the total synthesis of the relevant diastereomers. Of the four possible diastereomers of α-hydroxy-230 only αS-hydroxy-S230 has significant OXE receptor antagonist activity and only this diastereomer was found in significant amounts in blood following oral administration of S230. Other novel metabolites of S230 identified in plasma by LC-MS/MS were αS,ω2-dihydroxy-S230 and glucuronides of S230 and ω2-hydroxy-S230. Thus the alkyl side chain of S230, which is essential for its antagonist activity, is also the major target of the metabolic enzymes that terminate its antagonist activity. Modification of this side chain might result in the development of related antagonists with improved metabolic stability and efficacy.


Asunto(s)
Antiasmáticos/farmacocinética , Antiinflamatorios no Esteroideos/farmacocinética , Ácidos Araquidónicos/antagonistas & inhibidores , Factores Quimiotácticos/antagonistas & inhibidores , Indoles/farmacocinética , Cetoácidos/farmacocinética , Receptores Eicosanoides/antagonistas & inhibidores , Administración Oral , Alquilación , Animales , Antiasmáticos/administración & dosificación , Antiasmáticos/sangre , Antiasmáticos/farmacología , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/sangre , Antiinflamatorios no Esteroideos/farmacología , Ácidos Araquidónicos/metabolismo , Factores Quimiotácticos/metabolismo , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Eosinófilos/metabolismo , Femenino , Glucurónidos/sangre , Glucurónidos/química , Glucurónidos/farmacología , Humanos , Hidroxilación , Inactivación Metabólica , Indoles/administración & dosificación , Indoles/sangre , Indoles/química , Indoles/farmacología , Cetoácidos/administración & dosificación , Cetoácidos/sangre , Cetoácidos/química , Cetoácidos/farmacología , Macaca fascicularis , Estructura Molecular , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores Eicosanoides/agonistas , Receptores Eicosanoides/metabolismo , Estereoisomerismo
6.
Nat Commun ; 8: 14252, 2017 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-28165452

RESUMEN

Novel therapeutics are required for improving the management of chronic inflammatory diseases. Aptamers are single-stranded RNA or DNA molecules that have recently shown utility in a clinical setting, as they can specifically neutralize biomedically relevant proteins, particularly cell surface and extracellular proteins. The nuclear chromatin protein DEK is a secreted chemoattractant that is abundant in the synovia of patients with juvenile idiopathic arthritis (JIA). Here, we show that DEK is crucial to the development of arthritis in mouse models, thus making it an appropriate target for aptamer-based therapy. Genetic depletion of DEK or treatment with DEK-targeted aptamers significantly reduces joint inflammation in vivo and greatly impairs the ability of neutrophils to form neutrophil extracellular traps (NETs). DEK is detected in spontaneously forming NETs from JIA patient synovial neutrophils, and DEK-targeted aptamers reduce NET formation. DEK is thus key to joint inflammation, and anti-DEK aptamers hold promise for the treatment of JIA and other types of arthritis.


Asunto(s)
Aptámeros de Nucleótidos/uso terapéutico , Artritis Juvenil/terapia , Factores Quimiotácticos/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Trampas Extracelulares/inmunología , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Proteínas de Unión a Poli-ADP-Ribosa/genética , Adulto , Animales , Artritis Juvenil/inmunología , Factores Quimiotácticos/genética , Factores Quimiotácticos/inmunología , Factores Quimiotácticos/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/inmunología , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Trampas Extracelulares/metabolismo , Femenino , Voluntarios Sanos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Proteínas Oncogénicas/inmunología , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/inmunología , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Cultivo Primario de Células , Líquido Sinovial/química , Líquido Sinovial/citología , Líquido Sinovial/inmunología , Zimosan/inmunología
7.
Viral Immunol ; 29(2): 105-11, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26771290

RESUMEN

Hepatitis E virus (HEV) is one of the primary causative agents of acute hepatitis. It is noteworthy that HEV can develop chronic infection and even lead to liver cirrhosis; however, the mechanism has not been revealed. In this study, the ELISA assay was used to detect protein levels, and we found that HEV open reading frame 3 (ORF3) protein inhibited the expression of proinflammatory cytokines (tumor necrosis factor-alpha [TNF-α], interleukin [IL]-1ß, IL-6, IL-8, IL-12p40, and IL-18) and chemotactic factors (nitric oxide [NO], interferon-inducible protein-10 (IP-10), macrophage inflammatory protein (MIP)-1α, monocyte chemoattractant protein-1 (MCP-1), granulocyte colony-stimulating factor (G-CSF), granulocyte macrophage colony-stimulating factor (GM-CSF)] in lipopolysaccharide (LPS)-stimulated human PMA-THP1 cells. Further study showed that mRNA and protein levels of pattern recognition receptors (PRRs), such as Toll-like receptor 4 (TLR4), TNF receptor-associated factor 6 (TRAF6), and nucleotide-binding oligomerization domain containing 2 (NOD2), decreased after infection of pLL3.7-ORF3 (pORF3); moreover, the inhibition produced corresponding upregulation of IκBα and downregulation of phosphorylated IκB kinase IKKɛ (p-IKKɛ) and phosphorylated nuclear factor (NF)-κB (p-NF-κB), but little variation was found in the concentration of IKKɛ and NF-κB. Taken together, our results demonstrated that HEV ORF3 attenuated LPS-induced cytokine production and chemotactic factors, predominantly by inhibiting various PRRs-mediated NF-κB signaling pathways. The anti-inflammatory properties might be of great importance to clarify the role and mechanism of macrophages in chronic HEV infection and cirrhosis.


Asunto(s)
Factores Quimiotácticos/antagonistas & inhibidores , Citocinas/antagonistas & inhibidores , Virus de la Hepatitis E/inmunología , Virus de la Hepatitis E/patogenicidad , Evasión Inmune , FN-kappa B/antagonistas & inhibidores , Proteínas Virales/metabolismo , Línea Celular , Perfilación de la Expresión Génica , Humanos , Lipopolisacáridos/inmunología , Proteoma/análisis
8.
J Dent Res ; 94(1): 166-74, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25359783

RESUMEN

Complement system, a major component of the natural immunity, has been recently identified as an important mediator of the dentin-pulp regeneration process through STRO-1 pulp cell recruitment by the C5a active fragment. Moreover, it has been shown recently that under stimulation with lipoteichoic acid, a complex component of the Gram-positive bacteria cell wall, human pulp fibroblasts are able to synthesize all proteins required for complement activation. However, Gram-negative bacteria, which are also involved in tooth decay, are known as powerful activators of complement system and inflammation. Here, we investigated the role of Gram-negative bacteria-induced complement activation on the pulp progenitor cell recruitment using lipopolysaccharide (LPS), a major component of all Gram-negative bacteria. Our results show that incubating pulp fibroblasts with LPS induced membrane attack complex formation and C5a release in serum-free fibroblast cultures. The produced C5a binds to the pulp progenitor cells' membrane and induces their migration toward the LPS stimulation chamber, as revealed by the dynamic transwell migration assays. The inhibition of this migration by the C5aR-specific antagonist W54011 indicates that the pulp progenitor migration is mediated by the interaction between C5a and C5aR. Our findings demonstrate, for the first time, a direct interaction between the recruitment of progenitor pulp cells and the activation of complement system generated by pulp fibroblast stimulation with LPS.


Asunto(s)
Quimiotaxis/efectos de los fármacos , Activación de Complemento/efectos de los fármacos , Complemento C5a/efectos de los fármacos , Pulpa Dental/citología , Lipopolisacáridos/farmacología , Células Madre/efectos de los fármacos , Compuestos de Anilina/farmacología , Técnicas de Cultivo de Célula , Factores Quimiotácticos/antagonistas & inhibidores , Factores Quimiotácticos/inmunología , Complemento C5a/antagonistas & inhibidores , Complemento C5a/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/efectos de los fármacos , Caries Dental/microbiología , Pulpa Dental/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Bacterias Gramnegativas/fisiología , Humanos , Factores Inmunológicos/antagonistas & inhibidores , Factores Inmunológicos/inmunología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Tetrahidronaftalenos/farmacología
9.
Stem Cells ; 31(10): 2172-82, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23766088

RESUMEN

Bone is a dynamic tissue that is continuously remodeled through the action of formative osteoblasts and resorptive osteoclasts. Chemerin is a secreted protein that activates chemokine-like receptor 1 (CMKLR1), a G protein-coupled receptor expressed by various cell types including adipocytes, osteoblasts, mesenchymal stem cells (MSCs), and macrophages. Previously, we identified chemerin as a regulator of adipocyte and osteoblast differentiation of MSCs. Herein we examined the role of chemerin in Lin(-) Sca1(+) c-kit(+) CD34(+) hematopoietic stem cell (HSC) osteoclastogenesis. We found that HSCs expressed both chemerin and CMKLR1 mRNA and secreted chemerin protein into the extracellular media. Neutralization of chemerin with a blocking antibody beginning prior to inducing osteoclast differentiation resulted in a near complete loss of osteoclastogenesis as evidenced by reduced marker gene expression and matrix resorption. This effect was conserved in an independent model of RAW264.7 cell osteoclastogenesis. Reintroduction of chemerin by reversal of neutralization rescued osteoclast differentiation indicating that chemerin signaling is essential to permit HSC differentiation into osteoclasts but following blockade the cells maintained the potential to differentiate into osteoclasts. Mechanistically, neutralization of chemerin blunted the early receptor activator of nuclear factor-kappa B ligand induction of nuclear factor of activated T-cells 2 (NFAT2), Fos, Itgb3, and Src associated with preosteoclast formation. Consistent with a central role for NFAT2, induction or activation of NFAT2 by forced expression or stimulation of intracellular calcium release rescued the impairment of HSC osteoclastogenesis caused by chemerin neutralization. Taken together, these data support a novel autocrine/paracrine role for chemerin in regulating osteoclast differentiation of HSCs through modulating intracellular calcium and NFAT2 expression/activation.


Asunto(s)
Diferenciación Celular , Factores Quimiotácticos/fisiología , Células Madre Hematopoyéticas/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Animales , Anticuerpos/farmacología , Línea Celular , Quimiocinas , Factores Quimiotácticos/antagonistas & inhibidores , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Osteoclastos/metabolismo , Ligando RANK/fisiología , Receptores de Quimiocina , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
10.
Clin Cancer Res ; 19(14): 3764-75, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23719262

RESUMEN

PURPOSE: S100B is member of a multigenic family of Ca(2+)-binding proteins, which is overexpressed by gliomas. Recently, we showed that low concentrations of S100B attenuated microglia activation through the induction of Stat3. We hypothesized that overexpression of S100B in gliomas could promote tumor growth by modulating the activity of tumor-associated macrophages (TAM). EXPERIMENTAL DESIGN: We stably transfected GL261 glioma cell lines with constructs that overexpressed (S100B(high)) or underexpressed (S100B(low)) S100B and compared their growth characteristics to intracranial wild-type (S100B(wt)) tumors. RESULTS: Downregulation of S100B in gliomas had no impact on cell division in vitro but abrogated tumor growth in vivo. Interestingly, compared to S100B(low) tumors, S100B(wt) and S100B(high) intracranial gliomas exhibited higher infiltration of TAMs, stronger inflammatory cytokine expression, and increased vascularity. To identify the potential mechanisms involved, the expression of the S100B receptor, receptor for advanced glycation end products (RAGE), was evaluated in gliomas. Although S100B expression induced RAGE in vivo, RAGE ablation in mice did not significantly inhibit TAM infiltration into gliomas, suggesting that other pathways were involved in this process. To evaluate other mechanisms responsible for TAM chemoattraction, we then examined chemokine pathways and found that C-C motif ligand 2 (CCL2) was upregulated in S100B(high) tumors. Furthermore, analysis of The Cancer Genome Atlas's glioma data bank showed a positive correlation between S100B and CCL2 expression in human proneural and neural glioma subtypes, supporting our finding. CONCLUSIONS: These observations suggest that S100B promotes glioma growth by TAM chemoattraction through upregulation of CCL2 and introduces the potential utility of S100B inhibitors for glioma therapy.


Asunto(s)
Neoplasias Encefálicas/inmunología , Factores Quimiotácticos/metabolismo , Glioma/inmunología , Macrófagos/inmunología , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Caprilatos/farmacología , Línea Celular Tumoral , Proliferación Celular , Quimiocina CCL2/metabolismo , Factores Quimiotácticos/antagonistas & inhibidores , Factores Quimiotácticos/fisiología , Quimiotaxis , Activación Enzimática , Glioma/tratamiento farmacológico , Glioma/metabolismo , Glioma/patología , Humanos , Macrófagos/metabolismo , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/inmunología , Trasplante de Neoplasias , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100/antagonistas & inhibidores , Subunidad beta de la Proteína de Unión al Calcio S100/fisiología , Carga Tumoral , Regulación hacia Arriba
11.
Biochem J ; 447(1): 81-91, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22747445

RESUMEN

S100A2, an EF hand calcium-binding protein, is a potential biomarker in several cancers and is also a TGF-ß (transforming growth factor-ß)-regulated gene in melanoma and lung cancer cells. However, the mechanism of S100A2 regulation by TGF-ß and its significance in cancer progression remains largely unknown. In the present study we report the mechanism of S100A2 regulation by TGF-ß and its possible role in TGF-ß-mediated tumour promotion. Characterization of the S100A2 promoter revealed an AP-1 (activator protein-1) element at positions -1161 to -1151 as being the most critical factor for the TGF-ß1 response. Chromatin immunoprecipitation and electrophoretic mobility-shift assays confirmed the functional binding of the AP-1 complex, predominantly JunB, to the S100A2 promoter in response to TGF-ß1 in HaCaT keratinocytes. JunB overexpression markedly stimulated the S100A2 promoter which was blocked by the dominant-negative JunB and MEK1 [MAPK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) kinase 1] inhibitor, PD98059. Intriguingly, despite the presence of a putative SMAD-binding element, S100A2 regulation by TGF-ß1 was found to be SMAD3 independent. Interestingly, p53 protein and TGF-ß1 show synergistic regulation of the S100A2 promoter. Finally, knockdown of S100A2 expression compromised TGF-ß1-induced cell migration and invasion of Hep3B cells. Together our findings highlight an important link between the TGF-ß1-induced MAPK and p53 signalling pathways in the regulation of S100A2 expression and pro-tumorigenic actions.


Asunto(s)
Factores Quimiotácticos/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Invasividad Neoplásica/fisiopatología , Proteínas S100/genética , Factor de Crecimiento Transformador beta1/farmacología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Factores Quimiotácticos/antagonistas & inhibidores , Factores Quimiotácticos/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Interferente Pequeño/genética , Proteínas S100/antagonistas & inhibidores , Proteínas S100/fisiología , Factor de Transcripción AP-1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
12.
Int Immunopharmacol ; 12(1): 158-68, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22155625

RESUMEN

A recombinant complement anaphylatoxin, C4a, inhibited chemotaxis, respiratory burst and histamine release in mast cell-like HMC-1 cells that were treated with recombinant C5a anaphylatoxin. C4a also inhibited histamine release from HMC-1 cells that were induced by recombinant C3a. The inhibition of C5a- and C3a-induced leukocyte reactions by C4a was recapitulated in peripheral blood CD133(+) cell-derived differentiated mast cells. In HMC-1 cells, C4a inhibited cytoplasmic Ca(2+) influx, an event that precedes anaphylatoxin-induced chemotactic and secretary responses. A conditioned medium of HMC-1 cells after shortly treated with C4a also inhibited the anaphylatoxin-induced Ca(2+) influx even after removal of C4a, indicating that the effect of C4a is to liberate an autocrine inhibitor from the mast cells. The inhibitor secretion by C4a was prevented with pertussis toxin or with a phosphodiesterase inhibitor. Conversely, an adenylyl cyclase inhibitor reproduced the effect of C4a. C4a decreased the intracellular cyclic AMP concentration of HMC-1 cells, indicating that C4a elicited the Gi protein-adenylyl cyclase inhibition pathway. Neither C4a nor the conditioned medium, however, inhibited Ca(2+) influx and respiratory burst in C5a- or C3a-stimulated peripheral neutrophils, suggesting that these cells lack this inhibitory system. Additionally, in HMC-1 cells, C4a did not inhibit Ca(2+)-independent, Leu72Gln-C5a-stimulated chemotactic response. In agreement with this finding, C4a treatment inhibited ERK1/2 phosphorylation in HMC-1 cells stimulated with other anaphylatoxins but did not inhibit p38MAPK phosphorylation in cells stimulated with Leu72Gln-C5a. Taken together, these findings suggest that the autocrine inhibitory effect elicited by C4a is attributed to interruption of Ca(2+)-dependent intracellular signaling pathway.


Asunto(s)
Inhibidores de Adenilato Ciclasa , Anafilatoxinas/farmacología , Factores Quimiotácticos/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Factores Inmunológicos/farmacología , Adenilil Ciclasas/metabolismo , Anafilatoxinas/antagonistas & inhibidores , Calcio/metabolismo , Línea Celular , Células Cultivadas , Factores Quimiotácticos/antagonistas & inhibidores , Quimiotaxis/efectos de los fármacos , AMP Cíclico/metabolismo , Liberación de Histamina/efectos de los fármacos , Humanos , Iminas/farmacología , Factores Inmunológicos/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mastocitos/efectos de los fármacos , Mastocitos/fisiología , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/farmacología
13.
Clin Cancer Res ; 17(9): 2702-11, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21349996

RESUMEN

PURPOSE: To elucidate the role of Semaphorin-3F (SEMA3F), originally described as an axon guiding chemorepulsant implicated in nerve development, in the progression of colorectal carcinoma. EXPERIMENTAL DESIGN: SEMA3F and its receptor NRP2 were examined in 72 cases of human colorectal carcinoma specimens and cell lines LoVo, SW480, and SW620 with immunohistochemistry and Western blotting. SEMA3F mRNA expression in the frozen tissue specimens and cell lines was examined with quantitative reverse transcriptase-PCR. Confocal laser scanning microscopy was used for detection of cellular localization of the proteins by immunofluorescent staining. MTT assay, flow cytometry, cell adhesion and migration, and xenografts were used to evaluate biological significance of SEMA3F. RESULTS: SEMA3F was significantly reduced in colorectal carcinoma tissues and cell lines. Overexpression of SEMA3F resulted in reduced proliferation, adhesion to fibronectin, and migratory capability as well as reduced S-phase population and integrin αvß3 expression of SW480 colon cancer cells. In addition, SEMA3F-overexpressing cells exhibited diminished tumorigenesis when transplanted orthotopically in nude mice and reduced liver metastases. Moreover, transfection of siRNA targeting SEMA3F in colon cancer cells increased their tumorigenicity in vivo. CONCLUSIONS: Endogenous SEMA3F acts as a suppressor of the growth and metastasis of human colorectal cancer cells.


Asunto(s)
Carcinoma/patología , Proliferación Celular , Neoplasias Colorrectales/patología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Axones/metabolismo , Axones/fisiología , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/prevención & control , Línea Celular Tumoral , Inhibición de Migración Celular/genética , Proliferación Celular/efectos de los fármacos , Factores Quimiotácticos/antagonistas & inhibidores , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/prevención & control , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Terapia Genética , Humanos , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , ARN Interferente Pequeño/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Planta Med ; 77(7): 698-704, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21058242

RESUMEN

Flavonoids are polyphenols that are ubiquitous in plants and frequently consumed in the diet. They are suggested to have many beneficial actions on human health, including anti-inflammatory activity. Their properties have been studied in a number of cell types, but little is known about their effects on neutrophil biology. Consequently, we selected 25 flavonoids with different structural features to evaluate their in vitro inhibition of rat polymorphonuclear neutrophil (PMN) chemotaxis, employing a modified Boyden chamber. Migratory activity was measured towards a chemotactic stimulant, formyl-Met-Leu-Phe or lipopolysaccharide. Furthermore, the cytotoxic effect of flavonoids on PMNs was determined by the release of cytosolic lactate dehydrogenase (LDH). Ten flavonoids significantly retarded the migration of PMNs with at least one of the concentrations tested in a range between 0.625 and 100 µM; the best antichemotactic agents were flavone, flavonol, quercetin and rutin. None of the flavanones evaluated presented any significant inhibition of migration in this assay. Our findings indicated that non-hydroxylated flavones possess a better antichemotactic activity when compared to flavones with hydroxy groups. The presence of a sugar moiety in rutin did not produce any increase in this effect, when compared to the respective aglycone analogue. Finally, none of the flavonoids exhibited cell toxicity and for many of these flavonoids this is the first report of the inhibition of PMN chemotaxis.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Flavonoides/farmacología , Neutrófilos/efectos de los fármacos , Extractos Vegetales/farmacología , Achyrocline/química , Animales , Antiinflamatorios/farmacología , Inhibición de Migración Celular/efectos de los fármacos , Factores Quimiotácticos/antagonistas & inhibidores , Factores Quimiotácticos/farmacología , Citotoxinas/farmacología , Flavonoides/química , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , N-Formilmetionina Leucil-Fenilalanina/antagonistas & inhibidores , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/inmunología , Extractos Vegetales/química , Ratas
15.
Sci Rep ; 1: 123, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22355640

RESUMEN

Although various function of chemerin have been suggested, its physiological role remains to be elucidated. Here we show that chemerin-deficient mice are glucose intolerant irrespective of exhibiting reduced macrophage accumulation in adipose tissue. The glucose intolerance was mainly due to increased hepatic glucose production and impaired insulin secretion. Chemerin and its receptor ChemR23 were expressed in ß-cell. Studies using isolated islets and perfused pancreas revealed impaired glucose-dependent insulin secretion (GSIS) in chemerin-deficient mice. Conversely, chemerin transgenic mice revealed enhanced GSIS and improved glucose tolerance. Expression of MafA, a pivotal transcriptional factor for ß-cell function, was downregulated in chemerin-deficient islets and a chemerin-ablated ß-cell line and rescue of MafA expression restored GSIS, indicating that chemerin regulates ß-cell function via maintaining MafA expression. These results indicate that chemerin regulates ß-cell function and plays an important role in glucose homeostasis in a tissue-dependent manner.


Asunto(s)
Factores Quimiotácticos/fisiología , Células Secretoras de Insulina/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Tejido Adiposo/patología , Tejido Adiposo/fisiopatología , Animales , Línea Celular , Quimiocinas , Factores Quimiotácticos/antagonistas & inhibidores , Factores Quimiotácticos/deficiencia , Factores Quimiotácticos/genética , Dieta Alta en Grasa/efectos adversos , Técnicas de Silenciamiento del Gen , Prueba de Tolerancia a la Glucosa , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Macrófagos/patología , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Quimiocina , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
16.
J Pharm Pharmacol ; 61(12): 1699-707, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19958594

RESUMEN

OBJECTIVES: Huang-Lian-Jie-Du-Tang (HLJDT) is a traditional Chinese medicine with a long history of anti-inflammatory use, but its pharmacological effects have not been thoroughly investigated. This study aimed to evaluate the anti-inflammatory activity of HLJDT in vivo and in vitro. METHODS: The carrageenan rat air pouch model was used to investigate the anti-inflammatory action of HLJDT after oral administration. Moreover, we exploited a modified method based on liquid chromatography-tandem mass spectrometry (LC-MS/MS) technique to assay the effects of HLJDT on arachidonic acid metabolites. KEY FINDINGS: Our data demonstrate that oral administration of HLJDT significantly inhibited the inflammatory responses in carrageenan-injected rat air pouches, and also significantly reduced the production of nitric oxide (NO) and leukotriene B(4) (LTB(4)) in vivo, without any influence on biosynthesis of cyclooxygenase (COX)-derived eicosanoids. Similar behaviour of HLJDT was also observed by using calcium ionophore A23187-stimulated peritoneal macrophages, where HLJDT markedly inhibited eicosanoids derived from different lipoxygenases. The NO production and the mRNA expression of inducible nitric oxide synthase (iNOS) and chemotactic factors (CCL3, CCL4, CCL5 and CXCL2) were also inhibited by HLJDT in RAW 264.7 macrophages stimulated by lipopolysaccharide. CONCLUSIONS: Our data revealed, for the first time, that HLJDT could inhibit biosynthesis of eicosanoids derived from different lipoxygenases. Also, HLJDT may exert its anti-inflammatory effects by its suppression on eicosanoid generation, NO production and gene transcription of chemotactic factors, which supports its effectiveness in the treatment of inflammatory diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Medicamentos Herbarios Chinos/uso terapéutico , Eicosanoides/biosíntesis , Leucotrieno B4/biosíntesis , Lipooxigenasa/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Calcimicina , Carragenina , Línea Celular , Factores Quimiotácticos/antagonistas & inhibidores , Factores Quimiotácticos/metabolismo , Cromatografía Líquida de Alta Presión , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/farmacología , Inflamación/tratamiento farmacológico , Lipopolisacáridos , Macrófagos/efectos de los fármacos , Redes y Vías Metabólicas/efectos de los fármacos , Modelos Animales , Óxido Nítrico Sintasa de Tipo II/genética , Fitoterapia , Prostaglandina-Endoperóxido Sintasas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Eicosanoides/metabolismo , Espectrometría de Masas en Tándem
17.
Curr Pharm Des ; 15(16): 1854-67, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19519428

RESUMEN

Vascular system plays critical roles in tumor progression and metastasis. Tumor vessels generally sprout from preexisting vascular cells. In addition, pluripotent progenitor cells also participate in tumor neovascularization. The latter populations include endothelial progenitor cells, hematopoietic stem cells and mesenchymal stem cells that are stimulated and attracted into the lesion. Recent studies on tumor microenvironment have disclosed that BM (bone marrow)-derived progenitor cells contain unique subpopulations that do not become fully-differentiated vascular constituents; instead, they show the nature of immature myeloid or mesenchymal lineage, and they enhance tumor angiogenic milieu in close contact with tumor vessels. BM-derived cells also migrate into pre-metastatic niche and stimulate vascular beds of distant organ for attracting circulating tumor cells. Currently, several antiangiogenic molecules are under clinical trials and they are expected to improve overall prognosis. Humanized monoclonal antibody bevacizumab specifically targeting VEGF (vascular endothelial growth factor), and several tyrosine kinase inhibitors targeting VEGF receptors-mediated pathways are the most widely studied agents in several types of advanced cancers. It is obvious that VEGF contributes to tumor neovascularization as a mastermind molecule. On the other hand, the mechanism has also been elucidated how tumors evade VEGF targeting therapies. To establish safer and more effective antiangiogenic therapies, it is important to understand the cross-communication between tumors and hosts in proinflammatory milieu. In this review, we discuss features of tumor angiogenic vessels and their microenvironment. Recent topics on the contribution of BM-derived cells, complexities of VEGF-targeting approaches, and chemoattractants that activate tumor vascular beds are summarized.


Asunto(s)
Neoplasias/irrigación sanguínea , Neovascularización Patológica/fisiopatología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Factores Quimiotácticos/antagonistas & inhibidores , Factores Quimiotácticos/fisiología , Humanos , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo , Células Madre/patología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/fisiología
18.
Adv Drug Deliv Rev ; 61(4): 303-9, 2009 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-19385090

RESUMEN

Thioredoxin 1 (Trx 1) is a redox-active small protein ubiquitously present in human body. It is one of the defensive proteins induced in response to various stress conditions. In addition to its anti-oxidative effect by dithiol-disulfide exchange in its active site, Trx 1 has anti-apoptotic and anti-inflammatory effects. Trx 1 overexpression has been shown to be effective in a wide variety of animal models for oxidative and inflammatory disorders. An administration of recombinant Trx 1 protein is also effective in animal models especially for severe acute lung diseases where Trx 1 is likely to act with its anti-inflammatory properties. Trx 1 in circulation shows anti-chemotactic effects for neutrophils and inhibitory effects against macrophage migration inhibitory factor (MIF). Neovascularization is also suppressed by Trx 1 via inhibition of the complement activation. Here we discuss precise mechanisms of Trx 1 and potential therapeutic approach of this molecule.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Tiorredoxinas/administración & dosificación , Tiorredoxinas/uso terapéutico , Animales , Factores Quimiotácticos/antagonistas & inhibidores , Proteínas Inactivadoras de Complemento/administración & dosificación , Proteínas Inactivadoras de Complemento/fisiología , Proteínas Inactivadoras de Complemento/uso terapéutico , Citoprotección/fisiología , Modelos Animales de Enfermedad , Humanos , Neutrófilos/patología , Neutrófilos/fisiología , Tiorredoxinas/fisiología
19.
Respir Med ; 101(11): 2386-94, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17761412

RESUMEN

The driving force in the progression of COPD is the development of exacerbations which are mostly the result of excessive inflammation. Bronchodilatators play an important role in the treatment of COPD. The reported reduction in exacerbation rates in COPD is due to the inhibition of vagal-mediated bronchoconstriction and mucus secretion. However, recent studies have highlighted the existence of muscarinic receptors on inflammatory cells and we have explored the possibility that tiotropium bromide might also inhibit neutrophil migration. We analysed the influence of tiotropium on the release of neutrophil chemotactic activity in response to acetylcholine (ACh) and the expression of muscarinic receptors on human alveolar macrophages (AM), A549 cells, MonoMac6 cells, and human lung fibroblasts. We found significant levels of all muscarinic receptor subtypes on all analysed cells except the fibroblasts. Fibroblasts expressed predominantly M2, receptors and did not release chemotactic activity. AM, A549 cells, and MonoMac6 cells released chemotactic active mediators after incubation with ACh. The secretion could be suppressed by more than 70% after coincubation with tiotropium. Tiotropium alone did not influence the granulocyte migration. Most of the chemotactic activity could be attributed to leukotriene B4 (LTB4). The release of interleukin-8 (IL-8) and monocyte chemotactic protein-1 (MCP-1) was not induced by ACh. From this, we suggest that the suppression of the Ach-mediated release of chemotactic substances like LTB4 modulates the inflammatory reaction. This may contribute to the decreased rate of exacerbations in COPD, which was observed in clinical trials.


Asunto(s)
Factores Quimiotácticos/antagonistas & inhibidores , Antagonistas Colinérgicos/farmacología , Macrófagos Alveolares/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Derivados de Escopolamina/farmacología , Acetilcolina/fisiología , Anciano , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Técnicas In Vitro , Leucotrieno B4/fisiología , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Receptores Muscarínicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bromuro de Tiotropio
20.
J Immunol ; 178(12): 8148-57, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17548653

RESUMEN

The severe and protracted inflammation that characterizes acute lung injury (ALI) is driven by the ongoing recruitment of neutrophils to the lung. Although much of the cytokine signaling responsible for the initial phase of ALI has been elaborated, relatively little is known about the mechanisms governing the recruitment of neutrophils from the bone marrow to the lung in the later period of this disease. Given its previously described chemoattractant effects on marrow neutrophils, we investigated whether stromal-derived factor-1 (SDF-1) (CXCL12) might participate in this later phase of recruitment. Using immunohistochemistry to examine both banked human lung specimens from patients with ALI and lungs from mice with LPS-induced pneumonitis, we found that pulmonary SDF-1 expression increases during ALI. We further determined that both lung SDF-1 protein expression and mRNA expression rise in a delayed but sustained pattern in this mouse model and that the major source of the increase in expression appears to be the lung epithelium. Lastly, we found that expression of the SDF-1 receptor CXCR4 rises in a similar temporal pattern on neutrophils in both the blood and airspace of LPS-injured mice and that Ab-mediated SDF-1 blockade significantly attenuates late but not early pulmonary neutrophilia in this model. These results implicate SDF-1 in neutrophil recruitment to the lung in the later period of acute lung injury and suggest a novel role for this cytokine in coordinating the transition from the inflammatory response to the initiation of tissue repair.


Asunto(s)
Quimiocinas CXC/metabolismo , Factores Quimiotácticos/metabolismo , Pulmón/inmunología , Síndrome de Dificultad Respiratoria/inmunología , Animales , Membrana Celular/inmunología , Movimiento Celular , Quimiocina CXCL12 , Quimiocinas CXC/análisis , Quimiocinas CXC/genética , Factores Quimiotácticos/antagonistas & inhibidores , Factores Quimiotácticos/genética , Epitelio/química , Epitelio/inmunología , Femenino , Humanos , Lipopolisacáridos/toxicidad , Pulmón/química , Pulmón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Neumonía/inducido químicamente , Neumonía/inmunología , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Receptores CXCR4/análisis , Síndrome de Dificultad Respiratoria/inducido químicamente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...