Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Molecules ; 26(24)2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34946686

RESUMEN

Glioblastoma is an aggressive cancer, against which medical professionals are still quite helpless, due to its resistance to current treatments. Scorpion toxins have been proposed as a promising alternative for the development of effective targeted glioblastoma therapy and diagnostic. However, the exploitation of the long peptides could present disadvantages. In this work, we identified and synthetized AaTs-1, the first tetrapeptide from Androctonus australis scorpion venom (Aa), which exhibited an antiproliferative effect specifically against human glioblastoma cells. Both the native and synthetic AaTs-1 were endowed with the same inhibiting effect on the proliferation of U87 cells with an IC50 of 0.56 mM. Interestingly, AaTs-1 was about two times more active than the anti-glioblastoma conventional chemotherapeutic drug, temozolomide (TMZ), and enhanced its efficacy on U87 cells. AaTs-1 showed a significant similarity with the synthetic peptide WKYMVm, an agonist of a G-coupled formyl-peptide receptor, FPRL-1, known to be involved in the proliferation of glioma cells. Interestingly, the tetrapeptide triggered the dephosphorylation of ERK, p38, and JNK kinases. It also enhanced the expression of p53 and FPRL-1, likely leading to the inhibition of the store operated calcium entry. Overall, our work uncovered AaTs-1 as a first natural potential FPRL-1 antagonist, which could be proposed as a promising target to develop new generation of innovative molecules used alone or in combination with TMZ to improve glioblastoma treatment response. Its chemical synthesis in non-limiting quantity represents a valuable advantage to design and develop low-cost active analogues to treat glioblastoma cancer.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma , Oligopéptidos/farmacología , Receptores de Formil Péptido/biosíntesis , Receptores de Lipoxina/biosíntesis , Venenos de Escorpión/química , Proteína p53 Supresora de Tumor/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Animales , Antineoplásicos/química , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Humanos , Oligopéptidos/química , Escorpiones
2.
Ann Diagn Pathol ; 37: 62-66, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30286327

RESUMEN

Astrocytomas represent the majority of cerebral gliomas. Studies show that the anti-inflammatory protein Annexin-A1 (ANXA1) is associated with the tumor invasion process and that its actions can be mediated by the receptor for formylated peptides (FPR). Therefore, we evaluated the expression of ANXA1, the receptor FPR2 and matrix metalloproteinases 2 and 9 (MMP-2 and MMP-9) in brain astrocytomas. Detection of proteins was performed in sections of diffuse astrocytomas (grade II), anaplastic astrocytomas (grade III) and glioblastomas (GBM, grade IV) and quantifications were made by densitometry. Our analyses showed increased expression of ANXA1 in astrocytomas of all grades, but especially in GBM. The expression of FPR2 is similar to that found for ANXA1, being higher in GBM. Immunostaining for MMPs is also stronger as the degree of malignancy increases, especially with respect to MMP-9. The positive correlation between ANXA1/FPR2 and ANXA1/MMP-9 was observed in all tumors studied. The data indicate the possible action of ANXA1 and FPR2 on the development and progression of astrocytomas, related to increased expression of MMP-9. Thereby, ANXA1 and FPR2 are involved in the biology and malignancy of diffuse astrocytic tumors.


Asunto(s)
Anexina A1/biosíntesis , Astrocitoma/patología , Biomarcadores de Tumor/biosíntesis , Neoplasias Encefálicas/patología , Receptores de Formil Péptido/biosíntesis , Receptores de Lipoxina/biosíntesis , Adulto , Anciano , Femenino , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Persona de Mediana Edad
3.
Biochim Biophys Acta ; 1861(9 Pt A): 1025-1035, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27317426

RESUMEN

Resolution of inflammation is an active process involving a novel category of lipid factors known as specialized pro-resolving lipid mediators, which includes Resolvin D1 (RvD1). While accumulating evidence suggests that RvD1 counteracts proinflammatory signaling and promotes resolution, the specific cellular targets and mechanisms of action of RvD1 remain largely unknown. In the present study, we investigated the role and molecular mechanisms of RvD1 in ischemia/reperfusion (IR)-induced sterile liver inflammation. Male C57BL/6 mice underwent 70% hepatic ischemia for 60min, followed by reperfusion. RvD1 (5, 10, and 15µg/kg, i.p.) was administered to the mice 1h before ischemia and then immediately prior to reperfusion. RvD1 attenuated IR-induced hepatocellular damage and the proinflammatory response. In purified Kupffer cells (KCs) from mice exposed to IR, the levels of M1 marker genes (Nos2a and Cd40) increased, while those of M2 marker genes (Arg1, Cd206, and Mst1r) decreased, demonstrating a proinflammatory shift. RvD1 markedly attenuated these changes. Depletion of KCs by liposome clodronate abrogated the effects of RvD1 on proinflammatory mediators and macrophage polarization. In addition, RvD1 attenuated increases in myeloperoxidase activity and Cxcl1 and Cxcl2 mRNA expression. RvD1 markedly augmented the efferocytic activity of KCs, as indicated by increases in F4/80(+)Gr-1(+) cells in the liver. However, antagonist pretreatment or gene silencing of the RvD1 receptor, ALX/FPR2, abrogated the anti-inflammatory and pro-resolving actions of RvD1. These data indicate that RvD1 ameliorates IR-induced liver injury, and this protection is associated with enhancement of M2 polarization and efferocytosis via ALX/FPR2 activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Ácidos Docosahexaenoicos/administración & dosificación , Inflamación/tratamiento farmacológico , Receptores de Formil Péptido/genética , Daño por Reperfusión/tratamiento farmacológico , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Animales , Arginasa/genética , Quimiocina CXCL1/genética , Quimiocina CXCL2/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/metabolismo , Inflamación/patología , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/patología , Lectinas Tipo C/genética , Hígado/lesiones , Hígado/patología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Ratones , Fagocitosis/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Superficie Celular/genética , Receptores de Formil Péptido/biosíntesis , Daño por Reperfusión/patología , Transducción de Señal
4.
J Immunol ; 195(6): 2852-60, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26276873

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a common, progressive, and invariably lethal interstitial lung disease with no effective therapy. The key cell driving the development of fibrosis is the myofibroblast. Lipoxin A4 (LXA4) is an anti-inflammatory lipid, important in the resolution of inflammation, and it has potential antifibrotic activity. However, the effects of LXA4 on primary human lung myofibroblasts (HLMFs) have not previously been investigated. Therefore, the aim of this study was to examine the effects of LXA4 on TGF-ß1-dependent responses in IPF- and nonfibrotic control (NFC)-derived HLMFs. HLMFs were isolated from IPF and NFC patients and grown in vitro. The effects of LXA4 on HLMF proliferation, collagen secretion, α-smooth muscle actin (αSMA) expression, and Smad2/3 activation were examined constitutively and following TGF-ß1 stimulation. The LXA4 receptor (ALXR) was expressed in both NFC- and IPF-derived HLMFs. LXA4 (10(-10) and 10(-8) mol) reduced constitutive αSMA expression, actin stress fiber formation, contraction, and nuclear Smad2/3, indicating regression from a myofibroblast to fibroblast phenotype. LXA4 also significantly inhibited FBS-dependent proliferation and TGF-ß1-dependent collagen secretion, αSMA expression, and Smad2/3 nuclear translocation in IPF-derived HLMFs. LXA4 did not inhibit Smad2/3 phosphorylation. In summary, LXA4 attenuated profibrotic HLMF activity and promoted HLMF regression to a quiescent fibroblast phenotype. LXA4 or its stable analogs delivered by aerosol may offer a novel approach to the treatment of IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática/patología , Lipoxinas/farmacología , Miofibroblastos/metabolismo , Receptores de Formil Péptido/biosíntesis , Receptores de Lipoxina/biosíntesis , Factor de Crecimiento Transformador beta1/farmacología , Actinas/biosíntesis , Proliferación Celular , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Fibrosis Pulmonar Idiopática/inmunología , Inflamación/inmunología , Inflamación/patología , Pulmón/citología , Pulmón/patología , Fosforilación/efectos de los fármacos , ARN Mensajero/biosíntesis , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
5.
Anticancer Res ; 35(5): 2769-74, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25964556

RESUMEN

BACKGROUND/AIM: Formyl-peptide receptors (FPRs) are expressed in several tissues and cell types. The identification of markers involved in cell growth may further allow for molecular profiling of lung cancer. We investigated the possible role of FPRs as molecular markers in several types of lung carcinomas which is the main cause of cancer death worldwide. MATERIALS AND METHODS: Tumor tissue samples were collected from six patients affected by lung cancer. Biopsies were analyzed for expression of FPR isoforms both in tumoral and peritumoral tissue by real-time polymerase chain reaction (PCR), western blot and immunofluorescence. RESULTS: Real-time PCR, western blot and immunofluorescence analyses showed that FPR expression is lower in types of human lung cancer tissues when compared to the surrounding peritumoral tissues. CONCLUSION: The study of the mechanistic basis for the control of FPR expression in normal peritumoral versus tumoral tissues could provide the basis for new diagnostic and therapeutic interventions.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Neoplasias Pulmonares/genética , Receptores de Formil Péptido/biosíntesis , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/patología , Isoformas de Proteínas/biosíntesis , ARN Mensajero/biosíntesis , Receptores de Formil Péptido/genética
6.
Anticancer Res ; 34(5): 2223-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24778024

RESUMEN

BACKGROUND: Formyl peptide receptor 1 (FPR1) as a regulator of innate inflammatory response has been implicated in tumor progression of gliomas. The purpose of the present study was to evaluate the prognostic significance and the ligand-receptor interaction of FPR1 in gastric cancer (GC). PATIENTS AND METHODS: FPR1 was immunohistochemically-analyzed in tissue sections originating from 116 GC patients. Reverse transcription-polymerase chain reaction (RT-PCR) was used for the assessment of interaction between FPR1 and the FPR1 ligand annexin A1 (AnxA1) in GC cells. RESULTS: High FPR1 expression was significantly associated with stage IV disease, submucosal invasion, serosal invasion, and clinical outcome of GC. Multivariate analysis showed that high FPR1 expression was an independent risk factor of poor overall survival in GC patients. FPR1 expression increased significantly when AnxA1 overexpression was present in GC cells. A positive feedback regulation of FPR1 was involved in the AnxA1-FPR1 signal transduction. CONCLUSION: FPR1 expression may be used as a novel indicator to predict outcome in GC patients after gastrectomy.


Asunto(s)
Biomarcadores de Tumor/análisis , Receptores de Formil Péptido/biosíntesis , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Adulto , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Femenino , Gastrectomía/mortalidad , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Riesgo
7.
Ann Otol Rhinol Laryngol ; 122(11): 683-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24358628

RESUMEN

OBJECTIVES: The resolution of inflammation is an active process controlled by several anti-inflammatory and pro-resolution mediators. Lipoxin A4, an endogenous lipid mediator, is a potential pro-resolution mediator that could attenuate inflammation. This study was conducted to elucidate the role of lipoxin A4 in upper airway inflammation. METHODS: Nasal secretions were collected from patients with chronic rhinosinusitis with nasal polyposis, patients with allergic rhinitis, and control subjects. The concentration of lipoxin A4 was measured by enzyme-linked immunosorbent assay. Nasal tissues were obtained from nasal polyps and inferior turbinates during endonasal surgery. The mRNA expressions of lipoxygenases (LOXs), lipoxin receptor (formyl peptide receptor-like 1; FPRL-1), and cysteinyl leukotriene type 1 receptor (CysLT1R) in nasal tissues were examined by reverse-transcription polymerase chain reaction. Tissue localization of FPRL-1 was determined by immunohistochemical staining. The in vitro effect of lipoxin A4 on airway epithelial cells was also examined. RESULTS: A significant concentration of lipoxin A4 was found in nasal secretions, and the concentration was increased in patients with allergic rhinitis. The mRNA expressions of 5-LOX, 15-LOX-1, FPRL-1, and CysLT1R were significantly greater in nasal polyps than in inferior turbinates. FPRL-1 was localized in nasal epithelial cells. Lipoxin A4 inhibited tumor necrosis factor alpha-induced interleukin 8 release from airway epithelial cells via its receptor FPRL-1. CONCLUSIONS: These results indicate that lipoxin A4 may play a role in the resolution of upper airway inflammation. A low concentration of lipoxin A4 may be involved in chronic inflammation of the upper airways.


Asunto(s)
Regulación de la Expresión Génica , Lipoxinas/genética , Pólipos Nasales/genética , ARN Mensajero/genética , Rinitis/genética , Sinusitis/genética , Adulto , Anciano , Células Cultivadas , Eicosanoides , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunohistoquímica , Lipoxinas/biosíntesis , Masculino , Persona de Mediana Edad , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Pólipos Nasales/metabolismo , Receptores de Formil Péptido/biosíntesis , Receptores de Formil Péptido/genética , Receptores de Lipoxina/biosíntesis , Receptores de Lipoxina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rinitis/metabolismo , Rinitis/patología , Sinusitis/metabolismo , Sinusitis/patología
8.
Biochem Pharmacol ; 85(12): 1795-802, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23643932

RESUMEN

Lipoxin A4 (LXA4) has been described as an anti-inflammatory mediator, which exerts its effects through the formyl peptide receptor FPR2, also known as ALX. However, there has been a controversy whether or not cells expressing FPR2/ALX, such as neutrophils, respond to LXA4. We, therefore, systematically examined the ability of the human and murine forms of the receptor to respond to LXA4. We show that both receptor orthologues responded to the FPR2/ALX peptide agonist WKYMVM when expressed heterologously. In contrast, LXA4 from different sources neither increased [Ca²âº](i) and extracellular-signal-regulated kinase (ERK) phosphorylation, nor did it induce a decrease in cAMP levels or a translocation of ß-arrestin. Also, several LXA4 analogs were found to be unable to signal through FPR2/ALX. We conclude that FPR2/ALX is not activated by LXA4 and that the molecular mechanism by which LXA4 functions still needs to be identified.


Asunto(s)
Regulación de la Expresión Génica , Lipoxinas/fisiología , Receptores de Formil Péptido/administración & dosificación , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/antagonistas & inhibidores , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Lipoxinas/metabolismo , Ratones , Receptores de Formil Péptido/biosíntesis , Receptores de Formil Péptido/fisiología , Receptores de Lipoxina/biosíntesis
9.
Am J Dermatopathol ; 35(2): 184-90, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23147350

RESUMEN

Melanoma, due to its metastatic rate, is among the most aggressive forms of skin cancer. Human formyl peptide receptor (FPR) and its variant FPR-like 1 (FPRL1) have been associated with cell migration and invasiveness in neoplasms. We have studied the in situ expression of these receptors in a large series of melanocytic lesions and correlated the expression with clinicopathological features and prognosis. Tissue microarray blocks of 141 cases including nevi (31 cases), primary (84 cases), and metastatic melanomas (26 cases) were semiquantitatively evaluated by immunohistochemistry for the expression of FPR and FPRL1 proteins. A significant association was observed regarding diagnosis and percentage of cells showing expression of FPR (P = 0.0311) and FPRL1 (P = 0.0053). A gain of FPR immunoreactivity was observed in the lesions having ulceration (P = 0.0194) and Breslow thickness (P = 0.044). Also, high FPRL1 cytoplasmic immunoreactivity was seen in lesions without tumor regression (P = 0.04). In addition, in patients with increased cytoplasmic staining for FPR, the probability of disease-specific survival was significantly lower (log rank test, P = 0.0089). Our findings reveal that FPR and FPRL1 are overexpressed in primary melanoma and correlate with aggressive tumor characteristics, underscoring them as potential therapeutic targets.


Asunto(s)
Melanoma/metabolismo , Receptores de Formil Péptido/biosíntesis , Receptores de Lipoxina/biosíntesis , Neoplasias Cutáneas/metabolismo , Biomarcadores de Tumor/análisis , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Masculino , Melanoma/mortalidad , Melanoma/patología , Persona de Mediana Edad , Fenotipo , Receptores de Formil Péptido/análisis , Receptores de Lipoxina/análisis , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Análisis de Matrices Tisulares
10.
J Immunol ; 188(4): 1799-808, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22262660

RESUMEN

Formyl peptide receptors (FPRs) are chemoattractant receptors that mediate inflammatory cell responses to infection. Recent evidence indicates that noneosinophilic asthma phenotypes can be developed by both Th1 and Th17 cell responses when exposed to LPS-containing allergens. In this study, we evaluated the effects of airway activation of FPRs by their synthetic agonist, Trp-Lys-Tyr-Met-Val-D-Met (W-peptide), on the development of Th1 and Th17 cell responses in a noneosinophilic asthma mouse model. A noneosinophilic asthma mouse model was generated by intranasal sensitization with 10 µg of LPS plus 75 µg of OVA on days 0, 1, 2, and 7. Mice were then challenged with 50 µg of OVA alone on days 14, 15, 21, and 22. W-peptide was administered during the sensitization period, and immune and inflammatory responses were evaluated after OVA challenge. Lung inflammation after OVA challenge was partly abolished by airway activation of FPRs during sensitization. Maturation of dendritic cells (DCs) and migration of DCs from the lung to lung-draining lymph nodes were inhibited by FPR activation. In addition, airway activation of FPRs inhibited allergen-specific T cell proliferation in the lymph nodes. Production of IL-12 and IL-6 (Th1- and Th17-polarizing cytokines) from lung DCs was decreased by airway activation of FPRs. This effect resulted in the inhibition of allergen-specific Th1 and Th17 cell responses. Airway activation of FPRs during sensitization effectively prevents the development of Th1 and Th17 cell responses induced by LPS-containing allergens via multiple mechanisms, such as inhibition of DC maturation and migration and the production of Th1- and Th7-polarizing cytokines.


Asunto(s)
Asma/inmunología , Células Dendríticas/inmunología , Pulmón/inmunología , Receptores de Formil Péptido/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Animales , Asma/metabolismo , Asma/patología , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Modelos Animales de Enfermedad , Interleucina-12/biosíntesis , Interleucina-6/biosíntesis , Pulmón/metabolismo , Pulmón/patología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacología , Ovalbúmina/inmunología , Receptores de Formil Péptido/biosíntesis , Células TH1/efectos de los fármacos , Células Th17/efectos de los fármacos
11.
Neurosci Lett ; 506(2): 266-70, 2012 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-22133809

RESUMEN

Pattern recognition receptors (PRRs) play important roles in the inflammatory responses to Alzheimer's disease (AD). Our previous study indicated that soybean isoflavone (SIF) exhibited anti-inflammatory effect in rats treated by ß-amyloid peptides1-42 (Aß1-42). In present study, we further detected the effects of SIF against inflammation caused by Aß1-42 treatment in rats. Serum inflammatory mediators and neurotrophic factors including transforming growth factor-ß (TGF-ß), inducible nitric oxide synthase (iNOS), brain-derived neurotrophic factor (BDNF) and S100ß were detected by enzyme-like immunosorbent assay (ELISA). Reverse transcription-polymerase chain reaction (RT-PCR) and western blot methods were applied for detecting mRNA and protein expression of interleukin-1ß (IL-1ß), iNOS, tumor necrosis factor-α (TNF-α), TGF-ß, BDNF, S100ß, myeloid differentiation factor88 (Myd88), Toll-like receptor2 (TLR2), formyl peptide receptors (FPRs), inhibitor κB kinase (IKK) and inhibitor κB-α (IκB-α) in rat's brain tissue. Our results indicated that SIF could reduce the production of IL-1ß, TNF-α and iNOS induced by Aß1-42 in serum and brain of rats. SIF also significantly reversed Aß1-42-induced up-regulation of TLR2, FPR, Myd88, IKK and decreased IκB-α mRNA and protein expressions in rats. These results suggested that TLR2 and FPR might involve in the inflammatory process induced by Aß1-42 treatment, and SIF was an efficiency compound in reversing the inflammation caused by Aß1-42 treatment.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Glycine max/química , Isoflavonas/farmacología , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Inflamación/inducido químicamente , Inflamación/metabolismo , Masculino , Ratas , Ratas Wistar , Receptores de Formil Péptido/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 2/biosíntesis
12.
PLoS One ; 6(12): e28712, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22174875

RESUMEN

The gene encoding the human formyl peptide receptor 1 (FPR1) is heterogeneous, containing numerous single nucleotide polymorphisms (SNPs). Here, we examine the effect of these SNPs on gene transcription and protein translation. We also identify gene promoter sequences and putative FPR1 transcription factors. To test the effect of codon bias and codon pair bias on FPR1 expression, four FPR1 genetic variants were expressed in human myeloid U937 cells fused to a reporter gene encoding firefly luciferase. No significant differences in luciferase activity were detected, suggesting that the translational regulation and protein stability of FPR1 are modulated by factors other than the SNP codon bias and the variant amino acid properties. Deletion and mutagenesis analysis of the FPR1 promoter showed that a CCAAT box is not required for gene transcription. A -88/41 promoter construct resulted in the strongest transcriptional activity, whereas a -72/41 construct showed large reduction in activity. The region between -88 and -72 contains a consensus binding site for the transcription factor PU.1. Mutagenesis of this site caused significant reduction in reporter gene expression. The PU.1 binding was confirmed in vivo by chromatin immunoprecipitation, and the binding to nucleotides -84 to -76 (TTCCTATTT) was confirmed in vitro by an electrophoretic mobility shift assay. Thus, similar to many other myeloid genes, FPR1 promoter activity requires PU.1. Two single nucleotide polymorphisms at -56 and -54 did not significantly affect FPR1 gene expression, despite differences in binding of transcription factor IRF1 in vitro. Inflammatory mediators such as interferon-γ, tumor necrosis factor-α, and lipopolysaccharide did not increase FPR1 promoter activity in myeloid cells, whereas differentiation induced by DMSO and retinoic acid enhanced the activity. This implies that the expression of FPR1 in myeloid cells is developmentally regulated, and that the differentiated cells are equipped for immediate response to microbial infections.


Asunto(s)
Mediadores de Inflamación/metabolismo , Polimorfismo de Nucleótido Simple/genética , Receptores de Formil Péptido/biosíntesis , Receptores de Formil Péptido/genética , Factores de Transcripción/metabolismo , Secuencia de Bases , Sitios de Unión , Diferenciación Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Codón/genética , Dimetilsulfóxido/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Genotipo , Humanos , Datos de Secuencia Molecular , Mutagénesis/efectos de los fármacos , Mutagénesis/genética , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Reproducibilidad de los Resultados , Transcripción Genética/efectos de los fármacos , Tretinoina/farmacología , Células U937
13.
J Immunol ; 187(3): 1475-85, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21709160

RESUMEN

The peptide F2L was previously characterized as a high-affinity natural agonist for the human formyl peptide receptor (FPR) 3. F2L is an acetylated 21-aa peptide corresponding with the N terminus of the intracellular heme-binding protein 1 (HEBP1). In the current work, we have investigated which proteases were able to generate the F2L peptide from its precursor HEBP1. Structure-function analysis of F2L identified three amino acids, G(3), N(7), and S(8), as the most important for interaction of the peptide with FPR3. We expressed a C-terminally His-tagged form of human HEBP1 in yeast and purified it to homogeneity. The purified protein was used as substrate to identify proteases generating bioactive peptides for FPR3-expressing cells. A conditioned medium from human monocyte-derived macrophages was able to generate bioactivity from HEBP1, and this activity was inhibited by pepstatin A. Cathepsin D was characterized as the protease responsible for HEBP1 processing, and the bioactive product was identified as F2L. We have therefore determined how F2L, the specific agonist of FPR3, is generated from the intracellular protein HEBP1, although it is unknown in which compartment the processing by cathepsin D occurs in vivo.


Asunto(s)
Proteínas Portadoras/metabolismo , Catepsina D/fisiología , Factores Quimiotácticos/agonistas , Hemoproteínas/metabolismo , Péptidos/agonistas , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional/inmunología , Receptores de Formil Péptido/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Proteínas Portadoras/biosíntesis , Catepsina D/deficiencia , Células Cultivadas , Factores Quimiotácticos/biosíntesis , Factores Quimiotácticos/metabolismo , Cricetinae , Cricetulus , Proteínas de Unión al Hemo , Hemoproteínas/biosíntesis , Humanos , Ligandos , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neutrófilos/enzimología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Péptidos/metabolismo , Unión Proteica/inmunología , Precursores de Proteínas/biosíntesis , Receptores de Formil Péptido/biosíntesis
14.
Am J Physiol Gastrointest Liver Physiol ; 301(3): G487-97, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21659618

RESUMEN

The macrophage plays a major role in the induction and resolution phases of inflammation; however, how lipid mediator-derived signals may modulate macrophage function in the resolution of inflammation driven by microbes (e.g., in inflammatory bowel disease) is not well understood. We examined the effects of aspirin-triggered lipoxin (ATL), a stable analog of lipoxin A(4), on the antimicrobial responses of human peripheral blood mononuclear cell-derived macrophages and the monocytic THP-1 cell line. Additionally, we assessed the expression and localization of the lipoxin receptor, formyl peptide receptor 2 (FPR2), in colonic mucosal biopsies from patients with Crohn's disease to determine whether the capacity for lipoxin signaling is altered in inflammatory bowel disease. We found that THP-1 cells treated with ATL (100 nM) displayed increased phagocytosis of inert fluorescent beads and Escherichia coli in a scavenger receptor- and PI3K-dependent, opsonization-independent manner. This ATL-induced increase in phagocytosis was also observed in primary human macrophages, where it was associated with an inhibition of E. coli-induced IL-1ß and IL-8 production. Finally, we found that FPR2 gene expression was increased approximately sixfold in the colon of patients with Crohn's disease, a finding reproduced in vitro by the treatment of THP-1 cells with interferon-γ or lipopolysaccharide. These results suggest that lipoxin signaling is upregulated in inflammatory environments, and, in addition to their known role in tissue resolution following injury, lipoxins can enhance macrophage clearance of invading microbes.


Asunto(s)
Lipoxinas/farmacología , Macrófagos/inmunología , Aspirina/farmacología , Fosfatidilinositol 3-Quinasa Clase Ib/fisiología , Enfermedad de Crohn/genética , Enfermedad de Crohn/fisiopatología , Humanos , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Leucocitos Mononucleares/metabolismo , Macrófagos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Receptores CCR7/biosíntesis , Receptores de Formil Péptido/biosíntesis , Receptores de Lipoxina/biosíntesis , Regulación hacia Arriba
15.
Int Immunopharmacol ; 10(12): 1602-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20933627

RESUMEN

Endothelial progenitor cells (EPCs) are involved in tumor neovascularization with undefined mechanisms. In this study, we explored the role of formylpeptide receptor, a G protein-coupled receptor, expressed by human malignant glioma cells in neovascularization of malignant glioma. EPCs were isolated from human umbilical cord blood and their migratory capability and tubulogenesis induced by the supernatant of U87 glioblastoma (GBM) cell line were examined. We also assessed the recruitment and incorporation of EPCs into orthotopic intracranial tumors formed by implanted U87 GBM cells. The supernatant of control U87 cells induced high levels of migration and tubule-formation in vitro by EPCs. In contrast, the chemotactic and tubule-stimulating activities on EPCs in the supernatant of U87 cells with FPR knocking down by small interference (si) RNA were significantly attenuated. In addition, the number of EPCs recruited and incorporated into intracranial glioma xenografts was significantly higher in tumors formed by control U87 cells than tumors formed by U87 cells containing FPR-siRNA. Our results suggest that expression of functional FPR in glioma cells plays an important role in regulating vasculogenesis by EPCs, which constitute a novel target for anti-angiogenic therapy in gliomas.


Asunto(s)
Endotelio Vascular/patología , Glioma/irrigación sanguínea , Neovascularización Patológica , Receptores de Formil Péptido/biosíntesis , Células Madre/patología , Animales , Comunicación Celular , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Movimiento Celular , Quimiotaxis , Medios de Cultivo Condicionados , Femenino , Glioma/metabolismo , Glioma/patología , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neovascularización Patológica/metabolismo , ARN Interferente Pequeño/farmacología , Receptores de Formil Péptido/genética
16.
J Neurochem ; 113(3): 749-60, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20141570

RESUMEN

Recent studies suggest that the chemotactic G protein-coupled receptor formyl-peptide-receptor-like-1 (FPRL1) or the scavenger receptor MARCO (macrophage receptor with collagenous structure) plays an essential role in the inflammatory response of host defense mechanisms and neurodegenerative disorders such as Alzheimer's disease. We therefore analyzed the involvement of FPRL1 and MARCO in amyloid beta1-42 (Abeta1-42)-induced signalling by extracellular-signal regulated kinases 1/2 (ERK1/2) phosphorylation and cAMP level measurement in glial cells (astrocytes and microglia) and in transfected HEK293 cells. Receptors were inhibited by small interference RNA and the consequences in Abeta1-42- and MARCO agonist fucoidan-induced signal transduction were determined. Receptor deactivation by antagonists or small interference RNA verified the importance of FPRL1 for Abeta1-42-mediated signal transduction by ERK1/2 phosphorylation and cAMP level measurement in glial cells. Furthermore, for the first time, we have demonstrated a functional interaction between FPRL1 and scavenger receptors in fucoidan-mediated signalling by ERK1/2 phosphorylation and cAMP level measurement. In addition, co-immunoprecipitation data and fluorescence microscopy measurements revealed a physical interaction between FPR, FPRL1 and MARCO. These results suggest that FPRL1 plays a pivotal role for Abeta1-42-induced signal transduction in glial cells and the interaction with MARCO could explain the broad ligand spectrum of formyl peptide receptors.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Neuroglía/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores de Formil Péptido/fisiología , Receptores Inmunológicos/fisiología , Transducción de Señal/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Western Blotting , Células Cultivadas , AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Inmunoprecipitación , Microscopía Fluorescente , Fosforilación , Plásmidos/genética , Polisacáridos/farmacología , ARN/biosíntesis , ARN/aislamiento & purificación , ARN Interferente Pequeño/genética , Receptores de Formil Péptido/biosíntesis , Receptores de Formil Péptido/efectos de los fármacos , Receptores de Formil Péptido/genética , Receptores Inmunológicos/efectos de los fármacos , Receptores Inmunológicos/genética , Receptores de Lipoxina/biosíntesis , Receptores de Lipoxina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
17.
J Immunol ; 183(9): 5511-7, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19843937

RESUMEN

We determined the expression of the formyl peptide receptor (FPR) family and the functional roles of the FPR family in NK cells. All tested human NK cells express two members of the FPR family (FPR1 and FPR2). The expression of FPR3 was noted to occur in a donor-specific manner. The stimulation of NK cells with FPR family-selective agonists (fMLF (N-formyl-Met-Leu-Phe), MMK-1, F2L, and WKYMVm (Trp-Lys-Tyr-Met-Val-d-Met)) elicited cytolytic activity in resting NK cells, but not in IL-2-activated NK cells; the cytolytic activity was not inhibited by pertussis toxin. The FPR family agonists also stimulated chemotactic migration of IL-2-activated NK cells, but not resting NK cells; the chemotactic migration was completely inhibited by pertussis toxin. WKYMVm stimulates ERK, p38 MAPK, and JNK activities in both resting and IL-2-activated NK cells. WKYMVm-induced chemotactic migration was partially inhibited by PD98059 (2'-amino-3'-methoxyflavone); however, the inhibition of JNK by its selective inhibitor (SP600125, anthra[1,9-cd]pyrazol-6(2H)-one) dramatically inhibited the WKYMVm-induced cytolytic activity. Furthermore, WKYMVm-induced chemotactic migration and cytolytic activity were partly inhibited by FPR family-selective antagonists (cyclosporin H and WRWWWW). Taken together, our findings indicate that human NK cells express functional members of the FPR family, and in turn the activation of the three members of the FPR receptor family elicit cytolytic activity in NK cells, thus suggesting that the receptors are potentially important therapeutic targets for the modulation of NK cell-mediated immune responses.


Asunto(s)
Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Familia de Multigenes/inmunología , Receptores de Formil Péptido/biosíntesis , Quimiotaxis de Leucocito/inmunología , Citotoxicidad Inmunológica , Humanos , Activación de Linfocitos/inmunología , Oligopéptidos/agonistas , Oligopéptidos/biosíntesis , Oligopéptidos/fisiología , Receptores de Formil Péptido/agonistas , Receptores de Formil Péptido/fisiología , Receptores de Lipoxina/agonistas , Receptores de Lipoxina/biosíntesis , Receptores de Lipoxina/fisiología , Transducción de Señal/inmunología
18.
J Immunol ; 183(3): 2089-96, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19597002

RESUMEN

Aspirin is a unique nonsteroidal anti-inflammatory drug; at high doses (aspirin(high), 1g), it is anti-inflammatory stemming from the inhibition of cyclooxygenase and proinflammatory signaling pathways including NF-kappaB, but is cardioprotective at lower doses (aspirin(low), 75 mg). The latter arises from the inhibition of thromboxane (Tx) B(2), a prothrombotic eicosanoid also implicated in polymorphonuclear leukocyte trafficking. As a result, aspirin(low) is widely used as a primary and secondary preventative against vascular disease. Despite this and its ability to synthesize proresolution 15-epi-lipoxin A(4) it is not known whether aspirin(low) is anti-inflammatory in humans. To address this, we generated skin blisters by topically applying cantharidin on the forearm of healthy male volunteers, causing an acute inflammatory response including dermal edema formation and leukocyte trafficking. Although not affecting blister fluid volume, aspirin(low) (75 mg, oral, once daily/10 days) reduced polymorphonuclear leukocyte and macrophage accumulation independent of NF-kappaB-regulated gene expression and inhibition of conventional prostanoids. However, aspirin(low) triggered 15-epi-lipoxin A(4) synthesis and up-regulated its receptor (FPRL1, ALX). From complimentary in vitro experiments, we propose that 15-epi-lipoxin A(4) exerts its protective effects by triggering antiadhesive NO, thereby dampening leukocyte/endothelial cell interaction and subsequent extravascular leukocyte migration. Since similar findings were obtained from murine zymosan-induced peritonitis, we suggest that aspirin(low) possesses the ability to inhibit mammalian innate immune-mediated responses. This highlights 15-epi-lipoxin A(4) as a novel anti-inflammatory working through a defined receptor and suggests that mimicking its mode of action represents a new approach to treating inflammation-driven diseases.


Asunto(s)
Aspirina/administración & dosificación , Inflamación/tratamiento farmacológico , Lipoxinas/biosíntesis , Enfermedad Aguda , Aspirina/farmacología , Vesícula/inducido químicamente , Cantaridina , Adhesión Celular/efectos de los fármacos , Quimiotaxis de Leucocito/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Macrófagos , Masculino , Óxido Nítrico/biosíntesis , Receptores de Formil Péptido/biosíntesis , Receptores de Lipoxina/biosíntesis , Regulación hacia Arriba/efectos de los fármacos
19.
Mol Cancer Res ; 7(6): 907-15, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19491199

RESUMEN

Emerging evidence suggests that the antimicrobial peptide, leucine leucine-37 (LL-37), could play a role in the progression of solid tumors. LL-37 is expressed as the COOH terminus of human cationic antimicrobial protein-18 (hCAP-18) in ovarian, breast, and lung cancers. Previous studies have shown that the addition of LL-37 to various cancer cell lines in vitro stimulates proliferation, migration, and invasion. Similarly, overexpression of hCAP-18/LL-37 in vivo accelerates tumor growth. However, the receptor or receptors through which these processes are mediated have not been thoroughly examined. In the present study, expression of formyl peptide receptor-like 1 (FPRL1) was confirmed on ovarian cancer cells. Proliferation assays indicated that LL-37 does not signal through a G protein-coupled receptor, such as FPRL1, to promote cancer cell growth. By contrast, FPRL1 was required for LL-37-induced invasion through Matrigel. The peptide stimulated mitogen-activated protein kinase and Janus-activated kinase/signal transducers and activators of transcription signaling cascades and led to the significant activation of several transcription factors, through both FPRL1-dependent and FPRL1-independent pathways. Likewise, expression of some LL-37-stimulated genes was attenuated by the inhibition of FPRL1. Increased expression of CXCL10, EGF, and PDGF-BB as well as other soluble factors was confirmed from conditioned medium of LL-37-treated cells. Taken together, these data suggest that LL-37 potentiates a more aggressive behavior from ovarian cancer cells through its interaction with FPRL1.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Oncogenes , Neoplasias Ováricas/metabolismo , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Péptidos Catiónicos Antimicrobianos/biosíntesis , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Línea Celular Tumoral , Femenino , Citometría de Flujo , Humanos , Invasividad Neoplásica , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , ARN Interferente Pequeño/genética , Receptores de Formil Péptido/antagonistas & inhibidores , Receptores de Formil Péptido/biosíntesis , Receptores de Formil Péptido/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lipoxina/antagonistas & inhibidores , Receptores de Lipoxina/biosíntesis , Receptores de Lipoxina/genética , Proteínas Recombinantes/farmacología , Catelicidinas
20.
Carcinogenesis ; 30(2): 348-55, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19037090

RESUMEN

The G protein-coupled formylpeptide receptor (FPR), known to mediate phagocytic leucocyte chemotaxis in response to bacterial- and host-derived agonists, was expressed by tumor cells in specimens of surgically removed more highly malignant human gliomas. In human glioblastoma cell lines, FPR activation increased cell motility, tumorigenicity and production of angiogenic factors. In studies of the mechanistic basis for the selective expression of FPR in more highly malignant gliomas, we found that the DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine (Aza), while promoting the differentiation of human glioblastoma cells, downregulated FPR expression. Aza also reduced the global methylation levels in glioblastoma cells and activated the pathway of p53 tumor suppressor. Methylation-specific polymerase chain reaction revealed that Aza treatment of tumor cells reduced the methylation of p53 promoter, which was accompanied by increased expression of p53 gene and protein. In addition, overexpression of p53 in glioblastoma cells mimicked the effect of Aza treatment as shown by increased cell differentiation but reduction in FPR expression, the capacity of tumor sphere formation in soft agar and tumorigenesis in nude mice. Furthermore, Aza treatment or overexpression of the wild-type p53 in glioblastoma cells increased the binding of p53 to FPR promoter region shown by chromatin immunoprecipitation. These results indicate that increased methylation of p53 gene retains human glioblastoma cells at a more poorly differentiated phase associated with the aberrant expression of FPR as a tumor-promoting cell surface receptor.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Receptores de Formil Péptido/biosíntesis , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Neoplasias Encefálicas/patología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Quimiotaxis , Metilasas de Modificación del ADN/antagonistas & inhibidores , Decitabina , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Humanos , Metilación , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Regiones Promotoras Genéticas , Trasplante Heterólogo , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA