Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 73(6): 108, 2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38642131

RESUMEN

Tumor-associated macrophages (TAMs) are abundant in tumors and interact with tumor cells, leading to the formation of an immunosuppressive microenvironment and tumor progression. Although many studies have explored the mechanisms underlying TAM polarization and its immunosuppressive functions, understanding of its progression remains limited. TAMs promote tumor progression by secreting cytokines, which subsequently recruit immunosuppressive cells to suppress the antitumor immunity. In this study, we established an in vitro model of macrophage and non-small cell lung cancer (NSCLC) cell co-culture to explore the mechanisms of cell-cell crosstalk. We observed that in NSCLC, the C-X-C motif chemokine ligand 5 (CXCL5) was upregulated in macrophages because of the stimulation of A2AR by adenosine. Adenosine was catalyzed by CD39 and CD73 in macrophages and tumor cells, respectively. Nuclear factor kappa B (NFκB) mediated the A2AR stimulation of CXCL5 upregulation in macrophages. Additionally, CXCL5 stimulated NETosis in neutrophils. Neutrophil extracellular traps (NETs)-treated CD8+ T cells exhibited upregulation of exhaustion-related and cytosolic DNA sensing pathways and downregulation of effector-related genes. However, A2AR inhibition significantly downregulated CXCL5 expression and reduced neutrophil infiltration, consequently alleviating CD8+ T cell dysfunction. Our findings suggest a complex interaction between tumor and immune cells and its potential as therapeutic target.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Quimiocina CXCL5 , Neoplasias Pulmonares , Macrófagos , Humanos , Adenosina/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Linfocitos T CD8-positivos , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Microambiente Tumoral , Regulación hacia Arriba , Receptor de Adenosina A2A/metabolismo , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo
2.
BMC Cancer ; 24(1): 140, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38287266

RESUMEN

Glioblastoma multiforme (GBM) is the most aggressive brain cancer with a poor prognosis. Therefore, the correlative molecular markers and molecular mechanisms should be explored to assess the occurrence and treatment of glioma.WB and qPCR assays were used to detect the expression of CXCL5 in human GBM tissues. The relationship between CXCL5 expression and clinicopathological features was evaluated using logistic regression analysis, Wilcoxon symbolic rank test, and Kruskal-Wallis test. Univariate, multivariate Cox regression and Kaplan-Meier methods were used to assess CXCL5 and other prognostic factors of GBM. Gene set enrichment analysis (GSEA) was used to identify pathways associated with CXCL5. The correlation between CXCL5 and tumor immunoinfiltration was investigated using single sample gene set enrichment analysis (ssGSEA) of TCGA data. Cell experiments and mouse subcutaneous transplanted tumor models were used to evaluate the role of CXCL5 in GBM. WB, qPCR, immunofluorescence, and immunohistochemical assays showed that CXCL5 expression was increased in human GBM tissues. Furthermore, high CXCL5 expression was closely related to poor disease-specific survival and overall survival of GBM patients. The ssGSEA suggested that CXCL5 is closely related to the cell cycle and immune response through PPAR signaling pathway. GSEA also showed that CXCL5 expression was positively correlated with macrophage cell infiltration level and negatively correlated with cytotoxic cell infiltration level. CXCL5 may be associated with the prognosis and immunoinfiltration of GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Animales , Ratones , Humanos , Glioblastoma/patología , Pronóstico , Procesos Neoplásicos , Neoplasias Encefálicas/metabolismo , Transducción de Señal , Quimiocina CXCL5/genética
3.
J Neuroinflammation ; 20(1): 105, 2023 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-37138312

RESUMEN

BACKGROUND: Chronic cerebral ischemia induces white matter injury (WMI) contributing to cognitive decline. Both astrocytes and microglia play vital roles in the demyelination and remyelination processes, but the underlying mechanism remains unclear. This study aimed to explore the influence of the chemokine CXCL5 on WMI and cognitive decline in chronic cerebral ischemia and the underlying mechanism. METHODS: Bilateral carotid artery stenosis (BCAS) model was constructed to mimic chronic cerebral ischemia in 7-10 weeks old male mice. Astrocytic Cxcl5 conditional knockout (cKO) mice were constructed and mice with Cxcl5 overexpressing in astrocytes were generated by stereotactic injection of adeno-associated virus (AAV). WMI was evaluated by magnetic resonance imaging (MRI), electron microscopy, histological staining and western blotting. Cognitive function was examined by a series of neurobehavioral tests. The proliferation and differentiation of oligodendrocyte progenitor cells (OPCs), phagocytosis of microglia were analyzed via immunofluorescence staining, western blotting or flow cytometry. RESULTS: CXCL5 was significantly elevated in the corpus callosum (CC) and serum in BCAS model, mainly expressed in astrocytes, and Cxcl5 cKO mice displayed improved WMI and cognitive performance. Recombinant CXCL5 (rCXCL5) had no direct effect on the proliferation and differentiation of OPCs in vitro. Astrocytic specific Cxcl5 overexpression aggravated WMI and cognitive decline induced by chronic cerebral ischemia, while microglia depletion counteracted this effect. Recombinant CXCL5 remarkably hindered microglial phagocytosis of myelin debris, which was rescued by inhibition of CXCL5 receptor C-X-C motif chemokine receptor 2 (CXCR2). CONCLUSION: Our study revealed that astrocyte-derived CXCL5 aggravated WMI and cognitive decline by inhibiting microglial phagocytosis of myelin debris, suggesting a novel astrocyte-microglia circuit mediated by CXCL5-CXCR2 signaling in chronic cerebral ischemia.


Asunto(s)
Isquemia Encefálica , Estenosis Carotídea , Quimiocina CXCL5 , Sustancia Blanca , Animales , Masculino , Ratones , Astrocitos/patología , Isquemia Encefálica/patología , Estenosis Carotídea/patología , Quimiocina CXCL5/genética , Microglía , Vaina de Mielina/patología , Fagocitosis , Sustancia Blanca/patología
4.
Cardiovasc Diabetol ; 22(1): 172, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37420254

RESUMEN

BACKGROUND: Higher chemokine C-X-C motif ligand 5 (CXCL5) level was observed in type 2 diabetes mellitus (DM) patients; however, its role in diabetic vasculopathy was not clarified. This study aimed to explore the impacts and mechanistic insights of CXCL5 in neovasculogenesis and wound healing in DM. METHODS: Endothelial progenitor cells (EPCs) and human aortic endothelial cells (HAECs) were used in vitro. Streptozotocin-induced diabetic mice and Leprdb/JNarl mice were used as type 1 and type 2 DM models. Moreover, CXCL5 knockout mice were used to generate diabetic mice. Hindlimb ischemia surgery, aortic ring assays, matrigel plug assay, and wound healing assay were conducted. RESULTS: CXCL5 concentrations were increased in plasma and EPCs culture medium from type 2 DM patients. CXCL5 neutralizing antibody upregulated vascular endothelial growth factor (VEGF)/stromal cell-derived factor-1 (SDF-1) and promoted cell function in EPCs from type 2 DM patients and high glucose-treated EPCs from non-DM subjects as well as HAECs. CXCL5 directly up-regulated interleukin (IL)-1ß/IL-6/tumor necrosis factor-α and down-regulated VEGF/SDF-1 via ERK/p65 activation through chemokine C-X-C motif receptor 2 (CXCR2). CXCL5 neutralizing antibody recovered the blood flow after hindlimb ischemia, increased circulating EPC number, and enhanced VEGF and SDF-1 expression in ischemic muscle. CXCL5 suppression promoted neovascularization and wound healing in different diabetic animal models. The above observation could also be seen in streptozotocin-induced CXCL5 knockout diabetic mice. CONCLUSIONS: CXCL5 suppression could improve neovascularization and wound healing through CXCR2 in DM. CXCL5 may be regarded as a potential therapeutic target for vascular complications of DM.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Células Progenitoras Endoteliales , Humanos , Ratones , Animales , Diabetes Mellitus Tipo 2/metabolismo , Factor A de Crecimiento Endotelial Vascular , Diabetes Mellitus Experimental/metabolismo , Estreptozocina/metabolismo , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Células Progenitoras Endoteliales/metabolismo , Quimiocina CXCL12/metabolismo , Ratones Noqueados , Cicatrización de Heridas , Isquemia , Neovascularización Fisiológica/fisiología , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo
5.
Mol Biol Rep ; 50(10): 8015-8023, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37541997

RESUMEN

BACKGROUND: The tumor microenvironment contains chemokines that play a crucial role in various processes, such as tumorigenesis, inflammation, and therapy resistance, in different types of cancer. CXCL5 is a significant chemokine that has been shown to promote tumor proliferation, invasion, angiogenesis, and therapy resistance when overexpressed in various types of cancer. This research aims to investigate the impact of CXCL5 on the biological functions of glioblastoma (GBM). METHODS: The TCGA GBM and GEO databases were utilized to perform transcriptome microarray analysis and oncogenic signaling pathway analysis of CXCL5 in GBM. Validation of CXCL5 expression was performed using RT-qPCR and Western Blot. The impact of CXCL5 on cell proliferation, tumorigenesis, and angiogenesis in GBM was assessed through various methods, including cell proliferation assay, cloning assay, intracranial xenograft tumor models, and tube formation assay. Clinical prognosis was evaluated in 59 samples of gliomas with varying degrees of malignancy (grades 2, 3, and 4) and the TCGA GBM database, based on CXCL5 expression levels. The activities of the JAK-STAT and NF-κB signaling pathways were detected using Western Blot. RESULTS: The expression of CXCL5 was highly enriched in GBM. Moreover, the inhibition of CXCL5 showed a significant efficacy in suppressing cellular proliferation and angiogenesis, resulting in extended survival rates in xenograft mouse models in comparison to the control group. Notably, pretreatment with dapsone exhibited a reversal of the impact of CXCL5 on the formation of colonies and tubes in GBM cells. Elevated expression of CXCL5 was correlated with poor outcomes in GBM patients. Furthermore, the overexpression of CXCL5 has been associated with the activation of JAK-STAT and NF-κB signaling pathways. CONCLUSIONS: CXCL5 plays an important role in tumorigenesis and angiogenesis, indicating the potential for novel therapies targeting CXCL5 in GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Animales , Ratones , FN-kappa B/metabolismo , Glioblastoma/metabolismo , Transducción de Señal , Carcinogénesis/genética , Transformación Celular Neoplásica , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Microambiente Tumoral , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo
6.
Dig Dis Sci ; 68(3): 841-851, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35650416

RESUMEN

BACKGROUND: Pancreatic cancer (PC) is the most lethal malignant tumor, with average survival period of about 10 months. C-X-C ligand 5 (CXCL5), an important chemokine for immune cell accumulation in tumor tissues, has been reported to be involved in a variety of human cancers. However, the exact role of CXCL5 in PC progression has not been well defined. METHODS: The expression of CXCL5 in PC was analyzed based on online databases and clinical specimens immunohistochemical staining, and Western blotting of CXCL5 in PC cell lines and patient samples. The correlation between CXCL5 expression and prognosis in PC was explored. The role of CXCL5 in PC was investigated through in vitro and in vivo experiments. RESULTS: The expression of CXCL5 was significantly increased in PC tissues compared with that in pancreas tissues, and CXCL5 high expression predicts poor prognosis in PC patients. Further analyses demonstrated that overexpression of CXCL5 in PC cells was positively related to higher proliferation rate, higher migration ability, and higher EMT markers including SNAI2 and TWIST1 of tumor cells in vitro. Consistently, the knockdown of CXCL5 in PC cells harmed the proliferation rate, migration ability, and expression of EMT indexes of tumor cells in vitro. Importantly, knockdown of CXCL5 inhibited the growth of xenograft tumors in vivo. CONCLUSION: CXCL5 high expression predicts poor prognosis in PC patients. CXCL5 promotes PC cell growth and EMT process. Inhibition of CXCL5 may be a potential therapeutic approach for PC.


Asunto(s)
Transición Epitelial-Mesenquimal , Neoplasias Pancreáticas , Humanos , Xenoinjertos , Proliferación Celular , Línea Celular Tumoral , Neoplasias Pancreáticas/metabolismo , Páncreas/patología , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Neoplasias Pancreáticas
7.
Mol Ther ; 30(6): 2327-2341, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35283273

RESUMEN

CXCL5 is overexpressed in colorectal cancer (CRC) and promotes distant metastasis and angiogenesis of tumors; however, the underlying mechanism that mediates CXCL5 overexpression in CRC remains unclear. Here, we successfully extracted and identified primary mesenchymal stromal cells (MSCs) and verified the promoting effects of tumor-associated MSCs on CRC proliferation and metastasis in vivo and in vitro. We found that MSCs not only promoted the expression of CXCL5 by secreting CCL7 but also secreted TGF-ß to inhibit this process. After secretion, CCL7/CCR1 activated downstream CBP/P300 to acetylate KLF5 to promote CXCL5 transcription, while TGF-ß reversed the effect of KLF5 on transcription activation by regulating SMAD4. Taken together, our results indicate that MSCs in the tumor microenvironment promoted the progression and metastasis of CRC and regulated the expression of CXCL5 in CRC cells by secreting CCL7 and TGF-ß. KLF5 is the key site of these processes and plays a dual role in CXCL5 regulation. MSCs and their secreted factors may serve as potential therapeutic targets in the tumor environment.


Asunto(s)
Neoplasias Colorrectales , Células Madre Mesenquimatosas , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Quimiocina CCL7 , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Quimiocina CXCL5/farmacología , Neoplasias Colorrectales/patología , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Madre Mesenquimatosas/metabolismo , Metástasis de la Neoplasia , Neovascularización Patológica/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/genética
8.
Proc Natl Acad Sci U S A ; 117(22): 12281-12287, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32424099

RESUMEN

Sepsis is a life-threatening organ dysfunction condition caused by a dysregulated host response to an infection. Here we report that the circulating levels of growth and differentiation factor-15 (GDF15) are strongly increased in septic shock patients and correlate with mortality. In mice, we find that peptidoglycan is a potent ligand that signals through the TLR2-Myd88 axis for the secretion of GDF15, and that Gdf15-deficient mice are protected against abdominal sepsis due to increased chemokine CXC ligand 5 (CXCL5)-mediated recruitment of neutrophils into the peritoneum, leading to better local bacterial control. Our results identify GDF15 as a potential target to improve sepsis treatment. Its inhibition should increase neutrophil recruitment to the site of infection and consequently lead to better pathogen control and clearance.


Asunto(s)
Bacteriemia/inmunología , Quimiocina CXCL5/inmunología , Factor 15 de Diferenciación de Crecimiento/inmunología , Neutrófilos/inmunología , Animales , Bacteriemia/genética , Bacteriemia/microbiología , Bacteriemia/prevención & control , Quimiocina CXCL5/genética , Femenino , Factor 15 de Diferenciación de Crecimiento/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Cavidad Peritoneal/microbiología
9.
FASEB J ; 35(1): e21181, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33231340

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) metastasizes to the locoregional lymph nodes at high rates and is related to poor clinical outcomes. However, the mechanism by which cancer cells migrate to the lymph nodes is unclear. To address this, we established a conditioned medium culture system for HNSCC cells and lymphatic endothelial cells (LECs) and investigated their crosstalk. Stimulation with tumor-conditioned medium (TCM) activated LECs, resulting in a robust increase in cell proliferation to induce lymphatic hyperplasia. Further, stimulation of HNSCC cells with activated LEC Conditioned media (TCM-LEC CM) induced cell invasion. Among various chemokines, CXCL5 promoted the invasion of TCM-LEC CM-treated HNSCC cells. The level of CXCL5 protein was higher in cancer tissues than those in normal tissues from HNSCC patients. Furthermore, treatment with SB225002, a CXCR2 (CXCL5 receptor) inhibitor, resulted in decreased lymph node metastasis in vivo. In conclusion, inhibition of CXCL5-CXCR2 signaling between cancer cells and LECs suppresses cancer cell invasion and metastasis in vitro and in vivo. This novel therapeutic strategy might be a practical approach to the clinical management of HNSCC.


Asunto(s)
Comunicación Celular , Quimiocina CXCL5/metabolismo , Células Endoteliales/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Interleucina-8B/metabolismo , Transducción de Señal , Animales , Línea Celular Tumoral , Quimiocina CXCL5/genética , Células Endoteliales/patología , Femenino , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Receptores de Interleucina-8B/genética
10.
Anticancer Drugs ; 33(1): e103-e112, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34407043

RESUMEN

In our previous studies, we found that T24 lung metastatic cancer cells showed high invasion and metastasis abilities and cancer stem cell characteristics compared with T24 primary cancer cells. By screening for the expression of CXC chemokines in both cell lines, we found that CXCL5 is highly expressed in T24-L cells. The aim of this study is to shed light on the relationship of CXCL5 with epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs). RNAi technology was used to decrease CXCL5 expression in the T24-L cell line, and the EMT and CSCs of the shCXCL5 group and the control group were compared. The CXCR2 inhibitor SB225002 was used to inhibit the receptor of CXCL5 to determine the effect of the CXCL5/CXCR2 axis. The knockdown of CXCL5 expression in T24-L cells reduced their EMT and CSC characteristics. RT-PCR and Western blot analyses revealed the downregulation of N-cadherin, Vimentin and CD44. In addition, when CD44 expression was knocked down, the EMT ability of the cells was also inhibited. This phenomenon was most pronounced when both CXCL5 and CD44 were knocked down. CXCL5 and CD44 can affect the EMT and stem cell capacity of T24-L cells through some interaction.


Asunto(s)
Quimiocina CXCL5/genética , Receptores de Hialuranos/genética , Neoplasias Pulmonares/patología , Neoplasias de la Vejiga Urinaria/secundario , Cadherinas/fisiología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Vimentina/fisiología
11.
Eur J Immunol ; 50(5): 712-724, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31981231

RESUMEN

Unlike hematological malignancies, solid tumors have proved to be less susceptible to chimeric antigen receptor (CAR)-T cell therapy, which is partially caused by reduced accumulation of therapeutic T cells in tumor site. Since efficient trafficking is the precondition and pivotal step for infused CAR-T cells to exhibit their anti-tumor function, strategies are highly needed to improve the trafficking ability of CAR-T cells for solid tumor treatment. Here, based on natural lymphocyte chemotaxis theory and characteristics of solid tumor microenvironments, we explored the possibility of enhancing CAR-T cell trafficking by using chemokine receptors. Our study found that compared with other chemokines, several CXCR2 ligands showed relatively high expression level in human hepatocellular carcinoma tumor tissues and cell lines. However, both human peripheral T cells and hepatocellular carcinoma tumor infiltrating T cells lacked expression of CXCR2. CXCR2-expressing CAR-T cells exhibited identical cytotoxicity but displayed significantly increased migration ability in vitro. In a xenograft tumor model, we found that expressing CXCR2 in CAR-T cells could significantly accelerate in vivo trafficking and tumor-specific accumulation, and improve anti-tumor effect of these cells.


Asunto(s)
Carcinoma Hepatocelular/terapia , Inmunoterapia Adoptiva/métodos , Neoplasias Hepáticas/terapia , Receptores Quiméricos de Antígenos/genética , Receptores de Interleucina-8B/genética , Linfocitos T Citotóxicos/inmunología , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Quimiocina CXCL5/genética , Quimiocina CXCL5/inmunología , Citotoxicidad Inmunológica , Expresión Génica , Humanos , Interleucina-8/genética , Interleucina-8/inmunología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Ratones , Receptores Quiméricos de Antígenos/inmunología , Receptores de Interleucina-8B/inmunología , Linfocitos T Citotóxicos/citología , Carga Tumoral , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Blood ; 133(12): 1335-1345, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30723078

RESUMEN

Neutrophil migration to the site of bacterial infection is a critical step in host defense. Exclusively produced in the bone marrow, neutrophil release into the blood is tightly controlled. Although the chemokine CXCL1 induces neutrophil influx during bacterial infections, its role in regulating neutrophil recruitment, granulopoiesis, and neutrophil mobilization in response to lung infection-induced sepsis is unclear. Here, we used a murine model of intrapulmonary Streptococcus pneumoniae infection to investigate the role of CXCL1 in host defense, granulopoiesis, and neutrophil mobilization. Our results demonstrate that CXCL1 augments neutrophil influx to control bacterial growth in the lungs, as well as bacterial dissemination, resulting in improved host survival. This was shown in Cxcl1 -/- mice, which exhibited defective amplification of early neutrophil precursors in granulocytic compartments, and CD62L- and CD49d-dependent neutrophil release from the marrow. Administration of recombinant CXCL2 and CXCL5 after infection rescues the impairments in neutrophil-dependent host defense in Cxcl1 -/- mice. Taken together, these findings identify CXCL1 as a central player in host defense, granulopoiesis, and mobilization of neutrophils during Gram-positive bacterial pneumonia-induced sepsis.


Asunto(s)
Quimiocina CXCL1/fisiología , Homeostasis , Pulmón/inmunología , Infiltración Neutrófila/inmunología , Infecciones Neumocócicas/complicaciones , Neumonía Bacteriana/complicaciones , Sepsis/inmunología , Animales , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Femenino , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/patología , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Sepsis/metabolismo , Sepsis/microbiología , Serogrupo , Streptococcus pneumoniae/fisiología
13.
Clin Exp Dermatol ; 46(8): 1462-1470, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34050991

RESUMEN

BACKGROUND: Behçet disease (BD) is associated with the immune system, especially neutrophilic activity. The CXCR1, CXCR2 and CXCL5 genes mediate the activation and migration of neutrophils. AIM: To investigate CXCR1, CXCR2 and CXCL5 single nucleotide polymorphisms (SNPs) and examine their association with BD. METHODS: We studied polymorphic sites in CXCR1 (four sites: rs16858811, rs9282752, rs16858809 and rs16858808), CXCR2 (three sites: rs2230054, rs1126579 and rs1126580) and CXCL5 (one site: rs352046) in 87 patients with BD and 111 healthy controls (HCs), using a PCR restriction-fragment length polymorphism-based approach for genotyping. RESULTS: We found that the CXCR2 rs2230054 TT genotype and the CXCL5 rs352046 polymorphism might be possible genetic factors responsible for BD. We did not find any association between the development of BD and any of the four CXCR1 polymorphisms or the other two CXCR2 SNPs. In addition, our haplotype analysis results indicated that the haplotypes of the CXCR2 and CXCR1-CXCR2 polymorphic loci were different between the BD and HC groups. CONCLUSION: Our study suggests that polymorphisms of CXCR1, CXCR2 and CXCL5 may affect susceptibility to BD and increase the risk of developing the disease. These loci need to be studied in larger groups of patients from different geographical areas around the world in order to clarify the genetic background for BD pathogenesis.


Asunto(s)
Síndrome de Behçet/genética , Quimiocina CXCL5/genética , Polimorfismo de Nucleótido Simple , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8B/genética , Femenino , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Masculino , Turquía
14.
J Biol Chem ; 294(48): 18192-18206, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31636124

RESUMEN

GNA13, the α subunit of a heterotrimeric G protein, mediates signaling through G-protein-coupled receptors (GPCRs). GNA13 is up-regulated in many solid tumors, including prostate cancer, where it contributes to tumor initiation, drug resistance, and metastasis. To better understand how GNA13 contributes to tumorigenesis and tumor progression, we compared the entire transcriptome of PC3 prostate cancer cells with those cells in which GNA13 expression had been silenced. This analysis revealed that GNA13 levels affected multiple CXC-family chemokines. Further investigation in three different prostate cancer cell lines singled out pro-tumorigenic CXC motif chemokine ligand 5 (CXCL5) as a target of GNA13 signaling. Elevation of GNA13 levels consistently induced CXCL5 RNA and protein expression in all three cell lines. Analysis of the CXCL5 promoter revealed that the -505/+62 region was both highly active and influenced by GNA13, and a single NF-κB site within this region of the promoter was critical for GNA13-dependent promoter activity. ChIP experiments revealed that, upon induction of GNA13 expression, occupancy at the CXCL5 promoter was significantly enriched for the p65 component of NF-κB. GNA13 knockdown suppressed both p65 phosphorylation and the activity of a specific NF-κB reporter, and p65 silencing impaired the GNA13-enhanced expression of CXCL5. Finally, blockade of Rho GTPase activity eliminated the impact of GNA13 on NF-κB transcriptional activity and CXCL5 expression. Together, these findings suggest that GNA13 drives CXCL5 expression by transactivating NF-κB in a Rho-dependent manner in prostate cancer cells.


Asunto(s)
Quimiocina CXCL5/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Activación Transcripcional , Quimiocina CXCL5/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Humanos , Masculino , Proteínas de Neoplasias/genética , Células PC-3 , Neoplasias de la Próstata/genética , Factor de Transcripción ReIA/genética
15.
J Cell Physiol ; 235(5): 4756-4765, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31667838

RESUMEN

CXCL3 belongs to the CXC-type chemokine family and is known to play a multifaceted role in various human malignancies. While its clinical significance and mechanisms of action in uterine cervical cancer (UCC) remain unclear. This investigation demonstrated that the UCC cell line HeLa expressed CXCL3, and strong expression of CXCL3 was detected in UCC tissues relative to nontumor tissues. In addition, CXCL3 expression was strongly correlated with CXCL5 expression in UCC tissues. In vitro, HeLa cells overexpressing CXCL3, HeLa cells treated with exogenous CXCL3 or treated with conditioned medium from WPMY cells overexpressing CXCL3, exhibited enhanced proliferation and migration activities. In agreement with these findings, CXCL3 overexpression was also associated with the generation of HeLa cell tumor xenografts in athymic nude mice. Subsequent mechanistic studies demonstrated that CXCL3 overexpressing influenced the expression of extracellular signal-regulated kinase (ERK) signaling pathway associated genes, including ERK1/2, Bcl-2, and Bax, whereas the CXCL3-induced proliferation and migration effects were attenuated by exogenous administration of the ERK1/2 blocker PD98059. The data of the current investigation support that CXCL3 appears to hold promise as a potential tumor marker and interference target for UCC.


Asunto(s)
Quimiocinas CXC/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Neoplasias del Cuello Uterino/enzimología , Adulto , Anciano , Animales , Apoptosis , Movimiento Celular , Proliferación Celular , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Quimiocinas CXC/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Comunicación Paracrina , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Regulación hacia Arriba , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
16.
Hepatology ; 70(4): 1214-1230, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30933361

RESUMEN

Tumor-associated neutrophils (TANs) play a crucial role in tumor development and progression in the cancer microenvironment. Despite increased understanding of TAN contributions to hepatocellular carcinoma (HCC) progression and prognosis, the direct interaction between TANs and HCC cells is not fully understood. In this study, we tested the effect of TANs on HCC cells in vitro and in vivo and investigated the mechanism of interaction between them. Our results showed that TANs secreted bone morphogenetic protein 2 and transforming growth factor beta 2 and triggered microRNA 301b-3p (miR-301-3p) expression in HCC cells, subsequently suppressed gene expression of limbic system-associated membrane protein (LSAMP) and CYLD lysine 63 deubiquitinase (CYLD), and increased stem cell characteristics in HCC cells. These TAN-induced HCC stem-like cells were hyperactive in nuclear factor kappa B signaling, secreted higher levels of chemokine (C-X-C motif) ligand 5 (CXCL5), and recruited more TAN infiltration, suggesting a positive feedback loop. In clinical HCC samples, increased TANs correlated with elevated miR-301b-3p, decreased LSAMP and CYLD expression, and increased nuclear p65 accumulation and CXCL5 expression, all of which predicted patient outcome. Conclusion: Our work identified a positive feedback loop governing cancer stem-like cells and TANs in HCC that controls tumor progression and patient outcome.


Asunto(s)
Carcinoma Hepatocelular/genética , Quimiocina CXCL5/genética , Progresión de la Enfermedad , Neoplasias Hepáticas/genética , Neutrófilos/metabolismo , Animales , Biopsia con Aguja , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Supervivencia Celular/genética , Retroalimentación , Humanos , Inmunohistoquímica , Técnicas In Vitro , Neoplasias Hepáticas/patología , Ratones , Ratones SCID , MicroARNs/genética , Células Madre Neoplásicas/metabolismo , Pronóstico , Transducción de Señal/genética , Microambiente Tumoral
17.
Cytokine ; 126: 154873, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31629113

RESUMEN

Type 1 reactions (T1R) an inflammatory condition, of local skin patches in 30-40% leprosy patients during the course of MDT. IL-17A and IL-17F play an important role in regulating skin inflammation through neutrophils. In the present study, we have analyzed 18 of each T1R and Non-reactions (NR) patients through flow cytometry and qPCR. Interestingly we found that, CD3+CD4+ gated IL-17A+IL-17F+ cells were significantly high in T1R in both MLSA stimulated PBMCs and skin lesions as compared to NR leprosy patients. Hierarchical clustering analysis of gene expression showed that CXCL6, CXCL5, CCL20, CCL7, MMP13 and IL-17RB expression were significantly associated with IL-17A and IL-17F expression (Spearman r2 = 0.77 to 0.98), neutrophils and monocyte markers respectively. In this study, the inflammation noted in lesions of T1R is a different phenotype of Th17 which produce double positive IL-17A+IL17F+ and also contributes IL-17 producing neutrophils and thus would be useful for monitoring, diagnosis and treatment response before reactions episodes.


Asunto(s)
Citocinas/metabolismo , Interleucina-17/metabolismo , Lepra/inmunología , Mycobacterium leprae/inmunología , Neutrófilos/metabolismo , Células Th17/metabolismo , Adulto , Complejo CD3/metabolismo , Antígenos CD4/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Quimiocina CCL7/genética , Quimiocina CCL7/metabolismo , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Quimiocina CXCL6/genética , Quimiocina CXCL6/metabolismo , Citocinas/genética , Quimioterapia Combinada , Femenino , Citometría de Flujo , Humanos , Inflamación/genética , Inflamación/metabolismo , Lepra/patología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Persona de Mediana Edad , Familia de Multigenes , Reacción en Cadena en Tiempo Real de la Polimerasa
18.
Biotechnol Lett ; 42(12): 2569-2580, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32803430

RESUMEN

OBJECTIVES: The objective of this study was to discover unknown differentially expressed genes (DEGs) associated with bronchopulmonary dysplasia (BPD), analyze their functions and enriched signaling pathways, and identify hub genes correlating with BPD incidence and evolvement. RESULTS: Of 1289 DEGs identified, 568 were downregulated and 721 were upregulated. The DEGs were mainly associated with oxidative stress, angiogenesis, extracellular matrix, inflammation, cell cycle, and protein binding. Eight DEGs were identified as hub genes, including C-X-C motif chemokine ligand 5 (Cxcl5), connective tissue growth factor (Ctgf), interleukin 6 (IL6), matrix metallopeptidase 9 (Mmp9), mitogen-activated protein kinase 14 (Mapk14), platelet and endothelial cell adhesion molecule 1 (Pecam1), TIMP metallopeptidase inhibitor 1 (Timp1), and TIMP metallopeptidase inhibitor 2 (Timp2). IL6 mRNA and protein expression levels were significantly increased in the peripheral blood of neonates with BPD. CONCLUSIONS: Hence, BPD involves complex biological changes. Our findings indicate that inflammation and angiogenesis may play major roles in BPD pathogenesis and that IL6 has the potential to serve as a biomarker for early BPD diagnosis.


Asunto(s)
Biomarcadores/metabolismo , Displasia Broncopulmonar/genética , Biología Computacional , Interleucina-6/genética , Displasia Broncopulmonar/patología , Quimiocina CXCL5/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Regulación de la Expresión Génica/genética , Humanos , Metaloproteinasa 9 de la Matriz/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Transducción de Señal/genética , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-2/genética
19.
Infect Immun ; 87(3)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30559223

RESUMEN

The W-Beijing strain family is globally distributed and is associated with multidrug-resistant tuberculosis (TB) and treatment failure. Therefore, in this study, we examined the contribution of Toll-like receptor 2 (TLR2) to host resistance against Mycobacterium tuberculosis HN878, a clinical isolate belonging to the W-Beijing family. We show that TLR2 knockout (TLR2KO) mice infected with M. tuberculosis HN878 exhibit increased bacterial burden and are unable to control tissue-damaging, pulmonary neutrophilic inflammation. Consistent with a critical role for CXCL5 in regulating neutrophil influx, expression of epithelial cell-derived CXCL5 is significantly enhanced in TLR2KO mice prior to their divergence from wild-type (WT) mice in M. tuberculosis replication and neutrophilic inflammation. Depletion of neutrophils in TLR2KO mice by targeting Ly6G reverts lung inflammation and bacterial burden to levels comparable to those of WT mice. Together, the results establish that TLR2 controls neutrophil-driven immunopathology during infection with M. tuberculosis HN878 infection, likely by curtailing CXCL5 production.


Asunto(s)
Quimiocina CXCL5/metabolismo , Mycobacterium tuberculosis/clasificación , Neutrófilos/fisiología , Receptor Toll-Like 2/metabolismo , Tuberculosis/inmunología , Animales , Quimiocina CXCL5/genética , Femenino , Regulación de la Expresión Génica/inmunología , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organismos Libres de Patógenos Específicos , Receptor Toll-Like 2/genética
20.
Am J Physiol Gastrointest Liver Physiol ; 317(4): G398-G407, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31314571

RESUMEN

Initial colonizing bacteria play a critical role in completing the development of the immune system in the gastrointestinal tract of infants. Yet, the interaction of colonizing bacterial organisms with the developing human intestine favors inflammation over immune homeostasis. This characteristic of bacterial-intestinal interaction partially contributes to the pathogenesis of necrotizing enterocolitis (NEC), a devastating premature infant intestinal inflammatory disease. However, paradoxically some unique pioneer bacteria (initial colonizing species) have been shown to have a beneficial effect on the homeostasis of the immature intestine and the prevention of inflammation. We have reported that one such pioneer bacterium, Bacteroides fragilis (B. fragilis), and its surface component polysaccharide A (PSA) inhibit IL-1ß-induced inflammation in a human primary fetal small intestinal cell line (H4 cells). In this study, using transcription profiling of H4 cellular RNA after pretreatment with or without PSA before an inflammatory stimulation of IL-1ß, we have begun to further determine the cellular mechanism for anti-inflammation. We show that a developmentally regulated gene, zona pellucida protein 4 (ZP4), is uniquely elevated after IL-1ß stimulation and reduced with PSA exposure. ZP4 was known as a sperm receptor-mediating species-specific binding protein in the initial life of mammals. However, its intestinal epithelial function is unclear. We found that ZP4 is a developmentally regulated gene involved with immune function and regulated by both Toll-like receptor 2 and 4. Knockdown of ZP4-affected PSA inhibited IL-8 mRNA expression in response to IL-1ß. This represents an initial study of ZP4 innate immune function in immature enterocytes. This study may lead to new opportunity for efficient treatment of NEC.NEW & NOTEWORTHY This study extends previous observations to define the cellular mechanisms of polysaccharide A-induced anti-inflammation in immature enterocytes using transcription profiling of enterocyte genes after preexposure to polysaccharide A before an inflammatory stimulus with IL-1ß.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Bacteroides fragilis/química , Enterocitos/metabolismo , Polisacáridos/farmacología , Glicoproteínas de la Zona Pelúcida/genética , Glicoproteínas de la Zona Pelúcida/metabolismo , Antiinflamatorios no Esteroideos/química , Línea Celular , Quimiocina CXCL5/biosíntesis , Quimiocina CXCL5/genética , Enterocitos/efectos de los fármacos , Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-1beta/biosíntesis , Interleucina-8/biosíntesis , Interleucina-8/genética , Polisacáridos/química , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA