Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Redox Biol ; 54: 102359, 2022 08.
Article in English | MEDLINE | ID: mdl-35696764

ABSTRACT

Chemokines have been well-documented as a major factor in immune cell migration and the regulation of immune responses. However, recent studies have reported that chemokines have diverse roles, both in immune cells and other cell types, including adipocytes. This study investigated the molecular functions of C-X-C motif chemokine ligand 5 (CXCL5) in white adipose cells using Cxcl5 knock-out (KO) mice fed a high-fat diet (HFD). The expression of Cxcl5 decreased by 90% during adipocyte differentiation and remained at a low level in mature adipocytes. Moreover, adipogenesis was enhanced when adipocytes were differentiated from the stromal vascular fraction (SFV) of Cxcl5 KO mice. Feeding an HFD increased the generation of reactive oxygen species (ROS) and promoted abnormal adipogenesis in Cxcl5 KO mice. Oxidative stress and insulin resistance occurred in Cxcl5 KO mice due to decreased antioxidant enzymes and failure to remove ROS. These results indicate the principal roles of CXCL5 in adipogenesis and ROS regulation in adipose tissue, further suggesting that CXCL5 is a valuable chemokine for metabolic disease research.


Subject(s)
Adipocytes, White , Cell Differentiation , Chemokine CXCL5 , Diet, High-Fat , Oxidative Stress , Adipocytes, White/physiology , Adipogenesis , Animals , Chemokine CXCL5/physiology , Fatty Acids/metabolism , Insulin Resistance , Mice, Knockout , Oxidation-Reduction , Reactive Oxygen Species
2.
Microbiol Immunol ; 65(1): 40-47, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33026667

ABSTRACT

The ischemia-reperfusion-induced damage in human brain microvascular endothelial cells (BMECs) is associated with disruption of the blood-brain barrier. CXC chemokine ligand 5 (CXCL5) is reported to be up-regulated in ischemic stroke. However, the detailed function of CXCL5 in this pathological process remains largely unclear. To further analyze the function of CXCL5 in ischemic stroke, an oxygen-glucose deprivation model on human BMECs was constructed to mimic the ischemic stroke condition in vitro. Cell proliferation was analyzed using a cell counting kit-8 (CCK-8) assay. Quantitative real-time polymerase chain reaction and western blot were utilized to determine gene expression. The barrier function of BMECs was assessed using a fluorescently labeled dextran assay and a trans-epithelial/endothelial electrical resistance (TEER) technique. The results indicated that CXCL5 antibody (anti-CXCL5) promoted the proliferation of model cells, whereas it reduced the permeability. Moreover, the TEER value of model cells was enhanced in the presence of anti-CXCL5. Therefore, these findings demonstrated that CXCL5 silencing attenuated the ischemic/hypoxic-induced injury in human BMECs. Importantly, human recombinant protein CXCL5 (Re-CXCL5) deeply disrupted the function of BMECs in the normoxic condition. Furthermore, the p38 inhibitor SB203580 significantly abolished the function of CXCL5 in model cells. More importantly, similar results were also obtained in BMECs under normoxic conditions in the presence of Re-CXCL5. These results indicated that CXCL5 might regulate the function of BMECs by mediating the p38 pathway. This investigation not only enhanced the understanding of the biological effect of CXCL5 in human BMECs under ischemic/hypoxic conditions but also indicated its potential value as a therapeutic target for ischemic-induced brain disease.


Subject(s)
Brain , Chemokine CXCL5/physiology , Endothelial Cells , p38 Mitogen-Activated Protein Kinases/physiology , Brain/cytology , Cells, Cultured , Endothelial Cells/cytology , Humans , Ligands , Permeability
3.
Cancer Commun (Lond) ; 40(2-3): 69-80, 2020 03.
Article in English | MEDLINE | ID: mdl-32237072

ABSTRACT

The components of the tumor microenvironment (TME) in solid tumors, especially chemokines, are currently attracting much attention from scientists. C-X-C motif chemokine ligand 5 (CXCL5) is one of the important chemokines in TME. Overexpression of CXCL5 is closely related to the survival time, recurrence and metastasis of cancer patients. In TME, CXCL5 binds to its receptors, such as C-X-C motif chemokine receptor 2 (CXCR2), to participate in the recruitment of immune cells and promote angiogenesis, tumor growth, and metastasis. The CXCL5/CXCR2 axis can act as a bridge between tumor cells and host cells in TME. Blocking the transmission of CXCL5/CXCR2 signals can increase the sensitivity and effectiveness of immunotherapy and slow down tumor progression. CXCL5 and CXCR2 are also regarded as biomarkers for predicting prognosis and molecular targets for customizing the treatment. In this review, we summarized the current literature regarding the biological functions and clinical significance of CXCL5/CXCR2 axis in TME. The possibility to use CXCL5 and CXCR2 as potential prognostic biomarkers and therapeutic targets in cancer is also discussed.


Subject(s)
Chemokine CXCL5/physiology , Neoplasms/drug therapy , Receptors, Interleukin-8B/physiology , Tumor Microenvironment/physiology , Biomarkers, Tumor , Chemokine CXCL5/antagonists & inhibitors , Disease Progression , Humans , Neoplasms/diagnosis , Prognosis , Receptors, Interleukin-8B/antagonists & inhibitors
4.
Hepatology ; 69(1): 222-236, 2019 01.
Article in English | MEDLINE | ID: mdl-30014484

ABSTRACT

Transforming growth factor (TGF)-ß suppresses early hepatocellular carcinoma (HCC) development but triggers pro-oncogenic abilities at later stages. Recent data suggest that the receptor tyrosine kinase Axl causes a TGF-ß switch toward dedifferentiation and invasion of HCC cells. Here, we analyzed two human cellular HCC models with opposing phenotypes in response to TGF-ß. Both HCC models showed reduced proliferation and clonogenic growth behavior following TGF-ß stimulation, although they exhibited differences in chemosensitivity and migratory abilities, suggesting that HCC cells evade traits of anti-oncogenic TGF-ß. Transcriptome profiling revealed differential regulation of the chemokine CXCL5, which positively correlated with TGF-ß expression in HCC patients. The expression and secretion of CXCL5 was dependent on Axl expression, suggesting that CXCL5 is a TGF-ß target gene collaborating with Axl signaling. Loss of either TGF-ß or Axl signaling abrogated CXCL5-dependent attraction of neutrophils. In mice, tumor formation of transplanted HCC cells relied on CXCL5 expression. In HCC patients, high levels of Axl and CXCL5 correlated with advanced tumor stages, recruitment of neutrophils into HCC tissue, and reduced survival. Conclusion: The synergy of TGF-ß and Axl induces CXCL5 secretion, causing the infiltration of neutrophils into HCC tissue. Intervention with TGF-ß/Axl/CXCL5 signaling may be an effective therapeutic strategy to combat HCC progression in TGF-ß-positive patients.


Subject(s)
Carcinoma, Hepatocellular/immunology , Chemokine CXCL5/physiology , Liver Neoplasms/immunology , Neutrophil Infiltration , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Transforming Growth Factor beta/physiology , Animals , Humans , Mice , Tumor Cells, Cultured , Axl Receptor Tyrosine Kinase
6.
J. physiol. biochem ; 74(2): 313-324, mayo 2018. ilus, tab, graf
Article in English | IBECS | ID: ibc-178987

ABSTRACT

Epithelial neutrophil-activating peptide-78 (CXCL5), a member of the subgroup of CXC-type chemokine family, is an inflammatory factor involved in the progression of lung cancer, but the underlying mechanism remains unclear. In this study, we investigated the effects of CXCL5 on proliferation and migration in non-small cell lung cancer (NSCLC) using tissue microarrays from NSCLC patients and H460 cells transfected with a CXCL5-interfered lentivirus vector or stimulated with recombinant CXCL5. We observed that the expression of CXCL5 was significantly higher in lung cancer cell lines, and high CXCL5 was associated with high chemokine (C-X-C motif) receptor 2 expression and was significantly associated with poor differentiation. The high expression of CXCL5 was associated with poor NSCLC prognosis and was an independent predictive factor. Furthermore, downregulation of CXCL5 in H460 cells significantly reduced proliferation and migration. Recombinant CXCL5 promoted H460 cell proliferation and movement by activating MAPK/ERK1/2 and PI3K/AKT signaling. Our study elucidates the important role of CXCL5 in the progression and prognosis of NSCLC. These findings suggested that CXCL5 might be a potential biomarker and novel therapeutic target for lung cancer


Subject(s)
Humans , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/physiology , Chemokine CXCL5/physiology , Lung Neoplasms/pathology , Biomarkers, Tumor/physiology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Transformed , Cell Line, Tumor , Chemokine CXCL5/genetics , Disease Progression , Lung Neoplasms/metabolism , Prognosis
7.
Biochem Biophys Res Commun ; 498(4): 862-868, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29545183

ABSTRACT

The emergence of chemoresistance greatly increases the recurrence risk for non-muscle invasive bladder cancer (NMIBC) patients, which is still a big concern of clinicians. Understanding the mechanisms of drug resistance is of great significance for preventing and reversing it. We showed here that CXC motif chemokine ligand 5 (CXCL5) was overexpressed in mitomycin C-resistant bladder cancer cell line M-RT4. Meanwhile, parental RT4 cell treated with recombinant human CXCL5 (rhCXCL5) reduced its sensitivity to mitomycin C. Conversely, knockdown CXCL5 sensitized M-RT4 cell. We further investigated the molecular mechanisms finding that epithelial mesenchymal transition (EMT) and NF-κB pathway were activated in M-RT4 cell, which could be attenuated by knockdown CXCL5. All these data indicated that CXCL5 may promote mitomycin resistance by activating EMT and NF-κB pathway. Thus, our study identifies CXCL5 as a novel chemoresistance-related marker in NMIBC, thereby providing new strategies to overcome chemoresistance for NMIBC patients.


Subject(s)
Chemokine CXCL5/physiology , Drug Resistance, Neoplasm , Mitomycin/pharmacology , Cell Line, Tumor , Chemokine CXCL5/genetics , Chemokine CXCL5/metabolism , Epithelial-Mesenchymal Transition , Gene Knockdown Techniques , Humans , NF-kappa B/metabolism , Urinary Bladder Neoplasms
8.
J Physiol Biochem ; 74(2): 313-324, 2018 May.
Article in English | MEDLINE | ID: mdl-29526026

ABSTRACT

Epithelial neutrophil-activating peptide-78 (CXCL5), a member of the subgroup of CXC-type chemokine family, is an inflammatory factor involved in the progression of lung cancer, but the underlying mechanism remains unclear. In this study, we investigated the effects of CXCL5 on proliferation and migration in non-small cell lung cancer (NSCLC) using tissue microarrays from NSCLC patients and H460 cells transfected with a CXCL5-interfered lentivirus vector or stimulated with recombinant CXCL5. We observed that the expression of CXCL5 was significantly higher in lung cancer cell lines, and high CXCL5 was associated with high chemokine (C-X-C motif) receptor 2 expression and was significantly associated with poor differentiation. The high expression of CXCL5 was associated with poor NSCLC prognosis and was an independent predictive factor. Furthermore, downregulation of CXCL5 in H460 cells significantly reduced proliferation and migration. Recombinant CXCL5 promoted H460 cell proliferation and movement by activating MAPK/ERK1/2 and PI3K/AKT signaling. Our study elucidates the important role of CXCL5 in the progression and prognosis of NSCLC. These findings suggested that CXCL5 might be a potential biomarker and novel therapeutic target for lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/physiology , Chemokine CXCL5/physiology , Lung Neoplasms/pathology , Neoplasm Metastasis/physiopathology , Biomarkers, Tumor/physiology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Transformed , Cell Line, Tumor , Chemokine CXCL5/genetics , Disease Progression , Gene Silencing , Humans , Lung Neoplasms/metabolism , Prognosis , Protein Kinases/metabolism , Up-Regulation
9.
Oncol Res ; 25(2): 177-186, 2017 Jan 26.
Article in English | MEDLINE | ID: mdl-28277189

ABSTRACT

CXCL5, a CXC-type chemokine, is an important attractant for granulocytic immune cells by binding to its receptor CXCR2. Recently, CXCL5/CXCR2 has been found to play an oncogenic role in many human cancers. However, the exact role of CXCL5 in osteosarcoma cell migration and invasion has not been revealed. Here we found that the protein expression of CXCL5 was significantly increased in osteosarcoma tissues compared with that in matched adjacent nontumor tissues. Moreover, the expression of CXCL5 was significantly associated with advanced clinical stage and metastasis. Further investigation showed that the CXCL5 expression levels were also significantly increased in osteosarcoma cell lines, including Saos-2, MG63, U2OS, and SW1353, when compared with those in normal osteoblast hFoB1.19 cells. U2OS cells were further transfected with CXCL5-specific siRNA or overexpression plasmid. Knockdown of CXCL5 significantly suppressed U2OS cell migration and invasion. On the contrary, overexpression of CXLC5 remarkably promoted the migration and invasion of U2OS cells. Interestingly, both exogenous CXCL5 treatment and the conditioned medium of CXCL5-overexpressing hFoB1.19 cells could also enhance the migration and invasion of U2OS cells, suggesting that the promoting role of CXCL5 in U2OS cell migration and invasion is also in a paracrine-dependent manner. According to these data, our study demonstrates that CXCL5 is upregulated in osteosarcoma and may play an oncogenic role in osteosarcoma metastasis. Therefore, CXCL5 may become a potential therapeutic target for osteosarcoma treatment.


Subject(s)
Autocrine Communication/physiology , Cell Movement/physiology , Chemokine CXCL5/physiology , Neoplasm Invasiveness/pathology , Osteosarcoma/pathology , Paracrine Communication/physiology , Cell Line, Tumor , Humans , Osteosarcoma/metabolism
10.
Oncol Rep ; 36(6): 3303-3310, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27748886

ABSTRACT

CXCL5 and its receptor CXCR2 have been found to be involved in tumorigenesis and cancer progression. Recent studies have shown that CXCR2 is upregulated in glioma tissues, and associated with poor prognosis and recurrence. However, the role of CXCL5/CXCR2 signaling in mediating the malignant phenotypes of glioma cells, as well as the underlying mechanism, still remains unclear. In the present study, we found that CXCL5 was upregulated in glioma tissues compared to that noted in normal brain tissues. High CXCL5 levels were significantly associated with higher tumor grade, advanced clinical stage, and shorter survival time of glioma patients. In vitro studies indicated that the protein expression levels of CXCL5 and CXCR2 were markedly higher in human glioma cell lines (U87, U251, U373 and A172), when compared with those in normal human gliocyte HEB cells. Overexpression of CXLC5 significantly promoted the proliferation and migration of U87 cells, while knockdown of CXCL5 by small interfering RNA markedly inhibited U87 cell proliferation and migration. Moreover, both exogenous CXCL5 treatment and the conditioned medium of CXCL5-overexpressing HEB cells also enhanced the proliferation and migration of U87 cells. Molecular mechanism investigation revealed that CXLC5 activated the ERK, JNK, p38 MAPK signaling pathways, which play key roles in tumor growth and metastasis. According to these data, our study suggests that CXCL5 plays a promoting role in glioma in autocrine- and paracrine-dependent manners.


Subject(s)
Brain Neoplasms/metabolism , Cell Proliferation , Chemokine CXCL5/physiology , Glioma/metabolism , Autocrine Communication , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Female , Glioma/pathology , Humans , Male , Middle Aged , Paracrine Communication , Signal Transduction
11.
J Transl Med ; 12: 193, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25011526

ABSTRACT

Inflammatory factors play a vital role in the progression of liver cancer, although exact factors and related mechanisms still remain unclear. The present study aimed at screening inflammatory factors related to liver cancer metastasis and investigating the potential mechanism by which cancer cells are recruited. We screened and validated inflammatory factors by microarray and RT-PCR. Small interfering RNA (siRNA) and recombinant protein were used to assess CXCL5 effects on the movement of liver cancer cells (LCCs). Our screening microarray demonstrated over-expression of CXCL5 in LCCs with high metastatic potentials. CXCL5 increased LCCs migration and invasion, probably through autocrine and paracrine mechanisms. CXCL5-CXCR2 and ERK1/2 pathways could play critical roles in the regulation of LCCs migration. Our data indicates that LCCs per se may act as the producer and receptor of CXCL5 responsible for liver cancer migration and invasion.


Subject(s)
Chemokine CXCL5/physiology , Liver Neoplasms/pathology , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Blotting, Western , Humans , Liver Neoplasms/physiopathology , Polymerase Chain Reaction
12.
Biochem Biophys Res Commun ; 446(1): 18-24, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24583128

ABSTRACT

CXCL5 (epithelial neutrophil activating peptide-78) which acts as a potent chemoattractant and activator of neutrophil function was reported to play a multifaceted role in tumorigenesis. To investigate the role of CXCL5 in bladder cancer progression, we examined the CXCL5 expression in bladder cancer tissues by real-time PCR and Western blot, additionally, we used shRNA-mediated silencing to generate stable CXCL5 silenced bladder cancer T24 cells and defined its biological functions. Our results demonstrated that mRNA and protein of CXCL5 is increased in human bladder tumor tissues and cell lines, down-regulation of CXCL5 in T24 cells resulted in significantly decreased cell proliferation, migration and increased cell apoptosis in vitro through Snail, PI3K-AKT and ERK1/2 signaling pathways. These data suggest that CXCL5 is critical for bladder tumor growth and progression, it may represent a potential application in cancer diagnosis and therapy.


Subject(s)
Chemokine CXCL5/antagonists & inhibitors , Chemokine CXCL5/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/therapy , Apoptosis/genetics , Apoptosis/physiology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation , Chemokine CXCL5/physiology , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/metabolism , Urinary Bladder Neoplasms/pathology
13.
J Clin Invest ; 124(3): 1268-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24509076

ABSTRACT

Successful host defense against numerous pulmonary infections depends on bacterial clearance by polymorphonuclear leukocytes (PMNs); however, excessive PMN accumulation can result in life-threatening lung injury. Local expression of CXC chemokines is critical for PMN recruitment. The impact of chemokine-dependent PMN recruitment during pulmonary Mycobacterium tuberculosis infection is not fully understood. Here, we analyzed expression of genes encoding CXC chemokines in M. tuberculosis-infected murine lung tissue and found that M. tuberculosis infection promotes upregulation of Cxcr2 and its ligand Cxcl5. To determine the contribution of CXCL5 in pulmonary PMN recruitment, we generated Cxcl5(-/-) mice and analyzed their immune response against M. tuberculosis. Both Cxcr2(-/-) mice and Cxcl5(-/-) mice, which are deficient for only one of numerous CXCR2 ligands, exhibited enhanced survival compared with that of WT mice following high-dose M. tuberculosis infection. The resistance of Cxcl5(-/-) mice to M. tuberculosis infection was not due to heightened M. tuberculosis clearance but was the result of impaired PMN recruitment, which reduced pulmonary inflammation. Lung epithelial cells were the main source of CXCL5 upon M. tuberculosis infection, and secretion of CXCL5 was reduced by blocking TLR2 signaling. Together, our data indicate that TLR2-induced epithelial-derived CXCL5 is critical for PMN-driven destructive inflammation in pulmonary tuberculosis.


Subject(s)
Alveolar Epithelial Cells/immunology , Chemokine CXCL5/physiology , Mycobacterium tuberculosis/immunology , Neutrophils/immunology , Tuberculosis, Pulmonary/immunology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/microbiology , Animals , Cell Line , Inflammation/metabolism , Inflammation/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils/microbiology , Receptors, Interleukin-8B/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Toll-Like Receptor 2/metabolism , Transcriptional Activation , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/pathology
14.
Carcinogenesis ; 35(3): 597-605, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24293410

ABSTRACT

CXCL5 is a member of the CXC-type chemokine family that may play a role in carcinogenesis and cancer progression. This study investigates the biological function and clinical significance of CXCL5 in intrahepatic cholangiocarcinoma (ICC). We demonstrated that CXCL5 was overexpressed in ICC cell lines and tumor samples compared with paired normal tissues. CXCL5 had a direct chemoattractant effect on neutrophils in vitro through PI3K-Akt and extracellular signal-regulated kinase 1/2 signaling pathways. In animal studies, CXCL5 promoted tumor growth and metastasis without altering in vitro proliferative and invasive ability of ICC cells, and this effect was mediated by the recruitment of intratumoral infiltrative neutrophils by tumor-derived CXCL5. Immunohistochemical analysis of ICC samples showed that overexpression of CXCL5 correlated strongly with intratumoral neutrophil infiltration, shorter overall survival and high tumor recurrence. Multivariate analysis revealed that CXCL5 overexpression alone, or combined with the presence of intratumoral neutrophils, was an independent prognostic indicator for ICC. In conclusion, our data showed that CXCL5 promotes ICC growth and metastasis by recruiting intratumoral neutrophils. CXCL5 alone or combined with intratumoral neutrophils is a novel prognostic predictor for ICC patients and a potential therapeutic target.


Subject(s)
Chemokine CXCL5/physiology , Cholangiocarcinoma/physiopathology , Liver Neoplasms/physiopathology , Lymphocytes, Tumor-Infiltrating/pathology , Neoplasm Metastasis , Neutrophils/physiology , Cholangiocarcinoma/pathology , Humans , Liver Neoplasms/pathology , Up-Regulation
15.
Int J Cancer ; 133(12): 2872-83, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23737434

ABSTRACT

Increasing evidence indicates that myeloid-derived suppressor cells (MDSCs) negatively regulate immune responses during tumor progression, inflammation and infection. However, the underlying molecular mechanisms of their development and mobilization remain to be fully delineated. Kruppel-like factor KLF4 is a transcription factor that has an oncogenic function in breast cancer development, but its function in tumor microenvironment, a critical component for tumorigenesis, has not been examined. By using a spontaneously metastatic 4T1 breast cancer mouse model and an immunodeficient NOD/SCID mouse model, we demonstrated that KLF4 knockdown delayed tumor development and inhibited pulmonary metastasis, which accompanied by decreased accumulation of MDSCs in bone marrow, spleens and primary tumors. Mechanistically, we found that KLF4 knockdown resulted in a significant decrease of circulating GM-CSF, an important cytokine for MDSC biology. Consistently, recombinant GM-CSF restored the frequency of MDSCs in purified bone marrow cells incubated with conditioned medium from KLF4 deficient cells. In addition, we identified CXCL5 as a critical mediator to enhance the expression and function of GM-CSF. Reduced CXCL5 expression by KLF4 knockdown in primary tumors and breast cancer cells was correlated with a decreased GM-CSF expression in our mouse models. Finally, we found that CXCL5/CXCR2 axis facilitated MDSC migration and that anti-GM-CSF antibodies neutralized CXCL5-induced accumulation of MDSCs. Taken together, our data suggest that KLF4 modulates maintenance of MDSCs in bone marrow by inducing GM-CSF production via CXCL5 and regulates recruitment of MDSCs into the primary tumors through the CXCL5/CXCR2 axis, both of which contribute to KLF4-mediated mammary tumor development.


Subject(s)
Breast Neoplasms/pathology , Kruppel-Like Transcription Factors/physiology , Lung Neoplasms/secondary , Myeloid Cells/physiology , Breast Neoplasms/immunology , Cell Line, Tumor , Cell Movement , Chemokine CXCL5/physiology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Humans , Immune Tolerance , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/antagonists & inhibitors , Lung Neoplasms/prevention & control , Tumor Burden
16.
J Clin Invest ; 123(3): 1343-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23376791

ABSTRACT

The ELR(+)-CXCL chemokines have been described typically as potent chemoattractants and activators of neutrophils during the acute phase of inflammation. Their role in atherosclerosis, a chronic inflammatory vascular disease, has been largely unexplored. Using a mouse model of atherosclerosis, we found that CXCL5 expression was upregulated during disease progression, both locally and systemically, but was not associated with neutrophil infiltration. Unexpectedly, inhibition of CXCL5 was not beneficial but rather induced a significant macrophage foam cell accumulation in murine atherosclerotic plaques. Additionally, we demonstrated that CXCL5 modulated macrophage activation, increased expression of the cholesterol efflux regulatory protein ABCA1, and enhanced cholesterol efflux activity in macrophages. These findings reveal a protective role for CXCL5, in the context of atherosclerosis, centered on the regulation of macrophage foam cell formation.


Subject(s)
Atherosclerosis/immunology , Chemokine CXCL5/physiology , Foam Cells/immunology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Aorta/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Chemokine CXCL5/genetics , Chemokine CXCL5/metabolism , Cholesterol/metabolism , Endothelial Cells/metabolism , Foam Cells/metabolism , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , RNA, Messenger/blood , RNA, Messenger/genetics , Receptors, Interleukin-8B/metabolism , Transcriptional Activation , Up-Regulation
17.
Am J Physiol Gastrointest Liver Physiol ; 303(1): G93-102, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22538401

ABSTRACT

Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants. In tissue samples of NEC, we identified numerous macrophages and a few neutrophils but not many lymphocytes. We hypothesized that these pathoanatomic characteristics of NEC represent a common tissue injury response of the gastrointestinal tract to a variety of insults at a specific stage of gut development. To evaluate developmental changes in mucosal inflammatory response, we used trinitrobenzene sulfonic acid (TNBS)-induced inflammation as a nonspecific insult and compared mucosal injury in newborn vs. adult mice. Enterocolitis was induced in 10-day-old pups and adult mice (n = 25 animals per group) by administering TNBS by gavage and enema. Leukocyte populations were enumerated in human NEC and in murine TNBS-enterocolitis using quantitative immunofluorescence. Chemokine expression was measured using quantitative polymerase chain reaction, immunoblots, and immunohistochemistry. Macrophage recruitment was investigated ex vivo using intestinal tissue-conditioned media and bone marrow-derived macrophages in a microchemotaxis assay. Similar to human NEC, TNBS enterocolitis in pups was marked by a macrophage-rich leukocyte infiltrate in affected tissue. In contrast, TNBS-enterocolitis in adult mice was associated with pleomorphic leukocyte infiltrates. Macrophage precursors were recruited to murine neonatal gastrointestinal tract by the chemokine CXCL5, a known chemoattractant for myeloid cells. We also demonstrated increased expression of CXCL5 in surgically resected tissue samples of human NEC, indicating that a similar pathway was active in NEC. We concluded that gut mucosal injury in the murine neonate is marked by a macrophage-rich leukocyte infiltrate, which contrasts with the pleomorphic leukocyte infiltrates in adult mice. In murine neonatal enterocolitis, macrophages were recruited to the inflamed gut mucosa by the chemokine CXCL5, indicating that CXCL5 and its cognate receptor CXCR2 merit further investigation as potential therapeutic targets in NEC.


Subject(s)
Intestinal Mucosa/pathology , Macrophages/physiology , Aging/physiology , Animals , Animals, Newborn , Blotting, Western , Chemokine CXCL5/physiology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , Denaturing Gradient Gel Electrophoresis , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , In Vitro Techniques , Infant, Newborn , Infant, Premature , Inflammation/pathology , Intestinal Diseases/chemically induced , Intestinal Diseases/pathology , Intestinal Mucosa/cytology , Mice , Neutrophil Infiltration/physiology , Polymerase Chain Reaction , Risk Factors , Trinitrobenzenesulfonic Acid
18.
J Sex Med ; 8(2): 437-46, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21114767

ABSTRACT

INTRODUCTION: Previously we reported that paracrine actions likely mediated the therapeutic effects of adipose tissue-derived stem cells (ADSCs) on a rat model of cavernous nerve (CN) injury. AIM: To identify potential neurotrophic factors in ADSC's secretion, test the most promising one, and identify the molecular mechanism of its neurotrophic action. METHODS: Rat major pelvic ganglia (MPG) were cultured in conditioned media of ADSC and penile smooth muscle cells (PSMCs). Cytokine expression in these two media was probed with a cytokine antibody array. CXCL5 cytokine was quantified in these two media by enzyme-linked immunosorbent assay (ELISA). Activation of Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) by CXCL5 was tested in neuroblastoma cell lines BE(2)C and SH-SY5Y as well as in Schwann cell line RT4-D6P2T by Western blot. Involvement of CXCL5 and JAK/STAT in ADSC-conditioned medium's neurotrophic effects was confirmed with anti-CXCL5 antibody and JAK inhibitor AG490, respectively. MAIN OUTCOME MEASURES: Neurotrophic effects of ADSC and PSMC-conditioned media were quantified by measuring neurite length in MPG cultures. Secretion of CXCL5 in these two media was quantified by ELISA. Activation of JAK/STAT by CXCL5 was quantified by densitometry on Western blots for STAT1 and STAT3 phosphorylation. RESULTS: MPG neurite length was significantly longer in ADSC than in PSMC-conditioned medium. CXCL5 was secreted eight times higher in ADSC than in PSMC-conditioned medium. Anti-CXCL5 antibody blocked the neurotrophic effects of ADSC-conditioned medium. CXCL5 activated JAK/STAT concentration-dependently from 0 to 50 ng/mL in RT4-D6P2T Schwann cells. At 50 ng/mL, CXCL5 activated JAK/STAT time-dependently, peaking at 45 minutes. AG490 blocked these activities as well as the neurotrophic effects of ADSC-conditioned medium. CONCLUSIONS: CXCL5 was secreted by ADSC at a high level, promoted MPG neurite growth, and activated JAK/STAT in Schwann cells. CXCL5 may contribute to ADSC's therapeutic efficacy on CN injury-induced ED.


Subject(s)
Adipose Tissue/cytology , Chemokine CXCL5/physiology , Nerve Regeneration/physiology , Penis/innervation , Stem Cells/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Chemokine CXCL5/pharmacology , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay , Janus Kinases/metabolism , Male , Muscle, Smooth, Vascular/innervation , Muscle, Smooth, Vascular/physiology , Nerve Regeneration/drug effects , Neurites/drug effects , Neurites/physiology , Penis/physiology , Rats , Rats, Sprague-Dawley , STAT Transcription Factors/metabolism , Schwann Cells/drug effects , Schwann Cells/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/physiology
19.
Eur Respir J ; 34(1): 184-90, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19047312

ABSTRACT

The aim of this study was to investigate the presence of epithelial neutrophil-activating peptide (ENA)-78 in pleural effusions, as well as the chemoattractant activity of pleural ENA-78 on neutrophils. Pleural effusion and serum samples were collected from 75 patients who presented to the respiratory institute (19 with malignant pleural effusion, 21 with tuberculous pleural effusion, 18 with infectious pleural effusion and 17 with transudative pleural effusion). The concentrations of ENA-78, myeloperoxidase and neutrophil elastase were determined, and the chemoattractant activity of ENA-78 for neutrophils both in vitro and in vivo was also observed. The concentrations of ENA-78, myeloperoxidase and neutrophil elastase in infectious pleural effusion were significantly higher than those in malignant, tuberculous and transudative groups, respectively (all p<0.01). Infectious pleural fluid was chemotactic for neutrophils in vitro and anti-ENA-78 antibody could partly inhibit these chemotactic effects. Intrapleural administration of ENA-78 produced a marked progressive influx of neutrophils into pleural space. Compared with noninfectious pleural effusion, ENA-78 appeared to be increased in infectious pleural effusion. Our data suggested that ENA-78 was able to induce neutrophil infiltration into pleural space and might be responsible for pleural neutrophil degranulation.


Subject(s)
Chemokine CXCL5/physiology , Neutrophils/immunology , Pleural Effusion/immunology , Adolescent , Adult , Aged , Chemokine CXCL5/metabolism , Chemotactic Factors/metabolism , Female , Humans , Leukocyte Elastase/metabolism , Male , Middle Aged , Models, Biological , Neutrophils/metabolism , Peroxidase/metabolism , Pleural Effusion/metabolism , Prospective Studies
20.
Clin Cancer Res ; 14(8): 2276-84, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18413816

ABSTRACT

PURPOSE: We isolated a subline (CC531M) from the CC531S rat colon carcinoma cell line, which grows and metastasizes much more rapidly than CC531S. We found, using RNA expression profiling, that one of the major changes in the CC531M cell line was a 5.8-fold reduction of the chemokine CXCL5. The purpose of this study was to determine the effect of CXCL5 expression on colorectal tumor growth and metastasis. EXPERIMENTAL DESIGN: CC531 clones were generated with either knockdown or restored expression of CXCL5. These clones were inoculated in the liver of rats. In addition, in two independent cohorts of colorectal cancer patients, the level of CXCL5 expression was determined and associated to clinical variables. RESULTS: Knockdown of CXCL5 expression in CC531S resulted in rapid tumor growth and increased number of metastasis, whereas restored expression of CXCL5 in CC531M resulted in a return of the "mild" tumor growth pattern of the parental cell line CC531S. In vitro, no difference was found in proliferation rate between clones with either high or low expression of CXCL5, suggesting that environmental interactions directed by CXCL5 determine tumor outgrowth. Finally, the importance of our findings was established for patients with colorectal cancer. We found that low expression of CXCL5 was significantly associated with poor prognosis for colorectal cancer patients. CXCL5 showed a trend (P = 0.05) for a positive correlation with intratumoral CD8(+) T-cell infiltration, suggesting a possible explanation for the observed poorer prognosis. CONCLUSIONS: Our results show that CXCL5 is important in growth and development of colorectal cancer, implicating a future role in both cancer therapy and diagnosis.


Subject(s)
Chemokine CXCL5/physiology , Colorectal Neoplasms/etiology , Aged , Animals , Cell Line, Tumor , Chemokine CXCL5/analysis , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Humans , Male , Prognosis , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...