Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 193(3): 1416-26, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24981451

ABSTRACT

Airway smooth muscle cell (ASMC) migration is an important mechanism postulated to play a role in airway remodeling in asthma. CXCL1 chemokine has been linked to tissue growth and metastasis. In this study, we present a detailed examination of the inhibitory effect of CXCL1 on human primary ASMC migration and the role of the decoy receptor, Duffy AgR for chemokines (DARC), in this inhibition. Western blots and pathway inhibitors showed that this phenomenon was mediated by activation of the ERK-1/2 MAPK pathway, but not p38 MAPK or PI3K, suggesting a biased selection in the signaling mechanism. Despite being known as a nonsignaling receptor, small interference RNA knockdown of DARC showed that ERK-1/2 MAPK activation was significantly dependent on DARC functionality, which, in turn, was dependent on the presence of heat shock protein 90 subunit α. Interestingly, DARC- or heat shock protein 90 subunit α-deficient ASMCs responded to CXCL1 stimulation by enhancing p38 MAPK activation and ASMC migration through the CXCR2 receptor. In conclusion, we demonstrated DARC's ability to facilitate CXCL1 inhibition of ASMC migration through modulation of the ERK-1/2 MAPK-signaling pathway.


Subject(s)
Airway Remodeling/immunology , Cell Migration Inhibition/immunology , Chemokine CXCL1/physiology , Duffy Blood-Group System/physiology , Receptors, Cell Surface/physiology , Receptors, Interleukin-8B/physiology , Biomarkers/metabolism , Chemokine CXCL1/metabolism , Chemokine CXCL2/physiology , Duffy Blood-Group System/metabolism , Humans , MAP Kinase Signaling System/immunology , Primary Cell Culture , Receptors, Cell Surface/metabolism , Receptors, Interleukin-8B/metabolism
2.
J Immunol ; 191(5): 2731-41, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23904157

ABSTRACT

Structural cell migration plays a central role in the pathophysiology of several diseases, including asthma. Previously, we established that IL-17-induced (CXCL1, CXCL2, and CXCL3) production promoted airway smooth muscle cell (ASMC) migration, and consequently we sought to investigate the molecular mechanism of CXC-induced ASMC migration. Recombinant human CXCL1, CXCL2, and CXCL3 were used to assess migration of human primary ASMCs from normal and asthmatic subjects using a modified Boyden chamber. Neutralizing Abs or small interfering RNA (siRNA) knockdown and pharmacological inhibitors of PI3K, ERK1/2, and p38 MAPK pathways were used to investigate the receptors and the signaling pathways involved in CXC-induced ASMC migration, respectively. We established the ability of CXCL2 and CXCL3, but not CXCL1, to induce ASMC migration at the tested concentrations using normal ASMCs. We found CXCL2-induced ASMC migration to be dependent on p38 MAPK and CXCR2, whereas CXCL3-induced migration was dependent on p38 and ERK1/2 MAPK pathways via CXCR1 and CXCR2. While investigating the effect of CXCL2 and CXCL3 on asthmatic ASMC migration, we found that they induced greater migration of asthmatic ASMCs compared with normal ones. Interestingly, unlike normal ASMCs, CXCL2- and CXCL3-induced asthmatic ASMC migration was mainly mediated by the PI3K pathway through CXCR1. In conclusion, our results establish a new role of CXCR1 in ASMC migration and demonstrate the diverse mechanisms by which CXCL2 and CXCL3 mediate normal and asthmatic ASMC migration, suggesting that they may play a role in the pathogenesis of airway remodeling in asthma.


Subject(s)
Airway Remodeling/physiology , Asthma/metabolism , Chemokine CXCL2/metabolism , Chemokines, CXC/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Interleukin-8A/metabolism , Asthma/pathology , Blotting, Western , Bronchi/metabolism , Bronchi/pathology , Cell Movement/physiology , Chemokine CXCL1/metabolism , Flow Cytometry , Humans , RNA, Small Interfering , Signal Transduction/physiology , Transfection
3.
J Assist Reprod Genet ; 30(3): 341-52, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23381551

ABSTRACT

PURPOSE: Human chorionic gonadotrophin (hCG) has been used to induce ovulation and oocyte maturation. Although the most common dose of hCG used in IVF is 10,000 IU, there are reports that suggest 5,000 IU is sufficient to yield similar results. The objective of this study is to evaluate the dose dependent differences in gene expression of granulosa cells following various doses of hCG treatment. METHODS: Patients with polycystic ovarian syndrome (PCOS) were stimulated for IVF treatment. The hCG injection was either withheld or given at 5,000 or 10,000 IU. Granulosa cells from the follicular fluids have been collected for RNA isolation and analyzed using Affymetrix genechip arrays. RESULTS: Unsupervised hierarchical clustering based on whole gene expression revealed two distinct groups of patients in this experiment. All untreated patients were clustered together whereas hCG-treated patients separated to a different group regardless of the dose. A large number of the transcripts were similarly up- or down-regulated across both hCG doses (2229 and 1945 transcripts, respectively). However, we observed dose-dependent statistically significant differences in gene expression in only 15 transcripts. CONCLUSIONS: Although hCG injection caused a major change in the gene expression profile of granulosa cells, 10,000 IU hCG resulted in minimal changes in the gene expression profiles of granulosa cells as compared with 5,000 IU. Thus, based on our results, we suggest the use of 10,000 IU hCG should be reconsidered in PCOS patients.


Subject(s)
Chorionic Gonadotropin/administration & dosage , Fertilization in Vitro , Oocytes , Polycystic Ovary Syndrome/pathology , Adult , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Granulosa Cells/metabolism , Humans , Microarray Analysis , Middle Aged , Oocytes/drug effects , Oocytes/growth & development , Ovulation/drug effects , Ovulation Induction , Polycystic Ovary Syndrome/drug therapy , Pregnancy , Pregnancy Rate
4.
J Allergy Clin Immunol ; 130(4): 977-85.e6, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22698519

ABSTRACT

BACKGROUND: Airway smooth muscle cell (ASMC) migration is one of the proposed mechanisms underlying the increased airway smooth muscle mass seen in airway remodeling of patients with severe asthma. IL-17-related cytokines are a new subgroup of inflammatory mediators that have been suggested to play a role in regulating smooth muscle function. We hypothesized that IL-17-induced chemokine production from smooth muscle cells can contribute to migration of additional smooth muscle cells in the airways of asthmatic patients. OBJECTIVE: We sought to investigate the effect of IL-17 on smooth muscle-derived chemokines and to examine the mechanisms involved in their production and contribution to the increase in airway smooth muscle migration. METHODS: The effect of IL-17-induced supernatants on human ASMC migration was investigated. IL-17-induced growth-related oncogene (GRO) production and mRNA expression was assessed by using ELISA and RT-PCR, respectively. The direct effect of GROs on ASMC migration and the involvement of the CXCR2 receptor were also examined. RESULTS: IL-17-induced supernatants promoted ASMC migration. After IL-17 stimulation, GROs were the most abundant chemokines produced from ASMCs, and blocking their effect by using neutralizing antibodies significantly inhibited ASMC migration. In addition, a combination of recombinant human GRO-α, GRO-ß, and GRO-γ was able to promote significant migration of ASMCs that was mediated through the CXCR2 receptor. CONCLUSION: These findings suggest that IL-17-induced GROs can be an important mediator of ASMC migration and therefore might contribute to the pathogenesis of airway remodeling in asthmatic patients.


Subject(s)
Autocrine Communication , Bronchi/cytology , Cell Movement , Chemokine CXCL1/physiology , Interleukin-17/pharmacology , Myocytes, Smooth Muscle/physiology , Cell Movement/drug effects , Chemokine CXCL1/genetics , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Receptors, Interleukin-8B/physiology , p38 Mitogen-Activated Protein Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...