Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Int Immunopharmacol ; 133: 112044, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38648716

ABSTRACT

BACKGROUND: The prevalence of type 2 diabetic nephropathy (T2DN) ranges from 20 % to 40 % among individuals with type 2 diabetes. Multiple immune pathways play a pivotal role in the pathogenesis of T2DN. This study aimed to investigate the immunomodulatory effects of active ingredients derived from 14 traditional Chinese medicines (TCMs) on T2DN. METHODS: By removing batch effect on the GSE30528 and GSE96804 datasets, we employed a combination of weighted gene co-expression network analysis, least absolute shrinkage and selection operator analysis, protein-protein interaction network analysis, and the CIBERSORT algorithm to identify the active ingredients of TCMs as well as potential hub biomarkers associated with immune cells. Functional analysis was conducted using Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO), and gene set variation analysis (GSVA). Additionally, molecular docking was employed to evaluate interactions between active ingredients and potential immunotherapy targets. RESULTS: A total of 638 differentially expressed genes (DEGs) were identified in this study, comprising 5 hub genes along with 4 potential biomarkers. Notably, CXCR1, CXCR2, and FOS exhibit significant associations with immune cells while displaying robust or favorable affinities towards the active ingredients kaempferol, quercetin, and luteolin. Furthermore, functional analysis unveiled intricate involvement of DEGs, hub genes and potential biomarkers in pathways closely linked to immunity and diabetes. CONCLUSION: The potential hub biomarkers and immunotherapy targets associated with immune cells of T2DN comprise CXCR1, CXCR2, and FOS. Furthermore, kaempferol, quercetin, and luteolin demonstrate potential immunomodulatory effects in modulating T2DN through the regulation of CXCR1, CXCR2, and FOS expression.


Subject(s)
Computational Biology , Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Drugs, Chinese Herbal , Medicine, Chinese Traditional , Molecular Docking Simulation , Network Pharmacology , Protein Interaction Maps , Receptors, Interleukin-8B , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/genetics , Diabetic Nephropathies/immunology , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/genetics , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism , Gene Regulatory Networks/drug effects
2.
Zhen Ci Yan Jiu ; 49(4): 331-340, 2024 Apr 25.
Article in English, Chinese | MEDLINE | ID: mdl-38649200

ABSTRACT

OBJECTIVES: To observe whether acupuncture up-regulates chemokine CXC ligand 1 (CXCL1) in the brain to play an analgesic role through CXCL1/chemokine CXC receptor 2 (CXCR2) signaling in adjuvant induced arthritis (AIA) rats, so as to reveal its neuro-immunological mechanism underlying improvement of AIA. METHODS: BALB/c mice with relatively stable thermal pain reaction were subjected to planta injection of complete Freund adjuvant (CFA) for establishing AIA model, followed by dividing the AIA mice into simple AF750 (fluorochrome) and AF750+CXCL1 groups (n=2 in each group). AF750 labeled CXCL1 recombinant protein was then injected into the mouse's tail vein to induce elevation of CXCL1 level in blood for simulating the effect of acupuncture stimulation which has been demonstrated by our past study. In vivo small animal imaging technology was used to observe the AF750 and AF750+CXCL1-labelled target regions. After thermal pain screening, the Wistar rats with stable pain reaction were subjected to AIA modeling by injecting CFA into the rat's right planta, then were randomized into model and manual acupuncture groups (n=12 in each group). Other 12 rats that received planta injection of saline were used as the control group. Manual acupuncture (uniform reinforcing and reducing manipulations) was applied to bilateral "Zusanli" (ST36) for 4×2 min, with an interval of 5 min between every 2 min, once daily for 7 days. The thermal pain threshold was assessed by detecting the paw withdrawal latency (PWL) using a thermal pain detector. The contents of CXCL1 in the primary somatosensory cortex (S1), medial prefrontal cortex, nucleus accumbens, amygdala, periaqueductal gray and rostroventromedial medulla regions were assayed by using ELISA, and the expression levels of CXCL1, CXCR2 and mu-opioid receptor (MOR) mRNA in the S1 region were detected using real time-quantitative polymerase chain reaction. The immune-fluorescence positive cellular rate of CXCL1 and CXCR2 in S1 region was observed after immunofluorescence stain. The immunofluorescence double-stain of CXCR2 and astrocyte marker glial fibrillary acidic protein (GFAP) or neuron marker NeuN or MOR was used to determine whether there is a co-expression between them. RESULTS: In AIA mice, results of in vivo experiments showed no obvious enrichment signal of AF750 or AF750+CXCL1 in any organ of the body, while in vitro experiments showed that there was a stronger fluorescence signal of CXCL1 recombinant protein in the brain. In rats, compared with the control group, the PWL from day 0 to day 7 was significantly decreased (P<0.01) and the expression of CXCR2 mRNA in the S1 region significantly increased in the model group (P<0.05), while in comparison with the model group, the PWL from day 2 to day 7, CXCL1 content, CXCR2 mRNA expression and CXCR2 content, and MOR mRNA expression in the S1 region were significantly increased in the manual acupuncture group (P<0.05, P<0.01). Immunofluorescence stain showed that CXCR2 co-stained with NeuN and MOR in the S1 region, indicating that CXCR2 exists in neurons and MOR-positive neurons but not in GFAP positive astrocytes. CONCLUSIONS: Acupuncture can increase the content of CXCL1 in S1 region, up-regulate CXCR2 on neurons in the S1 region and improve MOR expression in S1 region of AIA rats, which may contribute to its effect in alleviating inflammatory pain.


Subject(s)
Acupuncture Therapy , Arthritis, Experimental , Chemokine CXCL1 , Receptors, Interleukin-8B , Somatosensory Cortex , Animals , Humans , Male , Mice , Rats , Acupuncture Points , Arthritis, Experimental/therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/genetics , Chemokine CXCL1/metabolism , Chemokine CXCL1/genetics , Inflammation/therapy , Inflammation/metabolism , Inflammation/genetics , Mice, Inbred BALB C , Pain/metabolism , Pain/genetics , Pain Management , Rats, Wistar , Receptors, Interleukin-8B/metabolism , Receptors, Interleukin-8B/genetics , Signal Transduction , Somatosensory Cortex/metabolism
3.
Cytokine ; 153: 155828, 2022 05.
Article in English | MEDLINE | ID: mdl-35247648

ABSTRACT

Early in the 1980s several laboratories mistakenly reported that partially purified interleukin-1 (IL-1) was chemotactic for neutrophils. However, further investigations by us, revealed that our purified IL-1 did not have neutrophil chemotactic activity and this activity in the LPS-stimulated human monocyte conditioned media could clearly be separated from IL-1 activity on HPLC gel filtration. This motivated Teizo Yoshimura and Kouji Matsushima to purify the monocyte-derived neutrophil chemotactic factor (MDNCF), present in LPS conditioned media and molecularly clone the cDNA for MDNCF. They found that MDNCF protein (later renamed IL-8, and finally termed CXCL8) is first translated as a precursor form consisting of 99 amino acid residues and the signal peptide is then removed, leading to the secretion and processing of biologically active IL-8 of 72 amino acid form (residues 28-99). There are four cysteine residues forming two disulfide linkage and 14 basic amino acid residues which result in a very basic property for the binding of IL-8 to heparan sulfate-proteoglycan. The IL-8 gene consists of 4 exons and 3 introns. IL-8 is produced by various types of cells in inflammation. The 5'-flanking region of IL-8 gene contains several nuclear factor binding sites, and NF-κB in combination with AP-1 or C/EBP synergistically activates IL-8 gene in response to IL-1 and TNFα. Two receptors exist for IL-8, CXCR1 and CXCR2 in humans, which belong to γ subfamily of GTP binding protein (G-protein) coupled rhodopsin-like 7 transmembrane domain receptors. Rodents express CXCR2 and do not produce IL-8, but produce numerous homologues instead. Once IL-8 binds to the receptor, ß and γ subunits of G-protein are released from Gα (Gαi2 in neutrophils) and activate PI3Kγ, PLCß2/ß3, PLA2 and PLD. Gαi2 inhibits adenyl cyclase to decrease cAMP levels. Small GTPases Ras/Rac/Rho/cdc42/Rap1, PKC and AKT (PKB) exist down-stream of ß and γ subunits and regulate cell adhesion, actin polymerization, membrane protrusion, and eventually cell migration. PLCß activation generates IP3 and induces Ca++ mobilization, DAG generation to activate protein kinase C to lead granule exocytosis and respiratory burst. MDNCF was renamed interleukin 8 (IL-8) at the International Symposium on Novel Neutrophil Chemotactic Activating Polypeptides, London, UK in 1989. The discovery of IL-8 prompted us to also purify and molecularly clone the cDNA of MCAF/MCP-1 responsible for monocyte chemotaxis, and other groups to identify a large family of chemotactic cytokines capable of attracting other types of leukocytes. In 1992, most of the investigators contributing to the discovery of this new family of chemotactic cytokines gathered in Baden, Austria and agreed to name this family "chemokines" and subsequently established the CXCL/CCL and CXCR/CCR nomenclature. The discovery of chemokines resulted in solving the long-time enigma concerning the mechanism of cell type specific leukocyte infiltration into inflamed tissues and provided a molecular basis for immune and hematopoietic cell migration and interactions under physiological as well as pathological conditions. To our surprise based on its recently identified multifunctional activities, IL-8 has evolved from a neutrophil chemoattractant to a promising therapeutic target for a wide range of inflammatory and neoplastic diseases. IL-8 was initially characterized as a chemoattractant of neutrophils engaged in acute inflammation and then discovered to also be chemotactic for endothelial cells with a major role in angiogenesis. These two activities of IL-8 foster its stimulatory effect on tumor growth. This is abetted by recent additional discoveries showing that IL-8 has stimulatory effects on stem cells and can therefore directly promote the growth of receptor expressing cancer stem cells. IL-8 by interacting with bone marrow stem/progenitor cells has also the capacity to mobilize and release hematopoietic cells into the peripheral circulation. This includes the mobilization of neutrophilic myeloid-derived suppressor cells (N-MDSC) to infiltrate into tumors and thus further promotes the immune escape of tumors. Finally, the capacity of IL-8 to induce trans-differentiation of epithelial cancer cells into mesenchymal phenotype (EMT) increases the malignancy of tumors by promoting their metastatic spread and resistance to chemotherapeutics and cytotoxic immune cells. These observations have stimulated considerable current efforts to develop receptor antagonists for IL-8 and humanized anti-IL-8 antibody for the therapy of cancer, particularly in combination with immune checkpoint inhibitors, such as anti-PD-1/PD-L1 antibodies.


Subject(s)
Interleukin-8 , Lipopolysaccharides , Amino Acids/metabolism , Chemokine CCL2/metabolism , Chemokines/metabolism , Culture Media, Conditioned/metabolism , DNA, Complementary , Endothelial Cells , Humans , Inflammation/metabolism , Interleukin-1/metabolism , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Neutrophils/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism
4.
Tumour Biol ; 43(1): 37-55, 2021.
Article in English | MEDLINE | ID: mdl-33935122

ABSTRACT

BACKGROUND: Green synthesized nanoparticles have been earmarked for use in nanomedicine including for the development of better anticancer drugs. OBJECTIVE: The aim of this study was to undertake biochemical evaluation of anticancer activities of green synthesized silver nanoparticles (AgNPs) from ethanolic extracts of fruits (AgNPs-F) and leaves (AgNPs-L) of Annona muricata. METHODS: Previously synthesized silver nanoparticles were used for the study. The effects of the AgNPs and 5-Fluorouracil were studied on PC3, HeLa and PNT1A cells. The resazurin, migration and colonogenic assays as well as qRT-PCR were employed. RESULTS: The AgNPs-F displayed significant antiproliferative effects against HeLa cells with an IC50 of 38.58µg/ml and PC3 cells with an IC50 of 48.17µg/ml but selectively spared normal PNT1A cells (selectivity index of 7.8), in comparison with first line drug 5FU and AgNPs-L whose selectivity index were 3.56 and 2.26 respectively. The migration assay revealed potential inhibition of the metastatic activity of the cells by the AgNPs-F while the colonogenic assay indicated the permanent effect of the AgNPs-F on the cancer cells yet being reversible on the normal cells in contrast with 5FU and AgNPs-L. CASP9 was significantly over expressed in all HeLa cells treated with the AgNPs-F (1.53-fold), AgNPs-L (1.52-fold) and 5FU (4.30-fold). CXCL1 was under expressed in HeLa cells treated with AgNPs-F (0.69-fold) and AgNPs-L (0.58-fold) and over expressed in cells treated with 5FU (4.95-fold), but the difference was not statistically significant. CXCR2 was significantly over expressed in HeLa cells treated with 5FU (8.66-fold) and AgNPs-F (1.12-fold) but under expressed in cells treated with AgNPs-L (0.76-fold). CONCLUSIONS: Here we show that biosynthesized AgNPs especially AgNPs-F can be used in the development of novel and better anticancer drugs. The mechanism of action of the AgNPs involves activation of the intrinsic apoptosis pathway through upregulation of CASP9 and concerted down regulation of the CXCL1/ CXCR2 gene axis.


Subject(s)
Annona/chemistry , Antineoplastic Agents/pharmacology , Caspase 9/genetics , Chemokine CXCL1/genetics , Metal Nanoparticles , Receptors, Interleukin-8B/genetics , Silver/pharmacology , Adenocarcinoma/pathology , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Green Chemistry Technology , Humans , Male , Plant Extracts/chemistry , Plant Extracts/pharmacology , Prostatic Neoplasms/pathology , Uterine Cervical Neoplasms/pathology
5.
PLoS Pathog ; 15(6): e1007877, 2019 06.
Article in English | MEDLINE | ID: mdl-31226163

ABSTRACT

Rapid bone destruction often leads to permanent joint dysfunction in patients with septic arthritis, which is mainly caused by Staphylococcus aureus (S. aureus). Staphylococcal cell wall components are known to induce joint inflammation and bone destruction. Here, we show that a single intra-articular injection of S. aureus lipoproteins (Lpps) into mouse knee joints induced chronic destructive macroscopic arthritis through TLR2. Arthritis was characterized by rapid infiltration of neutrophils and monocytes. The arthritogenic effect was mediated mainly by macrophages/monocytes and partially via TNF-α but not by neutrophils. Surprisingly, a S. aureus mutant lacking Lpp diacylglyceryl transferase (lgt) caused more severe joint inflammation, which coincided with higher bacterial loads of the lgt mutant in local joints than those of its parental strain. Coinjection of pathogenic S. aureus LS-1 with staphylococcal Lpps into mouse knee joints caused improved bacterial elimination and diminished bone erosion. The protective effect of the Lpps was mediated by their lipid moiety and was fully dependent on TLR2 and neutrophils. The blocking of CXCR2 on neutrophils resulted in total abrogation of the protective effect of the Lpps. Our data demonstrate that S. aureus Lpps elicit innate immune responses, resulting in a double-edged effect. On the one hand, staphylococcal Lpps boost septic arthritis. On the other hand, Lpps act as adjuvants and activate innate immunity, which could be useful for combating infections with multiple drug-resistant strains.


Subject(s)
Arthritis/immunology , Bacterial Proteins/immunology , Lipoproteins/immunology , Neutrophils/immunology , Staphylococcus aureus/immunology , Animals , Arthritis/genetics , Arthritis/microbiology , Arthritis/pathology , Bacterial Proteins/genetics , Female , Lipoproteins/genetics , Mice , Neutrophils/pathology , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/immunology , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
6.
Mol Med Rep ; 17(1): 179-185, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115434

ABSTRACT

Neutrophils, immune cells crucial for protecting against invading pathogens, are important in sepsis. Neutrophil migration is regulated by chemokine receptors and their cognate ligands. Our previous study investigated the effect of n­butanol extract from Folium isatidis on lipopolysaccharide (LPS)­induced septic shock. The present study stimulated neutrophils with LPS to explore the influence of LPS on cell. Neutrophils were then pretreated with n­butanol extract from Folium isatidis followed by LPS to examine the effect of this extract on neutrophil chemotaxis. The results showed that LPS decreased the expression levels of CXC­chemokine receptor (CXCR)1, CXCR2 and L­selectin (CD62L), and increased the expression of interleukin­8 (IL­8) by neutrophils. The addition of n­butanol extract from Folium isatidis inhibited this LPS­induced downregulation of CXCR1, CXCR2 and CD62L, and decreased the expression of IL­8 on neutrophils. In addition, n­butanol extract promoted myeloperoxidase activity in neutrophils. Taken together, LPS downregulated the expression of chemokine receptors, leading to the failure of neutrophils to migrate to sites of infection. The addition of n­butanol extract, which promoted the ability of neutrophils to migrate, is a natural product and potential therapeutic agent with which to target neutrophil chemotaxis during LPS stimulation.


Subject(s)
1-Butanol/pharmacology , Gene Expression Regulation/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Plant Extracts/pharmacology , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8B/genetics , 1-Butanol/chemistry , Adult , Biomarkers , Chemotaxis/drug effects , Chemotaxis/immunology , Female , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Male , Neutrophils/immunology , Peroxidase/genetics , Peroxidase/metabolism , Plant Extracts/chemistry , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Young Adult
7.
BMC Infect Dis ; 17(1): 94, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28114888

ABSTRACT

BACKGROUND: Fasciolosis remains a significant food-borne trematode disease causing high morbidity around the world and affecting grazing animals and humans. A deeper understanding concerning the molecular mechanisms by which Fasciola hepatica infection occurs, as well as the molecular basis involved in acquiring protection is extremely important when designing and selecting new vaccine candidates. The present study provides a first report of microarray-based technology for describing changes in the splenic gene expression profile for mice immunised with a highly effective, protection-inducing, multi-epitope, subunit-based, chemically-synthesised vaccine candidate against F. hepatica. METHODS: The mice were immunised with synthetic peptides containing B- and T-cell epitopes, which are derived from F. hepatica cathepsin B and amoebapore proteins, as novel vaccine candidates against F. hepatica formulated in an adjuvant adaptation vaccination system; they were experimentally challenged with F. hepatica metacercariae. Spleen RNA from mice immunised with the highest protection-inducing synthetic peptides was isolated, amplified and labelled using Affymetrix standardised protocols. Data was then background corrected, normalised and the expression signal was calculated. The Ingenuity Pathway Analysis tool was then used for analysing differentially expressed gene identifiers for annotating bio-functions and constructing and visualising molecular interaction networks. RESULTS: Mice immunised with a combination of three peptides containing T-cell epitopes induced high protection against experimental challenge according to survival rates and hepatic damage scores. It also induced differential expression of 820 genes, 168 genes being up-regulated and 652 genes being down-regulated, p value <0.05, fold change ranging from -2.944 to 7.632. A functional study of these genes revealed changes in the pathways related to nitric oxide and reactive oxygen species production, Interleukin-12 signalling and production in macrophages and Interleukin-8 signalling with up-regulation of S100 calcium-binding protein A8, Matrix metallopeptidase 9 and CXC chemokine receptor 2 genes. CONCLUSION: The data obtained in the present study provided us with a more comprehensive overview concerning the possible molecular pathways implied in inducing protection against F. hepatica in a murine model, which could be useful for evaluating future vaccine candidates.


Subject(s)
Fasciola hepatica/immunology , Fascioliasis/prevention & control , Gene Expression/drug effects , Protozoan Vaccines/pharmacology , Spleen/drug effects , Animals , Antibodies, Helminth/immunology , Calgranulin A/drug effects , Calgranulin A/genetics , Epitopes/immunology , Female , Gene Expression Profiling , Interleukin-12/genetics , Interleukin-8/drug effects , Interleukin-8/genetics , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/genetics , Mice , Peptides/immunology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Interleukin-8B/drug effects , Receptors, Interleukin-8B/genetics , Spleen/metabolism , Up-Regulation , Vaccination
8.
Int Immunopharmacol ; 39: 377-382, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27543854

ABSTRACT

Astragaloside IV (AS-IV), an active saponin purified from Astragali Radix, has been identified with broad biological and pharmacological activities. In the present study, we continue to explore the potential effect of AS-IV on antibacterial response using an acute E. coli peritoneal infection model. Our findings implied that administration of AS-IV decreases mortality in mice challenged by lethal E. coli infection. The protection of AS-IV was related to promotion of neutrophil extravasation into the peritoneum and bacterial clearance. Toll-like receptor (TLR) activation in neutrophils has been reported to reduce CXCR2 expression and subsequent neutrophil migration. Our data indicated that AS-IV prevented the reduction of CXCR2 expression and neutrophil migration induced by LPS, the activator for TLR4. Moreover, we found that AS-IV blocks LPS-induced suppression of CXCR2 on neutrophils by inhibiting the expression of G protein-coupled receptor kinase-2 (GRK2), an agonist that regulates desensitization and internalization of chemokine receptors. Taken together, these data propose that AS-IV, through modulating GRK2-CXCR2 signal in neutrophils, offers an essential efficacy on host antibacterial immunity.


Subject(s)
Anti-Infective Agents/therapeutic use , Astragalus propinquus/immunology , Cell Movement , Escherichia coli Infections/drug therapy , Escherichia coli , G-Protein-Coupled Receptor Kinase 2/metabolism , Neutrophils/drug effects , Peritonitis/drug therapy , Saponins/therapeutic use , Triterpenes/therapeutic use , Animals , Cell Movement/drug effects , G-Protein-Coupled Receptor Kinase 2/genetics , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred BALB C , Neutrophils/immunology , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism
9.
Cell Physiol Biochem ; 36(5): 2003-11, 2015.
Article in English | MEDLINE | ID: mdl-26202360

ABSTRACT

BACKGROUND/AIMS: Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are lethal diseases in humans, and the current treatments have limited therapeutic effects. Cordyceps militaris (CM) is a caterpillar-grown traditional medicinal mushroom, and has been used as a natural invigorant for longevity, endurance, and vitality in China. Recently, purified extracts from CM have been shown to have beneficial effects on various diseases including cancer. Nevertheless, a role of CM in ALI has not been examined previously. METHODS: Here, we used a bleomycin-induced ALI model to study the effects of CM on the severity of ALI in mice. The levels of CXCR2, a receptor for Interleukin 8 (IL-8) in pulmonary microvascular endothelial cells, were examined in different experimental groups. The levels of microRNA (miR)-1321 and miR-3188 were also examined in lung samples and in CM. Adeno-associated viruses carrying miR-1321 and miR-3188 were injected into bleomycin-treated mice for evaluation their effects on the severity of ALI. RESULTS: CM treatment significantly alleviated the severity of bleomycin-induced ALI in mice. The increases in lung CXCR2 by bleomycin were significantly reduced by CM at protein level, but not at mRNA level. CM contained high levels of 2 miRNAs (miR-1321 and miR-3188) that target 3'-UTR of CXCR2 mRNA to inhibit its expression. Overexpression of miR-1321 and miR-3188 in mouse lung through AAV-mediated gene therapy mimicked the effects of CM. CONCLUSION: CM may alleviate severity of murine ALI through miRNAs-mediated CXCR2 inhibition.


Subject(s)
Acute Lung Injury/prevention & control , Cordyceps/chemistry , MicroRNAs/genetics , Plant Extracts/pharmacology , Receptors, Interleukin-8B/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Bleomycin/toxicity , Female , Mice , Mice, Inbred BALB C , Receptors, Interleukin-8B/genetics
10.
J Huazhong Univ Sci Technolog Med Sci ; 34(1): 91-98, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24496685

ABSTRACT

To observe the effect of acupuncture on CXCL8 receptors (CXCR1 and CXCR2) in rat endometrium experiencing embryo implantation failure, 72 pregnant rats were randomly divided into four groups: normal group (N), embryo implantation failure group (M), acupuncture treatment group (A), and progestin treatment group (W). Then the rats in each group were equally randomized into a day-6 (D6) group, a day-8 (D8) group, and a day-10 (D10) group. The rats in group M, group A, and group W were treated with mifepristone-sesame oil solution on day 1, while the rats in group N were injected with the same amount of sesame oil. Meanwhile, "Housanli" and "Sanyinjiao" were selected for acupuncture. From day 1 to the time of death, the rats in group A were fastened up and then acupuncture was administered while the rats in group N and group M were only fixed, and the rats in group W were given progestin. The number of implanted embryos was calculated. The expression of CXCR1 and CXCR2 in rat endometrium was detected by immunohistochemistry, Western blotting and real-time PCR. Compared to group N, the average number of implanted embryos, the protein and mRNA expression of CXCR1 (D6, D8 and D10), and the protein and mRNA expression of CXCR2 (D8 and D10) in rat endometrium were significantly decreased in group M. Compared to group M, there was significant elevation in the average number of implanted embryos, the protein expression (D6, D8 and D10) and mRNA expression (D8) of CXCR1 in rat endometrium of group A, and the protein expression (D8 and D10) and mRNA expression (D8) of CXCR2 in rat endometrium of group W. These findings indicated that acupuncture can increase the number of implanted embryos in rats of embryo implantation failure, which may be relevant with up-regulation the expression of CXCR1 and CXCR2 at maternal-fetal interface of rats with embryo implantation failure.


Subject(s)
Acupuncture Therapy/methods , Embryo Implantation/genetics , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8B/genetics , Animals , Blotting, Western , Embryo Implantation/drug effects , Endometrium/drug effects , Endometrium/metabolism , Female , Gene Expression Regulation, Developmental , Hormone Antagonists/pharmacology , Immunohistochemistry , Mifepristone/pharmacology , Pregnancy , Progestins/pharmacology , Random Allocation , Rats , Rats, Wistar , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Treatment Outcome , Up-Regulation/drug effects
11.
J Clin Invest ; 122(10): 3647-51, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22996693

ABSTRACT

Although long considered a promising treatment option for type 1 diabetes, pancreatic islet cell transformation has been hindered by immune system rejection of engrafted tissue. The identification of pathways that regulate post-transplant detrimental inflammatory events would improve management and outcome of transplanted patients. Here, we found that CXCR1/2 chemokine receptors and their ligands are crucial negative determinants for islet survival after transplantation. Pancreatic islets released abundant CXCR1/2 ligands (CXCL1 and CXCL8). Accordingly, intrahepatic CXCL1 and circulating CXCL1 and CXCL8 were strongly induced shortly after islet infusion. Genetic and pharmacological blockade of the CXCL1-CXCR1/2 axis in mice improved intrahepatic islet engraftment and reduced intrahepatic recruitment of polymorphonuclear leukocytes and NKT cells after islet infusion. In humans, the CXCR1/2 allosteric inhibitor reparixin improved outcome in a phase 2 randomized, open-label pilot study with a single infusion of allogeneic islets. These findings indicate that the CXCR1/2-mediated pathway is a regulator of islet damage and should be a target for intervention to improve the efficacy of transplantation.


Subject(s)
Chemokine CXCL1/physiology , Diabetes Mellitus, Type 1/surgery , Interleukin-8/physiology , Islets of Langerhans Transplantation/immunology , Islets of Langerhans/metabolism , Receptors, Interleukin-8A/physiology , Receptors, Interleukin-8B/physiology , Sulfonamides/therapeutic use , Adult , Animals , Blood Glucose/analysis , Cell Survival/drug effects , Chemokine CXCL1/biosynthesis , Chemokine CXCL1/genetics , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 1/immunology , Drug Evaluation, Preclinical , Female , Graft Rejection/prevention & control , Graft Survival/drug effects , Humans , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Middle Aged , Natural Killer T-Cells/immunology , Neutrophils/immunology , Pilot Projects , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/deficiency , Receptors, Interleukin-8B/genetics , Sulfonamides/pharmacology , Treatment Outcome
12.
Shock ; 30(5): 563-70, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18317407

ABSTRACT

Ischemic gut contributes to the development of sepsis and organ failure in critically ill patients. Toll-like receptors (TLRs) have been reported to mediate the pathophysiology of organ damage following ischemia/reperfusion (I/R) injury. We hypothesize that LPS, a ligand for TLR4, decreases mesenteric I/R injury-induced gut damage through tumor necrosis factor alpha (TNF-alpha) signaling. First, wild-type (WT) mice were fed with oral antibiotics for 4 weeks to deplete the intestinal commensal microflora. At week 3, drinking water was supplemented with LPS (10 microg/microL) to trigger TLRs. The intestinal mucosa was harvested for TLR4 protein, caspase 3 activity, and terminal deoxynucleotide transferase labeling assay. Second, WT and Tnfrsf1a mice received 30-min ischemia and 30-min reperfusion (30I-30R) or 30I-180R of the intestine; intestinal permeability and lipid peroxidation of the intestine were examined. Third, WT and Tnfrsf1a mice were fed with oral antibiotics with or without LPS and received 30I-180R of the intestine. The intestinal mucosa was harvested for lipid peroxidation; glutathione (GSH) level; nuclear factor kappaB (NF-kappaB) and AP-1 DNA-binding activity; Bcl-w, TNF-alpha, and CXCR2 mRNA expression; and HSP70 protein assay. Commensal depletion increased caspase 3 activity as well as villi apoptosis and decreased TLR4 expression of the intestinal mucosa. LPS increased TLR4 expression and decreased villi apoptosis. Commensal depletion augmented 30I-180R-induced intestine permeability as well as lipid peroxidation and decreased GSH level in WT mice but not in Tnfrsf1a mice. LPS decreased 30I-180R-induced intestinal permeability as well as lipid peroxidation and increased GSH level of the intestinal mucosa in WT mice but not in Tnfrsf1a mice. Commensal depletion with 30I-180R increased NF-kappaB and AP-1 DNA-binding activity, HSP70 protein expression, and decreased Bcl-w and TNF-alpha mRNA expression of the intestinal mucosa in WT mice but not in Tnfrsf1a mice. Collectively, commensal microflora induces TLR4 expression and decreases apoptosis of the intestinal mucosa. Commensal depletion enhances I/R-induced gut damage. LPS prevents I/R-induced intestinal permeability, lipid peroxidation, and decrease in GSH level. Given that the preventive effect of LPS on I/R-induced gut damage and NF-kappaB activity of the intestine is abolished in Tnfrsf1a mice, we conclude that TLR ligand decreases mesenteric I/R injury-induced gut damage through TNF-alpha signaling.


Subject(s)
Reperfusion Injury/physiopathology , Signal Transduction/physiology , Toll-Like Receptor 4/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Blotting, Western , Electrophoretic Mobility Shift Assay , Gene Expression/drug effects , Glutathione/metabolism , In Situ Nick-End Labeling , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipid Peroxidation/drug effects , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , NF-kappa B/metabolism , Polymerase Chain Reaction , Protein Binding/drug effects , Proteins/genetics , Receptors, Interleukin-8B/genetics , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/genetics
13.
Circulation ; 110(10): 1276-83, 2004 Sep 07.
Article in English | MEDLINE | ID: mdl-15337697

ABSTRACT

BACKGROUND: Cardiac myosin-induced myocarditis is an experimental autoimmune myocarditis (EAM) model used to investigate autoimmunological mechanisms in inflammatory heart diseases and resembles fulminant myocarditis in humans. We investigated the therapeutic role of thioredoxin-1 (TRX-1), a redox-regulatory protein with antioxidant and antiinflammatory effects, in murine EAM. METHODS AND RESULTS: EAM was generated in 5-week-old male BALB/c mice by immunization with porcine cardiac myosin at days 0 and 7. Recombinant human TRX-1 (rhTRX-1), C32S/C35S mutant rhTRX-1, or saline was administered intraperitoneally every second day from day 0 to 20. In addition, rabbit anti-mouse TRX-1 serum or normal rabbit serum was administered intraperitoneally on days -1, 2, and 6. Animals were euthanized on day 21. Histological analysis of the heart showed that TRX-1 significantly reduced the severity of EAM, whereas mutant TRX-1 failed to have such an effect, and anti-TRX-1 antibody enhanced the disease markedly. Immunohistochemical analysis showed that TRX-1 significantly suppressed cardiac macrophage inflammatory protein (MIP)-1alpha, MIP-2, and 8-hydroxydeoxyguanosine expression and macrophage infiltration into the heart in EAM. Although serum levels of MIP-1alpha were not suppressed by TRX-1 until day 21, both an in vitro chemotaxis chamber assay and an in vivo air pouch model showed that TRX-1 significantly suppressed MIP-1alpha- or MIP-2-induced leukocyte chemotaxis. However, real-time reverse transcription-polymerase chain reaction showed that TRX-1 failed to decrease chemokine receptor expression increased in the bone marrow cells of EAM mice. CONCLUSIONS: TRX-1 attenuates EAM by suppressing chemokine expressions and leukocyte chemotaxis in mice.


Subject(s)
Autoimmune Diseases/drug therapy , Chemotaxis, Leukocyte/drug effects , Myocarditis/drug therapy , Thioredoxins/therapeutic use , Amino Acid Substitution , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Binding Sites , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Chemokine CCL3 , Chemokine CCL4 , Chemokine CXCL2 , Chemokines/biosynthesis , Chemokines/blood , Chemokines/genetics , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Humans , Injections, Intraperitoneal , Lymphocytes/drug effects , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/blood , Macrophage Inflammatory Proteins/genetics , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Myocarditis/immunology , Myocarditis/metabolism , Myocarditis/pathology , Myosins/immunology , Neutrophils/drug effects , Receptors, CCR1 , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/genetics , Receptors, Interleukin-8B/biosynthesis , Receptors, Interleukin-8B/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thioredoxins/administration & dosage , Thioredoxins/genetics , Thioredoxins/pharmacology
14.
Oncogene ; 23(41): 6924-32, 2004 Sep 09.
Article in English | MEDLINE | ID: mdl-15286717

ABSTRACT

Interleukin (IL)-8 produced from glioblastoma is suggested to contribute to its own proliferation and progression. Since various external stimuli have been shown to increase intracellular Ca(2+) in glioma cells, we investigated Ca(2+) mobilization-dependent IL-8 expression and effect of cyclosporin A (CsA), an inhibitor of calcineurin (Cn), on the expression and invasive potential of human glioblastoma U251MG cells. Combined treatment with Ca(2+)-ionophore and phorbol-myristate-acetate (A23187/PMA) increased IL-8 mRNA and protein levels. This increase was suppressed by CsA and by another Cn inhibitor FK506. Luciferase reporter gene assay and electrophoretic mobility shift assay revealed that activation of p65-containing nuclear factor-kappaB was essential for A23187/PMA-dependent activation of IL-8 promoter. CsA suppressed the promoter activity by attenuating IkappaB-alpha degradation. U251MG cells expressed IL-8 receptors CXCR-1 and -2, and Matrigel invasion assay revealed that CsA attenuated A23187/PMA-dependent stimulation of invasive potential, probably by inhibiting IL-8 production. In addition, IL-8-dependent proliferation was also suppressed by CsA. Taken together, these results demonstrate the novel inhibitory effects of CsA on glioblastoma cell functions, suggesting CsA as a potential therapeutic adjuvant for glioma treatment.


Subject(s)
Calcium/metabolism , Cyclosporine/pharmacology , Glioblastoma/pathology , Interleukin-8/genetics , Calcimycin/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Glioblastoma/metabolism , Humans , I-kappa B Kinase , NF-kappa B/metabolism , Neoplasm Invasiveness , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/analysis , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8B/genetics , Tetradecanoylphorbol Acetate/pharmacology
15.
J Immunol ; 169(11): 6435-44, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12444152

ABSTRACT

Much evidence implicates IL-8 as a major mediator of inflammation and joint destruction in rheumatoid arthritis. The effects of IL-8 and its related ligands are mediated via two receptors, CXCR1 and CXCR2. In the present study, we demonstrate that a potent and selective nonpeptide antagonist of human CXCR2 potently inhibits (125)I-labeled human IL-8 binding to, and human IL-8-induced calcium mobilization mediated by, rabbit CXCR2 (IC(50) = 40.5 and 7.7 nM, respectively), but not rabbit CXCR1 (IC(50) = >1000 and 2200 nM, respectively). These data suggest that the rabbit is an appropriate species in which to examine the anti-inflammatory effects of a human CXCR2-selective antagonist. In two acute models of arthritis in the rabbit induced by knee joint injection of human IL-8 or LPS, and a chronic Ag (OVA)-induced arthritis model, administration of the antagonist at 25 mg/kg by mouth twice a day significantly reduced synovial fluid neutrophils, monocytes, and lymphocytes. In addition, in the more robust LPS- and OVA-induced arthritis models, which were characterized by increased levels of proinflammatory mediators in the synovial fluid, TNF-alpha, IL-8, PGE(2), leukotriene B(4), and leukotriene C(4) levels were significantly reduced, as was erythrocyte sedimentation rate, possibly as a result of the observed decreases in serum TNF-alpha and IL-8 levels. In vitro, the antagonist potently inhibited human IL-8-induced chemotaxis of rabbit neutrophils (IC(50) = 0.75 nM), suggesting that inhibition of leukocyte migration into the knee joint is a likely mechanism by which the CXCR2 antagonist modulates disease.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/immunology , Arthritis, Experimental/prevention & control , Receptors, Interleukin-8B/antagonists & inhibitors , Urea/pharmacology , Acute Disease , Animals , Arthritis, Experimental/etiology , Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/immunology , Chemotaxis, Leukocyte/drug effects , Chronic Disease , Female , Humans , In Vitro Techniques , Interleukin-8/administration & dosage , Interleukin-8/immunology , Interleukin-8/metabolism , Lipopolysaccharides/toxicity , Neutrophils/drug effects , Neutrophils/immunology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Rabbits , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Urea/analogs & derivatives
16.
J Neuroimmunol ; 129(1-2): 66-73, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12161022

ABSTRACT

Experiments were conducted in both HEK cells and cerebellar neurons to investigate whether CXC chemokine receptor 2 (CXCR2) is functionally coupled to GluR1. The co-expression of CXCR2 with GluR1 in HEK cells increased (i) the GluR1 "apparent" affinity for the transmitter; (ii) the GluR1 channel open probability; and (iii) GluR1 binding site cooperativity upon CXCR2 stimulation with CXC chemokine ligand 2 (CXCL2). The affinity of C-terminal-deleted GluR1 for glutamate (Glu) remained stable instead. Furthermore, CXCL2 increased the binding site cooperativity of AMPA receptors in rat cerebellar granule cells; and the amplitude of spontaneous excitatory postsynaptic current (sEPSCs) in Purkinje neurons (PNs). Our findings indicate that the coupling of CXCR2 with GluR1 may modulate glutamatergic synaptic transmission.


Subject(s)
Central Nervous System/metabolism , Chemokines, CXC/metabolism , Glutamic Acid/metabolism , Receptors, AMPA/metabolism , Receptors, Interleukin-8B/metabolism , Synapses/metabolism , Synaptic Transmission/immunology , Animals , Binding Sites/drug effects , Binding Sites/immunology , Cells, Cultured , Central Nervous System/immunology , Cerebellar Cortex/drug effects , Cerebellar Cortex/immunology , Cerebellar Cortex/metabolism , Chemokines, CXC/immunology , Chemokines, CXC/pharmacology , DNA, Complementary/genetics , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Glutamic Acid/pharmacology , Humans , Ion Channels/genetics , Ion Channels/immunology , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, AMPA/genetics , Receptors, AMPA/immunology , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/immunology , Synapses/immunology
18.
J Biomol Screen ; 7(5): 451-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-14599361

ABSTRACT

A novel cell-based functional assay to directly monitor G protein-coupled receptor (GPCR) activation in a high-throughput format, based on a common GPCR regulation mechanism, the interaction between beta-arrestin and ligand-activated GPCR, is described. A protein-protein interaction technology, the InteraX trade mark system, uses a pair of inactive beta-galactosidase (beta-gal) deletion mutants as fusion partners to the protein targets of interest. To monitor GPCR activation, stable cell lines expressing both GPCR- and beta-arrestin-beta-gal fusion proteins are generated. Following ligand stimulation, beta-arrestin binds to the activated GPCR, and this interaction drives functional complementation of the beta-gal mutant fragments. GPCR activation is measured directly by quantitating restored beta-gal activity. The authors have validated this assay system with two functionally divergent GPCRs: the beta2-adrenergic amine receptor and the CXCR2 chemokine-binding receptor. Both receptors are activated or blocked with known agonists and antagonists in a dose-dependent manner. The beta2-adrenergic receptor cell line was screened with the LOPAC trade mark compound library to identify both agonists and antagonists, validating this system for high-throughput screening performance in a 96-well microplate format. Hit specificity was confirmed by quantitating the level of cAMP. This assay system has also been performed in a high-density (384-well) microplate format. This system provides a specific, sensitive, and robust methodology for studying and screening GPCR-mediated signaling pathways.


Subject(s)
Biological Assay/methods , Drug Evaluation, Preclinical/methods , Protein Interaction Mapping/methods , Receptors, G-Protein-Coupled/metabolism , beta-Galactosidase/metabolism , Arrestins/genetics , Arrestins/metabolism , Biological Assay/instrumentation , Cells, Cultured , Combinatorial Chemistry Techniques/methods , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/instrumentation , Genes, erbB-1 , Humans , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Interaction Mapping/instrumentation , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Interleukin-8B/drug effects , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , beta-Galactosidase/genetics
19.
Cytokine ; 12(9): 1368-73, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10975996

ABSTRACT

Rat cytokine-induced neutrophil chemoattractant-1 (CINC-1), CINC-2 and CINC-3/macrophage inflammatory protein-2 (MIP-2), members of the CXC chemokine family, are potent chemotactic factors for neutrophils. In order to identify the receptor for CINCs, rat CXC chemokine receptor 2 (CXCR2) was cloned and expressed in HEK293 cells. CINC-1, CINC-2 and CINC-3 induced calcium mobilizations dose-dependently in CXCR2-transfected cells, whereas formyl-methionyl-leucyl-phenylalanine (FMLP) did not. CINC-3 induced enhancement of cytoplasmic calcium concentration more potently than CINC-1 and CINC-2, and desensitized calcium transients induced by CINC-1 and CINC-2, which were essentially identical to those observed in rat neutrophils. In addition, anti-CXCR2 serum inhibited neutrophil chemotactic activities of three types of CINCs almost completely. The mutant CINC-3, whose amino-terminal amino acid sequence (SELR) was replaced to AAR, lost chemotactic activity of its own but inhibited that of CINC-1 and CINC-2 potently, and that of CINC-3 weakly. The results indicate that rat CXCR2 on neutrophils is the unique receptor for all three types of CINCs, and CINC-1/-2 and CINC-3 exert different biological activities through the common receptor.


Subject(s)
Chemokines, CXC , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Growth Substances/genetics , Growth Substances/metabolism , Intercellular Signaling Peptides and Proteins , Animals , Blotting, Northern , Calcium/metabolism , Cell Line , Chemokine CXCL1 , Chemokine CXCL2 , Chemotactic Factors/pharmacology , Chemotactic Factors/physiology , Chemotaxis/drug effects , Cloning, Molecular , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Growth Substances/pharmacology , Growth Substances/physiology , Humans , Liver/metabolism , Mutagenesis , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/metabolism , Polymerase Chain Reaction , Rats , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Spectrophotometry , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL