Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Publication year range
1.
Anticancer Drugs ; 33(1): e166-e177, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34387598

ABSTRACT

Circular RNAs (circRNAs) are reported to be related to cancer chemoresistance. However, the role of circ_0085495 in adriamycin (ADM) and its action mechanism has not been elucidated in breast cancer. Cell counting kit-8 was employed to detect cell viability. Quantitative real-time-PCR and western blot were performed to examine the gene and protein expression level. Flow cytometry and colony formation assay were conducted to measure cell apoptosis and proliferation. Cell migration and invasion were evaluated via transwell assay. The target association between molecules was confirmed by dual-luciferase reporter, RNA immunoprecipitation and RNA pull-down assays. Tumor xenograft assay was implemented to explore the role of circ_0085495 in vivo. Circ_0085495 and Integrin ß1 were upregulated, while miR-873-5p was downregulated in ADM-resistant cells. Circ_0085495 was a stable circRNA, mainly located in the cytoplasm. Depletion of circ_0085495 repressed ADM resistance, proliferation and metastasis of ADM-resistant breast cancer cells, which was weakened by miR-873-5p inhibition or integrin ß1 overexpression. Circ_0085495 sponged miR-873-5p to positively regulate integrin ß1 expression. Integrin ß1 knockdown also inhibited ADM resistance. Furthermore, circ_0085495 knockdown inhibited tumor growth in vivo. Circ_0085495 knockdown reduced ADM resistance in ADM-resistant cells through modulating miR-873-5p/integrin ß1 axis, indicating circ_0085495 as a promising target for overcoming ADM resistance in breast cancer patients.


Subject(s)
Breast Neoplasms/pathology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Integrin beta1/drug effects , MicroRNAs/drug effects , RNA, Circular/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Female , Humans , Integrin beta1/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Up-Regulation , Xenograft Model Antitumor Assays
2.
Invest New Drugs ; 39(2): 377-385, 2021 04.
Article in English | MEDLINE | ID: mdl-32989643

ABSTRACT

Purpose Our study aimed to investigate the antitumor effects of rAj-Tspin on BEL-7402 hepatocellular carcinoma cells and to explore the underlying mechanism. Method For the in vivo experiment, BEL-7402 cells were inoculated subcutaneously into the axilla of nude mice to generate a BEL-7402 cell-bearing model, and model mice were then treated with different doses of rAj-Tspin. A CCK-8 assay was used to evaluate the in vitro viability of BEL-7402 and LO2 cells after treatment with different concentrations of rAj-Tspin. The effects of rAj-Tspin on BEL-7402 cell apoptosis, migration and invasion were evaluated by AO/EB and Hoechst fluorescent staining and by scratch and Transwell assays, and the tumor-suppressive mechanism of rAj-Tspin was explored by Western blotting. Results rAj-Tspin suppressed the proliferation of BEL-7402 cells with an IC50 of 0.89 µM. The results of both microscopic analysis and Western blotting suggested that rAj-Tspin induced the apoptosis of BEL-7402 cells through a mitochondria-dependent pathway. Furthermore, rAj-Tspin disrupted EMT; this disruption ultimately caused BEL-7402 cells to lose their shape and decreased their migration and invasion. Moreover, rAj-Tspin might inhibit the proliferation and metastasis of BEL-7402 cells through the ITGB1-FAK-AKT pathway. Conclusion rAj-Tspin exerts an antitumor effect through the ITGB1-FAK-Akt signaling pathway and exhibits low toxicity at an effective dose.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Stichopus , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Focal Adhesion Kinase 1/drug effects , Integrin beta1/drug effects , Liver Neoplasms/pathology , Male , Marine Toxins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/prevention & control , Peptides/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
3.
Nature ; 519(7544): 425-30, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25799996

ABSTRACT

Cell migration is a stepwise process that coordinates multiple molecular machineries. Using in vitro angiogenesis screens with short interfering RNA and chemical inhibitors, we define here a MAP4K4-moesin-talin-ß1-integrin molecular pathway that promotes efficient plasma membrane retraction during endothelial cell migration. Loss of MAP4K4 decreased membrane dynamics, slowed endothelial cell migration, and impaired angiogenesis in vitro and in vivo. In migrating endothelial cells, MAP4K4 phosphorylates moesin in retracting membranes at sites of focal adhesion disassembly. Epistasis analyses indicated that moesin functions downstream of MAP4K4 to inactivate integrin by competing with talin for binding to ß1-integrin intracellular domain. Consequently, loss of moesin (encoded by the MSN gene) or MAP4K4 reduced adhesion disassembly rate in endothelial cells. Additionally, α5ß1-integrin blockade reversed the membrane retraction defects associated with loss of Map4k4 in vitro and in vivo. Our study uncovers a novel aspect of endothelial cell migration. Finally, loss of MAP4K4 function suppressed pathological angiogenesis in disease models, identifying MAP4K4 as a potential therapeutic target.


Subject(s)
Cell Movement , Endothelial Cells/cytology , Endothelial Cells/metabolism , Integrins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Motifs , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Shape/drug effects , Endothelial Cells/drug effects , Epistasis, Genetic , Focal Adhesions/metabolism , Humans , Integrin alpha1/drug effects , Integrin alpha1/metabolism , Integrin beta1/chemistry , Integrin beta1/drug effects , Integrin beta1/metabolism , Integrins/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neovascularization, Pathologic , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Talin/chemistry , Talin/metabolism
4.
Am J Obstet Gynecol ; 223(5): 733.e1-733.e14, 2020 11.
Article in English | MEDLINE | ID: mdl-32417359

ABSTRACT

BACKGROUND: Uterine leiomyomas, the most common tumors of the female reproductive system, are characterized by excessive deposition of disordered stiff extracellular matrix and fundamental alteration in the mechanical signaling pathways. Specifically, these alterations affect the normal dynamic state of responsiveness to mechanical cues in the extracellular environment. These mechanical cues are converted through integrins, cell membrane receptors, to biochemical signals including cytoskeletal signaling pathways to maintain mechanical homeostasis. Leiomyoma cells overexpress ß1 integrin and other downstream mechanical signaling proteins. We previously reported that simvastatin, an antihyperlipidemic drug, has antileiomyoma effects through cellular, animal model, and epidemiologic studies. OBJECTIVE: This study aimed to examine the hypothesis that simvastatin might influence altered mechanotransduction in leiomyoma cells. STUDY DESIGN: This is a laboratory-based experimental study. Primary leiomyoma cells were isolated from 5 patients who underwent hysterectomy at the Department of Gynecology and Obstetrics of the Johns Hopkins University Hospital. Primary and immortalized human uterine leiomyoma cells were treated with simvastatin at increasing concentrations (0.001, 0.01, 0.1, and 1 µM, or control) for 48 hours. Protein and mRNA levels of ß1 integrin and extracellular matrix components involved in mechanical signaling were quantified by quantitative real-time polymerase chain reaction, western blotting, and immunofluorescence. In addition, we examined the effect of simvastatin on the activity of Ras homolog family member A using pull-down assay and gel contraction. RESULTS: We found that simvastatin significantly reduced the protein expression of ß1 integrin by 44% and type I collagen by 60% compared with untreated leiomyoma cells. Simvastatin-treated cells reduced phosphorylation of focal adhesion kinase down to 26%-60% of control, whereas it increased total focal adhesion kinase protein expression. Using a Ras homolog family member A pull-down activation assay, we observed reduced levels of active Ras homolog family member A in simvastatin-treated cells by 45%-85% compared with control. Consistent with impaired Ras homolog family member A activation, simvastatin treatment reduced tumor gel contraction where gel area was 122%-153% larger than control. Furthermore, simvastatin treatment led to reduced levels of mechanical signaling proteins involved in ß1 integrin downstream signaling, such as A-kinase anchor protein 13, Rho-associated protein kinase 1, myosin light-chain kinase, and cyclin D1. CONCLUSION: The results of this study suggest a possible therapeutic role of simvastatin in restoring the altered state of mechanotransduction signaling in leiomyoma. Collectively, these findings are aligned with previous epidemiologic studies and other reports and support the need for clinical trials.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Leiomyoma/genetics , Mechanotransduction, Cellular/drug effects , Simvastatin/pharmacology , Uterine Neoplasms/genetics , A Kinase Anchor Proteins/drug effects , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Collagen Type I/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Cyclin D1/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/drug effects , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Integrin beta1/drug effects , Integrin beta1/genetics , Integrin beta1/metabolism , Leiomyoma/metabolism , Mechanotransduction, Cellular/genetics , Minor Histocompatibility Antigens/drug effects , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Myosin-Light-Chain Kinase/drug effects , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Phosphorylation , Primary Cell Culture , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Uterine Neoplasms/metabolism , rho-Associated Kinases/drug effects , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/drug effects , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
5.
Wound Repair Regen ; 28(1): 16-25, 2020 01.
Article in English | MEDLINE | ID: mdl-31270882

ABSTRACT

Reepithelialization is an important step of wound healing, which is mainly completed by proliferation and migration of epidermal cells. Akermanite is a Ca-, Mg-, and Si-containing bioceramic. This study evaluated the effects of Akermanite on wound healing and investigated the mechanisms. Using scald burn mice models, we demonstrated that local Akermanite treatment significantly accelerated wound healing by increasing reepithelialization and the stemness of epidermal cells. Epidermal cells were cultured in medium containing Akermanite extracts to explore the cellular mechanism of reepithelialization. Akermanite promoted the cell proliferation and migration, maintaining more cells in the S and G2 /M phases of the cell cycle. An additional study showed that Akermanite enhanced the expressions of integrinß1, Lgr4, Lgr5, and Lgr6, which are specific molecular markers of epidermal stem cells, accompanied by the activation of the Wnt/ß-catenin pathway. These results suggested that Akermanite accelerated reepithelialization by increasing the proliferation, migration, and stemness of epidermal cells in a manner related to the Wnt/ß-catenin pathway, which might contribute, at least partially, to accelerated wound healing by Akermanite therapy.


Subject(s)
Cell Proliferation/drug effects , Ceramics/pharmacology , Epidermal Cells/drug effects , Re-Epithelialization/drug effects , Stem Cells/metabolism , Animals , Biocompatible Materials , Cell Cycle/drug effects , Cell Movement/drug effects , Cells, Cultured , Epidermal Cells/metabolism , Humans , Integrin beta1/drug effects , Integrin beta1/genetics , Integrin beta1/metabolism , Mice , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Wnt Signaling Pathway , Wound Healing
6.
Exp Cell Res ; 362(2): 302-310, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29179977

ABSTRACT

Radioresistance (inherent or acquired) remains a major obstacle affecting the clinical outcome of radiotherapy for laryngeal carcinoma. Results from our laboratory and other groups suggest that aberrant glycosylation contributes to cancer acquired radioresistance. However, the role of glycosylation in inherent radioresistance of laryngeal carcinoma has not been fully uncovered. In this study, we investigated the glycan profiling of the inherent radioresistant (Hep-2max) and radiosensitive (Hep-2min) cell lines using lectin microarray analysis. The results revealed that the radioresistant cell line Hep-2max presented higher core 1-type O-glycans than the sensitive one. Further analysis of the O-glycan regulation by benzyl-α-GalNAc application in Hep-2max cells showed partial inhibition of the O-glycan biosynthesis and increased radiosensitivity. In addition, core 1 ß1, 3-galactosyltransferase (C1GALT1) overexpression in Hep-2min cells enhanced cell migration, invasion, and radioresistance. Conversely, knockdown of C1GALT1 in Hep-2max cells was able to suppress these malignant phenotypes. Moreover, mechanistic investigations showed that C1GALT1 modified the O-glycans on integrin ß1 and regulated its activity. The glycosylation-mediated radioresistance was further inhibited by anti-integrin ß1 blocking antibody. Importantly, we also observed that core 1-type O-glycans expression was correlated with advanced tumor stage, metastasis, and poor survival of laryngeal carcinoma patients. These findings suggest that altered O-glycosylation can lead to the inherent radioresistance and progression, and therefore may be important for enhancing the efficacy of radiotherapy in laryngeal carcinoma.


Subject(s)
Carcinoma/radiotherapy , Laryngeal Neoplasms/radiotherapy , Polysaccharides/genetics , Radiation Tolerance/genetics , Antibodies, Anti-Idiotypic/pharmacology , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Cell Movement/genetics , Galactosyltransferases/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Glycosylation , Humans , Integrin beta1/drug effects , Integrin beta1/genetics , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/pathology , Lectins/genetics , Microarray Analysis/methods , Polysaccharides/biosynthesis
7.
Biochem Biophys Res Commun ; 503(4): 2206-2211, 2018 09 18.
Article in English | MEDLINE | ID: mdl-29953857

ABSTRACT

Within the extracellular domains of metastasis suppressor CD82, the large extracellular loop (EC2) has received much of the attention and its structure and function have been studied in detail. However, little attention has been given to the small extracellular loop (EC1 domain). To investigate the function role of EC1 in metastasis suppression of CD82, the peptide mimicking EC1 amino acid sequence (EC1-mP) was synthesized and its effect on cancer cells behavior was examined. Here, we reported that EC1-mP strongly inhibited cancer cell migration in vitro, attnuated the ability of cancer cells adhesion to fibronectin, and induced the apoptosis. Furthermore, the EC1-mP was showed to supprese the expressions of integrins α5 and ß1, as well as decreased the phosphorylation of FAK and expression of ILK in SW620 cells. Taken together, these results demonstrate that this small peptide has the functional role of CD82 intact molecule. This novel finding will improve our understanding of the mechanism by which CD82 inhibits metastasis, and suggested that EC1 mimic peptide may be a promising candidate for developing anti-metastasis drugs.


Subject(s)
Kangai-1 Protein/genetics , Peptides/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Humans , Integrin alpha5/drug effects , Integrin beta1/drug effects , Molecular Mimicry , Neoplasm Metastasis , Protein Domains , Signal Transduction/drug effects
8.
Breast Cancer Res Treat ; 155(3): 431-40, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26860947

ABSTRACT

The central role of HER2 as the disease driver and HER3 as its essential partner has made them rational targets for the treatment of HER2-amplifed breast cancers, and there is considerable interest in developing highly effective treatment regimens for this disease that consist of targeted therapies alone. Much of these efforts are focused on dual targeting approaches, particularly dual targeting of the HER2-HER3 tumor driver complex itself, or vertical combinations that target downstream PI3K or Akt in addition to HER2. There is also potential in lateral combinations based on evidence implicating cross-talk with other membrane receptor systems, particularly integrins, and such lateral combinations can potentially involve either HER2 or HER3. We established a preclinical model of targeting HER3 using doxycycline-inducible shRNA and determined the efficacy of a ß1 integrin inhibitor in combination with targeting HER3. We report that targeting HER3 and ß1 integrin provides a particularly effective combination therapy approach for HER2-amplified cancers, surpassing the combination of HER2 and ß1 integrin targeting, and evading some of the safety concerns associated with direct HER2-targeting. This further validates HER3 as a major hub mediating the tumorigenic functions of HER2 and identifies it as a high value target for lateral combination therapy strategies.


Subject(s)
Breast Neoplasms/drug therapy , Doxycycline/administration & dosage , Integrin beta1/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Integrin beta1/drug effects , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Signal Transduction/drug effects , Treatment Outcome
9.
Ann Rheum Dis ; 75(3): 601-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25596157

ABSTRACT

OBJECTIVE: MicroRNAs (miRNAs) are small endogenous, non-coding RNAs that act as post-transcriptional regulators. We analysed the in vivo effect of miRNA-124 (miR-124, the rat analogue of human miR-124a) on adjuvant-induced arthritis (AIA) in rats. METHODS: AIA was induced in Lewis rats by injecting incomplete Freund's adjuvant with heat-killed Mycobacterium tuberculosis. Precursor (pre)-miR-124 was injected into the right hind ankle on day 9. Morphological changes in the ankle joint were assessed by micro-CT and histopathology. Cytokine expression was examined by western blotting and real-time RT-PCR. The effect of miR-124 on predicted target messenger RNAs (mRNAs) was examined by luciferase reporter assays. The effect of pre-miR-124 or pre-miR-124a on the differentiation of human osteoclasts was examined by tartrate-resistant acid phosphatase staining. RESULTS: We found that miR-124 suppressed AIA in rats, as demonstrated by decreased synoviocyte proliferation, leucocyte infiltration and cartilage or bone destruction. Osteoclast counts and expression level of receptor activator of the nuclear factor κB ligand (RANKL), integrin ß1 (ITGB1) and nuclear factor of activated T cells cytoplasmic 1 (NFATc1) were reduced in AIA rats treated with pre-miR-124. Luciferase analysis showed that miR-124 directly targeted the 3'UTR of the rat NFATc1, ITGB1, specificity protein 1 and CCAAT/enhancer-binding protein α mRNAs. Pre-miR-124 also suppressed NFATc1 expression in RAW264.7 cells. Both miR-124 and miR-124a directly targeted the 3'-UTR of human NFATc1 mRNA, and both pre-miR-124 and pre-miR-124a suppressed the differentiation of human osteoclasts. CONCLUSIONS: We found that miR-124 ameliorated AIA by suppressing critical prerequisites for arthritis development, such as RANKL and NFATc1. Thus, miR-124a is a candidate for therapeutic use for human rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , MicroRNAs/pharmacology , Osteoclasts/drug effects , RNA, Messenger/drug effects , Animals , CCAAT-Enhancer-Binding Protein-alpha/drug effects , CCAAT-Enhancer-Binding Protein-alpha/genetics , Disease Progression , Gene Expression Regulation/drug effects , Humans , Integrin beta1/drug effects , Integrin beta1/genetics , RANK Ligand/drug effects , RANK Ligand/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Sp1 Transcription Factor/drug effects , Sp1 Transcription Factor/genetics , Synovial Membrane/cytology , Transcription Factors/drug effects , Transcription Factors/genetics
10.
Environ Toxicol ; 31(7): 799-807, 2016 Jul.
Article in English | MEDLINE | ID: mdl-25534675

ABSTRACT

Bisphenol A (BPA) is a widely used industrial chemical and also an environmental endocrine disruptor (EED), which serves as a monomer in the manufacture of polycarbonate plastics. BPA enters human body mainly through oral intake, and has been reported as being linked to oncogenesis in many tissues. However, the association of BPA intake with gastrointestinal cancer, such as colon cancer, has received less attention. The present study was conducted to investigate the effects of BPA on the migration of normal colon epithelial cells (NCM460 cells) and further elucidate the underlying mechanisms. Our data showed that 1 × 10(-8) M (equivalent to environmental concentration) of BPA potently promoted the migration of NCM460 cells. Interestingly, BPA treatment induced an increase of integrin ß1 expression, and the functional blocking of integrin ß1 abolished the migration-promoting effects of BPA. Moreover, the results showed that it was estrogen receptor ß but not estrogen receptor α that was involved in this migration promotion. In addition, cellular exposure of BPA stimulated the expression and activity of MMP-9, a well-known factor of cell migration. Taken together, these results indicate that environmental concentration of BPA exposure promotes cell migration through activating ERß-mediated integrin ß1/MMP-9 pathway, suggesting exposure to BPA in the colon may present a potential cancer risk. © 2014 Wiley Periodicals, Inc. Environ Toxicol 31: 799-807, 2016.


Subject(s)
Benzhydryl Compounds/toxicity , Cell Movement/drug effects , Endocrine Disruptors/toxicity , Integrin beta1/drug effects , Matrix Metalloproteinase 9/drug effects , Phenols/toxicity , Receptors, Estrogen/drug effects , Cell Line , Cell Survival/drug effects , Epithelial Cells/drug effects , Estrogen Receptor alpha/drug effects , Estrogen Receptor beta/drug effects , Humans , Signal Transduction/drug effects , Wound Healing/drug effects
11.
J Periodontal Res ; 50(4): 479-85, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25244101

ABSTRACT

BACKGROUND AND OBJECTIVE: Migration of the junctional epithelium occurs in association with the formation of a periodontal pocket. Although the migration of junctional epithelium is known to be related to the proliferation and migration of gingival junctional epithelial cells, the mechanism has not been clarified. In patients with periodontitis, the levels of interleukin-8 (IL-8) in both gingival tissue and gingival crevicular fluid are dramatically increased. IL-8 has broad bioactive functions. In this study, we examined the role of IL-8 in DNA synthesis, migration and protection against apoptosis in cultured human gingival epithelial cells (HGEC). MATERIAL AND METHODS: DNA synthesis was estimated by measuring the incorporation of bromodeoxyuridine. The migration of gingival epithelial cells was assessed in a wound-healing assay. The expression of integrin beta-1 was analyzed using immunofluorescence confocal microscopy and western blotting. Cleaved caspase-3 was detected using western blotting and a Caspase-Glo assay kit. RESULTS: IL-8 increased the synthesis of DNA in HGEC, and the maximal effect was seen at 25 or 50 ng/mL of IL-8. In addition, 50 ng/mL of IL-8 induced cell migration, and a neutralizing antibody of integrin beta-1 inhibited the migration. IL-8 also activated expression of integrin beta-1. Furthermore, IL-8 reduced the Aggregatibacter actinomycetemcomitans-induced increase in caspase-3 expression in HGEC. CONCLUSION: IL-8 may facilitate the migration of gingival junctional epithelium by enhancing DNA synthesis, migration and preventing apoptosis of gingival epithelial cells.


Subject(s)
Caspase 3/drug effects , DNA/biosynthesis , Epithelial Attachment/drug effects , Gingiva/drug effects , Interleukin-8/pharmacology , Adult , Aggregatibacter actinomycetemcomitans/physiology , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , DNA/drug effects , Down-Regulation/drug effects , Epithelial Attachment/cytology , Epithelial Cells/drug effects , Female , Gingiva/cytology , Humans , Integrin beta1/drug effects , Male , Young Adult
12.
Am J Physiol Heart Circ Physiol ; 306(1): H101-8, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24163072

ABSTRACT

Previous studies have established integrins as cell surface receptors that mediate cardiomyocyte-extracellular matrix (ECM) attachments. This study sought to determine the contributions of the myocardial ß1- and ß3-integrin subunits to ventricular dilatation and coronary flow regulation using a blood-perfused isolated heart preparation. Furthermore, cardiomyocyte adhesion to collagen types I and IV, fibronectin, and laminin with and without a ß1-integrin subunit neutralizing antibody was assessed during the course of remodeling secondary to a sustained cardiac volume overload, including the onset of heart failure. Isolated cardiomyocytes were obtained during the initial, compensated, and decompensated phases of remodeling resulting from an aortocaval fistula created in 8-wk-old male Sprague-Dawley rats. Blocking the ß1-integrin subunit in isolated normal hearts produced ventricular dilatation, whereas this was not the case when the ß3-subunit was blocked. Substantial reductions in cardiomyocyte adhesion coincided with the previously documented development of ventricular dilatation and decreased contractility postfistula, with the ß1-integrin contribution to adhesion ranging from 28% to 73% over the course of remodeling being essentially substrate independent. In contrast, both integrin subunits were found to be involved in regulating coronary vascular resistance. It is concluded that marked reductions in integrin-mediated cardiomyocyte adhesion to the ECM play a significant role in the progression of adverse myocardial remodeling that leads to heart failure. Furthermore, although both the ß1- and ß3-integrin subunits were involved in regulating coronary vascular resistance, only inhibition of ß1-integrin-mediated adhesion resulted in ventricular dilatation of the normal heart.


Subject(s)
Cardiac Volume , Integrin beta1/metabolism , Integrin beta3/metabolism , Myocytes, Cardiac/physiology , Animals , Antibodies, Neutralizing/pharmacology , Cell Adhesion , Coronary Circulation , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , In Vitro Techniques , Integrin beta1/drug effects , Integrin beta3/drug effects , Male , Myocardial Contraction , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Ventricular Remodeling
13.
Toxicol Appl Pharmacol ; 259(3): 387-94, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22310179

ABSTRACT

Oroxylin A, a naturally occurring monoflavonoid extracted from Scutellariae radix, shows effective anticancer activities and low toxicities both in vivo and in vitro in previous studies. In this study, we investigated whether the CAM-DR model of HepG2 cells showed resistance to cytotoxic agents compared with normally cultured HepG2 cells. Furthermore, after the treatment of Paclitaxel, less inhibitory effects and decreased apoptosis rate were detected in the model. Data also revealed increased expression of Integrinß1 might be responsible for the resistance ability. Moreover, Integrinß1-siRNA-transfected CAM-DR HepG2 cells exhibited more inhibitory effects and higher levels of apoptosis than the non-transfected CAM-DR cells. The data corroborated that Integrinß1 played a significant role in CAM-DR. After the treatment of weakly-toxic concentrations of Oroxylin A, the apoptosis induced by Paclitaxel in the CAM-DR model increased dramatically. Western blot assay revealed Oroxylin A markedly down-regulated the expression of Integrinß1 and the activity of related pathway. As a conclusion, Oroxylin A can reverse the resistance of CAM-DR via inhibition of Integrinß1 and its related pathway. Oroxylin A may be a potential candidate of a CAM-DR reversal agent.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm/drug effects , Flavonoids/pharmacology , Integrin beta1/drug effects , Liver Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Blotting, Western , Carcinoma, Hepatocellular/pathology , Cell Adhesion , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Flavonoids/administration & dosage , Flavonoids/isolation & purification , Hep G2 Cells , Humans , Integrin beta1/metabolism , Liver Neoplasms/pathology , Paclitaxel/pharmacology , Scutellaria baicalensis/chemistry
14.
J Cell Biol ; 173(5): 767-80, 2006 Jun 05.
Article in English | MEDLINE | ID: mdl-16754960

ABSTRACT

Dynamic turnover of integrin cell adhesion molecules to and from the cell surface is central to cell migration. We report for the first time an association between integrins and Rab proteins, which are small GTPases involved in the traffic of endocytotic vesicles. Rab21 (and Rab5) associate with the cytoplasmic domains of alpha-integrin chains, and their expression influences the endo/exocytic traffic of integrins. This function of Rab21 is dependent on its GTP/GDP cycle and proper membrane targeting. Knock down of Rab21 impairs integrin-mediated cell adhesion and motility, whereas its overexpression stimulates cell migration and cancer cell adhesion to collagen and human bone. Finally, overexpression of Rab21 fails to induce cell adhesion via an integrin point mutant deficient in Rab21 association. These data provide mechanistic insight into how integrins are targeted to intracellular compartments and how their traffic regulates cell adhesion.


Subject(s)
Endosomes/metabolism , Integrin beta1/metabolism , rab GTP-Binding Proteins/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Endosomes/drug effects , Gene Expression Regulation , Green Fluorescent Proteins/drug effects , Green Fluorescent Proteins/metabolism , Humans , Integrin beta1/drug effects , Mutation , Protein Transport/physiology , Time Factors , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
15.
J Cell Biol ; 173(4): 601-13, 2006 May 22.
Article in English | MEDLINE | ID: mdl-16702230

ABSTRACT

Plexins are cell surface receptors for semaphorins and regulate cell migration in many cell types. We recently reported that the semaphorin 4D (Sema4D) receptor Plexin-B1 functions as a GTPase-activating protein (GAP) for R-Ras, a member of Ras family GTPases implicated in regulation of integrin activity and cell migration. We characterized the role of R-Ras downstream of Sema4D/Plexin-B1 in cell migration. Activation of Plexin-B1 by Sema4D suppressed the ECM-dependent R-Ras activation, R-Ras-mediated phosphatydylinositol 3-kinase activation, and beta(1) integrin activation through its R-Ras GAP domain, leading to inhibition of cell migration. In addition, inactivation of R-Ras by overexpression of the R-Ras-specific GAP or knockdown of R-Ras by RNA interference was sufficient for suppressing beta(1) integrin activation and cell migration in response to the ECM stimulation. Thus, we conclude that R-Ras activity is critical for ECM-mediated beta(1) integrin activation and cell migration and that inactivation of R-Ras by Sema4D/Plexin-B1-mediated R-Ras GAP activity controls cell migration by modulating the activity of beta(1) integrins.


Subject(s)
Antigens, CD/metabolism , Cell Movement/physiology , GTP Phosphohydrolases/metabolism , GTPase-Activating Proteins/metabolism , Integrin beta1/metabolism , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , ras Proteins/metabolism , Animals , Antigens, CD/pharmacology , COS Cells , Cell Movement/drug effects , Chlorocebus aethiops , Collagen/metabolism , Collagen/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Enzyme Activation/physiology , Extracellular Matrix/metabolism , GTP Phosphohydrolases/genetics , Glycine/analogs & derivatives , Integrin beta1/drug effects , Phosphatidylinositol 3-Kinases/metabolism , RNA Interference , Rats , Semaphorins/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , ras Proteins/genetics
16.
Anticancer Res ; 41(6): 2913-2923, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34083282

ABSTRACT

BACKGROUND/AIM: Epithelial to mesenchymal transition (EMT), and focal adhesion kinase (FAK) facilitate lung cancer cell motility and survival. We, therefore, investigated the antimigratory effect of 3,4-dihydroxy-5,4'-dimethoxybibenzyl (DS-1) on human lung cancer cells. MATERIALS AND METHODS: Cell viability and proliferation were examined by the 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide assay. Filopodia formation, migration, and anchorage-independent growth assays were performed to assess metastatic behaviors while EMT-related proteins, integrins, and FAK-RhoA pathway were evaluated by western blot analysis. RESULTS: We found that DS-1 significantly inhibited the proliferation of lung cancer cells compared to the control. The aggressive behavior of cancer cells, including migration and invasion, was significantly reduced by DS-1. Anchorage-independent growth analysis provided evidence that DS-1 suppressed the growth and survival of cancer cells in detached conditions as indicated by the significant reduction in size and number of colonies. With regard to the mechanisms involved, we found that DS-1-suppressed EMT, as indicated by the reduction of EMT markers, namely N-cadherin, SNAIL and SLUG, and increased levels of the epithelial marker, E-cadherin. In addition, DS-1 was shown to reduce the level of integrin ß1 protein and FAK activation. CONCLUSION: DS-1 suppressed lung cancer metastasis via suppressing EMT, integrin ß1 expression and FAK-related signaling.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Epithelial-Mesenchymal Transition/drug effects , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Integrin beta1/drug effects , Lung Neoplasms/pathology , Signal Transduction/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Integrin beta1/metabolism
17.
Glia ; 58(7): 790-801, 2010 May.
Article in English | MEDLINE | ID: mdl-20091784

ABSTRACT

Microglia are the primary immune surveillance cells in the brain, and when activated they play critical roles in inflammatory reactions and tissue repair in the damaged brain. Microglia rapidly extend their processes toward the damaged areas in response to stimulation of the metabotropic ATP receptor P2Y(12) by ATP released from damaged tissue. This chemotactic response is a highly important step that enables microglia to function properly at normal and pathological sites in the brain. To investigate the molecular pathways that underlie microglial process extension, we developed a novel method of modeling microglial process extension that uses transwell chambers in which the insert membrane is coated with collagen gel. In this study, we showed that ATP increased microglial adhesion to collagen gel, and that the ATP-induced process extension and increase in microglial adhesion were inhibited by integrin blocking peptides, RGD, and a functional blocking antibody against integrin-beta1. An immunoprecipitation analysis with an antibody against the active form of integrin-beta1 showed that P2Y(12) mediated the integrin-beta1 activation by ATP. In addition, time-lapse imaging of EGFP-labeled microglia in mice hippocampal slices showed that RGD inhibited the directional process extension toward the nucleotide source, and immunohistochemical staining showed that integrin-beta1 accumulated in the tips of the microglial processes in rat hippocampal slices stimulated with ADP. These findings indicate that ATP induces the integrin-beta1 activation in microglia through P2Y(12) and suggest that the integrin-beta1 activation is involved in the directional process extension by microglia in brain tissue.


Subject(s)
Adenosine Triphosphate/metabolism , Encephalitis/metabolism , Gliosis/metabolism , Integrin beta1/metabolism , Microglia/metabolism , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Antibodies, Blocking/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Chemotaxis/drug effects , Chemotaxis/physiology , Coculture Techniques , Collagen/metabolism , Encephalitis/pathology , Encephalitis/physiopathology , Extracellular Matrix/metabolism , Gliosis/pathology , Gliosis/physiopathology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Integrin beta1/drug effects , Microglia/drug effects , Microglia/ultrastructure , Organ Culture Techniques , Peptides/metabolism , Peptides/pharmacology , Rats , Rats, Wistar , Receptors, Purinergic P2Y12
18.
Medicine (Baltimore) ; 99(27): e20944, 2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32629699

ABSTRACT

BACKGROUND: Colorectal cancer is the second commonly seen cancer around the world and accounts for 13% of all human cancers. Among them, 25% of all case were diagnosed with metastasis and 50% occurs metastasis during the development of disease. Cetuximab is a chimeric monoclonal antibody against epidermal growth factor receptor, and is used for treatment of metastatic colorectal cancer alone or combined with chemotherapy or radiation therapy. Integrin-beta 1 (ITGB1), which is also known as CD29, and plays an important role in development of malignant cancers. However, the effect of ITGB1 in promoting the anti-tumor effect of cetuximab is not fully understand. METHODS: The model of ITGB1 inhibition and overexpression was firstly constructed in LS174T cells, and the viability of cells in each group was detected using CCK-8 assay. The expression of key factors in tumor formation process at transcription level was detected using real-time quantitative polymerase chain reaction method. The expression of key proteins in metastasis process, cell apoptosis and activation of Ras/Raf/MEK signaling pathway was detected using western blotting analysis. And the concentration of key factors of in tumor formation process in cultured medium of LS174T cells were detected using enzyme-linked immunosorbent assay method. RESULTS: We found that cetuximab could inhibit the proliferation of LS174T cells, and inhibition of ITGB1 enhanced this effect while overexpression of ITGB1 reduced this effect. We further found that cetuximab could inhibit the expression and secretion of extracellular matrix degradation related molecules in cultured medium and transcription level. Besides, we also found that the expression of key factors in angiogenesis and extracellular matrix degradation related proteins were also reduced after cetuximab treatment. These effects might be mediated by Ras/Raf/MAPK signaling pathway and enhanced after inhibition of ITGB1 expression. CONCLUSION: Inhibition of ITGB1 might be a new therapeutic method in colorectal cancer.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Cetuximab/pharmacology , Colorectal Neoplasms/drug therapy , Integrin beta1/drug effects , Antineoplastic Agents, Immunological/therapeutic use , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cetuximab/therapeutic use , Colorectal Neoplasms/pathology , Humans , Integrin beta1/metabolism
19.
Sci Rep ; 10(1): 11133, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32636409

ABSTRACT

Pancreatic cancer, one of the deadliest human malignancies, has a dismal 5-year survival rate of 9%. KRAS is the most commonly mutated gene in pancreatic cancer, but clinical agents that directly target mutant KRAS are not available. Several effector pathways are activated downstream of oncogenic Kras, including MAPK signaling. MAPK signaling can be inhibited by targeting MEK1/2; unfortunately, this approach has been largely ineffective in pancreatic cancer. Here, we set out to identify mechanisms of MEK inhibitor resistance in pancreatic cancer. We optimized the culture of pancreatic tumor 3D clusters that utilized Matrigel as a basement membrane mimetic. Pancreatic tumor 3D clusters recapitulated mutant KRAS dependency and recalcitrance to MEK inhibition. Treatment of the clusters with trametinib, a MEK inhibitor, had only a modest effect on these cultures. We observed that cells adjacent to the basement membrane mimetic Matrigel survived MEK inhibition, while the cells in the interior layers underwent apoptosis. Our findings suggested that basement membrane attachment provided survival signals. We thus targeted integrin ß1, a mediator of extracellular matrix contact, and found that combined MEK and integrin ß1 inhibition bypassed trametinib resistance. Our data support exploring integrin signaling inhibition as a component of combination therapy in pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Integrin beta1/metabolism , MAP Kinase Signaling System/drug effects , Pancreatic Neoplasms/drug therapy , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Integrin beta1/drug effects , Mice , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism
20.
J Recept Signal Transduct Res ; 29(2): 119-25, 2009.
Article in English | MEDLINE | ID: mdl-19519177

ABSTRACT

Gram-negative bacillus infection is an important risk factor of acute lung injury (ALI). Previous experiments have revealed that lipopolysaccharide (LPS), a primary component of endotoxin of gram-negative bacilli, stimulated the inflammatory reactions that contribute to ALI and pulmonary interstitial fibrosis, but the mechanisms were not well understood. We reported that LPS was able to directly induce secretion of collagen in mouse lung fibroblasts via activation of phosphoinositide3-kinase-Akt (PI3K-Akt) pathway through toll-like receptor 4 (TLR4) in vitro. We found that overexpression of TLR4, type I procollagen, alpha smooth muscle actin (alpha-SMA), and p-AKT in primary cultured mouse lung fibroblast stimulated by LPS were detected by real-time PCR or Western blots, and the contents of C-terminal propeptide of type I procollagen (PICP) in cell culture supernatants were increased simultaneously. The activation of TLR4 stimulated by LPS could also up-regulate the expression of integrin beta1 and TLR4 in mouse lung fibroblast, which could accelerate ALI and pulmonary interstitial fibrosis processes. All these changes could be inhabited by transfection of Lentivirus-TLR4-siRNA or application of PI3K inhibitor LY294002. Therefore, we infer that besides pulmonary macrophage, lung fibroblasts are also important target cells directly influenced by LPS, which may play an important role in ALI and pulmonary interstitial fibrosis.


Subject(s)
Acute Lung Injury/immunology , Collagen Type I/metabolism , Integrin beta1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Toll-Like Receptor 4/metabolism , Acute Lung Injury/microbiology , Animals , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblasts/immunology , Integrin beta1/drug effects , Lipopolysaccharides/immunology , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 4/agonists , Up-Regulation/drug effects , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL