Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 305
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 67(3): 512-527.e4, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28757207

ABSTRACT

Aberrant signaling by the mammalian target of rapamycin (mTOR) contributes to the devastating features of cancer cells. Thus, mTOR is a critical therapeutic target and catalytic inhibitors are being investigated as anti-cancer drugs. Although mTOR inhibitors initially block cell proliferation, cell viability and migration in some cancer cells are quickly restored. Despite sustained inhibition of mTORC1/2 signaling, Akt, a kinase regulating cell survival and migration, regains phosphorylation at its regulatory sites. Mechanistically, mTORC1/2 inhibition promotes reorganization of integrin/focal adhesion kinase-mediated adhesomes, induction of IGFR/IR-dependent PI3K activation, and Akt phosphorylation via an integrin/FAK/IGFR-dependent process. This resistance mechanism contributes to xenograft tumor cell growth, which is prevented with mTOR plus IGFR inhibitors, supporting this combination as a therapeutic approach for cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Cell Movement/drug effects , Drug Resistance, Neoplasm , Focal Adhesion Kinase 1/metabolism , Melanoma/drug therapy , Multiprotein Complexes/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Receptors, Somatomedin/antagonists & inhibitors , Skin Neoplasms/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Female , Focal Adhesion Kinase 1/genetics , Humans , Integrin alpha2/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Melanoma/enzymology , Melanoma/pathology , Mice, Nude , Multiprotein Complexes/metabolism , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptor, IGF Type 1 , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Signal Transduction/drug effects , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
2.
Carcinogenesis ; 45(4): 235-246, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38142122

ABSTRACT

Glioma is the most common malignant brain tumor in adults with a high mortality and recurrence rate. Integrin alpha 2 (ITGA2) is involved in cell adhesion, stem cell regulation, angiogenesis and immune cell function. The role of ITGA2 in glioma malignant invasion remains unknown. The function and clinical relevance of ITGA2 were analysed by bioinformatics databases. The expression of ITGA2 in parent cells and GSCs was detected by flow cytometry and immunofluorescence double staining. The role of ITGA2 on the malignant phenotype of GSCs and epithelial-mesenchymal transition (EMT) was identified by stem cell function assays and Western blot. The effect of ITGA2 on glioma progression in vivo was determined by the intracranial orthotopic xenograft model. Immunohistochemistry, Spearman correlation and Kaplan-Meier were used to analyse the relationship of ITGA2 with clinical features and glioma prognosis. Biological analysis showed that ITGA2 might be related to cell invasion and migration. ITGA2, enriched in GSCs and co-expressed with SOX2, promoted the invasion and migration of GSCs by activating STAT3 phosphorylation and enhancing EMT. ITGA2 knockout suppressed the intracranial orthotopic xenograft growth and prolonged the survival of xenograft mice. In addition, the expression level of ITGA2 was significantly correlated to the grade of malignancy, N-cadherin and Ki67. High expression of ITGA2 indicated a worse prognosis of glioma patients. As a biomarker for the prediction of prognosis, ITGA2 promotes the malignant invasion of GSCs by activating STAT3 phosphorylation and enhancing EMT, leading to tumor recurrence and poor prognosis.


Subject(s)
Brain Neoplasms , Glioma , Integrin alpha2 , Neoplastic Stem Cells , STAT3 Transcription Factor , Adult , Animals , Humans , Mice , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Glioma/pathology , Integrin alpha2/genetics , Integrin alpha2/metabolism , Phosphorylation , Prognosis , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Neoplastic Stem Cells/metabolism
3.
BMC Cancer ; 24(1): 559, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702644

ABSTRACT

In contrast to the decreasing trends in developed countries, the incidence and mortality rates of cervical squamous cell carcinoma in China have increased significantly. The screening and identification of reliable biomarkers and candidate drug targets for cervical squamous cell carcinoma are urgently needed to improve the survival rate and quality of life of patients. In this study, we demonstrated that the expression of MUC1 was greater in neoplastic tissues than in non-neoplastic tissues of the cervix, and cervical squamous cell carcinoma patients with high MUC1 expression had significantly worse overall survival than did those with low MUC1 expression, indicating its potential for early diagnosis of cervical squamous cell carcinoma. Next, we explored the regulatory mechanism of MUC1 in cervical squamous cell carcinoma. MUC1 could upregulate ITGA2 and ITGA3 expression via ERK phosphorylation, promoting the proliferation and metastasis of cervical cancer cells. Further knockdown of ITGA2 and ITGA3 significantly inhibited the tumorigenesis of cervical cancer cells. Moreover, we designed a combination drug regimen comprising MUC1-siRNA and a novel ERK inhibitor in vivo and found that the combination of these drugs achieved better results in animals with xenografts than did MUC1 alone. Overall, we discovered a novel regulatory pathway, MUC1/ERK/ITGA2/3, in cervical squamous cell carcinoma that may serve as a potential biomarker and therapeutic target in the future.


MUC1 is overexpressed in cervical squamous cell carcinoma. MUC1 regulates ERK phosphorylation, and subsequently upregulates ITGA2 and ITGA3 expression to promote tumorigenesis in cervical squamous cell carcinoma. A combination drug regimen targeting MUC1 and ERK achieved better results compared than MUC1 alone.


Subject(s)
Carcinoma, Squamous Cell , Cell Proliferation , Integrin alpha2 , Integrin alpha3 , Mucin-1 , Uterine Cervical Neoplasms , Humans , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/drug therapy , Female , Integrin alpha2/metabolism , Integrin alpha2/genetics , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/drug therapy , Mucin-1/metabolism , Mucin-1/genetics , Mice , Phosphorylation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Xenograft Model Antitumor Assays , MAP Kinase Signaling System , Mice, Nude , Extracellular Signal-Regulated MAP Kinases/metabolism
4.
Mol Cell Proteomics ; 21(4): 100213, 2022 04.
Article in English | MEDLINE | ID: mdl-35182768

ABSTRACT

Primary myelofibrosis (PMF) is a neoplasm prone to leukemic transformation, for which limited treatment is available. Among individuals diagnosed with PMF, the most prevalent mutation is the JAK2V617F somatic point mutation that activates the Janus kinase 2 (JAK2) enzyme. Our earlier reports on hyperactivity of ß1 integrin and enhanced adhesion activity of the α2ß1 complex in JAK2V617F megakaryocytes (MKs) led us to examine the new hypothesis that this mutation leads to posttranslational modification via changes in glycosylation. Samples were derived from immunoprecipitation of MKs obtained from Vav1-hJAK2V617F and WT mice. Immunoprecipitated fractions were separated by SDS-PAGE and analyzed using LC-MS/MS techniques in a bottom-up glycoproteomics workflow. In the immunoprecipitate, glycopeptiforms corresponding to 11 out of the 12 potential N-glycosylation sites of integrin ß1 and to all nine potential glycosylation sites of integrin α2 were observed. Glycopeptiforms were compared across WT and JAK2V617F phenotypes for both integrins. The overall trend observed is that JAK2V617F mutation in PMF MKs leads to changes in ß1 glycosylation; in most cases, it results in an increase in the integrated area of glycopeptiforms. We also observed that in mutated MKs, changes in integrin α2 glycosylation were more substantial than those observed for integrin ß1 glycosylation, a finding that suggests that altered integrin α2 glycosylation may also affect activation. Additionally, the identification of proteins associated to the cytoskeleton that were co-immunoprecipitated with integrins α2 and ß1 demonstrated the potential of the methodology employed in this study to provide some insight, at the peptide level, into the consequences of integrin activation in MKs. The extensive and detailed glycosylation patterns we uncovered provide a basis for future functional studies of each site in control cells as compared to JAK2V617F-mutated cells. Data are available via ProteomeXchange with identifier PXD030550.


Subject(s)
Janus Kinase 2/genetics , Megakaryocytes , Primary Myelofibrosis , Animals , Chromatography, Liquid , Integrin alpha2/genetics , Integrin alpha2/metabolism , Integrin beta1/genetics , Integrin beta1/metabolism , Megakaryocytes/metabolism , Mice , Mutation , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/genetics , Tandem Mass Spectrometry
5.
Am J Respir Crit Care Med ; 207(5): 553-565, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36170617

ABSTRACT

Rationale: Tissue-resident natural killer (trNK) cells have been identified in numerous organs, but little is known about their functional contribution to respiratory immunity, in particular during chronic lung diseases such as chronic obstructive pulmonary disease (COPD). Objectives: To investigate the phenotype and antiviral responses of trNK cells in murine cigarette smoke-induced experimental COPD and in human lung parenchyma from COPD donors. Methods: Mice were exposed to cigarette smoke for 12 weeks to induce COPD-like lung disease. Lung trNK cell phenotypes and function were analyzed by flow cytometry in both murine and human disease with and without challenge with influenza A virus. Measurements and Main Results: In the mouse lung, CD49a+CD49b+EOMES+ and CD49a+CD49b-EOMESlo NK cell populations had a distinct phenotype compared with CD49a- circulating NK cells. CD49a+ NK cells were more extensively altered earlier in disease onset than circulating NK cells, and increased proportions of CD49a+ NK cells correlated with worsening disease in both murine and human COPD. Furthermore, the presence of lung disease delayed both circulating and trNK cell functional responses to influenza infection. CD49a+ NK cells markedly increased their NKG2D, CD103, and CD69 expression in experimental COPD after influenza infection, and human CD49a+ NK cells were hyperactive to ex vivo influenza infection in COPD donors. Conclusions: Collectively, these results demonstrate that trNK cell function is altered in cigarette smoke-induced disease and suggests that smoke exposure may aberrantly prime trNK cell responsiveness to viral infection. This may contribute to excess inflammation during viral exacerbations of COPD.


Subject(s)
Influenza, Human , Lung Diseases , Pulmonary Disease, Chronic Obstructive , Humans , Mice , Animals , Integrin alpha1/metabolism , Influenza, Human/metabolism , Integrin alpha2/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Killer Cells, Natural , Lung/metabolism , Lung Diseases/metabolism , Antiviral Agents
6.
J Cell Biochem ; 124(7): 989-1001, 2023 07.
Article in English | MEDLINE | ID: mdl-37210732

ABSTRACT

Mutations in the αIIb ß-propeller domain have long been known to disrupt heterodimerization and intracellular trafficking of αIIbß3 complexes leading to diminished surface expression and/or function, resulting in Glanzmann thrombasthenia. Our previous study on three ß-propeller mutations, namely G128S, S287L, and G357S, showed variable defects in protein transport correlated with the patient's clinical phenotypes. Pulse-chase experiments revealed differences in αIIbß3 complex maturation among the three mutations. Hence, the current study aims to correlate conformational changes caused by each one of them. Evolutionary conservation analysis, stability analysis, and molecular dynamics simulations of the three mutant structures were carried out. Stability analysis revealed that, while G128S and G357S mutations destabilized the ß-propeller structure, S287L retained the stability. Wild-type and mutant ß-propeller structures, when subjected to molecular dynamics simulations, confirmed that G128S and G357S were both destabilizing in nature when compared with the wild-type and S287L based on several parameters studied, like RMSD, RMSF, Rg, FEL, PCA, secondary structure, and hydrogen bonds. In our previous study, we demonstrated that mutant S287L αIIbß3 complexes were more stable than the wild-type αIIbß3 complexes, as evidenced in pulse-chase experiments. These findings corroborate variable intracellular fates of mutant αIIbß3 complexes as a result of these ß-propeller mutations.


Subject(s)
Integrin alpha2 , Integrin beta3 , Platelet Glycoprotein GPIIb-IIIa Complex , Thrombasthenia , Humans , Integrin beta3/genetics , Molecular Dynamics Simulation , Mutation , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Structure, Secondary , Thrombasthenia/genetics , Thrombasthenia/metabolism , Integrin alpha2/genetics , Integrin alpha2/metabolism
7.
Int J Mol Sci ; 24(2)2023 Jan 04.
Article in English | MEDLINE | ID: mdl-36674478

ABSTRACT

The Src homology 2 domain-containing inositol 5-phosphatase 1 (SHIP1) is known to dephosphorylate PtdIns(3,4,5)P3 into PtdIns(3,4)P2 and to interact with several signaling proteins though its docking functions. It has been shown to negatively regulate platelet adhesion and spreading on a fibrinogen surface and to positively regulate thrombus growth. In the present study, we have investigated its role during the early phase of platelet activation. Using confocal-based morphometric analysis, we found that SHIP1 is involved in the regulation of cytoskeletal organization and internal contractile activity in thrombin-activated platelets. The absence of SHIP1 has no significant impact on thrombin-induced Akt or Erk1/2 activation, but it selectively affects the RhoA/Rho-kinase pathway and myosin IIA relocalization to the cytoskeleton. SHIP1 interacts with the spectrin-based membrane skeleton, and its absence induces a loss of sustained association of integrins to this network together with a decrease in αIIbß3 integrin clustering following thrombin stimulation. This αIIbß3 integrin dynamics requires the contractile cytoskeleton under the control of SHIP1. RhoA activation, internal platelet contraction, and membrane skeleton integrin association were insensitive to the inhibition of PtdIns(3,4,5)P3 synthesis or SHIP1 phosphatase activity, indicating a role for the docking properties of SHIP1 in these processes. Altogether, our data reveal a lipid-independent function for SHIP1 in the regulation of the contractile cytoskeleton and integrin dynamics in platelets.


Subject(s)
Integrin alpha2 , Integrin beta3 , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Platelet Activation , Blood Platelets/metabolism , Integrin beta3/metabolism , Phosphatidylinositols/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombin/pharmacology , Thrombin/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Integrin alpha2/metabolism
8.
Am J Physiol Gastrointest Liver Physiol ; 322(6): G583-G597, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35319286

ABSTRACT

Intestinal ganglionic cells in the adult enteric nervous system (ENS) are continually exposed to stimuli from the surrounding microenvironment and need at times to respond to disturbed homeostasis following acute intestinal injury. The kinase DCLK1 and intestinal Dclk1-positive cells have been reported to contribute to intestinal regeneration. Although Dclk1-positive cells are present in adult enteric ganglia, their cellular identity and response to acute injury have not been investigated in detail. Here, we reveal the presence of distinct Dclk1-tdTom+/CD49b+ glial-like and Dclk1-tdTom+/CD49b- neuronal cell types in adult myenteric ganglia. These ganglionic cells demonstrate distinct patterns of tracing over time yet show a similar expansion in response to elevated serotonergic signaling. Interestingly, Dclk1-tdTom+ glial-like and neuronal cell types appear resistant to acute irradiation injury-mediated cell death. Moreover, Dclk1-tdTom+/CD49b+ glial-like cells show prominent changes in gene expression profiles induced by injury, in contrast to Dclk1-tdTom+/CD49b- neuronal cell types. Finally, subsets of Dclk1-tdTom+/CD49b+ glial-like cells demonstrate prominent overlap with Nestin and p75NTR and strong responses to elevated serotonergic signaling or acute injury. These findings, together with their role in early development and their neural crest-like gene expression signature, suggest the presence of reserve progenitor cells in the adult Dclk1 glial cell lineage.NEW & NOTEWORTHY The kinase DCLK1 identifies glial-like and neuronal cell types in adult murine enteric ganglia, which resist acute injury-mediated cell death yet differ in their cellular response to injury. Interestingly, Dclk1-labeled glial-like cells show prominent transcriptional changes in response to injury and harbor features reminiscent of previously described enteric neural precursor cells. Our data thus add to recently emerging evidence of reserve cellular plasticity in the adult enteric nervous system.


Subject(s)
Enteric Nervous System , Neural Stem Cells , Animals , Enteric Nervous System/physiology , Integrin alpha2/metabolism , Mice , Mice, Transgenic , Neuroglia/metabolism , Neurons/metabolism
9.
Breast Cancer Res Treat ; 192(1): 89-100, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35098437

ABSTRACT

PURPOSE: ITGA2 encodes the integrin, α2 which mediates metastatic progression, and is a predictor of poor prognosis and chemoresistance in breast cancer. Decreased ITGA2 promoter methylation is implicated as a driver of increased gene expression in aggressive prostate and pancreatic tumours, however the contribution of altered methylation to ITGA2 expression changes in breast tumours has not been examined. METHODS: ITGA2 gene methylation and gene expression was examined in publicly available breast cancer datasets, and ITGA2 promoter methylation was mapped by targeted bisulphite sequencing analysis in breast tumour cell lines. The expression of a putative regulatory long noncoding RNA (lncRNA) was examined by qPCR and its' functionality was investigated using gene knockdown (antisense oligonucleotides) and over expression in breast cancer cell lines. RESULTS: In breast tumours and breast cancer cell lines the ITGA2 promoter is largely unmethylated, with gene expression variable in tumour subtypes, irrespective of promoter methylation. A novel lncRNA (AC025180.1;ENSG00000249899), named herein I2ALR, was identified at the ITGA2 gene locus, and was variably expressed in breast tumours and breast cancer cell subtypes. I2LAR knockdown resulted in upregulation of ITGA2 gene expression, whilst over-expression of I2ALR resulted in downregulation of ITGA2 mRNA. Further, examination of two downstream targets of ITGA2 associated with breast tumor stemness and metastasis (CCND1 and ACLY), revealed concomitant gene expression changes in response to I2ALR modulation. CONCLUSION: I2ALR represents a novel regulatory molecule targeting ITGA2 expression in breast tumours; a finding of significant and topical interest to the development of therapeutics targeting this integrin.


Subject(s)
Breast Neoplasms , RNA, Long Noncoding , Breast Neoplasms/genetics , Cell Line, Tumor , DNA Methylation , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha2/genetics , Integrin alpha2/metabolism , Integrins , Male , RNA, Long Noncoding/genetics
10.
Pediatr Crit Care Med ; 23(9): 727-735, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35687090

ABSTRACT

OBJECTIVES: Morbidity and mortality related to modified Blalock-Taussig shunt (mBTTS) thrombosis remain a significant risk. Platelet inhibition following mBTTS may reduce this risk. However, oral antiplatelet agents have variable absorption following surgery. We determine risk factors for mBTTS thrombosis and hypothesize that IV glycoprotein IIb/IIIa inhibitor (tirofiban) as a bridge to oral aspirin reduces the rate of shunt thrombosis in the immediate postoperative period. End points within the 14-day follow-up period include mBTTS thrombosis, overall thrombosis, bleeding, length of stay, and mortality. DESIGN: Retrospective, Institutional Review Board-approved cohort study. SETTING: Single-center cardiac ICU. PATIENTS: Patients under the age of 18 who had an mBTTS placed within the study period of January 2008 to December 2018 were included. INTERVENTIONS: Patients were divided into two groups: standard of care (SOC) anticoagulation alone and SOC with tirofiban as a bridge to oral aspirin. MEASUREMENTS AND MAIN RESULTS: Freedom from mBTTS thrombosis was estimated using the Kaplan-Meier method. A multivariable predictive model using the four most significant risk factors was developed using logistic regression. A total of 272 patients were included: 36 subjects in the SOC/tirofiban group and 236 in the SOC group. Shunt thrombosis occurred in 26 (11%) SOC group with zero in SOC/tirofiban group ( p = 0.03). The median time to thrombosis was 0 days (range, 0-12 d). The area under the curve for the predictive model (anticoagulation group, history of coagulopathy, intraoperative shunt clipping, and shunt size/weight ratio) is 0.790 ( p < 0.001). Prevalence of bleeding and mortality was not significantly different between the groups. CONCLUSIONS: Highest risk for shunt thrombosis following mBTTS occurs within the first few days after surgical procedure. Tirofiban is a safe addition to SOC and may be an effective strategy to prevent early mBTTS thrombosis.


Subject(s)
Blalock-Taussig Procedure , Fibrinolytic Agents , Integrin alpha2 , Integrin beta3 , Platelet Aggregation Inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex , Thrombosis , Tirofiban , Anticoagulants , Aspirin/therapeutic use , Blalock-Taussig Procedure/adverse effects , Cohort Studies , Fibrinolytic Agents/therapeutic use , Hemorrhage/etiology , Humans , Integrin alpha2/metabolism , Integrin beta3/metabolism , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Retrospective Studies , Thrombosis/etiology , Thrombosis/prevention & control , Tirofiban/therapeutic use , Treatment Outcome
11.
Int J Mol Sci ; 23(2)2022 Jan 06.
Article in English | MEDLINE | ID: mdl-35054807

ABSTRACT

Hemophilia A (HA) is caused by mutations in the coagulation factor VIII (FVIII) gene (F8). Gene therapy is a hopeful cure for HA; however, FVIII inhibitors formation hinders its clinical application. Given that platelets promote coagulation via locally releasing α-granule, FVIII ectopically expressed in platelets has been attempted, with promising results for HA treatment. The B-domain-deleted F8 (BDDF8), driven by a truncated ITGA2B promoter, was targeted at the ribosomal DNA (rDNA) locus of HA patient-specific induced pluripotent stem cells (HA-iPSCs). The F8-modified, human induced pluripotent stem cells (2bF8-iPSCs) were differentiated into induced hematopoietic progenitor cells (iHPCs), induced megakaryocytes (iMKs), and mesenchymal stem cells (iMSCs), and the FVIII expression was detected. The ITGA2B promoter-driven BDDF8 was site-specifically integrated into the rDNA locus of HA-iPSCs. The 2bF8-iPSCs were efficiently differentiated into 2bF8-iHPCs, 2bF8-iMKs, and 2bF8-iMSCs. FVIII was 10.31 ng/106 cells in lysates of 2bF8-iHPCs, compared to 1.56 ng/106 cells in HA-iHPCs, and FVIII was 3.64 ng/106 cells in 2bF8-iMSCs lysates, while 1.31 ng/106 cells in iMSCs with CMV-driven BDDF8. Our results demonstrated a high expression of FVIII in iHPCs and iMSCs derived from hiPSCs with site-specific integration of ITGA2B promoter-driven BDDF8, indicating potential clinical prospects of this platelet-targeted strategy for HA gene therapy.


Subject(s)
Ectopic Gene Expression , Factor VIII/genetics , Hematopoietic Stem Cells/metabolism , Hemophilia A/genetics , Induced Pluripotent Stem Cells/metabolism , Integrin alpha2/genetics , Mesenchymal Stem Cells/metabolism , Promoter Regions, Genetic , Base Sequence , DNA, Ribosomal/genetics , Factor VIII/chemistry , Factor VIII/metabolism , Gene Targeting , Genetic Loci , Genetic Vectors/metabolism , Humans , Integrin alpha2/metabolism , Megakaryocytes/metabolism , Protein Domains , Sequence Deletion , Transcription Activator-Like Effector Nucleases/metabolism
12.
J Cell Mol Med ; 25(4): 2013-2024, 2021 02.
Article in English | MEDLINE | ID: mdl-33369167

ABSTRACT

Sepsis is a systemic inflammatory response to invading pathogens, leading to high mortality rates in intensive care units worldwide. Krüppel-like factor 4 (KLF4) is an important anti-inflammatory transcription factor. In this study, we investigate the anti-inflammatory role of KLF4 in caecal ligation and puncture (CLP)-induced septic mice and lipopolysaccharide (LPS)-induced RAW264.7 cells and its potential mechanism. We found that KLF4 was down-regulated in CLP-induced septic mice and in LPS-induced RAW264.7 cells, and that its overexpression led to increased survival rates of septic mice along with inhibited inflammatory response in vivo and in vitro. ITGA2B was up-regulated in the setting of sepsis and was inhibited by KLF4 overexpression. ITGA2B knock-down mimicked the effects of KLF4 overexpression on septic mice and LPS-induced RAW264.7 cells. TLR4 promoted the phosphorylation of ERK1/2 and then up-regulated the ubiquitination and the degradation of KLF4, thereby elevating the expression of ITGA2B. Moreover, TLR4 knock-down or treatment with PD98059 (a MEK inhibitor) inhibited inflammatory response in the setting of sepsis in vivo and in vitro. Furthermore, this effect of PD98059 treatment was lost upon KLF4 knock-down. Collectively, these results explain the down-regulation of KLF4 in sepsis, namely via TLR4 promotion of ERK1/2 phosphorylation, and identify ITGA2B as the downstream gene of KLF4, thus highlighting the anti-inflammatory role of KLF4 in sepsis.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Sepsis/etiology , Sepsis/metabolism , Toll-Like Receptor 4/metabolism , Animals , Biomarkers , Disease Models, Animal , Gene Expression , Integrin alpha2/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Lipopolysaccharides/adverse effects , Male , Mice , Models, Biological , Phosphorylation , RAW 264.7 Cells , Sepsis/pathology
13.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G420-G438, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33470153

ABSTRACT

Intestinal epithelia are critical for maintaining gastrointestinal homeostasis. Epithelial barrier injury, causing inflammation and vascular damage, results in inflammatory hypoxia, and thus, healing occurs in an oxygen-restricted environment. The transcription factor hypoxia-inducible factor (HIF)-1 regulates genes important for cell survival and repair, including the cell adhesion protein ß1-integrin. Integrins function as αß-dimers, and α-integrin-matrix binding is critical for cell migration. We hypothesized that HIF-1 stabilization accelerates epithelial migration through integrin-dependent pathways. We aimed to examine functional and posttranslational activity of α-integrins during HIF-1-mediated intestinal epithelial healing. Wound healing was assessed in T84 monolayers over 24 h with/without prolyl-hydroxylase inhibitor (PHDi) (GB-004), which stabilizes HIF-1. Gene and protein expression were measured by RT-PCR and immunoblot, and α-integrin localization was assessed by immunofluorescence. α-integrin function was assessed by antibody-mediated blockade, and integrin α6 regulation was determined by HIF-1α chromatin immunoprecipitation. Models of mucosal wounding and 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis were used to examine integrin expression and localization in vivo. PHDi treatment accelerated wound closure and migration within 12 h, associated with increased integrin α2 and α6 protein, but not α3. Functional blockade of integrins α2 and α6 inhibited PHDi-mediated accelerated wound closure. HIF-1 bound directly to the integrin α6 promoter. PHDi treatment accelerated mucosal healing, which was associated with increased α6 immunohistochemical staining in wound-associated epithelium and wound-adjacent tissue. PHDi treatment increased α6 protein levels in colonocytes of TNBS mice and induced α6 staining in regenerating crypts and reepithelialized inflammatory lesions. Together, these data demonstrate a role for HIF-1 in regulating both integrin α2 and α6 responses during intestinal epithelial healing.NEW & NOTEWORTHY HIF-1 plays an important role in epithelial restitution, selectively inducing integrins α6 and α2 to promote migration and proliferation, respectively. HIF-stabilizing prolyl-hydroxylase inhibitors accelerate intestinal mucosal healing by inducing epithelial integrin expression.


Subject(s)
Colitis/prevention & control , Colon/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Integrin alpha Chains/metabolism , Intestinal Mucosa/drug effects , Prolyl-Hydroxylase Inhibitors/pharmacology , Wound Healing/drug effects , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Integrin alpha Chains/genetics , Integrin alpha2/metabolism , Integrin alpha6/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice, Inbred BALB C , Protein Stability , Signal Transduction , Trinitrobenzenesulfonic Acid
14.
Development ; 145(16)2018 04 16.
Article in English | MEDLINE | ID: mdl-29540503

ABSTRACT

During pregnancy the trophoblast cells of the placenta are the only fetal cells in direct contact with maternal blood and decidua. Their functions include transport of nutrients and oxygen, secretion of pregnancy hormones, remodelling of the uterine arteries, and communicating with maternal cells. Despite the importance of trophoblast cells in placental development and successful pregnancy, little is known about the identity, location and differentiation of human trophoblast progenitors. We identify a proliferative trophoblast niche at the base of the cytotrophoblast cell columns in first trimester placentas that is characterised by integrin α2 (ITGA2) expression. Pulse-chase experiments with 5-iodo-2'-deoxyuridine indicate that these cells might contribute to both villous (VCT) and extravillous (EVT) lineages. These proliferating trophoblast cells can be isolated by flow cytometry using ITGA2 as a marker and express genes from both VCT and EVT. Microarray expression analysis shows that ITAG2+ cells display a unique transcriptional signature, including genes involved in NOTCH signalling, and exhibit a combination of epithelial and mesenchymal characteristics. ITGA2 thus marks a niche allowing the study of pure populations of trophoblast progenitor cells.


Subject(s)
Integrin alpha2/metabolism , Placenta/cytology , Placentation/physiology , Receptor, Notch1/metabolism , Stem Cells/cytology , Trophoblasts/cytology , Biomarkers/metabolism , Cell Proliferation , Female , Humans , Placenta/metabolism , Pregnancy , Pregnancy Trimester, First , Signal Transduction
15.
BMC Cancer ; 21(1): 571, 2021 May 18.
Article in English | MEDLINE | ID: mdl-34006252

ABSTRACT

BACKGROUND: The mechanism of small-molecule stabilised protein-protein interactions is of growing interest in the pharmacological discovery process. A plethora of different substances including the aromatic sulphonamide E7820 have been identified to act by such a mechanism. The process of E7820 induced CAPERα degradation and the resultant transcriptional down regulation of integrin α2 expression has previously been described for a variety of different cell lines and been made responsible for E7820's antiangiogenic activity. Currently the application of E7820 in the treatment of various malignancies including pancreas carcinoma and breast cancer is being investigated in pre-clinical and clinical trials. It has been shown, that integrin α2 deficiency has beneficial effects on bone homeostasis in mice. To transfer E7820 treatment to bone-related pathologies, as non-healing fractures, osteoporosis and bone cancer might therefore be beneficial. However, at present no data is available on the effect of E7820 on osseous cells or skeletal malignancies. METHODS: Pre-osteoblastic (MC3T3 and Saos-2) cells and endothelial (eEnd2 cells and HUVECs) cells, each of human and murine origin respectively, were investigated. Vitality assay with different concentrations of E7820 were performed. All consecutive experiments were done at a final concentration of 50 ng/ml E7820. The expression and production of integrin α2 and CAPERα were investigated by quantitative real-time PCR and western blotting. Expression of CAPERα splice forms was differentiated by semi-quantitiative reverse transcriptase PCR. RESULTS: Here we present the first data showing that E7820 can increase integrin α2 expression in the pre-osteoblast MC3T3 cell line whilst also reproducing canonical E7820 activity in HUVECs. We show that the aberrant activity of E7820 in MC3T3 cells is likely due to differential activity of CAPERα at the integrin α2 promoter, rather than due to differential CAPERα degradation or differential expression of CAPERα spliceforms. CONCLUSION: The results presented here indicate that E7820 may not be suitable to treat certain malignancies of musculoskeletal origin, due to the increase in integrin α2 expression it may induce. Further investigation of the differential functioning of CAPERα and the integrin α2 promoter in cells of various origin would however be necessary to more clearly differentiate between cell lines that will positively respond to E7820 from those that will not.


Subject(s)
Indoles/pharmacology , Integrin alpha2/genetics , RNA-Binding Proteins/antagonists & inhibitors , Sulfonamides/pharmacology , Trans-Activators/antagonists & inhibitors , Animals , Cell Line , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Integrin alpha2/metabolism , Ligands , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Promoter Regions, Genetic/genetics , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteolysis/drug effects , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Up-Regulation/drug effects
16.
Arterioscler Thromb Vasc Biol ; 40(3): 624-637, 2020 03.
Article in English | MEDLINE | ID: mdl-31969014

ABSTRACT

OBJECTIVE: The αIIbß3 antagonist antiplatelet drug abciximab is the chimeric antigen-binding fragment comprising the variable regions of murine monoclonal antibody 7E3 and the constant domains of human IgG1 and light chain κ. Previous mutagenesis studies suggested that abciximab binds to the ß3 C177-C184 specificity-determining loop (SDL) and Trp129 on the adjacent ß1-α1 helix. These studies could not, however, assess whether 7E3 or abciximab prevents fibrinogen binding by steric interference, disruption of either the αIIbß3-binding pocket for fibrinogen or the ß3 SDL (which is not part of the binding pocket but affects fibrinogen binding), or some combination of these effects. To address this gap, we used cryo-electron microscopy to determine the structure of the αIIbß3-abciximab complex at 2.8 Å resolution. Approach and Results: The interacting surface of abciximab is comprised of residues from all 3 complementarity-determining regions of both the light and heavy chains, with high representation of aromatic residues. Binding is primarily to the ß3 SDL and neighboring residues, the ß1-α1 helix, and ß3 residues Ser211, Val212 and Met335. Unexpectedly, the structure also indicated several interactions with αIIb. As judged by the cryo-electron microscopy model, molecular-dynamics simulations, and mutagenesis, the binding of abciximab does not appear to rely on the interaction with the αIIb residues and does not result in disruption of the fibrinogen-binding pocket; it does, however, compress and reduce the flexibility of the SDL. CONCLUSIONS: We deduce that abciximab prevents ligand binding by steric interference, with a potential contribution via displacement of the SDL and limitation of the flexibility of the SDL residues.


Subject(s)
Abciximab/ultrastructure , Cryoelectron Microscopy , Integrin alpha2/ultrastructure , Integrin beta3/ultrastructure , Platelet Aggregation Inhibitors , Abciximab/metabolism , Binding Sites , Binding, Competitive , HEK293 Cells , Humans , Integrin alpha2/genetics , Integrin alpha2/metabolism , Integrin beta3/genetics , Integrin beta3/metabolism , Ligands , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Mutation , Platelet Aggregation Inhibitors/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins/ultrastructure , Structure-Activity Relationship
17.
Exp Cell Res ; 390(2): 111960, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32194036

ABSTRACT

Human leukocyte antigen (HLA) class I molecules present antigenic peptides to cytotoxic T cells, causing lysis of malignant cells. Transplantation biology studies have implicated HLA class I molecules in cell migration, but there has been little evidence presented that they influence cancer cell migration, a contributing factor in metastasis. In this study, we examined the effect of HLA-B on pancreatic cancer cell migration. HLA-B siRNA transfection increased the migration of the S2-013 pancreatic cancer cells but, in contrast, reduced migration of the PANC-1 and MIA PaCa-2 pancreatic cancer cell lines. Integrin molecules have previously been implicated in the upregulation of pancreatic cancer cell migration, and knockdown of HLA-B in S2-013 cells heightened the expression of integrin beta 1 (ITGB1), but in the PANC-1 and MIA PaCa-2 cells HLA-B knockdown diminished ITGB1 expression. A transmembrane sequence in an S2-013 HLA-B heavy chain matches a corresponding sequence in HLA-B in the BxPC-3 pancreatic cancer cell line, and knockdown of BxPC-3 HLA-B mimics the effect of S2-013 HLA-B knockdown on migration. In total, our findings indicate that HLA-B influences the expression of ITGB1 in pancreatic cancer cells, with concurrent distinctions in transmembrane sequences and effects on the migration of the cells.


Subject(s)
Cell Movement/genetics , Gene Expression Regulation, Neoplastic , HLA-B Antigens/genetics , Integrin beta1/genetics , Pancreas/metabolism , Cell Line, Tumor , Cell Proliferation , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , HLA-B Antigens/metabolism , Humans , Integrin alpha2/genetics , Integrin alpha2/metabolism , Integrin beta1/metabolism , Organ Specificity , Pancreas/pathology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
18.
Platelets ; 32(2): 238-242, 2021 Feb 17.
Article in English | MEDLINE | ID: mdl-32089034

ABSTRACT

Glanzmann's thrombasthenia (GT) is an autosomal recessive disorder in which the underlying problem is the lack or dysfunction of the GpIIb/IIIa receptor on the platelet surface. The present study determines the genetic mutation typology and analyzes the association between mutation types and clinical findings in patients diagnosed with GT who were followed up in Department of Pediatric Hematology of the Yüzüncü Yil University School of Medicine. The medical charts of 17 patients who underwent therapy and that were followed up in the Department of Pediatric Hematology of the Yüzüncü Yil University Dursun Odabas Medical Center between January 2008 and April 2018 were reviewed retrospectively. Data on such patient characteristics as present genetic mutations, age, gender, age at diagnosis, presenting symptoms, clinical findings, and platelet count and volume were garnered from the patient records. Of the 12 patients identified with genetic mutations, six had the same type of mutation, while four were identified with novel mutations that have to date not been defined in literature. Of these four mutations, three were located in the ITGA2B gene and one in the ITGB3 gene. The present study identified no significant association between the genetic and clinical findings of the patients. Novel mutations were identified in four cases in the present study. No association was found between the mutation type, and the bleeding scores and bleeding phenotypes of the patients. Further studies involving a larger number of patients are required to determine the relationship between the genotypes and clinical findings in patients with GT.


Subject(s)
Integrin alpha2/metabolism , Integrin beta3/metabolism , Thrombasthenia/genetics , Child , Child, Preschool , Female , Humans , Infant , Male , Mutation , Thrombasthenia/pathology , Turkey
19.
Acta Biochim Biophys Sin (Shanghai) ; 53(12): 1650-1661, 2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34687203

ABSTRACT

Papillary thyroid cancer (PTC) usually has favorable prognosis; however, distant metastasis is a leading cause of death associated with PTC. MicroRNA-99a-3p (miR-99a-3p) is a member of the miR-99 family that is shown to be a tumor suppressor in various human cancers including the anaplastic thyroid cancer, another type of thyroid cancer. The Cancer Genome Atlas database and our previous study reported that miR-99a-3p is downregulated in human PTC tissues as well as human papillary thyroid carcinoma B-CPAP and TPC-1 cell lines. However, its pathological role in PTC remains unclear, especially its impact on PTC metastasis. In the present study, the role of miR-99a-3p in PTC metastasis was molecularly evaluated in in vitro and in vivo models. Our functional study revealed that overexpressing miR-99a-3p significantly suppresses epithelial-mesenchymal transition (EMT) and anoikis resistance as well as migration and invasion of B-CPAP and TPC-1 cells. The mechanical study indicated that glucose-regulated protein 94 (GRP94) is the direct target of miR-99a-3p. Moreover, GRP94 overexpression reverses the inhibitory effect of miR-99a-3p on PTC metastasis. In addition, the miR-99a-3p/GRP94 axis exerts its effect via inhibiting the expression and cytoplasmic relocation of integrin 2α (ITGA2). Furthermore, in vivo experiments confirmed that miR-99a-3p significantly inhibits tumor growth and lung metastasis in PTC xenograft mice. Overall, our findings suggested that the miR-99a-3p/GRP94/ITGA2 axis may be a novel therapeutic target for the prevention of PTC metastasis.


Subject(s)
Integrin alpha2/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Animals , Anoikis/genetics , Cell Line, Tumor , Cell Movement/genetics , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Female , Heterografts/metabolism , Humans , Mice, Nude , Neoplasm Metastasis/genetics , Thyroid Cancer, Papillary/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
20.
Int J Mol Sci ; 22(7)2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33807403

ABSTRACT

Platelets play a crucial role in the physiology of primary hemostasis and pathological processes such as arterial thrombosis; thus, developing a therapeutic target that prevents platelet activation can reduce arterial thrombosis. Pterostilbene (PTE) has remarkable pharmacological activities, including anticancer and neuroprotection. Few studies have reported the effects of pterostilbene on platelet activation. Thus, we examined the inhibitory mechanisms of pterostilbene in human platelets and its role in vascular thrombosis prevention in mice. At low concentrations (2-8 µM), pterostilbene strongly inhibited collagen-induced platelet aggregation. Furthermore, pterostilbene markedly diminished Lyn, Fyn, and Syk phosphorylation and hydroxyl radical formation stimulated by collagen. Moreover, PTE directly hindered integrin αIIbß3 activation through interfering with PAC-1 binding stimulated by collagen. In addition, pterostilbene affected integrin αIIbß3-mediated outside-in signaling, such as integrin ß3, Src, and FAK phosphorylation, and reduced the number of adherent platelets and the single platelet spreading area on immobilized fibrinogen as well as thrombin-stimulated fibrin clot retraction. Furthermore, pterostilbene substantially prolonged the occlusion time of thrombotic platelet plug formation in mice. This study demonstrated that pterostilbene exhibits a strong activity against platelet activation through the inhibition of integrin αIIbß3-mediated inside-out and outside-in signaling, suggesting that pterostilbene can serve as a therapeutic agent for thromboembolic disorders.


Subject(s)
Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Stilbenes/metabolism , Animals , Blood Coagulation/drug effects , Blood Platelets/drug effects , Clot Retraction/drug effects , Collagen , Fibrinogen/metabolism , Hemostasis/drug effects , Humans , Integrin alpha2/drug effects , Integrin alpha2/metabolism , Integrin beta3/drug effects , Integrin beta3/metabolism , Integrins/drug effects , Integrins/metabolism , Mice , P-Selectin/metabolism , Phosphorylation , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Signal Transduction/drug effects , Stilbenes/pharmacology , Thrombosis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL