Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 795
Filter
1.
J Agric Food Chem ; 72(13): 7043-7054, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38509000

ABSTRACT

14-3-3ζ protein, the key target in the regulation and control of integrin ß3 outside-in signaling, is an attractive new strategy to inhibit thrombosis without affecting hemostasis. In this study, 4'-O-methylbavachalconeB (4-O-MB) in Psoraleae Fructus was identified as a 14-3-3ζ ligand with antithrombosis activity by target fishing combined with ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS) analysis. The competitive inhibition analysis showed that 4-O-MB targeted 14-3-3ζ and blocked the 14-3-3ζ/integrin ß3 interaction with inhibition constant (Ki) values of 9.98 ± 0.22 µM. Molecular docking and amino acid mutation experiments confirmed that 4-O-MB specifically bound to 14-3-3ζ through LSY9 and SER28 to regulate the 14-3-3ζ/integrin ß3 interaction. Besides, 4-O-MB affected the integrin ß3 early outside-in signal by inhibiting AKT and c-Src phosphorylation. Meanwhile, 4-O-MB could inhibit ADP-, collagen-, or thrombin-induced platelet aggregation function but had no effect on platelet adhesion to collagen-coated surfaces in vivo. Administration of 4-O-MB could significantly inhibit thrombosis formation without disturbing hemostasis in mice. These findings provide new prospects for the antithrombotic effects of Psoraleae Fructus and the potential application of 4-O-MB as lead compounds in the therapy of thrombosis by targeting 14-3-3ζ.


Subject(s)
Platelet Aggregation , Thrombosis , Mice , Animals , Integrin beta3/genetics , Integrin beta3/chemistry , Integrin beta3/metabolism , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/pharmacology , Molecular Docking Simulation , Thrombosis/drug therapy , Thrombosis/genetics , Thrombosis/metabolism , Collagen/metabolism , Blood Platelets/metabolism
2.
Endocrinology ; 165(3)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38195194

ABSTRACT

BACKGROUND: Repeated implantation failure (RIF) leads to a waste of high-quality embryos and remains a challenge in assisted reproductive technology. During early human placentation, the invasion of trophoblast cells into the decidua is an essential step for the establishment of maternal-fetal interactions and subsequent successful pregnancy. Bone morphogenetic protein 2 (BMP2) has been reported to regulate endometrial receptivity and promote trophoblast invasion. However, whether there is dysregulation of endometrial BMP2 expression in patients with RIF remains unknown. Additionally, the molecular mechanisms underlying the effects of BMP2 on human trophoblast invasion and early placentation remain to be further elucidated. METHODS: Midluteal phase endometrial samples were biopsied from patients with RIF and from routine control in vitro fertilization followed by quantitative polymerase chain reaction and immunoblotting analyses. Human trophoblast organoids, primary human trophoblast cells, and an immortalized trophoblast cell line (HTR8/SVneo) were used as study models. RESULTS: We found that BMP2 was aberrantly low in midluteal phase endometrial tissues from patients with RIF. Recombinant human BMP2 treatment upregulated integrin ß3 (ITGB3) in a SMAD2/3-SMAD4 signaling-dependent manner in both HTR8/SVneo cells and primary trophoblast cells. siRNA-mediated integrin ß3 downregulation reduced both basal and BMP2-upregulated trophoblast invasion and vascular mimicry in HTR8/SVneo cells. Importantly, shRNA-mediated ITGB3 knockdown significantly decreased the formation ability of human trophoblast organoids. CONCLUSION: Our results demonstrate endometrial BMP2 deficiency in patients with RIF. ITGB3 mediates both basal and BMP2-promoted human trophoblast invasion and is essential for early placentation. These findings broaden our knowledge regarding the regulation of early placentation and provide candidate diagnostic and therapeutic targets for RIF clinical management.


Subject(s)
Bone Morphogenetic Protein 2 , Integrin beta3 , Pregnancy , Humans , Female , Integrin beta3/genetics , Integrin beta3/metabolism , Bone Morphogenetic Protein 2/metabolism , Trophoblasts/metabolism , Cell Line , Placentation/physiology , RNA, Small Interfering/metabolism , Cell Movement
3.
Cell Biol Int ; 48(2): 216-228, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38081783

ABSTRACT

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer (BC) compared to other BC subtypes in clinical settings. Currently, there are no effective therapeutic strategies for TNBC treatment. Therefore, there is an urgent need to identify suitable biomarkers or therapeutic targets for TNBC patients. Thrombomodulin (TM) plays a role in cancer progression and metastasis in many different cancers. However, the role of TM in TNBC is not yet fully understood. First, silenced-TM in MDA-MB-231 cells caused an increase in proliferative and metastatic activity. In contrast, overexpression of TM in Hs578T cells caused a reduction in proliferation, invasion, and migration rate. Using RNA-seq analysis, we found that Integrin beta 3 (ITGB3) expression may be a downstream target of TM. Furthermore, we found an increase in ITGB3 levels in TM-KD cells by QPCR and western blot analysis but a decrease in ITGB3 levels in TM-overexpressing cells. We found phospho-smad2/3 levels were increased in TM-KD cells but decreased in TM-overexpressing cells. This implies that TM negatively regulates ITGB3 levels through the activation of the smad2/3 pathway. Silencing ITGB3 in TM-KD cells caused a decrease in proliferation and migration. Finally, we found that higher ITGB3 levels were correlated with poor overall survival and relapse-free survival in patients with TNBC. Our results indicated a novel regulatory relationship between TM and ITGB3 in TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Blotting, Western , Cell Line, Tumor , Cell Movement , Cell Proliferation , Integrin beta3/genetics , Thrombomodulin/genetics , Triple Negative Breast Neoplasms/metabolism
5.
Gene ; 888: 147805, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-37716584

ABSTRACT

BACKGROUND/AIM: Recently, it was reported that the non-synonymous c.1431C > T (p. G477=) mutation of the integrin subunit ß3 (ITGB3) gene is the cause of Glanzmann's thrombasthenia (GT). However, the functional consequences of this mutation on the ITGB3 gene and protein expression remain to be elucidated. Therefore, this study was conducted to cover this scientific shortage. METHODS: Peripheral blood samples were collected from Chinese family members (parents and proband and his sister), and DNA was extracted and sequenced using whole-exome and Sanger sequencing. The effect of c.1431C > T mutation on the splicing of mRNA was verified by the in vitro minigene assay and the three variants that resulted from the mutation were cloned into a phage vector and pEGFP-C1 vector, and ITGB3 gene and protein expression was detected in the transfected 293 T cells using qPCR and Western blotting. RESULTS: Minigene splicing assay showed that c.1431C > T mutation causes three kinds of alternative splicing; (1) a 95 bp deletion in the middle of exon10, (2) a 155 bp deletion (95 bp deletion in the middle of exon10 plus a 60 bp deletion in the right side of exon10), and (3) a 261 bp deletion in the right side of exon10. The in vitro expression assay showed that the c.1431C > T variant did not affect the ITGB3 mRNA levels, but directly led to protein truncation and declined expression. CONCLUSION: Due to its significant impact on protein expression, c.1431C > T mutation in ITGB3 could be considered a pathogenic variant of GT. This could enrich the ITGB3 mutation spectrum and provide a base for the genetic diagnosis of GT.


Subject(s)
Thrombasthenia , Humans , Thrombasthenia/genetics , Thrombasthenia/diagnosis , Mutation , RNA Splicing , Base Sequence , RNA, Messenger/genetics , Integrin beta3/genetics
6.
J Thromb Haemost ; 21(12): 3597-3607, 2023 12.
Article in English | MEDLINE | ID: mdl-37604334

ABSTRACT

BACKGROUND: Glanzmann thrombasthenia (GT) is an autosomal recessive platelet aggregation disorder caused by mutations in ITGA2B or ITGB3. OBJECTIVES: We aimed to assess the phenotype and investigate the genetic etiology of a GT pedigree. METHODS: A patient with bleeding manifestations and mild mental retardation was enrolled. Complete blood count, coagulation, and platelet aggregation tests were performed. Causal mutations were identified via whole exome and genome sequencing and subsequently confirmed through polymerase chain reaction and Sanger sequencing. The transcription of ITGB3 was characterized using RNA sequencing and reverse transcription polymerase chain reaction. The αⅡb and ß3 biosynthesis was investigated via whole blood flow cytometry and in vitro studies. RESULTS: GT was diagnosed in a patient with defective platelet aggregation. Novel compound heterozygous ITGB3 variants were identified, with a maternal nonsense mutation (c.2222G>A, p.Trp741∗) and a paternal SINE-VNTR-Alu (SVA) retrotransposon insertion. The 5' truncated SVA element was inserted in a sense orientation in intron 11 of ITGB3, resulting in aberrant splicing of ITGB3 and significantly reducing ß3 protein content. Meanwhile, both the expression and transportation of ß3 were damaged by the ITGB3 c.2222G>A. Almost no αⅡb and ß3 expressions were detected on the patient's platelets surface. CONCLUSION: Novel compound heterozygous ITGB3 mutations were identified in the GT pedigree, resulting in defects of αⅡbß3 biosynthesis. This is the first report of SVA retrotransposon insertion in the genetic pathogenesis of GT. Our study highlights the importance of combining multiple high-throughput sequencing technologies for the molecular diagnosis of genetic disorders.


Subject(s)
Thrombasthenia , Humans , Thrombasthenia/diagnosis , Thrombasthenia/genetics , Retroelements , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Phenotype , Blood Platelets/metabolism , Integrin beta3/genetics , Integrin beta3/metabolism
8.
Mol Brain ; 16(1): 49, 2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37296444

ABSTRACT

The relationship between autism spectrum disorder (ASD) and dendritic spine abnormalities is well known, but it is unclear whether the deficits relate to specific neuron types and brain regions most relevant to ASD. Recent genetic studies have identified a convergence of ASD risk genes in deep layer pyramidal neurons of the prefrontal cortex. Here, we use retrograde recombinant adeno-associated viruses to label specifically two major layer V pyramidal neuron types of the medial prefrontal cortex: the commissural neurons, which put the two cerebral hemispheres in direct communication, and the corticopontine neurons, which transmit information outside the cortex. We compare the basal dendritic spines on commissural and corticopontine neurons in WT and KO mice for the ASD risk gene Itgb3, which encodes for the cell adhesion molecule ß3 integrin selectively enriched in layer V pyramidal neurons. Regardless of the genotype, corticopontine neurons had a higher ratio of stubby to mushroom spines than commissural neurons. ß3 integrin affected selectively spine length in corticopontine neurons. Ablation of ß3 integrin resulted in corticopontine neurons lacking long (> 2 µm) thin dendritic spines. These findings suggest that a deficiency in ß3 integrin expression compromises specifically immature spines on corticopontine neurons, thereby reducing the cortical territory they can sample. Because corticopontine neurons receive extensive local and long-range excitatory inputs before relaying information outside the cortex, specific alterations in dendritic spines of corticopontine neurons may compromise the computational output of the full cortex, thereby contributing to ASD pathophysiology.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Mice , Animals , Dendritic Spines/metabolism , Autistic Disorder/genetics , Autistic Disorder/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Integrin beta3/genetics , Integrin beta3/metabolism , Pyramidal Cells/physiology
9.
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
10.
Front Endocrinol (Lausanne) ; 14: 1110266, 2023.
Article in English | MEDLINE | ID: mdl-37008940

ABSTRACT

Purpose: To investigate the effect of acupuncture for improving the pregnancy rate of COH rats from the viewpoint of regulating the opening time of the implantation window and endometrial receptivity. Methods: Experimental rats were randomly divided into normal group (N), model group (M) and acupuncture group(A), and samples were collected on Day 4, 5 and 6 after mating. COH rats were treated with acupuncture at SP6, LR3, and ST36 once a day for 7 times. The pinopodes were observed under a scanning electron microscope. Serum estrogen and progesterone levels were measured via ELISA. The protein and mRNA levels of estrogen receptor (ER), progesterone receptor (PR), leukemia inhibitory factor (LIF), integrin ß3, vascular endothelial growth factor (VEGF), and fibroblast growth factor 2 (FGF-2) in the endometrium were evaluated via West-blot, immunohistochemistry, and PCR. Results: Compared with group N, the pregnancy rate of group M was significantly decreased (P<0.05), and the abnormal serum hormone levels and implantation window advancement were observed. Compared with group M, the pregnancy rate of group A was significantly increased (P<0.05), the supraphysiological serum progesterone levels were restored to normalcy (P<0.05), and the advanced implantation window was restored to a certain extent. Further, the abnormal ER, PR, LIF, integrin ß3, VEGF, and FGF-2 expression levels of the endometrium got recovered to varying degrees. Conclusion: Acupuncture may restore the estrogen and progesterone balance in COH rats and the forward shift of the implantation window to a certain extent, improving the endometrial receptivity and finally improving the pregnancy rate of COH rats.


Subject(s)
Acupuncture Therapy , Ovarian Hyperstimulation Syndrome , Pregnancy , Humans , Female , Rats , Animals , Progesterone , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Integrin beta3/genetics , Integrin beta3/metabolism , Integrin beta3/pharmacology , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Endometrium , Estrogens/metabolism
11.
Hippocampus ; 33(8): 936-947, 2023 08.
Article in English | MEDLINE | ID: mdl-36967540

ABSTRACT

In excitatory hippocampal pyramidal neurons, integrin ß3 is critical for synaptic maturation and plasticity in vitro. Itgb3 is a potential autism susceptibility gene that regulates dendritic morphology in the cerebral cortex in a cell-specific manner. However, it is unknown what role Itgb3 could have in regulating hippocampal pyramidal dendritic morphology in vivo, a key feature that is aberrant in many forms of autism and intellectual disability. We found that Itgb3 mRNA is expressed in the stratum pyramidale of CA3. We examined the apical dendritic morphology of CA3 hippocampal pyramidal neurons in conditional Itgb3 knockouts and controls, utilizing the Thy1-GFP-M line. We fully reconstructed the apical dendrite of each neuron and determined each neuron's precise location along the dorsoventral, proximodistal, and radial axes of the stratum pyramidale. We found a very strong effect for Itgb3 expression on CA3 apical dendritic morphology: neurons from conditional Itgb3 knockouts had longer and thinner apical dendrites than controls, particularly in higher branch orders. We also assessed potential relationships between pairs of topographic or morphological variables, finding that most variable pairs were free from any linear relationships to each other. We also found that some neurons from controls, but not conditional Itgb3 knockouts, had a graded pattern of overall diameter along the dorsoventral and proximodistal axes of the stratum pyramidale of CA3. Taken together, Itgb3 is essential for constructing normal dendritic morphology in pyramidal neurons throughout CA3.


Subject(s)
Dendrites , Integrin beta3 , Integrin beta3/genetics , Dendrites/physiology , Hippocampus/physiology , Pyramidal Cells/physiology , Neurons
12.
J Med Life ; 16(2): 261-266, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36937472

ABSTRACT

Puberty menorrhagia is one of the urgent problems of modern reproductive medicine. The study aimed to investigate the relationship between polymorphism of the GP IIIa (PLA1/PLA2) gene and improve the diagnosis of puberty menorrhagia in girls with thyroid gland pathology. Ninety-seven girls at puberty age were divided into three groups: group 1 (main) - girls with puberty menorrhagia and thyroid gland pathology (30 individuals), group 2 (comparison) - 40 girls with puberty menorrhagia, group 3 (control) - 27 practically healthy girls. Polymorphism of the GP IIIa (PLA1/PLA2) gene was studied by isolating genomic DNA from peripheral blood leukocytes, followed by amplification with a polymerase chain reaction. Results showed that mutation in the 17th chromosome of q21.32 of the GP IIIa gene occurred in 8.6% of cases among adolescents with menorrhagia, in contrast to the control group, where it was not observed at all. The A1A1-genotype occurred by 11.7% (X2=4.01, p=0.041) more often in adolescents with menorrhagia than in girls with concomitant thyroid gland pathology and by 15.0% (X2=4.54, p=0.033) more often than in the control group. It was also found that the presence of the A1A2-genotype unreliably reduced the chances of uterine bleeding in adolescent girls by 1.45 times (OR=2.12) and was a protective factor in the puberty menorrhagia occurrence (OR=0.47). It may be concluded that the identification of a hereditary factor of the reproductive system diseases of adolescent girls fundamentally changes the point of view on the tactics of disease management and subsequent therapy.


Subject(s)
Integrin beta3 , Menorrhagia , Adolescent , Female , Humans , Genetic Testing , Genotype , Integrin beta3/genetics , Menorrhagia/genetics , Puberty
13.
Curr Med Sci ; 43(1): 48-57, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36680686

ABSTRACT

OBJECTIVE: Cardiac fibroblasts (CFs) proliferation and extracellular matrix deposition are important features of cardiac fibrosis. Various studies have indicated that vitamin D displays an anti-fibrotic property in chronic heart diseases. This study explored the role of vitamin D in the growth of CFs via an integrin signaling pathway. METHODS: MTT and 5-ethynyl-2'-deoxyuridine assays were performed to determine cell viability. Western blotting was performed to detect the expression of proliferating cell nuclear antigen (PCNA) and integrin signaling pathway. The fibronectin was observed by ELISA. Immunohistochemical staining was employed to evaluate the expression of integrin ß3. RESULTS: The PCNA expression in the CFs was enhanced after isoproterenol (ISO) stimulation accompanied by an elevated expression of integrin beta-3 (ß3). The blockade of the integrin ß3 with a specific integrin ß3 antibody reduced the PCNA expression induced by the ISO. Decreasing the integrin ß3 by siRNA reduced the ISO-triggered phosphorylation of FAK and Akt. Both the FAK inhibitor and Akt inhibitor suppressed the PCNA expression induced by the ISO in the CFs. Calcitriol (CAL), an active form of vitamin D, attenuated the ISO-induced CFs proliferation by downregulating the integrin ß3 expression, and phosphorylation of FAK and Akt. Moreover, CAL reduced the increased levels of fibronectin and hydroxyproline in the CFs culture medium triggered by the ISO. The administration of calcitriol decreased the integrin ß3 expression in the ISO-induced myocardial injury model. CONCLUSION: These findings revealed a novel role for CAL in suppressing the CFs growth by the downregulation of the integrin ß3/FAK/Akt pathway.


Subject(s)
Calcitriol , Humans , Calcitriol/metabolism , Calcitriol/pharmacology , Fibronectins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Isoproterenol , Proliferating Cell Nuclear Antigen/metabolism , Integrin beta3/genetics , Integrin beta3/metabolism , Vitamins , Cell Proliferation , Fibroblasts/metabolism
14.
J Neuroinflammation ; 20(1): 5, 2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36609298

ABSTRACT

BACKGROUND: In response to brain injury or inflammation, astrocytes undergo hypertrophy, proliferate, and migrate to the damaged zone. These changes, collectively known as "astrogliosis", initially protect the brain; however, astrogliosis can also cause neuronal dysfunction. Additionally, these astrocytes undergo intracellular changes involving alterations in the expression and localization of many proteins, including αvß3 integrin. Our previous reports indicate that Thy-1, a neuronal glycoprotein, binds to this integrin inducing Connexin43 (Cx43) hemichannel (HC) opening, ATP release, and astrocyte migration. Despite such insight, important links and molecular events leading to astrogliosis remain to be defined. METHODS: Using bioinformatics approaches, we analyzed different Gene Expression Omnibus datasets to identify changes occurring in reactive astrocytes as compared to astrocytes from the normal mouse brain. In silico analysis was validated by both qRT-PCR and immunoblotting using reactive astrocyte cultures from the normal rat brain treated with TNF and from the brain of a hSOD1G93A transgenic mouse model. We evaluated the phosphorylation of Cx43 serine residue 373 (S373) by AKT and ATP release as a functional assay for HC opening. In vivo experiments were also performed with an AKT inhibitor (AKTi). RESULTS: The bioinformatics analysis revealed that genes of the PI3K/AKT signaling pathway were among the most significantly altered in reactive astrocytes. mRNA and protein levels of PI3K, AKT, as well as Cx43, were elevated in reactive astrocytes from normal rats and from hSOD1G93A transgenic mice, as compared to controls. In vitro, reactive astrocytes stimulated with Thy-1 responded by activating AKT, which phosphorylated S373Cx43. Increased pS373Cx43 augmented the release of ATP to the extracellular medium and AKTi inhibited these Thy-1-induced responses. Furthermore, in an in vivo model of inflammation (brain damage), AKTi decreased the levels of astrocyte reactivity markers and S373Cx43 phosphorylation. CONCLUSIONS: Here, we identify changes in the PI3K/AKT molecular signaling network and show how they participate in astrogliosis by regulating the HC protein Cx43. Moreover, because HC opening and ATP release are important in astrocyte reactivity, the phosphorylation of Cx43 by AKT and the associated increase in ATP release identify a potential therapeutic window of opportunity to limit the adverse effects of astrogliosis.


Subject(s)
Brain Injuries , Connexin 43 , Animals , Mice , Rats , Adenosine Triphosphate/pharmacology , Adenosine Triphosphate/metabolism , Astrocytes/metabolism , Brain Injuries/metabolism , Connexin 43/metabolism , Gliosis/metabolism , Inflammation/metabolism , Integrin beta3/genetics , Integrin beta3/metabolism , Integrin beta3/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation , Thy-1 Antigens/metabolism , Integrin alpha5/metabolism
15.
J Drug Target ; 31(3): 269-277, 2023 03.
Article in English | MEDLINE | ID: mdl-36217893

ABSTRACT

Inevitable emergence of acquired resistance to EGFR TKIs including third-generation TKI osimertinib limits their long-term efficacy in treating EGFR-mutant lung cancer. A fuller investigation of novel molecular mechanisms underlying acquired resistance is essential to develop efficacious therapeutic strategies. Consequently, we have identified a novel TGFß1/integrin ß3 loop that contributes to the occurrence of EGFR TKI-acquired resistance. EGFR TKIs dramatically and sustainably increased the expression of both TGFß1 and integrin ß3 in in vitro and in vivo EGFR-mutant lung cancer models with acquired resistance to EGFR TKIs. Previously, we reported that integrin ß3 expression was partially induced by TGFß1 in these models. Moreover, elevated TGFß1 in these models was secreted mostly from lung cancer cells. Mechanistically, TGFß1 was induced and activated by overexpressed integrin ß3, forming a positive feedback loop. More importantly, the interruption of TGFß1/integrin ß3 positive feedback loop was shown to dramatically delay the occurrence of acquired resistance and greatly improve the efficacy of EGFR TKI in treating EGFR-mutant lung cancer. Taken together, our study first demonstrated the TGFß1/integrin ß3 loop a new mechanism and target for acquired EGFR TKI resistance in EGFR-mutant lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Integrin beta3/genetics , Integrin beta3/therapeutic use , Feedback , ErbB Receptors/genetics , ErbB Receptors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation
16.
Int J Biol Sci ; 18(15): 5858-5872, 2022.
Article in English | MEDLINE | ID: mdl-36263165

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a malignancy with high metastatic and invasive nature. Distant metastasis contributes substantially to treatment failure and mortality in NPC. Platelets are versatile blood cells and the number of platelets is positively associated with the distant metastasis of tumor cells. However, the role and underlying mechanism of platelets responsible for the metastasis of NPC cells remain unclear. Here we found that the distant metastasis of NPC patients was positively correlated with the expression levels of integrin ß3 (ITGB3) in platelet-derived extracellular vesicles (EVs) from NPC patients (P-EVs). We further revealed that EVs transfer occurred from platelets to NPC cells, mediating cell-cell communication and inducing the metastasis of NPC cells by upregulating ITGB3 expression. Mechanistically, P-EVs-upregulated ITGB3 increased SLC7A11 expression by enhancing protein stability and activating the MAPK/ERK/ATF4/Nrf2 axis, which suppressed ferroptosis, thereby facilitating the metastasis of NPC cells. NPC xenografts in mouse models further confirmed that P-EVs inhibited the ferroptosis of circulating NPC cells and promoted the distant metastasis of NPC cells. Thus, these findings elucidate a novel role of platelet-derived EVs in NPC metastasis, which not only improves our understanding of platelet-mediated tumor distant metastasis, but also has important implications in diagnosis and treatment of NPC.


Subject(s)
Extracellular Vesicles , Ferroptosis , Nasopharyngeal Neoplasms , Mice , Animals , Humans , Nasopharyngeal Carcinoma/genetics , Integrin beta3/genetics , Integrin beta3/metabolism , NF-E2-Related Factor 2/metabolism , Cell Line, Tumor , Extracellular Vesicles/metabolism , Nasopharyngeal Neoplasms/metabolism , Neoplasm Metastasis/pathology , Gene Expression Regulation, Neoplastic
17.
Cell Death Dis ; 13(9): 810, 2022 09 21.
Article in English | MEDLINE | ID: mdl-36130933

ABSTRACT

Migration and invasion inhibitory protein (MIIP) has been identified as a tumor suppressor in various cancer types. Although MIIP is reported to exert tumor suppressive functions by repressing proliferation and metastasis of cancer cells, the detailed mechanism is poorly understood. In the present study, we found MIIP is a favorable indicator of prognosis in triple-negative breast cancer. MIIP could inhibit tumor angiogenesis, proliferation, and metastasis of triple-negative breast cancer cells in vivo and in vitro. Mechanistically, MIIP directly interacted with ITGB3 and suppressed its downstream signaling. As a result, ß-catenin was reduced due to elevated ubiquitin-mediated degradation, leading to downregulated VEGFA production and epithelial mesenchymal transition. More importantly, we found RGD motif is essential for MIIP binding with ITGB3 and executing efficient tumor-suppressing effect. Our findings unravel a novel mechanism by which MIIP suppresses tumorigenesis in triple-negative breast cancer, and MIIP is thus a promising molecular biomarker or therapeutic target for the disease.


Subject(s)
Triple Negative Breast Neoplasms , beta Catenin , Carcinogenesis/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Epithelial-Mesenchymal Transition/genetics , Humans , Integrin beta3/genetics , Integrin beta3/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Triple Negative Breast Neoplasms/genetics , Ubiquitins/metabolism , beta Catenin/metabolism
18.
J Cell Physiol ; 237(11): 4251-4261, 2022 11.
Article in English | MEDLINE | ID: mdl-36108162

ABSTRACT

Integrins are transmembrane proteins that transmit bi-directional signals across the cell membrane through global structural rearrangement among three different conformational states: bent, extended- closed, and extended-open conformations. However, the ß8 integrin is distinctive and may adopt only one conformation, that is, extended-closed conformation, with high affinity for ligands under physiological conditions, and may not transmit bi-directional signals like other integrin members. It is unclear how different ß8 domains affect its unique conformation and signaling. We swapped different domains of integrin ß3 with those of ß8 and investigated how they affected integrin ligand binding, global conformation, and outside-in signaling. We found that the ß8 epidermal growth factor (EGF) domains increased integrin ligand binding ability and contributed to its extended conformation. By comparison, the ß8 transmembrane and cytoplasmic domains had little effect on ligand binding or global conformation. The ß8 EGF and transmembrane domains did not affect integrin-mediated cell adhesion, cell spreading, focal adhesion formation, or colocalization of integrin with other proteins, but the cytoplasmic domain had a defective effect on outside-in signaling. Our results showed that different domains of ß8 play various roles on its unique conformation, ligand binding, and signaling, which are considered atypical among integrin members.


Subject(s)
Epidermal Growth Factor , Integrins , Integrins/metabolism , Epidermal Growth Factor/metabolism , Ligands , Protein Binding , Protein Domains , Integrin beta3/genetics , Integrin beta3/metabolism
20.
Acta Histochem ; 124(6): 151926, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35777302

ABSTRACT

This study was conducted to investigate the impact of the microRNA (miR)-25-3p/ITGB3 axis on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) from patients with osteoporosis (OP). BMSCs isolated from the bone marrow of healthy controls and OP patients were identified by flow cytometry, in which ITGB3 mRNA and miR-25-3p expression was detected by RT-qPCR and ITGB3, Runx2, OPN, ALP, and OSX protein expression by western blot. The binding between ITGB3 and miR-25-3p was assessed by dual-luciferase reporter gene and Ago2-RIP assays. BMSC osteogenic differentiation was observed by alizarin red staining and ALP activity. The differentiation of BMSCs to adipocytes and chondrocytes was measured by oil red O staining and alcian blue staining, respectively. BMSCs were successfully isolated from the bone marrow of healthy controls (normal-BMSCs) and OP patients (OP-BMSCs). ITGB3, Runx2, OPN, ALP, and OSX expression was poorer and miR-25-3p expression was higher in OP-BMSCs than in normal-BMSCs. Mechanistically, ITGB3 was negatively targeted by miR-25-3p. After osteogenic, adipogenic, and chondrogenic differentiation of BMSCs were successfully induced, adipogenic differentiation increased and osteogenic and chondrogenic differentiation decreased in OP-BMSCs compared with normal-BMSCs. Overexpression of ITGB3 facilitated mineralized nodule formation and elevated ALP activity and Runx2, OPN, and ALP expression in OP-BMSCs. miR-25-3p upregulation diminished mineralized nodule formation, ALP activity, and Runx2, OPN, and ALP expression in OP-BMSCs and normal-BMSCs, which was annulled by additional ITGB3 overexpression. miR-25-3p targets ITGB3, thereby suppressing osteogenic differentiation of BMSCs from OP patients.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Osteoporosis , Cell Differentiation/genetics , Cells, Cultured , Core Binding Factor Alpha 1 Subunit , Humans , Integrin beta3/genetics , Integrin beta3/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Osteogenesis/genetics , Osteoporosis/genetics , Osteoporosis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...