Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.972
Filter
1.
Front Immunol ; 15: 1341745, 2024.
Article in English | MEDLINE | ID: mdl-38765012

ABSTRACT

Individuals with Kabuki syndrome present with immunodeficiency; however, how pathogenic variants in the gene encoding the histone-modifying enzyme lysine methyltransferase 2D (KMT2D) lead to immune alterations remain poorly understood. Following up on our prior report of KMT2D-altered integrin expression in B-cells, we performed targeted analyses of KMT2D's influence on integrin expression in T-cells throughout development (thymocytes through peripheral T-cells) in murine cells with constitutive- and conditional-targeted Kmt2d deletion. Using high-throughput RNA-sequencing and flow cytometry, we reveal decreased expression (both at the transcriptional and translational levels) of a cluster of leukocyte-specific integrins, which perturb aspects of T-cell activation, maturation, adhesion/localization, and effector function. H3K4me3 ChIP-PCR suggests that these evolutionary similar integrins are under direct control of KMT2D. KMT2D loss also alters multiple downstream programming/signaling pathways, including integrin-based localization, which can influence T-cell populations. We further demonstrated that KMT2D deficiency is associated with the accumulation of murine CD8+ single-positive (SP) thymocytes and shifts in both human and murine peripheral T-cell populations, including the reduction of the CD4+ recent thymic emigrant (RTE) population. Together, these data show that the targeted loss of Kmt2d in the T-cell lineage recapitulates several distinct features of Kabuki syndrome-associated immune deficiency and implicates epigenetic mechanisms in the regulation of integrin signaling.


Subject(s)
Integrins , Lymphocyte Activation , Animals , Mice , Integrins/metabolism , Integrins/genetics , Lymphocyte Activation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Mice, Knockout , Vestibular Diseases/genetics , Vestibular Diseases/immunology , Vestibular Diseases/metabolism , Face/abnormalities , Humans , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Mice, Inbred C57BL , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Signal Transduction , Gene Expression Regulation , Abnormalities, Multiple , Hematologic Diseases , Myeloid-Lymphoid Leukemia Protein
2.
Cells ; 13(9)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38727292

ABSTRACT

Integrin α4ß7+ T cells perpetuate tissue injury in chronic inflammatory diseases, yet their role in hepatic fibrosis progression remains poorly understood. Here, we report increased accumulation of α4ß7+ T cells in the liver of people with cirrhosis relative to disease controls. Similarly, hepatic fibrosis in the established mouse model of CCl4-induced liver fibrosis was associated with enrichment of intrahepatic α4ß7+ CD4 and CD8 T cells. Monoclonal antibody (mAb)-mediated blockade of α4ß7 or its ligand mucosal addressin cell adhesion molecule (MAdCAM)-1 attenuated hepatic inflammation and prevented fibrosis progression in CCl4-treated mice. Improvement in liver fibrosis was associated with a significant decrease in the infiltration of α4ß7+ CD4 and CD8 T cells, suggesting that α4ß7/MAdCAM-1 axis regulates both CD4 and CD8 T cell recruitment to the fibrotic liver, and α4ß7+ T cells promote hepatic fibrosis progression. Analysis of hepatic α4ß7+ and α4ß7- CD4 T cells revealed that α4ß7+ CD4 T cells were enriched for markers of activation and proliferation, demonstrating an effector phenotype. The findings suggest that α4ß7+ T cells play a critical role in promoting hepatic fibrosis progression, and mAb-mediated blockade of α4ß7 or MAdCAM-1 represents a promising therapeutic strategy for slowing hepatic fibrosis progression in chronic liver diseases.


Subject(s)
Cell Adhesion Molecules , Disease Progression , Integrins , Liver Cirrhosis , Liver , Mucoproteins , Animals , Liver Cirrhosis/pathology , Liver Cirrhosis/immunology , Liver Cirrhosis/metabolism , Cell Adhesion Molecules/metabolism , Mucoproteins/metabolism , Humans , Mice , Liver/pathology , Liver/metabolism , Integrins/metabolism , Male , Mice, Inbred C57BL , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Inflammation/pathology , CD8-Positive T-Lymphocytes/immunology , Immunoglobulins/metabolism , Disease Models, Animal , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Female , Antibodies, Monoclonal/pharmacology
3.
Front Immunol ; 15: 1387516, 2024.
Article in English | MEDLINE | ID: mdl-38784377

ABSTRACT

Background: It has been well documented that Takayasu arteritis (TAK) and ulcerative colitis (UC) coexist in the same patients. HLA-B*52 characterizes the co-occurrence, which is one of the common genetic features between these two diseases, indicating shared underlying pathologic mechanisms. Anti-integrin αvß6 antibody (Ab) is present in sera of UC patients in a highly specific manner. We investigated if there were any associations between anti-integrin αvß6 Ab and TAK, considering the risk HLA alleles. Methods: A total of 227 Japanese TAK patients were recruited in the current study and their serum samples were subjected to measurement of anti-integrin αvß6 Ab by ELISA. The clinical information, including the co-occurrence of UC, was collected. The HLA allele carrier status was determined by Luminex or genotype imputation. Results: The information about the presence of UC was available for 165 patients, among which eight (4.84%) patients had UC. Anti-integrin αvß6 antibody was identified in 7 out of 8 TAK subjects with UC (87.5%) while only 5 out of 157 (3.18%) TAK subjects without UC had the antibody (OR 121, p=7.46×10-8). A total of 99 out of 218 (45.4%) patients were HLA-B*52 carriers. There was no significant association between the presence of anti-integrin αvß6 Ab and HLA-B*52 carrier status in those without UC (OR 2.01, 95% CI 0.33-12.4, p = 0.189). Conclusions: The prevalence of anti-integrin αvß6 Ab was high in TAK patients with UC, but not in the absence of concomitant UC. The effect of HLA-B*52 on anti-integrin αvß6 Ab production would be minimal.


Subject(s)
Antigens, Neoplasm , Colitis, Ulcerative , Integrins , Takayasu Arteritis , Humans , Colitis, Ulcerative/immunology , Colitis, Ulcerative/genetics , Takayasu Arteritis/immunology , Takayasu Arteritis/genetics , Female , Integrins/immunology , Male , Adult , Middle Aged , Antigens, Neoplasm/immunology , Antigens, Neoplasm/genetics , HLA-B52 Antigen/immunology , HLA-B52 Antigen/genetics , Alleles , Young Adult , Japan/epidemiology , Genotype , Autoantibodies/blood , Autoantibodies/immunology
4.
Commun Biol ; 7(1): 629, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789481

ABSTRACT

Integrin trafficking to and from membrane adhesions is a crucial mechanism that dictates many aspects of a cell's behaviour, including motility, polarisation, and invasion. In endothelial cells (ECs), the intracellular traffic of α5 integrin is regulated by both neuropilin 1 (NRP1) and neuropilin 2 (NRP2), yet the redundancies in function between these co-receptors remain unclear. Moreover, the endocytic complexes that participate in NRP-directed traffic remain poorly annotated. Here we identify an important role for the GTPase-activating protein p120RasGAP in ECs, promoting the recycling of α5 integrin from early endosomes. Mechanistically, p120RasGAP enables transit of endocytosed α5 integrin-NRP1-NRP2 complexes to Rab11+ recycling endosomes, promoting cell polarisation and fibronectin (FN) fibrillogenesis. Silencing of both NRP receptors, or p120RasGAP, resulted in the accumulation of α5 integrin in early endosomes, a loss of α5 integrin from surface adhesions, and attenuated EC polarisation. Endothelial-specific deletion of both NRP1 and NRP2 in the postnatal retina recapitulated our in vitro findings, severely impairing FN fibrillogenesis and polarised sprouting. Our data assign an essential role for p120RasGAP during integrin traffic in ECs and support a hypothesis that NRP receptors co-traffic internalised cargoes. Importantly, we utilise comparative proteomics analyses to isolate a comprehensive map of NRP1-dependent and NRP2-dependent α5 integrin interactions in ECs.


Subject(s)
Endosomes , Endothelial Cells , Fibronectins , Integrin alpha5 , Neuropilin-1 , Neuropilin-2 , Proteomics , p120 GTPase Activating Protein , Neuropilin-1/metabolism , Neuropilin-1/genetics , Humans , Integrin alpha5/metabolism , Integrin alpha5/genetics , Endosomes/metabolism , Proteomics/methods , Neuropilin-2/metabolism , Neuropilin-2/genetics , Animals , Fibronectins/metabolism , Endothelial Cells/metabolism , p120 GTPase Activating Protein/metabolism , p120 GTPase Activating Protein/genetics , Protein Transport , Mice , Human Umbilical Vein Endothelial Cells/metabolism , Integrins
5.
Cell Commun Signal ; 22(1): 265, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741195

ABSTRACT

Cisplatin (CDDP) is a widely used first-line chemotherapeutic drug in various cancers. However, CDDP resistance is frequently observed in cancer patients. Therefore, it is required to evaluate the molecular mechanisms associated with CDDP resistance to improve prognosis among cancer patients. Integrins are critical factors involved in tumor metastasis that regulate cell-matrix and cell-cell interactions. They modulate several cellular mechanisms including proliferation, invasion, angiogenesis, polarity, and chemo resistance. Modification of integrin expression levels can be associated with both tumor progression and inhibition. Integrins are also involved in drug resistance of various solid tumors through modulation of the tumor cell interactions with interstitial matrix and extracellular matrix (ECM). Therefore, in the present review we discussed the role of integrin protein family in regulation of CDDP response in tumor cells. It has been reported that integrins mainly promoted the CDDP resistance through interaction with PI3K/AKT, MAPK, and WNT signaling pathways. They also regulated the CDDP mediated apoptosis in tumor cells. This review paves the way to suggest the integrins as the reliable therapeutic targets to improve CDDP response in tumor cells.


Subject(s)
Cisplatin , Drug Resistance, Neoplasm , Integrins , Neoplasms , Cisplatin/pharmacology , Cisplatin/therapeutic use , Humans , Integrins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Signal Transduction/drug effects
6.
Aging (Albany NY) ; 16(9): 7902-7914, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38709270

ABSTRACT

BACKGROUND: Traditional bandages, gauze, and cotton balls are increasingly insufficient for addressing complex war injuries characterized by severe bleeding and diverse wound conditions. The giant salamander, a species of high medical value, secretes a unique mucus when stimulated, which has potential applications in wound care. MATERIALS: Giant salamander skin mucus gel dressing wrapped with bone marrow mesenchymal stem cells (BMSCs-GSSM-gel) was prepared and validated. Skin wound injury of rabbit and mouse models were established. Hematoxylin and Eosin, Masson's trichrome, and Sirius red staining were performed. The platelet aggregation rate and coagulation items were measured. Transcriptome sequencing was performed to find potential differential expression genes. RESULTS: Preparation and characterization of BMSCs-GSSM-gel were performed, and BMSCs-GSSM-gel particles with a diameter of about 200 nm were obtained. BMSCs-GSSM-gel accelerated wound healing in both rabbit and mouse models. BMSCs-GSSM-gel significantly promoted hemostasis via increasing platelet aggregation rate and fibrinogen, but decreasing activated partial thromboplastin time, thrombin time, and prothrombin time. BMSCs-GSSM-gel treatment significantly impacted several genes associated with cell adhesion, inflammatory response, collagen-containing extracellular matrix, and the positive regulation of cell migration based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Integrin Subunit Beta 4 (ITGB4), Integrin Subunit Alpha 3 (ITGA3), and Laminin Subunit Beta 3 (LAMB3) might be involved in the wound healing process by BMSCs-GSSM-gel. CONCLUSIONS: We proved the BMSCs-GSSM-gel greatly improved the skin wound healing, and it might play a crucial role in the application fields of skin damage repair.


Subject(s)
Mesenchymal Stem Cells , Skin , Wound Healing , Animals , Rabbits , Mesenchymal Stem Cells/metabolism , Skin/injuries , Skin/metabolism , Mice , Mucus/metabolism , Integrins/metabolism , Integrins/genetics , Gels , Mesenchymal Stem Cell Transplantation/methods , Male
7.
Viruses ; 16(5)2024 05 13.
Article in English | MEDLINE | ID: mdl-38793651

ABSTRACT

Numerous human adenovirus (AdV) types are endowed with arginine-glycine-aspartic acid (RGD) sequences that enable them to recognize vitronectin-binding (αv) integrins. These RGD-binding cell receptors mediate AdV entry into host cells, a crucial early step in virus infection. Integrin interactions with adenoviruses not only initiate receptor-mediated endocytosis but also facilitate AdV capsid disassembly, a prerequisite for membrane penetration by AdV protein VI. This review discusses fundamental aspects of AdV-host interactions mediated by integrins. Recent efforts to re-engineer AdV vectors and non-viral nanoparticles to target αv integrins for bioimaging and the eradication of cancer cells will also be discussed.


Subject(s)
Genetic Therapy , Integrins , Virus Internalization , Humans , Genetic Therapy/methods , Integrins/metabolism , Genetic Vectors/genetics , Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Adenoviridae/genetics , Adenoviridae/physiology , Animals , Receptors, Virus/metabolism , Neoplasms/therapy , Neoplasms/virology , Integrin alphaV/metabolism , Integrin alphaV/genetics , Oligopeptides
8.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38749543

ABSTRACT

Phosphatidylcholine (PC) is the major membrane phospholipid in most eukaryotic cells. Bi-allelic loss of function variants in CHKB, encoding the first step in the synthesis of PC, is the cause of a rostrocaudal muscular dystrophy in both humans and mice. Loss of sarcolemma integrity is a hallmark of muscular dystrophies; however, how this occurs in the absence of choline kinase function is not known. We determine that in Chkb -/- mice there is a failure of the α7ß1 integrin complex that is specific to affected muscle. We observed that in Chkb -/- hindlimb muscles there is a decrease in sarcolemma association/abundance of the PI(4,5)P2 binding integrin complex proteins vinculin, and α-actinin, and a decrease in actin association with the sarcolemma. In cells, pharmacological inhibition of choline kinase activity results in internalization of a fluorescent PI(4,5)P2 reporter from discrete plasma membrane clusters at the cell surface membrane to cytosol, this corresponds with a decreased vinculin localization at plasma membrane focal adhesions that was rescued by overexpression of CHKB.


Subject(s)
Choline Kinase , Integrins , Mice, Knockout , Muscular Dystrophies , Sarcolemma , Vinculin , Animals , Mice , Vinculin/metabolism , Vinculin/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/genetics , Integrins/metabolism , Choline Kinase/metabolism , Choline Kinase/genetics , Sarcolemma/metabolism , Humans , Focal Adhesions/metabolism , Cell Membrane/metabolism , Actinin/metabolism , Actinin/genetics , Muscle, Skeletal/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Actins/metabolism , Disease Models, Animal
9.
Int J Med Sci ; 21(7): 1307-1320, 2024.
Article in English | MEDLINE | ID: mdl-38818471

ABSTRACT

Transforming growth factor-ß (TGF-ß) is strongly associated with the cell adhesion signaling pathway in cell differentiation, migration, etc. Mechanistically, TGF-ß is secreted in an inactive form and localizes to the extracellular matrix (ECM) via the latent TGF-ß binding protein (LTBP). However, it is the release of mature TGF-ß that is essential for the activation of the TGF-ß signaling pathway. This progress requires specific integrins (one of the main groups of cell adhesion molecules (CAMs)) to recognize and activate the dormant TGF-ß. In addition, TGF-ß regulates cell adhesion ability through modulating CAMs expression. The aberrant activation of the TGF-ß signaling pathway, caused by abnormal expression of key regulatory molecules (such as Smad proteins, certain transcription factors, and non-coding RNAs), promotes tumor invasive and metastasis ability via epithelial-mesenchymal transition (EMT) during the late stages of tumorigenesis. In this paper, we summarize the crosstalk between TGF-ß and cell adhesion signaling pathway in cancer and its underlying molecular mechanisms.


Subject(s)
Cell Adhesion , Epithelial-Mesenchymal Transition , Neoplasms , Signal Transduction , Transforming Growth Factor beta , Humans , Transforming Growth Factor beta/metabolism , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Integrins/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic
10.
Biomed Pharmacother ; 175: 116718, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744221

ABSTRACT

Advanced cholangiocarcinoma (CCA) presents a clinical challenge due to limited treatment options, necessitating exploration of innovative therapeutic approaches. Bispecific T cell engager (BTE)-armed T cell therapy shows promise in hematological and solid malignancies, offering potential advantages in safety over continuous BTE infusion. In this context, we developed a novel BTE, targeting CD3 on T cells and integrin αvß6, an antigen elevated in various epithelial malignancies, on cancer cells. The novel BTE was generated by fusing an integrin αvß6-binding peptide (A20) to an anti-CD3 (OKT3) single-chain variable fragment (scFv) through a G4S peptide linker (A20/αCD3 BTE). T cells were then armed with A20/αCD3 BTE (A20/αCD3-armed T cells) and assessed for antitumor activity. Our results highlight the specific binding of A20/αCD3 BTE to CD3 on T cells and integrin αvß6 on target cells, effectively redirecting T cells towards these targets. After co-culture, A20/αCD3-armed T cells exhibited significantly heightened cytotoxicity against integrin αvß6-expressing target cells compared to unarmed T cells in both KKU-213A cells and A375.ß6 cells. Moreover, in a five-day co-culture, A20/αCD3-armed T cells demonstrated superior cytotoxicity against KKU-213A spheroids compared to unarmed T cells. Importantly, A20/αCD3-armed T cells exhibited an increased proportion of the effector memory T cell (Tem) subset, upregulation of T cell activation markers, enhanced T cell proliferation, and increased cytolytic molecule/cytokine production, when compared to unarmed T cells in an integrin αvß6-dependent manner. These findings support the potential of A20/αCD3-armed T cells as a novel therapeutic approach for integrin αvß6-expressing cancers.


Subject(s)
Antigens, Neoplasm , Bile Duct Neoplasms , Cholangiocarcinoma , Integrins , T-Lymphocytes , Humans , Cholangiocarcinoma/immunology , Cholangiocarcinoma/therapy , Cholangiocarcinoma/pathology , Antigens, Neoplasm/immunology , T-Lymphocytes/immunology , Integrins/metabolism , Cell Line, Tumor , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/therapy , CD3 Complex/immunology , Single-Chain Antibodies/pharmacology , Coculture Techniques , Antibodies, Bispecific/pharmacology
11.
PLoS Comput Biol ; 20(5): e1012140, 2024 May.
Article in English | MEDLINE | ID: mdl-38768266

ABSTRACT

Apical-basal polarization in renal epithelial cells is crucial to renal function and an important trigger for tubule formation in kidney development. Loss of polarity can induce epithelial-to-mesenchymal transition (EMT), which can lead to kidney pathologies. Understanding the relative and combined roles of the involved proteins and their interactions that govern epithelial polarity may provide insights for controlling the process of polarization via chemical or mechanical manipulations in an in vitro or in vivo setting. Here, we developed a computational framework that integrates several known interactions between integrins, Rho-GTPases Rho, Rac and Cdc42, and polarity complexes Par and Scribble, to study their mutual roles in the emergence of polarization. The modeled protein interactions were shown to induce the emergence of polarized distributions of Rho-GTPases, which in turn led to the accumulation of apical and basal polarity complexes Par and Scribble at their respective poles, effectively recapitulating polarization. Our multiparametric sensitivity analysis suggested that polarization depends foremost on the mutual inhibition between Rac and Rho. Next, we used the computational framework to investigate the role of integrins and GTPases in the generation and disruption of polarization. We found that a minimum concentration of integrins is required to catalyze the process of polarization. Furthermore, loss of polarization was found to be only inducible via complete degradation of the Rho-GTPases Rho and Cdc42, suggesting that polarization is fairly stable once it is established. Comparison of our computational predictions against data from in vitro experiments in which we induced EMT in renal epithelial cells while quantifying the relative Rho-GTPase levels, displayed that EMT coincides with a large reduction in the Rho-GTPase Rho. Collectively, these results demonstrate the essential roles of integrins and Rho-GTPases in the establishment and disruption of apical-basal polarity and thereby provide handles for the in vitro or in vivo regulation of polarity.


Subject(s)
Cell Polarity , Epithelial Cells , Integrins , Kidney , rho GTP-Binding Proteins , Cell Polarity/physiology , Integrins/metabolism , Epithelial Cells/metabolism , rho GTP-Binding Proteins/metabolism , Kidney/metabolism , Kidney/cytology , Animals , Computational Biology , Models, Biological , Computer Simulation , Humans , Epithelial-Mesenchymal Transition/physiology
12.
Hepatol Commun ; 8(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38704651

ABSTRACT

BACKGROUND: Alcohol-associated liver disease is a complex disease regulated by genetic and environmental factors such as diet and sex. The combination of high-fat diet and alcohol consumption has synergistic effects on liver disease progression. Female sex hormones are known to protect females from liver disease induced by high-fat diet. In contrast, they promote alcohol-mediated liver injury. We aimed to define the role of female sex hormones on liver disease induced by a combination of high-fat diet and alcohol. METHODS: Wild-type and protein arginine methyltransferase (Prmt)6 knockout female mice were subjected to gonadectomy (ovariectomy, OVX) or sham surgeries and then fed western diet and alcohol in the drinking water. RESULTS: We found that female sex hormones protected mice from western diet/alcohol-induced weight gain, liver steatosis, injury, and fibrosis. Our data suggest that these changes are, in part, mediated by estrogen-mediated induction of arginine methyltransferase PRMT6. Liver proteome changes induced by OVX strongly correlated with changes induced by Prmt6 knockout. Using Prmt6 knockout mice, we confirmed that OVX-mediated weight gain, steatosis, and injury are PRMT6 dependent, while OVX-induced liver fibrosis is PRMT6 independent. Proteomic and gene expression analyses revealed that estrogen signaling suppressed the expression of several components of the integrin pathway, thus reducing integrin-mediated proinflammatory (Tnf, Il6) and profibrotic (Tgfb1, Col1a1) gene expression independent of PRMT6 levels. Integrin signaling inhibition using Arg-Gly-Asp peptides reduced proinflammatory and profibrotic gene expression in mice, suggesting that integrin suppression by estrogen is protective against fibrosis development. CONCLUSIONS: Taken together, estrogen signaling protects mice from liver disease induced by a combination of alcohol and high-fat diet through upregulation of Prmt6 and suppression of integrin signaling.


Subject(s)
Estradiol , Integrins , Mice, Knockout , Protein-Arginine N-Methyltransferases , Signal Transduction , Animals , Mice , Female , Signal Transduction/drug effects , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Integrins/metabolism , Diet, High-Fat/adverse effects , Ovariectomy , Ethanol/adverse effects , Liver Cirrhosis, Alcoholic/metabolism , Liver Cirrhosis, Alcoholic/prevention & control , Liver Cirrhosis, Alcoholic/pathology , Liver/metabolism , Liver/pathology , Liver/drug effects , Mice, Inbred C57BL , Disease Models, Animal
13.
Cells ; 13(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38727316

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a process during which epithelial cells lose epithelial characteristics and gain mesenchymal features. Here, we used several cell models to study migratory activity and redistribution of cell-cell adhesion proteins in cells in different EMT states: EGF-induced EMT of epithelial IAR-20 cells; IAR-6-1 cells with a hybrid epithelial-mesenchymal phenotype; and their more mesenchymal derivatives, IAR-6-1-DNE cells lacking adherens junctions. In migrating cells, the cell-cell adhesion protein α-catenin accumulated at the leading edges along with ArpC2/p34 and α-actinin. Suppression of α-catenin shifted cell morphology from fibroblast-like to discoid and attenuated cell migration. Expression of exogenous α-catenin in MDA-MB-468 cells devoid of α-catenin drastically increased their migratory capabilities. The Y654 phosphorylated form of ß-catenin was detected at integrin adhesion complexes (IACs). Co-immunoprecipitation studies indicated that α-catenin and pY654-ß-catenin were associated with IAC proteins: vinculin, zyxin, and α-actinin. Taken together, these data suggest that in cells undergoing EMT, catenins not participating in assembly of adherens junctions may affect cell migration.


Subject(s)
Actin Cytoskeleton , Cell Movement , Epithelial-Mesenchymal Transition , alpha Catenin , Humans , Actin Cytoskeleton/metabolism , alpha Catenin/metabolism , beta Catenin/metabolism , Vinculin/metabolism , Adherens Junctions/metabolism , Cell Adhesion , Actinin/metabolism , Cell Line, Tumor , Zyxin/metabolism , Phosphorylation , Integrins/metabolism , Animals , Epithelial Cells/metabolism
14.
Theranostics ; 14(7): 3014-3028, 2024.
Article in English | MEDLINE | ID: mdl-38773979

ABSTRACT

Background: Periostin (POSTN) is a critical extracellular matrix protein in various tumor microenvironments. However, the function of POSTN in thyroid cancer progression remains largely unknown. Methods: Postn and Rag1 knock-out mice and orthotopic mouse models were used to determine the role of POSTN on papillary thyroid tumor progression. Immunofluorescence, cell co-culture, fluorescence in situ hybridization, chromatin immunoprecipitation assay, recombinant protein and inhibitor treatment were performed to explore the underlying mechanisms of POSTN-promoted papillary thyroid tumor growth. Results: POSTN is up-regulated in papillary thyroid tumors and negatively correlates with the overall survival of patients with thyroid cancer. Cancer-associated fibroblast (CAF)-derived POSTN promotes papillary thyroid tumor growth in vivo and in vitro. POSTN deficiency in CAFs significantly impairs CAF-promoted papillary thyroid tumor growth. POSTN promotes papillary thyroid tumor cell proliferation and IL-4 expression through integrin-FAK-STAT3 signaling. In turn, tumor cell-derived IL-4 induces the activation of CAFs and stimulates POSTN expression by activating STAT6. We reveal the crucial role of CAF-derived POSTN and tumor cell-derived IL-4 in driving the development of papillary thyroid tumors through the POSTN-integrin-FAK-STAT3-IL-4 pathway in tumor cells and IL-4-STAT6-POSTN signaling in CAFs. Conclusion: Our findings underscore the significance of POSTN and IL-4 as critical molecular mediators in the dynamic interplay between CAFs and tumor cells, ultimately supporting the growth of papillary thyroid tumors.


Subject(s)
Cancer-Associated Fibroblasts , Cell Adhesion Molecules , Cell Proliferation , Mice, Knockout , STAT3 Transcription Factor , Signal Transduction , Thyroid Cancer, Papillary , Thyroid Neoplasms , Animals , Thyroid Cancer, Papillary/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/genetics , STAT3 Transcription Factor/metabolism , Cancer-Associated Fibroblasts/metabolism , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Mice , Humans , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Thyroid Neoplasms/genetics , Cell Line, Tumor , Tumor Microenvironment , Interleukin-4/metabolism , Integrins/metabolism , Focal Adhesion Kinase 1/metabolism , Periostin
15.
Acta Neuropathol Commun ; 12(1): 71, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38706008

ABSTRACT

Diffuse Intrinsic Pontine Glioma (DIPG) is a highly aggressive and fatal pediatric brain cancer. One pre-requisite for tumor cells to infiltrate is adhesion to extracellular matrix (ECM) components. However, it remains largely unknown which ECM proteins are critical in enabling DIPG adhesion and migration and which integrin receptors mediate these processes. Here, we identify laminin as a key ECM protein that supports robust DIPG cell adhesion and migration. To study DIPG infiltration, we developed a DIPG-neural assembloid model, which is composed of a DIPG spheroid fused to a human induced pluripotent stem cell-derived neural organoid. Using this assembloid model, we demonstrate that knockdown of laminin-associated integrins significantly impedes DIPG infiltration. Moreover, laminin-associated integrin knockdown improves DIPG response to radiation and HDAC inhibitor treatment within the DIPG-neural assembloids. These findings reveal the critical role of laminin-associated integrins in mediating DIPG progression and drug response. The results also provide evidence that disrupting integrin receptors may offer a novel therapeutic strategy to enhance DIPG treatment outcomes. Finally, these results establish DIPG-neural assembloid models as a powerful tool to study DIPG disease progression and enable drug discovery.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Integrins , Laminin , Humans , Laminin/metabolism , Integrins/metabolism , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/metabolism , Brain Stem Neoplasms/therapy , Diffuse Intrinsic Pontine Glioma/pathology , Diffuse Intrinsic Pontine Glioma/genetics , Cell Adhesion/drug effects , Cell Movement , Cell Line, Tumor , Glioma/pathology , Glioma/metabolism , Glioma/genetics , Glioma/therapy
16.
J Phys Condens Matter ; 36(29)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38574682

ABSTRACT

Cell-matrix adhesions connect the cytoskeleton to the extracellular environment and are essential for maintaining the integrity of tissue and whole organisms. Remarkably, cell adhesions can adapt their size and composition to an applied force such that their size and strength increases proportionally to the load. Mathematical models for the clutch-like force transmission at adhesions are frequently based on the assumption that mechanical load is applied tangentially to the adhesion plane. Recently, we suggested a molecular mechanism that can explain adhesion growth under load for planar cell adhesions. The mechanism is based on conformation changes of adhesion molecules that are dynamically exchanged with a reservoir. Tangential loading drives the occupation of some states out of equilibrium, which for thermodynamic reasons, leads to the association of further molecules with the cluster, which we refer to as self-stabilization. Here, we generalize this model to forces that pull at an oblique angle to the plane supporting the cell, and examine if this idealized model also predicts self-stabilization. We also allow for a variable distance between the parallel planes representing cytoskeletal F-actin and transmembrane integrins. Simulation results demonstrate that the binding mechanism and the geometry of the cluster have a strong influence on the response of adhesion clusters to force. For oblique angles smaller than about 40∘, we observe a growth of the adhesion site under force. However this self-stabilization is reduced as the angle between the force and substrate plane increases, with vanishing self-stabilization for normal pulling. Overall, these results highlight the fundamental difference between the assumption of pulling and shearing forces in commonly used models of cell adhesion.


Subject(s)
Extracellular Matrix , Focal Adhesions , Focal Adhesions/metabolism , Extracellular Matrix/metabolism , Cell Adhesion/physiology , Actins , Integrins/metabolism
17.
J Biochem Mol Toxicol ; 38(4): e23693, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38622980

ABSTRACT

IGSF10, a protein that belongs to the immunoglobulin superfamily, is involved in regulating the early migration of neurons that produce gonadotropin-releasing hormone and performs a fundamental function in development. Our previous study confirmed that the mRNA expression level of IGSF10 may be a protective prognosis factor for lung adenocarcinoma (LUAD) patients. However, the specific mechanisms of IGSF10 are still unclear. In this research, it was shown that the protein level of IGSF10 was down-modulated in LUAD tissues and had a link to the clinical and pathological characteristics as well as the patient's prognosis in LUAD. Importantly, IGSF10 regulates the metastatic ability of LUAD cells in vitro and in vivo. It was proven in a mechanistic sense that IGSF10 inhibits the capacity of LUAD cells to metastasize through the Spi-B/Integrin-ß1 signaling pathway. These findings gave credence to the premise that IGSF10 performed a crucial function in LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Adenocarcinoma of Lung/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Integrins/genetics , Integrins/metabolism , Lung Neoplasms/metabolism , Signal Transduction
18.
Int J Mol Sci ; 25(7)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38612378

ABSTRACT

Disturbed remodeling of the extracellular matrix (ECM) is frequently observed in several high-prevalence pathologies that include fibrotic diseases of organs such as the heart, lung, periodontium, liver, and the stiffening of the ECM surrounding invasive cancers. In many of these lesions, matrix remodeling mediated by fibroblasts is dysregulated, in part by alterations to the regulatory and effector systems that synthesize and degrade collagen, and by alterations to the functions of the integrin-based adhesions that normally mediate mechanical remodeling of collagen fibrils. Cell-matrix adhesions containing collagen-binding integrins are enriched with regulatory and effector systems that initiate localized remodeling of pericellular collagen fibrils to maintain ECM homeostasis. A large cadre of regulatory molecules is enriched in cell-matrix adhesions that affect ECM remodeling through synthesis, degradation, and contraction of collagen fibrils. One of these regulatory molecules is Transient Receptor Potential Vanilloid-type 4 (TRPV4), a mechanically sensitive, Ca2+-permeable plasma membrane channel that regulates collagen remodeling. The gating of Ca2+ across the plasma membrane by TRPV4 and the consequent generation of intracellular Ca2+ signals affect several processes that determine the structural and mechanical properties of collagen-rich ECM. These processes include the synthesis of new collagen fibrils, tractional remodeling by contractile forces, and collagenolysis. While the specific mechanisms by which TRPV4 contributes to matrix remodeling are not well-defined, it is known that TRPV4 is activated by mechanical forces transmitted through collagen adhesion receptors. Here, we consider how TRPV4 expression and function contribute to physiological and pathological collagen remodeling and are associated with collagen adhesions. Over the long-term, an improved understanding of how TRPV4 regulates collagen remodeling could pave the way for new approaches to manage fibrotic lesions.


Subject(s)
Extracellular Matrix , TRPV Cation Channels , Cell Membrane , Cell-Matrix Junctions , Collagen , Integrins , TRPV Cation Channels/genetics , Humans
19.
Int J Mol Sci ; 25(7)2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38612904

ABSTRACT

Cardiovascular diseases (CVDs) pose a significant global health threat due to their complex pathogenesis and high incidence, imposing a substantial burden on global healthcare systems. Integrins, a group of heterodimers consisting of α and ß subunits that are located on the cell membrane, have emerged as key players in mediating the occurrence and progression of CVDs by regulating the physiological activities of endothelial cells, vascular smooth muscle cells, platelets, fibroblasts, cardiomyocytes, and various immune cells. The crucial role of integrins in the progression of CVDs has valuable implications for targeted therapies. In this context, the development and application of various integrin antibodies and antagonists have been explored for antiplatelet therapy and anti-inflammatory-mediated tissue damage. Additionally, the rise of nanomedicine has enhanced the specificity and bioavailability of precision therapy targeting integrins. Nevertheless, the complexity of the pathogenesis of CVDs presents tremendous challenges for monoclonal targeted treatment. This paper reviews the mechanisms of integrins in the development of atherosclerosis, cardiac fibrosis, hypertension, and arrhythmias, which may pave the way for future innovations in the diagnosis and treatment of CVDs.


Subject(s)
Cardiovascular Diseases , Hypertension , Humans , Integrins , Endothelial Cells , Cell Membrane
20.
Pflugers Arch ; 476(6): 963-974, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38563997

ABSTRACT

Complex interactions of the branching ureteric bud (UB) and surrounding mesenchymal cells during metanephric kidney development determine the final number of nephrons. Impaired nephron endowment predisposes to arterial hypertension and chronic kidney disease. In the kidney, extracellular matrix (ECM) proteins are usually regarded as acellular scaffolds or as the common histological end-point of chronic kidney diseases. Since only little is known about their physiological role in kidney development, we aimed for analyzing the expression and role of fibronectin. In mouse, fibronectin was expressed during all stages of kidney development with significant changes over time. At embryonic day (E) 12.5 and E13.5, fibronectin lined the UB epithelium, which became less pronounced at E16.5 and then switched to a glomerular expression in the postnatal and adult kidneys. Similar results were obtained in human kidneys. Deletion of fibronectin at E13.5 in cultured metanephric mouse kidneys resulted in reduced kidney sizes and impaired glomerulogenesis following reduced cell proliferation and branching of the UB epithelium. Fibronectin colocalized with alpha 8 integrin and fibronectin loss caused a reduction in alpha 8 integrin expression, release of glial-derived neurotrophic factor and expression of Wnt11, both of which are promoters of UB branching. In conclusion, the ECM protein fibronectin acts as a regulator of kidney development and is a determinant of the final nephron number.


Subject(s)
Fibronectins , Kidney , Animals , Fibronectins/metabolism , Fibronectins/genetics , Mice , Humans , Kidney/metabolism , Kidney/embryology , Wnt Proteins/metabolism , Wnt Proteins/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , Cell Proliferation , Integrins/metabolism , Integrins/genetics , Mice, Inbred C57BL , Extracellular Matrix/metabolism , Integrin alpha Chains
SELECTION OF CITATIONS
SEARCH DETAIL
...