Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 650
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(20): e2214853120, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37155874

ABSTRACT

Gastric cancer is a dominating cause of cancer-associated mortality with limited therapeutic options. Here, we show that syndecan-4 (SDC4), a transmembrane proteoglycan, is highly expressed in intestinal subtype gastric tumors and that this signature associates with patient poor survival. Further, we mechanistically demonstrate that SDC4 is a master regulator of gastric cancer cell motility and invasion. We also find that SDC4 decorated with heparan sulfate is efficiently sorted in extracellular vesicles (EVs). Interestingly, SDC4 in EVs regulates gastric cancer cell-derived EV organ distribution, uptake, and functional effects in recipient cells. Specifically, we show that SDC4 knockout disrupts the tropism of EVs for the common gastric cancer metastatic sites. Our findings set the basis for the molecular implications of SDC4 expression in gastric cancer cells and provide broader perspectives on the development of therapeutic strategies targeting the glycan-EV axis to limit tumor progression.


Subject(s)
Stomach Neoplasms , Syndecan-4 , Humans , Heparitin Sulfate/metabolism , Neoplasm Invasiveness , Stomach Neoplasms/genetics , Syndecan-4/genetics , Syndecan-4/metabolism
2.
EMBO J ; 40(20): e106765, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34510494

ABSTRACT

The current pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and outbreaks of new variants highlight the need for preventive treatments. Here, we identified heparan sulfate proteoglycans as attachment receptors for SARS-CoV-2. Notably, neutralizing antibodies against SARS-CoV-2 isolated from COVID-19 patients interfered with SARS-CoV-2 binding to heparan sulfate proteoglycans, which might be an additional mechanism of antibodies to neutralize infection. SARS-CoV-2 binding to and infection of epithelial cells was blocked by low molecular weight heparins (LMWH). Although dendritic cells (DCs) and mucosal Langerhans cells (LCs) were not infected by SARS-CoV-2, both DC subsets efficiently captured SARS-CoV-2 via heparan sulfate proteoglycans and transmitted the virus to ACE2-positive cells. Notably, human primary nasal cells were infected by SARS-CoV-2, and infection was blocked by pre-treatment with LMWH. These data strongly suggest that heparan sulfate proteoglycans are important attachment receptors facilitating infection and transmission, and support the use of LMWH as prophylaxis against SARS-CoV-2 infection.


Subject(s)
COVID-19/transmission , Heparan Sulfate Proteoglycans/metabolism , Heparin, Low-Molecular-Weight/pharmacology , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/immunology , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Neutralizing/pharmacology , Chlorocebus aethiops , Dendritic Cells/metabolism , Dendritic Cells/virology , Epithelial Cells/metabolism , Epithelial Cells/virology , Host-Pathogen Interactions , Humans , Mucous Membrane/cytology , Mucous Membrane/virology , SARS-CoV-2/metabolism , Syndecan-1/metabolism , Syndecan-4/metabolism , Vero Cells , COVID-19 Drug Treatment
3.
Am J Physiol Cell Physiol ; 327(2): C380-C386, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38953842

ABSTRACT

Cell surface receptors play crucial roles in cellular responses to extracellular ligands, helping to modulate the functions of a cell based on information coming from outside the cell. Syndecan refers to a family of cell adhesion receptors that regulate both extracellular and cytosolic events. Alteration of syndecan expression disrupts regulatory mechanisms in a cell type-specific fashion, often leading to serious diseases, notably cancer. Given the multifaceted functions and distinct tissue distributions of syndecan, it will be important to unravel the gene-level intricacies of syndecan expression and thereby further understand its involvement in various carcinogenic processes. Although accumulating evidence indicates that the protein expression patterns of syndecan family members are significantly altered in cancer cells, the underlying gene-level mechanisms remain largely unknown. This review endeavors to explore syndecan gene expression levels across different cancer types by scrutinizing extensive cancer genome datasets using tools such as cBioPortal. Our analysis unveils that somatic mutations in SDC genes are rare occurrences, whereas copy number alterations are frequently observed across diverse cancers, particularly in SDC2 and SDC4. Notably, amplifications of SDC2 and SDC4 correlate with heightened metastatic potential and dismal prognosis. This underscores the recurrent nature of SDC2 and SDC4 amplifications during carcinogenesis and sheds light on their role in promoting cancer activity through augmented protein expression. The identification of these amplifications not only enriches our understanding of carcinogenic mechanisms but also hints at the potential therapeutic avenue of targeting SDC2 and SDC4 to curb cancer cell proliferation and metastasis.


Subject(s)
Gene Amplification , Humans , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis , Animals , Syndecan-4/genetics , Syndecan-4/metabolism , Syndecans/genetics , Syndecans/metabolism , Carcinoma/genetics , Carcinoma/pathology , Carcinoma/metabolism , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism
4.
Immunol Cell Biol ; 102(2): 97-116, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37982607

ABSTRACT

Reducing the activity of cytokines and leukocyte extravasation is an emerging therapeutic strategy to limit tissue-damaging inflammatory responses and restore immune homeostasis in inflammatory diseases. Proteoglycans embedded in the vascular endothelial glycocalyx, which regulate the activity of cytokines to restrict the inflammatory response in physiological conditions, are proteolytically cleaved in inflammatory diseases. Here we critically review the potential of proteolytically shed, soluble vascular endothelial glycocalyx proteoglycans to modulate pathological inflammatory responses. Soluble forms of the proteoglycans syndecan-1, syndecan-3 and biglycan exert beneficial anti-inflammatory effects by the removal of chemokines, suppression of proinflammatory cytokine expression and leukocyte migration, and induction of autophagy of proinflammatory M1 macrophages. By contrast, soluble versikine and decorin enhance proinflammatory responses by increasing inflammatory cytokine synthesis and leukocyte migration. Endogenous syndecan-2 and mimecan exert proinflammatory effects, syndecan-4 and perlecan mediate beneficial anti-inflammatory effects and glypican regulates Hh and Wnt signaling pathways involved in systemic inflammatory responses. Taken together, targeting the vascular endothelial glycocalyx-derived, soluble syndecan-1, syndecan-2, syndecan-3, syndecan-4, biglycan, versikine, mimecan, perlecan, glypican and decorin might be a potential therapeutic strategy to suppress overstimulated cytokine and leukocyte responses in inflammatory diseases.


Subject(s)
Glycocalyx , Syndecan-1 , Syndecan-1/metabolism , Glycocalyx/metabolism , Syndecan-3/metabolism , Syndecan-4/metabolism , Syndecan-2/metabolism , Biglycan/metabolism , Glypicans/metabolism , Decorin/metabolism , Chemokines/metabolism , Anti-Inflammatory Agents/metabolism
5.
Circ Res ; 130(1): 67-79, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34789007

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) are generated by back splicing of mostly mRNAs and are gaining increasing attention as a novel class of regulatory RNAs that control various cellular functions. However, their physiological roles and functional conservation in vivo are rarely addressed, given the inherent challenges of their genetic inactivation. Here, we aimed to identify locus conserved circRNAs in mice and humans, which can be genetically deleted due to retained intronic elements not contained in the mRNA host gene to eventually address functional conservation. METHODS AND RESULTS: Combining published endothelial RNA-sequencing data sets with circRNAs of the circATLAS databank, we identified locus-conserved circRNA retaining intronic elements between mice and humans. CRISPR/Cas9 mediated genetic depletion of the top expressed circRNA cZfp292 resulted in an altered endothelial morphology and aberrant flow alignment in the aorta in vivo. Consistently, depletion of cZNF292 in endothelial cells in vitro abolished laminar flow-induced alterations in cell orientation, paxillin localization and focal adhesion organization. Mechanistically, we identified the protein SDOS (syndesmos) to specifically interact with cZNF292 in endothelial cells by RNA-affinity purification and subsequent mass spectrometry analysis. Silencing of SDOS or its protein binding partner Syndecan-4, or mutation of the SDOS-cZNF292 binding site, prevented laminar flow-induced cytoskeletal reorganization thereby recapitulating cZfp292 knockout phenotypes. CONCLUSIONS: Together, our data reveal a hitherto unknown role of cZNF292/cZfp292 in endothelial flow responses, which influences endothelial shape.


Subject(s)
DNA-Binding Proteins , Endothelial Cells , Endothelium, Vascular , RNA, Circular , Transcription Factors , Animals , Humans , Mice , Blood Circulation , DNA-Binding Proteins/genetics , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Protein Binding , RNA, Circular/genetics , RNA, Circular/metabolism , Syndecan-4/metabolism , Transcription Factors/genetics
6.
Cell Biol Int ; 48(6): 883-897, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38591778

ABSTRACT

Anoikis is a process of programmed cell death induced by the loss of cell/matrix interactions. In previous work, we have shown that the acquisition of anoikis resistance upregulates syndecan-4 (SDC4) expression in endothelial cells. In addition, SDC4 gene silencing by microRNA interference reverses the transformed phenotype of anoikis-resistant endothelial cells. Due to this role of SDC4 in regulating the behavior of anoikis-resistant endothelial cells, we have evaluated that the functional consequences of SDC4 silencing in the extracellular matrix (ECM) remodeling in anoikis-resistant rabbit aortic endothelial cells submitted to SDC4 gene silencing (miR-Syn4-Adh-1-EC). For this, we evaluated the expression of adhesive proteins, ECM receptors, nonreceptor protein-tyrosine kinases, and ECM-degrading enzymes and their inhibitors. Altered cell behavior was monitored by adhesion, migration, and tube formation assays. We found that SDC4 silencing led to a decrease in migration and angiogenic capacity of anoikis-resistant endothelial cells; this was accompanied by an increase in adhesion to fibronectin. Furthermore, after SDC4 silencing, we observed an increase in the expression of fibronectin, collagen IV, and vitronectin, and a decrease in the expression of integrin α5ß1 and αvß3, besides that, silenced cells show an increase in Src and FAK expression. Quantitative polymerase chain reaction and Western blot analysis demonstrated that SDC4 silencing leads to altered gene and protein expression of MMP2, MMP9, and HSPE. Compared with parental cells, SDC4 silenced cells showed a decrease in nitric oxide production and eNOS expression. In conclusion, these data demonstrate that SDC4 plays an important role in ECM remodeling. In addition, our findings represent an important step toward understanding the mechanism by which SDC4 can reverse the transformed phenotype of anoikis-resistant endothelial cells.


Subject(s)
Anoikis , Endothelial Cells , Extracellular Matrix , Gene Silencing , Syndecan-4 , Syndecan-4/metabolism , Syndecan-4/genetics , Animals , Extracellular Matrix/metabolism , Endothelial Cells/metabolism , Rabbits , Cell Adhesion , Cell Movement , Fibronectins/metabolism , Cells, Cultured
7.
Mol Biol Rep ; 51(1): 604, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38700644

ABSTRACT

BACKGROUND: The healing process after a myocardial infarction (MI) in humans involves complex events that replace damaged tissue with a fibrotic scar. The affected cardiac tissue may lose its function permanently. In contrast, zebrafish display a remarkable capacity for scar-free heart regeneration. Previous studies have revealed that syndecan-4 (SDC4) regulates inflammatory response and fibroblast activity following cardiac injury in higher vertebrates. However, whether and how Sdc4 regulates heart regeneration in highly regenerative zebrafish remains unknown. METHODS AND RESULTS: This study showed that sdc4 expression was differentially regulated during zebrafish heart regeneration by transcriptional analysis. Specifically, sdc4 expression increased rapidly and transiently in the early regeneration phase upon ventricular cryoinjury. Moreover, the knockdown of sdc4 led to a significant reduction in extracellular matrix protein deposition, immune cell accumulation, and cell proliferation at the lesion site. The expression of tgfb1a and col1a1a, as well as the protein expression of Fibronectin, were all down-regulated under sdc4 knockdown. In addition, we verified that sdc4 expression was required for cardiac repair in zebrafish via in vivo electrocardiogram analysis. Loss of sdc4 expression caused an apparent pathological Q wave and ST elevation, which are signs of human MI patients. CONCLUSIONS: Our findings support that Sdc4 is required to mediate pleiotropic repair responses in the early stage of zebrafish heart regeneration.


Subject(s)
Heart , Regeneration , Syndecan-4 , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Syndecan-4/genetics , Syndecan-4/metabolism , Regeneration/genetics , Heart/physiology , Heart/physiopathology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Cell Proliferation/genetics , Myocardium/metabolism , Myocardium/pathology , Gene Knockdown Techniques
8.
J Appl Toxicol ; 44(6): 908-918, 2024 06.
Article in English | MEDLINE | ID: mdl-38396353

ABSTRACT

Cadmium (Cd) is one of the most polluting heavy metal in the environment. Cd exposure has been elucidated to cause dysfunction of the glomerular filtration barrier (GFB). However, the underlying mechanism remains unclear. C57BL/6J male mice were administered with 2.28 mg/kg cadmium chloride (CdCl2) dissolved in distilled water by oral gavage for 14 days. The expression of SDC4 in the kidney tissues was detected. Human renal glomerular endothelial cells (HRGECs) were exposed to varying concentrations of CdCl2 for 24 h. The mRNA levels of SDC4, along with matrix metalloproteinase (MMP)-2 and 9, were analyzed by quantitative PCR. Additionally, the protein expression levels of SDC4, MMP-2/9, and both total and phosphorylated forms of Smad2/3 (P-Smad2/3) were detected by western blot. The extravasation rate of fluorescein isothiocyanate-dextran through the Transwell was used to evaluate the permeability of HRGECs. SB431542 was used as an inhibitor of transforming growth factor (TGF)-ß signaling pathway to further investigate the role of TGF-ß. Cd reduced SDC4 expression in both mouse kidney tissues and HRGECs. In addition, Cd exposure increased permeability and upregulated P-Smad2/3 levels in HRGECs. SB431542 treatment inhibited the phosphorylation of Smad2/3, Cd-induced SDC4 downregulation, and hyperpermeability. MMP-2/9 levels increased by Cd exposure was also blocked by SB431542, demonstrating the involvement of TGF-ß/Smad pathway in low-dose Cd-induced SDC4 reduction in HRGECs. Given that SDC4 is an essential component of glycocalyx, protection or repair of endothelial glycocalyx is a potential strategy for preventing or treating kidney diseases associated with environmental Cd exposure.


Subject(s)
Cadmium , Endothelial Cells , Glycocalyx , Kidney Glomerulus , Syndecan-4 , Animals , Humans , Male , Mice , Cadmium/toxicity , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glycocalyx/drug effects , Glycocalyx/metabolism , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Mice, Inbred C57BL , Signal Transduction/drug effects , Syndecan-4/metabolism , Syndecan-4/genetics , Transforming Growth Factor beta/metabolism
9.
Int J Mol Sci ; 25(14)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39062776

ABSTRACT

Antithrombin III (ATIII) is a potent endogenous anticoagulant that binds to heparan sulfate proteoglycans (HSPGs) on endothelial cells' surfaces. Among these HSPGs, syndecans (SDCs) are crucial as transmembrane receptors bridging extracellular ligands with intracellular signaling pathways. Specifically, syndecan-4 (SDC4) has been identified as a key receptor on endothelial cells for transmitting the signaling effects of ATIII. Meanwhile, SDCs have been implicated in facilitating the cellular internalization of SARS-CoV-2. Given the complex interactions between ATIII and SDC4, our study analyzed the impact of ATIII on the virus entry into host cells. While ATIII binds to all SDC isoforms, it shows the strongest affinity for SDC4. SDCs' heparan sulfate chains primarily influence ATIII's SDC attachment, although other parts might also play a role in ATIII's dominant affinity toward SDC4. ATIII significantly reduces SARS-CoV-2's cellular entry into cell lines expressing SDCs, suggesting a competitive inhibition mechanism at the SDC binding sites, particularly SDC4. Conversely, the virus or its spike protein decreases the availability of SDCs on the cell surface, reducing ATIII's cellular attachment and hence contributing to a procoagulant environment characteristic of COVID-19.


Subject(s)
Antithrombin III , COVID-19 , SARS-CoV-2 , Syndecan-4 , Virus Internalization , Humans , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , SARS-CoV-2/drug effects , Syndecan-4/metabolism , COVID-19/virology , COVID-19/metabolism , Virus Internalization/drug effects , Antithrombin III/metabolism , Antithrombin III/pharmacology , Protein Binding , COVID-19 Vaccines/immunology , COVID-19 Drug Treatment , Syndecans/metabolism , Animals
10.
J Biol Chem ; 298(6): 102029, 2022 06.
Article in English | MEDLINE | ID: mdl-35569509

ABSTRACT

Epidermal growth factor receptor (EGFR) is a causal factor in carcinoma, yet many carcinoma patients are resistant to EGFR inhibitors. Potential insight into this resistance stems from prior work that showed EGFR in normal epithelial cells docks to the extracellular domain of the plasma membrane proteoglycan syndecan-4 (Sdc4) engaged with α3ß1 and α6ß4 integrins. We now report that this receptor complex is modified by the recruitment of syndecan-2 (Sdc2), the Recepteur d'Origine Nantais (RON) tyrosine kinase, and the cellular signaling mediator Abelson murine leukemia viral oncogene homolog 1 (ABL1) in triple-negative breast carcinoma and head and neck squamous cell carcinoma, where it contributes to EGFR kinase-independent proliferation. Treatment with a peptide mimetic of the EGFR docking site in the extracellular domain of Sdc4 (called SSTNEGFR) disrupts the entire complex and causes a rapid, global arrest of the cell cycle. Normal epithelial cells do not recruit these additional receptors to the adhesion mechanism and are not arrested by SSTNEGFR. Although EGFR docking with Sdc4 in the tumor cells is required, cell cycle progression does not depend on EGFR kinase. Instead, progression depends on RON kinase, activated by its incorporation into the complex. RON activates ABL1, which suppresses p38 mitogen-activated protein kinase and prevents a p38-mediated signal that would otherwise arrest the cell cycle. These findings add to the growing list of receptor tyrosine kinases that support tumorigenesis when activated by their association with syndecans at sites of matrix adhesion and identify new potential targets for cancer therapy.


Subject(s)
Carcinoma , Cell Cycle , ErbB Receptors , Receptor Protein-Tyrosine Kinases , Syndecan-2 , Syndecan-4 , Carcinoma/pathology , Cell Membrane/metabolism , ErbB Receptors/metabolism , Humans , Proto-Oncogene Proteins c-abl/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Syndecan-2/metabolism , Syndecan-4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Cell Mol Life Sci ; 79(2): 122, 2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35128576

ABSTRACT

Skeletal muscle demonstrates a high degree of regenerative capacity repeating the embryonic myogenic program under strict control. Rhabdomyosarcoma is the most common sarcoma in childhood and is characterized by impaired muscle differentiation. In this study, we observed that silencing the expression of syndecan-4, the ubiquitously expressed transmembrane heparan sulfate proteoglycan, significantly enhanced myoblast differentiation, and fusion. During muscle differentiation, the gradually decreasing expression of syndecan-4 allows the activation of Rac1, thereby mediating myoblast fusion. Single-molecule localized superresolution direct stochastic optical reconstruction microscopy (dSTORM) imaging revealed nanoscale changes in actin cytoskeletal architecture, and atomic force microscopy showed reduced elasticity of syndecan-4-knockdown cells during fusion. Syndecan-4 copy-number amplification was observed in 28% of human fusion-negative rhabdomyosarcoma tumors and was accompanied by increased syndecan-4 expression based on RNA sequencing data. Our study suggests that syndecan-4 can serve as a tumor driver gene in promoting rabdomyosarcoma tumor development. Our results contribute to the understanding of the role of syndecan-4 in skeletal muscle development, regeneration, and tumorigenesis.


Subject(s)
Actins/metabolism , Rhabdomyosarcoma/pathology , Syndecan-4/metabolism , rac1 GTP-Binding Protein/metabolism , Actin Cytoskeleton , Animals , Cell Differentiation , Cell Line , DNA Copy Number Variations , Humans , Male , Mice , Muscle Development , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Rhabdomyosarcoma/metabolism , Syndecan-4/antagonists & inhibitors , Syndecan-4/genetics , T-Lymphoma Invasion and Metastasis-inducing Protein 1/metabolism
12.
Proc Natl Acad Sci U S A ; 117(11): 5913-5922, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32108028

ABSTRACT

Exosomes, extracellular vesicles (EVs) of endosomal origin, emerge as master regulators of cell-to-cell signaling in physiology and disease. Exosomes are highly enriched in tetraspanins (TSPNs) and syndecans (SDCs), the latter occurring mainly in proteolytically cleaved form, as membrane-spanning C-terminal fragments of the proteins. While both protein families are membrane scaffolds appreciated for their role in exosome formation, composition, and activity, we currently ignore whether these work together to control exosome biology. Here we show that TSPN6, a poorly characterized tetraspanin, acts as a negative regulator of exosome release, supporting the lysosomal degradation of SDC4 and syntenin. We demonstrate that TSPN6 tightly associates with SDC4, the SDC4-TSPN6 association dictating the association of TSPN6 with syntenin and the TSPN6-dependent lysosomal degradation of SDC4-syntenin. TSPN6 also inhibits the shedding of the SDC4 ectodomain, mimicking the effects of matrix metalloproteinase inhibitors. Taken together, our data identify TSPN6 as a regulator of the trafficking and processing of SDC4 and highlight an important physical and functional interconnection between these membrane scaffolds for the production of exosomes. These findings clarify our understanding of the molecular determinants governing EV formation and have potentially broad impact for EV-related biomedicine.


Subject(s)
Exosomes/metabolism , Syntenins/metabolism , Tetraspanins/metabolism , Cell Communication , Exosomes/genetics , Extracellular Vesicles/metabolism , Humans , Lysosomes/metabolism , MCF-7 Cells , Matrix Metalloproteinases/metabolism , Protein Transport , Syndecan-4/metabolism , Syndecans/metabolism
13.
Int J Mol Sci ; 24(8)2023 Apr 08.
Article in English | MEDLINE | ID: mdl-37108098

ABSTRACT

The remodelling of the extracellular matrix plays an important role in skeletal muscle development and regeneration. Syndecan-4 is a cell surface proteoglycan crucial for muscle differentiation. Syndecan-4-/- mice have been reported to be unable to regenerate following muscle damage. To investigate the consequences of the decreased expression of Syndecan-4, we have studied the in vivo and in vitro muscle performance and the excitation-contraction coupling machinery in young and aged Syndecan-4+/- (SDC4) mice. In vivo grip force was decreased significantly as well as the average and maximal speed of voluntary running in SDC4 mice, regardless of their age. The maximal in vitro twitch force was reduced in both EDL and soleus muscles from young and aged SDC4 mice. Ca2+ release from the sarcoplasmic reticulum decreased significantly in the FDB fibres of young SDC4 mice, while its voltage dependence was unchanged regardless of age. These findings were present in muscles from young and aged mice as well. On C2C12 murine skeletal muscle cells, we have also found altered calcium homeostasis upon Syndecan-4 silencing. The decreased expression of Syndecan-4 leads to reduced skeletal muscle performance in mice and altered motility in C2C12 myoblasts via altered calcium homeostasis. The altered muscle force performance develops at an early age and is maintained throughout the life course of the animal until old age.


Subject(s)
Muscle, Skeletal , Syndecan-4 , Animals , Mice , Calcium/metabolism , Muscle Contraction/physiology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Sarcoplasmic Reticulum/metabolism , Syndecan-4/genetics , Syndecan-4/metabolism
14.
Int J Mol Sci ; 24(4)2023 Feb 05.
Article in English | MEDLINE | ID: mdl-36834552

ABSTRACT

Due to their low pathogenicity, immunogenicity, and long-term gene expression, adeno-associated virus (AAV) vectors emerged as safe and efficient gene delivery tools, over-coming setbacks experienced with other viral gene delivery systems in early gene therapy trials. Among AAVs, AAV9 can translocate through the blood-brain barrier (BBB), making it a promising gene delivery tool for transducing the central nervous system (CNS) via systemic administration. Recent reports on the shortcomings of AAV9-mediated gene delivery into the CNS require reviewing the molecular base of AAV9 cellular biology. A more detailed understanding of AAV9's cellular entry would eradicate current hurdles and enable more efficient AAV9-based gene therapy approaches. Syndecans, the transmembrane family of heparan-sulfate proteoglycans, facilitate the cellular uptake of various viruses and drug delivery systems. Utilizing human cell lines and syndecan-specific cellular assays, we assessed the involvement of syndecans in AAV9's cellular entry. The ubiquitously expressed isoform, syndecan-4 proved its superiority in facilitating AAV9 internalization among syndecans. Introducing syndecan-4 into poorly transducible cell lines enabled robust AAV9-dependent gene transduction, while its knockdown reduced AAV9's cellular entry. Attachment of AAV9 to syndecan-4 is mediated not just by the polyanionic heparan-sulfate chains but also by the cell-binding domain of the extracellular syndecan-4 core protein. Co-immunoprecipitation assays and affinity proteomics also confirmed the role of syndecan-4 in the cellular entry of AAV9. Overall, our findings highlight the universally expressed syndecan-4 as a significant contributor to the cellular internalization of AAV9 and provide a molecular-based, rational explanation for the low gene delivery potential of AAV9 into the CNS.


Subject(s)
Dependovirus , Syndecan-4 , Humans , Dependovirus/metabolism , Heparan Sulfate Proteoglycans , Heparitin Sulfate/metabolism , Sulfates , Syndecan-1 , Syndecans/metabolism
15.
Molecules ; 28(23)2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38067582

ABSTRACT

Syndecan-4 (SDC4) consists of transmembrane heparan sulfate proteoglycan (HSPG) belonging to the syndecan family. It is present in most cell types of Mammalia. Its structure contains a heparan-sulfate-modified extracellular domain, a single transmembrane domain, and a short C-terminal cytoplasmic domain. Regarding the overall cellular function of SDC4, other cells or ligands can bind to its ecto-domain. In addition, 4,5-bisphosphate phosphatidylinositol (PIP2) or protein kinase Cα can bind to its cyto-domain to activate downstream signaling pathways. To understand the signal transduction mechanism of syndecan, it is important to know the interactions between their actual structure and function in vivo. Therefore, it is important to identify the structure of SDC4 to understand the ligand binding behavior of SDC4. In this study, expression and purification were performed to reveal structures of the short ecto-domain, the transmembrane domain, and the cytoplasmic domain of Syd4-eTC (SDC4). Solution-state NMR spectroscopy and solid-state NMR spectroscopy were used to study the structure of Syd4-eTC in membrane environments and to demonstrate the interaction between Syd4-eTC and PIP2.


Subject(s)
Signal Transduction , Syndecan-4 , Syndecan-4/metabolism , Cytoplasm/metabolism , Signal Transduction/physiology , Heparan Sulfate Proteoglycans/metabolism , Magnetic Resonance Spectroscopy
16.
Am J Physiol Cell Physiol ; 323(5): C1345-C1354, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36094435

ABSTRACT

Expression of the cell surface heparan sulfate proteoglycan syndecan-4 is dysregulated in breast cancer, the most frequent malignancy in women. High expression of syndecan-4 correlates with a worse survival in the subgroup of estrogen receptor negative and estrogen/progesterone-receptor negative patients. Aberrant expression of syndecan-4 in breast cancer involves both transcriptional and posttranscriptional mechanisms, including estrogen- and growth factor-dependent regulation, mutations in GAPVD1, NUP153, PDE4DIP, and RREB1, as well as targeting by microRNAs. At the functional level, syndecan-4 plays an important role in various stages of breast cancer progression by interacting with ligands as diverse as plasma proteins, extracellular matrix proteins, growth factors, and surface receptors, as well as members of the integrin family. Mechanisms including integrin recycling, ectodomain shedding, and crosstalk with other syndecans expand the repertoire of syndecan-4 function. Through these interactions, syndecan-4 regulates cellular processes such as adhesion, migration, and invasion. Additional possible functions of syndecan-4 in cells of the microenvironment contribute to the complexity of its pathophysiology. Notably, syndecan-4 expression is modulated by drugs used in breast cancer treatment, such as trastuzumab and zoledronate. Overall, these findings mark syndecan-4 as a novel pathogenesis factor and promising target for therapeutic interventions in breast cancer.


Subject(s)
Breast Neoplasms , MicroRNAs , Humans , Female , Syndecan-4/genetics , Syndecan-4/metabolism , Heparan Sulfate Proteoglycans/metabolism , Breast Neoplasms/pathology , Zoledronic Acid , Progesterone , Ligands , Receptors, Estrogen , Extracellular Matrix Proteins , Intercellular Signaling Peptides and Proteins , Trastuzumab , Integrins , Estrogens , Syndecan-1 , Tumor Microenvironment
17.
J Cell Sci ; 133(7)2020 04 08.
Article in English | MEDLINE | ID: mdl-32269093

ABSTRACT

ADAMTS-1 is an extracellular protease with critical roles in organogenesis and angiogenesis. Here we demonstrate a functional convergence of ADAMTS-1 and the transmembrane heparan sulfate proteoglycan syndecan-4 in influencing adhesion, migration and angiogenesis. Knockdown of ADAMTS-1 in endothelial cells resulted in a parallel reduction in cell surface syndecan-4, attributable to increased matrix metalloproteinase-9 (MMP9) activity. Knockdown of either ADAMTS-1 or syndecan-4 increased cellular responses to vascular endothelial growth factor A isoform VEGFA164, and increased ex vivo aortic ring microvessel sprouting. On fibronectin, knockdown of either protein enhanced migration and promoted formation of long α5 integrin-containing fibrillar adhesions. However, integrin α5 null cells still showed increased migration in response to ADAMTS-1 and syndecan-4 siRNA treatment. Plating of naïve endothelial cells on cell-conditioned matrix from ADAMTS-1 and syndecan-4 knockdown cells demonstrated that the altered adhesive behaviour was matrix dependent, and this correlated with a lack of expression of fibulin-1: an extracellular matrix co-factor for ADAMTS-1 that is known to inhibit migration. These findings support the notion that ADAMTS-1 and syndecan-4 are functionally interconnected in regulating cell migration and angiogenesis, via collaboration with MMP9 and fibulin-1.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Syndecan-4 , Vascular Endothelial Growth Factor A , ADAMTS1 Protein , Animals , Cell Adhesion , Cell Movement , Endothelial Cells , Humans , Mice , Neovascularization, Pathologic , Syndecan-1 , Syndecan-2 , Syndecan-4/genetics
18.
Am J Physiol Heart Circ Physiol ; 322(6): H1057-H1071, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35522553

ABSTRACT

Pathological myocardial hypertrophy in response to an increase in left ventricular (LV) afterload may ultimately lead to heart failure. Cell surface receptors bridge the interface between the cell and the extracellular matrix (ECM) in cardiac myocytes and cardiac fibroblasts and have been suggested to be important mediators of pathological myocardial hypertrophy. We identify for the first time that integrin α11 (α11) is preferentially upregulated among integrin ß1 heterodimer-forming α-subunits in response to increased afterload induced by aortic banding (AB) in wild-type (WT) mice. Mice were anesthetized in a chamber with 4% isoflurane and 95% oxygen before being intubated and ventilated with 2.5% isoflurane and 97% oxygen. For pre- and postoperative analgesia, animals were administered 0.02-mL buprenorphine (0.3 mg/mL) subcutaneously. Surprisingly, mice lacking α11 develop myocardial hypertrophy following AB comparable to WT. In the mice lacking α11, we further show a compensatory increase in the expression of another mechanoreceptor, syndecan-4, following AB compared with WT AB mice, indicating that syndecan-4 compensated for lack of α11. Intriguingly, mice lacking mechanoreceptors α11 and syndecan-4 show ablated myocardial hypertrophy following AB compared with WT mice. Expression of the main cardiac collagen isoforms col1a2 and col3a1 was significantly reduced in AB mice lacking mechanoreceptors α11 and syndecan-4 compared with WT AB.NEW & NOTEWORTHY Despite their putative importance in stress sensing, the specific integrin α-subunit(s) involved in cardiac hypertrophy has not been identified. Here, we show that α11 and syndecan-4 are critical and interdependent mediators of the hypertrophic response to increased LV afterload. We demonstrate in cells lacking both receptors an interdependent reduction in cell attachment to the major cardiac extracellular matrix components, suggesting that their interplay represents an important mechanism for stress sensing in cardiac cells.


Subject(s)
Isoflurane , Syndecan-4 , Animals , Cardiomegaly/metabolism , Integrin alpha Chains/metabolism , Integrins/metabolism , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Receptors, Collagen , Syndecan-4/genetics , Syndecan-4/metabolism
19.
J Transl Med ; 20(1): 319, 2022 07 16.
Article in English | MEDLINE | ID: mdl-35842658

ABSTRACT

BACKGROUND: Cardiovascular diseases (CVDs) are a significant cause of mortality worldwide and are characterized by severe atherosclerosis (AS) in patients. However, the molecular mechanism of AS formation remains elusive. In the present study, we investigated the role of syndecan-4 (SDC4), a member of the syndecan family, in atherogenesis. METHODS AND RESULTS: The expression of SDC4 decreased in mouse severe AS models. Moreover, knockout of SDC4 accelerated high-cholesterol diets (HCD)-induced AS in ApoE-/- mice. Mechanistically, the decrease of SDC4 increased macrophage proinflammatory capacity may be through the PKCα-ABCA1/ABCG1 signaling pathway. CONCLUSION: These findings provide evidence that SDC4 reduction links macrophages and inflammation to AS and that SDC4 in macrophages provides a therapeutic target for preventing AS formation.


Subject(s)
Atherosclerosis , Macrophages/metabolism , Syndecan-4/metabolism , Animals , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol/metabolism , Disease Models, Animal , Mice , Mice, Knockout , Syndecan-4/genetics
20.
IUBMB Life ; 74(10): 969-981, 2022 10.
Article in English | MEDLINE | ID: mdl-35833571

ABSTRACT

Radiotherapy is widely used for the treatment of breast cancer. However, we have shown that ionizing radiation can provoke premature senescence in breast stromal cells. In particular, breast stromal fibroblasts can become senescent after irradiation both in vitro and in vivo and they express an inflammatory phenotype and an altered profile of extracellular matrix components, thus facilitating tumor progression. Adipose-derived stem cells (ASCs) represent another major component of the breast tissue stroma. They are multipotent cells and due to their ability to differentiate in multiple cell lineages they play an important role in tissue maintenance and repair in normal and pathologic conditions. Here, we investigated the characteristics of human breast ASCs that became senescent prematurely after their exposure to ionizing radiation. We found decreased expression levels of the specific mesenchymal cell surface markers CD105, CD73, CD44, and CD90. In parallel, we demonstrated a significantly reduced expression of transcription factors regulating osteogenic (i.e., RUNX2), adipogenic (i.e., PPARγ), and chondrogenic (i.e., SOX9) differentiation; this was followed by an analogous reduction in their differentiation capacity. Furthermore, they overexpress inflammatory markers, that is, IL-6, IL-8, and ICAM-1, and a catabolic phenotype, marked by the reduction of collagen type I and the increase of MMP-1 and MMP-13 expression. Finally, we detected changes in proteoglycan expression, for example, the upregulation of syndecan 1 and syndecan 4 and the downregulation of decorin. Notably, all these alterations, when observed in the breast stroma, represent poor prognostic factors for tumor development. In conclusion, we showed that ionizing radiation-mediated prematurely senescent human breast ASCs have a decreased differentiation potential and express specific changes adding to the formation of a permissive environment for tumor growth.


Subject(s)
Core Binding Factor Alpha 1 Subunit , Syndecan-1 , Adipose Tissue/metabolism , Cell Differentiation/physiology , Cells, Cultured , Collagen Type I , Core Binding Factor Alpha 1 Subunit/metabolism , Decorin/metabolism , Extracellular Matrix/genetics , Humans , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 13/metabolism , PPAR gamma/metabolism , Stem Cells/metabolism , Syndecan-1/metabolism , Syndecan-4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL