Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.473
Filter
1.
Open Biol ; 14(6): 240035, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38862019

ABSTRACT

Neurodegenerative diseases, particularly Alzheimer's disease (AD), pose a significant challenge in ageing populations. Our current understanding indicates that the onset of toxic amyloid and tau protein pathologies initiates disease progression. However, existing treatments targeting these hallmark symptoms offer symptomatic relief without halting disease advancement. This review offers an alternative perspective on AD, centring on impaired adult hippocampal neurogenesis (AHN) as a potential early aetiological factor. By delving into the intricate molecular events during the initial stages of AD (Braak Stages I-III), a novel hypothesis is presented, interweaving the roles of Notch signalling and heparan sulfate proteoglycans (HSPGs) in compromised AHN. While acknowledging the significance of the amyloid and tau hypotheses, it calls for further exploration beyond these paradigms, suggesting the potential of altered HS sulfation patterns in AD initiation. Future directions propose more detailed investigations into early HS aggregation, aberrant sulfation patterns and examination of their temporal relationship with tau hyperphosphorylation. In challenging the conventional 'triggers' of AD and urging their reconsideration as symptoms, this review advocates an alternative approach to understanding this disease, offering new avenues of investigation into the intricacies of AD pathogenesis.


Subject(s)
Alzheimer Disease , tau Proteins , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/etiology , tau Proteins/metabolism , Animals , Neurogenesis , Hippocampus/metabolism , Heparan Sulfate Proteoglycans/metabolism , Phosphorylation , Signal Transduction , Amyloid beta-Peptides/metabolism , Receptors, Notch/metabolism
2.
Biomed Pharmacother ; 176: 116893, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38850653

ABSTRACT

Polymer-cationic mediated gene delivery is a well-stablished strategy of transient gene expression (TGE) in mammalian cell cultures. Nonetheless, its industrial implementation is hindered by the phenomenon known as cell density effect (CDE) that limits the cell density at which cultures can be efficiently transfected. The rise in personalized medicine and multiple cell and gene therapy approaches based on TGE, make more relevant to understand how to circumvent the CDE. A rational study upon DNA/PEI complex formation, stability and delivery during transfection of HEK293 cell cultures has been conducted, providing insights on the mechanisms for polyplexes uptake at low cell density and disruption at high cell density. DNA/PEI polyplexes were physiochemically characterized by coupling X-ray spectroscopy, confocal microscopy, cryo-transmission electron microscopy (TEM) and nuclear magnetic resonance (NMR). Our results showed that the ionic strength of polyplexes significantly increased upon their addition to exhausted media. This was reverted by depleting extracellular vesicles (EVs) from the media. The increase in ionic strength led to polyplex aggregation and prevented efficient cell transfection which could be counterbalanced by implementing a simple media replacement (MR) step before transfection. Inhibiting and labeling specific cell-surface proteoglycans (PGs) species revealed different roles of PGs in polyplexes uptake. Importantly, the polyplexes uptake process seemed to be triggered by a coalescence phenomenon of HSPG like glypican-4 around polyplex entry points. Ultimately, this study provides new insights into PEI-based cell transfection methodologies, enabling to enhance transient transfection and mitigate the cell density effect (CDE).


Subject(s)
DNA , Glypicans , Transfection , Humans , HEK293 Cells , Transfection/methods , Glypicans/metabolism , Glypicans/genetics , DNA/metabolism , DNA/genetics , Polyethyleneimine/chemistry , Heparan Sulfate Proteoglycans/metabolism , Osmolar Concentration
3.
Virology ; 596: 110120, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38805801

ABSTRACT

Glycoprotein C (gC), one of ∼12 HSV-1 envelope glycoproteins, carries out several important functions during infection, including the enhancement of virion attachment by binding to host cell heparan sulfate proteoglycans (HSPG). Here we report that gC can also enhance the release of cell-free progeny virions at the end of the infectious cycle. This activity was observed in multiple cellular contexts including Vero cells and immortalized human keratinocytes. In the absence of gC, progeny virions bound more tightly to infected cells, suggesting that gC promotes the detachment of virions from the infected cell surface. Given this finding, we analyzed the biochemical interactions that tether progeny virions to cells and report evidence for two distinct modes of binding. One is consistent with a direct interaction between gC and HSPG, whereas the other is gC-independent and likely does not involve HSPG. Together, our results i) identify a novel function for a long-studied HSV-1 glycoprotein, and ii) demonstrate that the extracellular release of HSV-1 virions is a dynamic process involving multiple viral and host components.


Subject(s)
Herpesvirus 1, Human , Viral Envelope Proteins , Virion , Virus Release , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/metabolism , Humans , Chlorocebus aethiops , Vero Cells , Animals , Virion/metabolism , Heparan Sulfate Proteoglycans/metabolism , Keratinocytes/virology , Keratinocytes/metabolism
4.
Mol Cell Neurosci ; 129: 103936, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38750678

ABSTRACT

Neurological disorders impact around one billion individuals globally (15 % approx.), with significant implications for disability and mortality with their impact in Australia currently amounts to 6.8 million deaths annually. Heparan sulfate proteoglycans (HSPGs) are complex extracellular molecules implicated in promoting Tau fibril formation resulting in Tau tangles, a hallmark of Alzheimer's disease (AD). HSPG-Tau protein interactions contribute to various AD stages via aggregation, toxicity, and clearance, largely via interactions with the glypican 1 and syndecan 3 core proteins. The tunnelling nanotubes (TNTs) pathway is emerging as a facilitator of intercellular molecule transport, including Tau and Amyloid ß proteins, across extensive distances. While current TNT-associated evidence primarily stems from cancer models, their role in Tau propagation and its effects on recipient cells remain unclear. This review explores the interplay of TNTs, HSPGs, and AD-related factors and proposes that HSPGs influence TNT formation in neurodegenerative conditions such as AD.


Subject(s)
Alzheimer Disease , Heparan Sulfate Proteoglycans , tau Proteins , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Heparan Sulfate Proteoglycans/metabolism , Animals , tau Proteins/metabolism , Nanotubes , Amyloid beta-Peptides/metabolism , Cell Membrane Structures
5.
Blood ; 143(25): 2571-2587, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38639475

ABSTRACT

ABSTRACT: From signaling mediators in stem cells to markers of differentiation and lineage commitment to facilitators for the entry of viruses, such as HIV-1, cell surface heparan sulfate (HS) glycans with distinct modification patterns play important roles in hematopoietic biology. In this review, we provide an overview of the importance of HS and the proteoglycans (HSPGs) to which they are attached within the major cellular subtypes of the hematopoietic system. We summarize the roles of HSPGs, HS, and HS modifications within each main hematopoietic cell lineage of both myeloid and lymphoid arms. Lastly, we discuss the biological advances in the detection of HS modifications and their potential to further discriminate cell types within hematopoietic tissue.


Subject(s)
Hematopoiesis , Heparan Sulfate Proteoglycans , Heparitin Sulfate , Humans , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/metabolism , Animals , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology
6.
FASEB J ; 38(7): e23609, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38593345

ABSTRACT

PTPRD, a well-established tumor suppressor gene, encodes the protein tyrosine phosphatase-type D. This protein consists of three immunoglobulin-like (Ig) domains, four to eight fibronectin type 3 (FN) domains, a single transmembrane segment, and two cytoplasmic tandem tyrosine phosphatase domains. PTPRD is known to harbor various cancer-associated point mutations. While it is assumed that PTPRD regulates cellular functions as a tumor suppressor through the tyrosine phosphatase activity in the intracellular region, the function of its extracellular domain (ECD) in cancer is not well understood. In this study, we systematically examined the impact of 92 cancer-associated point mutations within the ECD. We found that 69.6% (64 out of 92) of these mutations suppressed total protein expression and/or plasma membrane localization. Notably, almost all mutations (20 out of 21) within the region between the last FN domain and transmembrane segment affected protein expression and/or localization, highlighting the importance of this region for protein stability. We further found that some mutations within the Ig domains adjacent to the glycosaminoglycan-binding pocket enhanced PTPRD's binding ability to heparan sulfate proteoglycans (HSPGs). This interaction is proposed to suppress phosphatase activity. Our findings therefore suggest that HSPG-mediated attenuation of phosphatase activity may be involved in tumorigenic processes through PTPRD dysregulation.


Subject(s)
Heparan Sulfate Proteoglycans , Neoplasms , Humans , Heparan Sulfate Proteoglycans/metabolism , Point Mutation , Extracellular Matrix Proteins/genetics , Immunoglobulins , Protein Stability , Tyrosine/genetics , Phosphoric Monoester Hydrolases/genetics , Heparitin Sulfate , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism
7.
In Vitro Cell Dev Biol Anim ; 60(5): 441-448, 2024 May.
Article in English | MEDLINE | ID: mdl-38379096

ABSTRACT

Wnt, a family of secreted signaling proteins, serves diverse functions in embryogenesis, organogenesis, cancer, and stem cell functions. In the context of development, Wnt has been considered a representative morphogen, forming concentration gradients to give positional information to cells or tissues. However, although gradients are often illustrated in schemata, the reality of concentration gradients, or in other words, actual spatial distribution of Wnt ligands, and their behaviors in the extracellular space still remain poorly known. To understand extracellular behavior of Wnt ligands, quantitative analyses such as fluorescence correlation spectroscopy (FCS) and fluorescence recovery after photobleaching (FRAP) are highly informative because Wnt dispersal involves physical and biochemical processes, such as diffusion and binding to or dissociation from cell surface molecules, including heparan sulfate proteoglycans (HSPGs). Here, I briefly discuss representative methods to quantify morphogen dynamics. In addition, I discuss molecular manipulations of morphogens, mainly focusing on use of protein binders, and synthetic biology of morphogens as indicators of current and future directions in this field.


Subject(s)
Wnt Proteins , Ligands , Animals , Humans , Wnt Proteins/metabolism , Extracellular Space/metabolism , Fluorescence Recovery After Photobleaching , Heparan Sulfate Proteoglycans/metabolism , Wnt Signaling Pathway
8.
Matrix Biol ; 128: 39-64, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38387749

ABSTRACT

Collagen type XVIII (COL18) is an abundant heparan sulfate proteoglycan in vascular basement membranes. Here, we asked (i) if the loss of COL18 would result in blood-brain barrier (BBB) breakdown, pathological alterations of small arteries and capillaries and neuroinflammation as found in cerebral small vessel disease (CSVD) and (ii) if such changes may be associated with remodeling of synapses and neural extracellular matrix (ECM). We found that 5-month-old Col18a1-/- mice had elevated BBB permeability for mouse IgG in the deep gray matter, and intravascular erythrocyte accumulations were observed brain-wide in capillaries and arterioles. BBB permeability increased with age and affected cortical regions and the hippocampus in 12-month-old Col18a1-/- mice. None of the Col18a1-/- mice displayed hallmarks of advanced CSVD, such as hemorrhages, and did not show perivascular space enlargement. Col18a1 deficiency-induced BBB leakage was accompanied by activation of microglia and astrocytes, a loss of aggrecan in the ECM of perineuronal nets associated with fast-spiking inhibitory interneurons and accumulation of the perisynaptic ECM proteoglycan brevican and the microglial complement protein C1q at excitatory synapses. As the pathway underlying these regulations, we found increased signaling through the TGF-ß1/Smad3/TIMP-3 cascade. We verified the pivotal role of COL18 for small vessel wall structure in CSVD by demonstrating the protein's involvement in vascular remodeling in autopsy brains from patients with cerebral hypertensive arteriopathy. Our study highlights an association between the alterations of perivascular ECM, extracellular proteolysis, and perineuronal/perisynaptic ECM, as a possible substrate of synaptic and cognitive alterations in CSVD.


Subject(s)
Cerebral Small Vessel Diseases , Collagen Type XVIII , Neuroinflammatory Diseases , Animals , Humans , Infant , Mice , Cerebral Small Vessel Diseases/genetics , Cerebral Small Vessel Diseases/metabolism , Collagen Type XVIII/genetics , Collagen Type XVIII/metabolism , Endostatins , Extracellular Matrix/metabolism , Heparan Sulfate Proteoglycans/metabolism , Mice, Knockout
9.
Proc Natl Acad Sci U S A ; 121(3): e2316733121, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38215181

ABSTRACT

The epithelial-mesenchymal transition (EMT) program is crucial for transforming carcinoma cells into a partially mesenchymal state, enhancing their chemoresistance, migration, and metastasis. This shift in cell state is tightly regulated by cellular mechanisms that are not yet fully characterized. One intriguing EMT aspect is the rewiring of the proteoglycan landscape, particularly the induction of heparan sulfate proteoglycan (HSPG) biosynthesis. This proteoglycan functions as a co-receptor that accelerates cancer-associated signaling pathways through its negatively-charged residues. However, the precise mechanisms through which EMT governs HSPG biosynthesis and its role in cancer cell plasticity remain elusive. Here, we identified exostosin glycosyltransferase 1 (EXT1), a central enzyme in HSPG biosynthesis, to be selectively upregulated in aggressive tumor subtypes and cancer cell lines, and to function as a key player in breast cancer aggressiveness. Notably, ectopic expression of EXT1 in epithelial cells is sufficient to induce HSPG levels and the expression of known mesenchymal markers, subsequently enhancing EMT features, including cell migration, invasion, and tumor formation. Additionally, EXT1 loss in MDA-MB-231 cells inhibits their aggressiveness-associated traits such as migration, chemoresistance, tumor formation, and metastasis. Our findings reveal that EXT1, through its role in HSPG biosynthesis, governs signal transducer and activator of transcription 3 (STAT3) signaling, a known regulator of cancer cell aggressiveness. Collectively, we present the EXT1/HSPG/STAT3 axis as a central regulator of cancer cell plasticity that directly links proteoglycan synthesis to oncogenic signaling pathways.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , Heparan Sulfate Proteoglycans/metabolism , STAT3 Transcription Factor/metabolism , Cell Line , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Epithelial-Mesenchymal Transition , Cell Line, Tumor , Cell Movement
10.
Cell Rep ; 43(1): 113668, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38198277

ABSTRACT

Perlecan (HSPG2), a heparan sulfate proteoglycan similar to agrin, is key for extracellular matrix (ECM) maturation and stabilization. Although crucial for cardiac development, its role remains elusive. We show that perlecan expression increases as cardiomyocytes mature in vivo and during human pluripotent stem cell differentiation to cardiomyocytes (hPSC-CMs). Perlecan-haploinsuffient hPSCs (HSPG2+/-) differentiate efficiently, but late-stage CMs have structural, contractile, metabolic, and ECM gene dysregulation. In keeping with this, late-stage HSPG2+/- hPSC-CMs have immature features, including reduced ⍺-actinin expression and increased glycolytic metabolism and proliferation. Moreover, perlecan-haploinsuffient engineered heart tissues have reduced tissue thickness and force generation. Conversely, hPSC-CMs grown on a perlecan-peptide substrate are enlarged and display increased nucleation, typical of hypertrophic growth. Together, perlecan appears to play the opposite role of agrin, promoting cellular maturation rather than hyperplasia and proliferation. Perlecan signaling is likely mediated via its binding to the dystroglycan complex. Targeting perlecan-dependent signaling may help reverse the phenotypic switch common to heart failure.


Subject(s)
Agrin , Heparan Sulfate Proteoglycans , Humans , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Agrin/metabolism , Myocytes, Cardiac/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism
11.
J Biol Chem ; 300(2): 105660, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38242322

ABSTRACT

Persistent high-risk HPV infection is closely associated with cervical cancer development, and there is no drug targeting HPV on the market at present, so it is particularly important to understand the interaction mechanism between HPV and the host which may provide the novel strategies for treating HPV diseases. HPV can hijack cell surface heparan sulfate proteoglycans (HSPGs) as primary receptors. However, the secondary entry receptors for HPV remain elusive. We identify myosin-9 (NMHC-IIA) as a host factor that interacts with HPV L1 protein and mediates HPV internalization. Efficient HPV entry required myosin-9 redistribution to the cell surface regulated by HPV-hijacked MEK-MLCK signaling. Myosin-9 maldistribution by ML-7 or ML-9 significantly inhibited HPV pseudoviruses infection in vitro and in vivo. Meanwhile, N-glycans, especially the galactose chains, may act as the decoy receptors for HPV, which can block the interaction of HPV to myosin-9 and influence the way of HPV infection. Taken together, we identify myosin-9 as a novel functional entry receptor for high-risk HPV both in vitro and in vivo, and unravel the new roles of myosin-9 and N-glycans in HPV entry, which provides the possibilities for host targets of antiviral drugs.


Subject(s)
Human Papillomavirus Viruses , Papillomavirus Infections , Virus Internalization , Humans , Cytoskeletal Proteins , Heparan Sulfate Proteoglycans/metabolism , Myosins , Cell Line , Animals , Cricetinae , Cricetulus , Polysaccharides/metabolism
12.
J Biol Chem ; 300(1): 105544, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38072044

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) are composed of a core protein and glycosaminoglycan (GAG) chains and serve as coreceptors for many growth factors and morphogens. To understand the molecular mechanisms by which HSPGs regulate morphogen gradient formation and signaling, it is important to determine the relative contributions of the carbohydrate and protein moieties to the proteoglycan function. To address this question, we generated ΔGAG alleles for dally and dally-like protein (dlp), two Drosophila HSPGs of the glypican family, in which all GAG-attachment serine residues are substituted to alanine residues using CRISPR/Cas9 mutagenesis. In these alleles, the glypican core proteins are expressed from the endogenous loci with no GAG modification. Analyses of the dallyΔGAG allele defined Dally functions that do not require heparan sulfate (HS) chains and that need both core protein and HS chains. We found a new, dallyΔGAG-specific phenotype, the formation of a posterior ectopic vein, which we have never seen in the null mutants. Unlike dallyΔGAG, dlpΔGAG mutants do not show most of the dlp null mutant phenotypes, suggesting that HS chains are dispensable for these dlp functions. As an exception, HS is essentially required for Dlp's activity at the neuromuscular junction. Thus, Drosophila glypicans show strikingly different levels of HS dependency. The ΔGAG mutant alleles of the glypicans serve as new molecular genetic toolsets highly useful to address important biological questions, such as molecular mechanisms of morphogen gradient formation.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Glypicans , Heparitin Sulfate , Animals , Drosophila Proteins/metabolism , Glypicans/genetics , Glypicans/chemistry , Glypicans/metabolism , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/genetics , Heparitin Sulfate/metabolism , Membrane Glycoproteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism
13.
Alzheimers Dement ; 20(2): 819-836, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37791598

ABSTRACT

INTRODUCTION: We discovered that the APOE3 Christchurch (APOE3Ch) variant may provide resistance to Alzheimer's disease (AD). This resistance may be due to reduced pathological interactions between ApoE3Ch and heparan sulfate proteoglycans (HSPGs). METHODS: We developed and characterized the binding, structure, and preclinical efficacy of novel antibodies targeting human ApoE-HSPG interactions. RESULTS: We found that one of these antibodies, called 7C11, preferentially bound ApoE4, a major risk factor for sporadic AD, and disrupts heparin-ApoE4 interactions. We also determined the crystal structure of a Fab fragment of 7C11 and used computer modeling to predict how it would bind to ApoE. When we tested 7C11 in mouse models, we found that it reduced recombinant ApoE-induced tau pathology in the retina of MAPT*P301S mice and curbed pTau S396 phosphorylation in brains of systemically treated APOE4 knock-in mice. Targeting ApoE-HSPG interactions using 7C11 antibody may be a promising approach to developing new therapies for AD.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Mice , Humans , Animals , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Heparan Sulfate Proteoglycans/metabolism , Phosphorylation , Apolipoproteins E/metabolism , Alzheimer Disease/pathology , Immunologic Factors , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism
14.
Protein Sci ; 33(1): e4843, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37996967

ABSTRACT

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has caused a global health crisis with significant clinical morbidity and mortality. While angiotensin-converting enzyme 2 (ACE2) is the primary receptor for viral entry, other cell surface and extracellular matrix proteins may also bind to the viral receptor binding domain (RBD) within the SARS-CoV-2 spike protein. Recent studies have implicated heparan sulfate proteoglycans, specifically perlecan LG3, in facilitating SARS-CoV-2 binding to ACE2. However, the role of perlecan LG3 in SARS-CoV-2 pathophysiology is not well understood. In this study, we investigated the binding interactions between the SARS-CoV-2 spike protein RBD and perlecan LG3 through molecular modeling simulations and surface plasmon resonance (SPR) experiments. Our results indicate stable binding between LG3 and SARS-CoV-2 spike protein RBD, which may potentially enhance RBD-ACE2 interactions. These findings shed light on the role of perlecan LG3 in SARS-CoV-2 infection and provide insight into SARS-CoV-2 pathophysiology and potential therapeutic strategy for COVID-19.


Subject(s)
COVID-19 , Spike Glycoprotein, Coronavirus , Humans , Spike Glycoprotein, Coronavirus/chemistry , SARS-CoV-2/metabolism , Heparan Sulfate Proteoglycans/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Protein Binding
15.
J Control Release ; 365: 43-59, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37935257

ABSTRACT

Imprecise targeting of chemotherapeutic drugs often leads to severe toxicity during breast cancer therapy. To address this issue, we have devised a strategy to load dacarbazine (DC) into fucose-based carbon quantum dots (CQDs), which are subsequently coated with exosomes (Ex-DC@CQDs) derived from breast cancer cells. Nanoparticle tracking analysis and western blotting revealed that Ex-DC@CQDs retained the structural and functional characteristics of exosomes. We found that exosomes facilitated the transport of DC@CQDs to cancer cells via heparan sulfate proteoglycan (HSPG) receptors, followed by an augmented depolarization of the mitochondrial membrane potential, ROS generation, and induction of apoptosis leading to cell death. In vivo imaging and pharmacokinetic studies demonstrated enhanced antitumor targeting and efficacy compared to free DC which we attribute to an improved pharmacokinetic profile, a greater tumor accumulation via exosome-mediated- HSPG receptor-driven cell uptake, and sustained release of the Ex-DC@CQDs. Our findings may pave the way for the further development of biologically sourced nanocarriers for breast cancer targeting.


Subject(s)
Breast Neoplasms , Exosomes , Quantum Dots , Humans , Female , Quantum Dots/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Exosomes/metabolism , Dacarbazine , Heparan Sulfate Proteoglycans/metabolism , Carbon/chemistry
16.
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
17.
Immun Inflamm Dis ; 11(10): e1034, 2023 10.
Article in English | MEDLINE | ID: mdl-37904703

ABSTRACT

Heat stroke is a life-threatening disease with high mortality and complications. Endothelial glycocalyx (EGCX) is essential for maintaining endothelial cell structure and function as well as preventing the adhesion of inflammatory cells. Potential relationship that underlies the imbalance in inflammation and coagulation remains elusive. Moreover, the role of EGCX in heat stroke-induced organ injury remained unclear. Therefore, the current study aimed to illustrate if EGCX aggravates apoptosis, inflammation, and oxidative damage in human pulmonary microvascular endothelial cells (HPMEC). Heat stress and lipopolysaccharide (LPS) were employed to construct in vitro models to study the changes of glycocalyx structure and function, as well as levels of heparansulfate proteoglycan (HSPG), syndecan-1 (SDC-1), heparansulfate (HS), tumor necrosis factor-α (TNF-α), interleukin (IL)-6, Von Willebrand factor (vWF), endothelin-1 (ET-1), occludin, E-selectin, vascular cell adhesion molecule-1 (VCAM-1), and reactive oxygen species (ROS). Here, we showed that heat stress and LPS devastated EGCX structure, activated EGCX degradation, and triggered oxidative damage and apoptosis in HPMEC. Stimulation of heat stress and LPS decreased expression of HSPG, increased levels of SDC-1 and HS in culture supernatant, promoted the production and release of proinflammation cytokines (TNF-α and IL-6,) and coagulative factors (vWF and ET-1) in HPMEC. Furthermore, Expressions of E-selection, VCAM-1, and ROS were upregulated, while that of occludin was downregulated. These changes could be deteriorated by heparanase, whereas they meliorated by unfractionated heparin. This study indicated that EGCX may contribute to apoptosis and heat stroke-induced coagulopathy, and these effects may have been due to the decrease in the shedding of EGCX.


Subject(s)
Endothelial Cells , Heat Stroke , Humans , Endothelial Cells/metabolism , Glycocalyx/metabolism , Lipopolysaccharides/toxicity , Tumor Necrosis Factor-alpha/metabolism , Reactive Oxygen Species/metabolism , Heparin/metabolism , Heparin/pharmacology , von Willebrand Factor/metabolism , von Willebrand Factor/pharmacology , Heparan Sulfate Proteoglycans/metabolism , Heparan Sulfate Proteoglycans/pharmacology , Occludin/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/pharmacology , Inflammation/metabolism , Interleukin-6/pharmacology , Heat Stroke/metabolism , Heat-Shock Response
18.
J Control Release ; 362: 184-196, 2023 10.
Article in English | MEDLINE | ID: mdl-37648081

ABSTRACT

Growth factors are key molecules involved in angiogenesis, a process critical for tissue repair and regeneration. Despite the potential of growth factor delivery to stimulate angiogenesis, limited clinical success has been achieved with this approach. Growth factors interact with the extracellular matrix (ECM), and particularly heparan sulphate (HS), to bind and potentiate their signalling. Here we show that engineered short forms of perlecan, the major HS proteoglycan of the vascular ECM, bind and signal angiogenic growth factors, including fibroblast growth factor 2 and vascular endothelial growth factor-A. We also show that engineered short forms of perlecan delivered in porous chitosan biomaterial scaffolds promote angiogenesis in a rat full thickness dermal wound model, with the fusion of perlecan domains I and V leading to superior vascularisation compared to native endothelial perlecan or chitosan scaffolds alone. Together, this study demonstrates the potential of engineered short forms of perlecan delivered in chitosan scaffolds as next generation angiogenic therapies which exert biological activity via the potentiation of growth factors.


Subject(s)
Chitosan , Vascular Endothelial Growth Factor A , Rats , Animals , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Heparan Sulfate Proteoglycans/metabolism , Extracellular Matrix Proteins
19.
Int J Mol Sci ; 24(14)2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37511353

ABSTRACT

Retention of circulating lipoproteins by their interaction with extracellular matrix molecules has been suggested as an underlying mechanism for atherosclerosis. We investigated the role of glypican-4 (GPC4), a heparan sulfate (HS) proteoglycan, in the development of endothelial dysfunction and plaque progression; Expression of GPC4 and HS was investigated in human umbilical vein/artery endothelial cells (HUVECs/HUAECs) using flow cytometry, qPCR, and immunofluorescent staining. Leukocyte adhesion was determined in HUVECs in bifurcation chamber slides under dynamic flow. The association between the degree of inflammation and GPC4, HS, and syndecan-4 expressions was analyzed in human carotid plaques; GPC4 was expressed in HUVECs/HUAECs. In HUVECs, GPC4 protein expression was higher in laminar than in non-uniform shear stress regions after a 1-day or 10-day flow (p < 0.01 each). The HS expression was higher under laminar flow after a 1 day (p < 0.001). Monocytic THP-1 cell adhesion to HUVECs was facilitated by GPC4 knock-down (p < 0.001) without affecting adhesion molecule expression. GPC4 and HS expression was lower in more-inflamed than in less-inflamed plaque shoulders (p < 0.05, each), especially in vulnerable plaque sections; Reduced expression of GPC4 was associated with atherogenic conditions, suggesting the involvement of GPC4 in both early and advanced stages of atherosclerosis.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Humans , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cells, Cultured , Clinical Relevance , Glypicans/genetics , Glypicans/metabolism , Heparan Sulfate Proteoglycans/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism
20.
Biochem Soc Trans ; 51(3): 1083-1096, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37334846

ABSTRACT

Syndecans are transmembrane heparan sulfate proteoglycans present on most mammalian cell surfaces. They have a long evolutionary history, a single syndecan gene being expressed in bilaterian invertebrates. Syndecans have attracted interest because of their potential roles in development and disease, including vascular diseases, inflammation and various cancers. Recent structural data is providing important insights into their functions, which are complex, involving both intrinsic signaling through cytoplasmic binding partners and co-operative mechanisms where syndecans form a signaling nexus with other receptors such as integrins and tyrosine kinase growth factor receptors. While the cytoplasmic domain of syndecan-4 has a well-defined dimeric structure, the syndecan ectodomains are intrinsically disordered, which is linked to a capacity to interact with multiple partners. However, it remains to fully establish the impact of glycanation and partner proteins on syndecan core protein conformations. Genetic models indicate that a conserved property of syndecans links the cytoskeleton to calcium channels of the transient receptor potential class, compatible with roles as mechanosensors. In turn, syndecans influence actin cytoskeleton organization to impact motility, adhesion and the extracellular matrix environment. Syndecan clustering with other cell surface receptors into signaling microdomains has relevance to tissue differentiation in development, for example in stem cells, but also in disease where syndecan expression can be markedly up-regulated. Since syndecans have potential as diagnostic and prognostic markers as well as possible targets in some forms of cancer, it remains important to unravel structure/function relationships in the four mammalian syndecans.


Subject(s)
Heparan Sulfate Proteoglycans , Signal Transduction , Animals , Syndecans/chemistry , Syndecans/metabolism , Cell Membrane/metabolism , Heparan Sulfate Proteoglycans/metabolism , Receptors, Cell Surface/metabolism , Extracellular Matrix/metabolism , Mammals/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...