Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Matrix Biol ; 126: 25-42, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38232913

ABSTRACT

The inter-alpha-trypsin inhibitor (IαI) complex is composed of the bikunin core protein with a single chondroitin sulfate (CS) attached and one or two heavy chains (HCs) covalently linked to the CS chain. The HCs from IαI can be transferred to hyaluronan (HA) through a TNFα-stimulated gene-6 (TSG-6) dependent process to form an HC•HA matrix. Previous studies reported increased IαI, HA, and HC•HA complexes in mouse bronchoalveolar lavage fluid (BALF) post-influenza infection. However, the expression and incorporation of HCs into the HA matrix of the lungs during the clinical course of influenza A virus (IAV) infection and the biological significance of the HC•HA matrix are poorly understood. The present study aimed to better understand the composition of HC•HA matrices in mice infected with IAV and how these matrices regulate the host pulmonary immune response. In IAV infected mice bikunin, HC1-3, TSG-6, and HAS1-3 all show increased gene expression at various times during a 12-day clinical course. The increased accumulation of IαI and HA was confirmed in the lungs of infected mice using immunohistochemistry and quantitative digital pathology. Western blots confirmed increases in the IαI components in BALF and lung tissue at 6 days post-infection (dpi). Interestingly, HCs and bikunin recovered from BALF and plasma from mice 6 dpi with IAV, displayed differences in the HC composition by Western blot analysis and differences in bikunin's CS chain sulfation patterns by mass spectrometry analysis. This strongly suggests that the IαI components were synthesized in the lungs rather than translocated from the vascular compartment. HA was significantly increased in BALF at 6 dpi, and the HA recovered in BALF and lung tissues were modified with HCs indicating the presence of an HC•HA matrix. In vitro experiments using polyinosinic-polycytidylic acid (poly(I:C)) treated mouse lung fibroblasts (MLF) showed that modification of HA with HCs increased cell-associated HA, and that this increase was due to the retention of HA in the MLF glycocalyx. In vitro studies of leukocyte adhesion showed differential binding of lymphoid (Hut78), monocyte (U937), and neutrophil (dHL60) cell lines to HA and HC•HA matrices. Hut78 cells adhered to immobilized HA in a size and concentration-dependent manner. In contrast, the binding of dHL60 and U937 cells depended on generating a HC•HA matrix by MLF. Our in vivo findings, using multiple bronchoalveolar lavages, correlated with our in vitro findings in that lymphoid cells bound more tightly to the HA-glycocalyx in the lungs of influenza-infected mice than neutrophils and mononuclear phagocytes (MNPs). The neutrophils and MNPs were associated with a HC•HA matrix and were more readily lavaged from the lungs. In conclusion, this work shows increased IαI and HA accumulation and the formation of a HC•HA matrix in mouse lungs post-IAV infection. The formation of HA and HC•HA matrices could potentially create specific microenvironments in the lungs for immune cell recruitment and activation during IAV infection.


Subject(s)
Alpha-Globulins , Influenza, Human , Orthomyxoviridae , Mice , Animals , Humans , Hyaluronic Acid/metabolism , Chondroitin Sulfates/metabolism , Lung/metabolism , Orthomyxoviridae/metabolism , Immunity, Innate , Disease Progression
2.
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
3.
Chem Biol Drug Des ; 102(4): 782-792, 2023 10.
Article in English | MEDLINE | ID: mdl-37455326

ABSTRACT

Amentoflavone (AF) is a natural multifunctional biflavonoid that has been revealed to possess multiple biological activities, including anticancer activity. Here, this work focused on exploring the functions and mechanism of AF in gastric cancer (GC). Levels of genes and proteins were examined by quantitative real-time PCR and western blotting. Cell proliferation and cell death were analyzed using cell counting kit-8, colony formation, and lactate dehydrogenase (LDH) release assay, respectively. Cell ferroptosis was evaluated by detecting the levels of malondialdehyde (MDA), reduced glutathione (GSH), Fe2+ , and intracellular reactive oxygen species (ROS). The binding between miR-496 and activating transcription factor 2 (ATF2) was confirmed by using dual-luciferase reporter assay. Murine xenograft assay was conducted for in vivo experiments. The results showed that AF suppressed the proliferation and induced ferroptotic cell death in GC cells. MiR-496 expression was decreased in GC tissues and cells, and AF treatment increased miR-496 expression level in GC cells. Functionally, miR-496 inhibition reversed the inhibitory effects of AF on GC cell proliferation and promoting effects on ferroptotic cell death. Mechanistically, ATF2 was targeted by miR-496. ATF2 expression was increased in GC tissues and cells, which was decreased by AF treatment and subsequently rescued by miR-496 downregulation in GC cells. Moreover, miR-496 overexpression suppressed the proliferation and induced ferroptotic cell death in GC cells via targeting ATF2. In all, AF suppressed the proliferation and induced ferroptotic cell death in GC cells via miR-496/ATF2 axis, indicating a novel therapeutic approach for GC patients.


Subject(s)
Biflavonoids , Ferroptosis , MicroRNAs , Stomach Neoplasms , Humans , Animals , Mice , Biflavonoids/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Stomach Neoplasms/drug therapy , Activating Transcription Factor 2/genetics , Cell Line, Tumor , Cell Proliferation
4.
Am J Physiol Cell Physiol ; 323(2): C249-C276, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35649251

ABSTRACT

The extracellular matrix (ECM) imparts critical mechanical and biochemical information to cells in the lungs. Proteoglycans are essential constituents of the ECM and play a crucial role in controlling numerous biological processes, including regulating cellular phenotype and function. Versican, a chondroitin sulfate proteoglycan required for embryonic development, is almost absent from mature, healthy lungs and is reexpressed and accumulates in acute and chronic lung disease. Studies using genetically engineered mice show that the versican-enriched matrix can be pro- or anti-inflammatory depending on the cellular source or disease process studied. The mechanisms whereby versican develops a contextual ECM remain largely unknown. The primary goal of this review is to provide an overview of the interaction of versican with its many binding partners, the "versican interactome," and how through these interactions, versican is an integrator of complex extracellular information. Hopefully, the information provided in this review will be used to develop future studies to determine how versican and its binding partners can develop contextual ECMs that control select biological processes. Although this review focuses on versican and the lungs, what is described can be extended to other proteoglycans, tissues, and organs.


Subject(s)
Extracellular Matrix , Versicans , Animals , Extracellular Matrix/metabolism , Lung/metabolism , Mice , Versicans/genetics , Versicans/metabolism
5.
ACS Biomater Sci Eng ; 8(2): 512-525, 2022 02 14.
Article in English | MEDLINE | ID: mdl-34989230

ABSTRACT

Angiogenesis plays a key role in cancer progression, including transition to the metastatic phase via reactive oxygen species (ROS)-dependent pathways, among others. Antivascular endothelial growth factor (VEGF) antibodies have been trialed as an anti-angiogenic therapy for cancer but are associated with high cost, limited efficacy, and side effects. Cerium oxide nanoparticles (nanoceria) are promising nanomaterials for biomedical applications due to their ability to modulate intracellular ROS. Nanoceria can be produced by a range of synthesis methods, with chemical precipitation as the most widely explored. It has been reported that chemical precipitation can fine-tune primary particle size where a limited number of synthesis parameters were varied. Here, we explore the effect of temperature, precipitating agent concentration and rate of addition, stirring rate, and surfactant concentration on nanoceria primary particle size using a fractional factorial experimental design approach. We establish a robust synthesis method for faceted nanoceria with primary particle diameters of 5-6 nm. The nanoceria are not cytotoxic to a human melanoma cell line (Mel1007) at doses up to 400 µg/mL and are dose-dependently internalized by the cells. The intracellular ROS level for some cells that internalized the nanoceria is reduced, which correlates with a dose-dependent reduction in angiogenic gene expression including VEGF. These findings contribute to our knowledge of the anti-angiogenic effects of nanoceria and help to develop our understanding of potentially new anti-angiogenic agents for combination cancer therapies.


Subject(s)
Cerium , Melanoma , Nanoparticles , Cerium/pharmacology , Humans , Melanoma/drug therapy , Reactive Oxygen Species/metabolism
6.
Acta Biomater ; 132: 162-175, 2021 09 15.
Article in English | MEDLINE | ID: mdl-33588126

ABSTRACT

Blood compatible materials are required for the development of therapeutic and diagnostic blood contacting devices as blood-material interactions are a key factor dictating device functionality. In this work, we explored biofunctionalization of silk biomaterials with a recombinantly expressed domain V of the human basement membrane proteoglycan perlecan (rDV) towards the development of blood compatible surfaces. Perlecan and rDV are of interest in vascular device development as they uniquely support endothelial cell, while inhibiting smooth muscle cell and platelet interactions. rDV was covalently immobilized on silk biomaterials using plasma immersion ion implantation (PIII), a new method of immobilizing proteins on silk biomaterials that does not rely on modification of specific amino acids in the silk protein chain, and compared to physisorbed and carbodiimide immobilized rDV. Untreated and treated silk biomaterials were examined for interactions with blood components with varying degrees of complexity, including isolated platelets, platelet rich plasma, blood plasma, and whole blood, both under agitated and flow conditions. rDV-biofunctionalized silk biomaterials were shown to be blood compatible in terms of platelet and whole blood interactions and the PIII treatment was shown to be an effective and efficient means of covalently immobilizing rDV in its bioactive form. These biomimetic silk biomaterials are a promising platform toward development of silk-based blood-contacting devices for therapeutic, diagnostic, and research applications. STATEMENT OF SIGNIFICANCE: Blood compatible materials are required for the development of therapeutic and diagnostic blood contacting devices as blood-material interactions are a key factor dictating device functionality. In this work, we explored biofunctionalization of silk biomaterials with a recombinantly expressed domain V (rDV) of the human basement membrane proteoglycan perlecan towards the development of blood compatible surfaces. Perlecan and rDV are of interest in vascular device development as they uniquely support endothelial cell, while inhibiting smooth muscle cell and platelet interactions. rDV was covalently immobilized on silk biomaterials using plasma immersion ion implantation (PIII), a new method of immobilizing proteins on silk biomaterials that does not rely on modification of specific amino acids in the silk protein chain. These biomimetic silk biomaterials are a promising platform toward development of silk-based blood-contacting devices for therapeutic, diagnostic, and research applications.


Subject(s)
Fibroins , Silk , Biocompatible Materials , Biomimetics , Heparan Sulfate Proteoglycans , Humans
7.
Biomater Sci ; 8(24): 7093-7105, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33079079

ABSTRACT

The biophysical properties of biomaterials are key to directing the biological responses and biomaterial integration and function in in situ tissue engineering approaches. We present silk photo-lyogels, a biomaterial format fabricated using a new combinatorial approach involving photo-initiated crosslinking of silk fibroin via di-tyrosine bonds followed by lyophilization to generate 3D, porous lyogels showing physical properties distinct to those of lyophilized silk sponges or silk hydrogels. This fabrication approach allowed introduction of microchannels into 3D constructs via biofabrication approaches involving silk crosslinking around an array of 3D printed photocurable resin pillars to generate parallel channels or around a 3D printed sacrificial thermosensitive gel to generate interconnected channels in a rapid manner and without the need for chemical modification of silk fibroin. The presence of interconnected microchannels significantly improved migration of endothelial cells into 3D photo-lyogels in vitro, and tissue infiltration, photo-lyogel integration, and vascularization when implanted in vivo in a mouse subcutaneous model. Taken together, these findings demonstrate the feasibility and utility of a new combinatorial fabrication approach for generation of silk biomaterials that support cell interactions and implant integration for in situ tissue engineering approaches.


Subject(s)
Fibroins , Animals , Biocompatible Materials , Endothelial Cells , Mice , Silk , Tissue Engineering , Tissue Scaffolds
8.
Adv Sci (Weinh) ; 7(17): 2000900, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32995122

ABSTRACT

Angiogenic therapy involving delivery of pro-angiogenic growth factors to stimulate new blood vessel formation in ischemic disease is promising but has seen limited clinical success due to issues associated with the need to deliver supra-physiological growth factor concentrations. Bio-inspired growth factor delivery utilizing the native growth factor signaling roles of the extracellular matrix proteoglycans has the potential to overcome many of the drawbacks of angiogenic therapy. In this study, the potential of the recombinantly expressed domain V (rDV) of human perlecan is investigated as a means of promoting growth factor signaling toward enhanced angiogenesis and vascularization of implanted biomaterials. rDV is found to promote angiogenesis in established in vitro and in vivo angiogenesis assays by potentiating endogenous growth factor signaling via its glycosaminoglycan chains. Further, rDV is found to potentiate fibroblast growth factor 2 (FGF2) signaling at low concentrations that in the absence of rDV are not biologically active. Finally, rDV immobilized on 3D porous silk fibroin biomaterials promotes enhanced vascular ingrowth and integration of the implanted scaffolds with the surrounding tissue. Together, these studies demonstrate the important role of this biologically active perlecan fragment and its potential in the treatment of ischemia in both native and bioengineered tissues.

9.
ACS Biomater Sci Eng ; 6(3): 1476-1486, 2020 03 09.
Article in English | MEDLINE | ID: mdl-33455399

ABSTRACT

Functional integration of implanted biomaterials and bioengineered tissues in vivo requires effective and timely vascular ingrowth. While many vascularization strategies rely on delivery of angiogenic growth factors or endothelial cells to promote vascular ingrowth, the effect of physical and architectural features of biomaterials on the vascularization process is less well understood. Microchannels are a simple, accessible architectural feature frequently engineered into 3D biomaterials to promote mass transfer. In this study, the effect of microchannels on the integration and vascularization of 3D porous silk scaffolds was explored over a 14 week period. An array of 508 µm diameter microchannels spanning the length of critically sized, porous silk scaffolds significantly improved tissue ingrowth into the constructs. At week 6, all silk scaffolds (n = 8) with microchannels showed complete tissue infiltration throughout the construct, while only one of eight (12.5%) did so in the absence of microchannels. The presence of microchannels improved silk scaffold vascularization with significantly more vessels per unit area in the presence of microchannels. The vessel size distribution was similar in both scaffold types, but a shift in distribution toward smaller vessels was observed in the presence of microchannels. The blood vessels in silk scaffolds were perfused, functional and connected to the animal's cardiovascular system, as demonstrated by the presence of red blood cells in the vessel lumens, and effective delivery of a contrast agent the vessels inside the scaffold. This study demonstrates the utility of microchannels as a simple architectural feature that significantly improves vascularization and integration of implanted biomaterials.


Subject(s)
Biocompatible Materials , Silk , Animals , Cues , Endothelial Cells , Tissue Scaffolds
10.
Cell Prolif ; 52(2): e12547, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30657238

ABSTRACT

OBJECTIVE: Chinese Herb QingBai decoction (QBD) has been approved affective in the treatment of IBD patients in clinic. However, the underlying mechanism remains unknown. We aim to investigate the effect of QBD on the mouse model of ulcerative colitis and its possible mechanism. METHODS: C57/bL mice were given 5% DSS to induce colitis and were divided as QBD and mesalazine group. Weight, faeces and mental status were recorded each day and the histopathological changes (goblet cells etc) of the colon were observed after sacrificed. Fluorescein isothiocyanate-dextran 4000 was measured to reflect the intestinal mucosal permeability. In addition, cell junction-related proteins and possible signal pathways were investigated. RESULTS: QingBai decoction could significantly alleviate the inflammation and the protection effect of colitis is comparable as those in mesalazine enema group. It was found that the permeability reduced significantly with QBD treatment vs the control group, while no significant difference between the mesalazine and QBD groups. QBD treatment could upregulate the expression of tight junction complex(ZO-1, claudin-1 and occludin)and muc-2 expression. It significantly reduced the production and secretion of serials proinflammatory cytokines (IL-1ß, IL-6, Kc and TNF-α) compared with the control group. Meanwhile, NF-κB and Notch pathways were regulated. CONCLUSION: QingBai decoction can effectively alleviate intestinal inflammation and mucosal barrier function in colitis mice, and the mechanism may be related to the inhibition of inflammatory cascade as well as enhanced mucus layer barrier and mechanical barrier function by NF-κB and Notch signalling.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis, Ulcerative/drug therapy , Drugs, Chinese Herbal/therapeutic use , Intestinal Absorption/drug effects , Intestines/drug effects , NF-kappa B/immunology , Animals , Apoptosis/drug effects , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Cytokines/analysis , Cytokines/immunology , Dextran Sulfate , Disease Models, Animal , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestines/immunology , Intestines/pathology , Mice, Inbred C57BL , NF-kappa B/analysis , Permeability/drug effects , Receptors, Notch/analysis , Receptors, Notch/immunology , Signal Transduction/drug effects
11.
ACS Appl Mater Interfaces ; 10(21): 17605-17616, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29733628

ABSTRACT

Silk fibroin isolated from Bombyx mori cocoons is a promising material for a range of biomedical applications, but it has no inherent cell-interactive domains, necessitating functionalization with bioactive molecules. Here we demonstrate significantly enhanced cell interactions with silk fibroin biomaterials in the absence of biofunctionalization following surface modification using plasma immersion ion implantation (PIII). Further, PIII treated silk fibroin biomaterials supported direct covalent immobilization of proteins on the material surface in the absence of chemical cross-linkers. Surface analysis after nitrogen plasma and PIII treatment at 20 kV revealed that the silk macromolecules are significantly fragmented, and at the higher fluences of implanted ions, surface carbonization was observed to depths corresponding to that of the ion penetration. Consistent with the activity of radicals created in the treated surface layer, oxidation was observed on contact with atmospheric oxygen and the PIII treated surfaces were capable of direct covalent immobilization of bioactive macromolecules. Changes in thickness, amide and nitrile groups, refractive index, and extinction coefficient in the wavelength range 400-1000 nm as a function of ion fluence are presented. Reactions responsible for the restructuring of the silk surface under ion beam treatment that facilitate covalent binding of proteins and a significant improvement in cell interactions on the modified surface are proposed.


Subject(s)
Silk , Animals , Biocompatible Materials , Bombyx , Fibroins , Ions , Prostheses and Implants
12.
Matrix Biol ; 68-69: 150-166, 2018 08.
Article in English | MEDLINE | ID: mdl-29475023

ABSTRACT

Perlecan, or heparan sulfate proteoglycan 2 (HSPG2), is a ubiquitous heparan sulfate proteoglycan that has major roles in tissue and organ development and wound healing by orchestrating the binding and signaling of mitogens and morphogens to cells in a temporal and dynamic fashion. In this review, its roles in fibrosis are reviewed by drawing upon evidence from tissue and organ systems that undergo fibrosis as a result of an uncontrolled response to either inflammation or traumatic cellular injury leading to an over production of a collagen-rich extracellular matrix. This review focuses on examples of fibrosis that occurs in lung, liver, kidney, skin, kidney, neural tissues and blood vessels and its link to the expression of perlecan in that particular organ system.


Subject(s)
Collagen/metabolism , Heparan Sulfate Proteoglycans/metabolism , Regeneration , Animals , Cell Adhesion , Cell Movement , Cell Proliferation , Extracellular Matrix/metabolism , Fibrosis , Heparan Sulfate Proteoglycans/chemistry , Humans
13.
J Biochem ; 163(5): 399-412, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29462330

ABSTRACT

Chondroitin sulphate proteoglycan 4 (CSPG4) is a cell surface proteoglycan highly expressed by tumour, perivascular and oligodendrocyte cells and known to be involved cell adhesion and migration. This study showed that CSPG4 was present as a proteoglycan on the cell surface of two melanoma cell lines, MM200 and Me1007, as well as shed into the conditioned medium. CSPG4 from the two melanoma cell lines differed in the amount of chondroitin sulphate (CS) decoration, as well as the way the protein core was fragmented. In contrast, the CSPG4 expressed by a colon carcinoma cell line, WiDr, was predominantly as a protein core on the cell surface lacking glycosaminoglycan (GAG) chains. This study demonstrated that CSPG4 immunopurified from the melanoma cell lines formed a complex with perlecan synthesized by the same cultured cells. Mechanistic studies showed that CSPG4 bound to perlecan via hydrophobic protein-protein interactions involving multiple sites on perlecan including the C-terminal region. Furthermore, this study revealed that CSPG4 interacted with perlecan to support cell adhesion and actin polymerization. Together these data suggest a novel mechanism by which CSPG4 expressing cells might attach to perlecan-rich matrices so as those found in connective tissues and basement membranes.


Subject(s)
Cell Adhesion , Chondroitin Sulfate Proteoglycans/metabolism , Heparan Sulfate Proteoglycans/metabolism , Membrane Proteins/metabolism , Binding Sites , Chondroitin Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/chemistry , Humans , Membrane Proteins/chemistry , Protein Binding , Tumor Cells, Cultured
14.
Adv Healthc Mater ; 7(6): e1701042, 2018 03.
Article in English | MEDLINE | ID: mdl-29210510

ABSTRACT

Proteoglycans and their glycosaminoglycans (GAG) are essential for life as they are responsible for orchestrating many essential functions in development and tissue homeostasis, including biophysical properties and roles in cell signaling and extracellular matrix assembly. In an attempt to capture these biological functions, a range of biomaterials are designed to incorporate off-the-shelf GAGs, typically isolated from animal sources, for tissue engineering, drug delivery, and regenerative medicine applications. All GAGs, with the exception of hyaluronan, are present in the body covalently coupled to the protein core of proteoglycans, yet the incorporation of proteoglycans into biomaterials remains relatively unexplored. Proteoglycan-based biomaterials are more likely to recapitulate the unique, tissue-specific GAG profiles and native GAG presentation in human tissues. The protein core offers additional biological functionality, including cell, growth factor, and extracellular matrix binding domains, as well as sites for protein immobilization chemistries. Finally, proteoglycans can be recombinantly expressed in mammalian cells and thus offer genetic manipulation and metabolic engineering opportunities for control over the protein and GAG structures and functions. This Progress Report summarizes current developments in GAG-based biomaterials and presents emerging research and future opportunities for the development of biomaterials that incorporate GAGs presented in their native proteoglycan form.


Subject(s)
Biocompatible Materials , Glycosaminoglycans , Metabolic Engineering/methods , Proteoglycans , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Glycosaminoglycans/biosynthesis , Glycosaminoglycans/chemistry , Glycosaminoglycans/genetics , Humans , Proteoglycans/biosynthesis , Proteoglycans/chemistry , Proteoglycans/genetics
15.
Biomed Pharmacother ; 96: 877-883, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29145164

ABSTRACT

Long non-coding RNAs (lncRNAs) have been identified to play critical roles in tumorigenesis. LncRNA HNF1A-AS1 has been suggested to act as an oncogene and serves as a novel prognostic biomarker for various cancer. However, the biological role and clinical significance of lncRNA HNF1A-AS1 in colorectal cancer (CRC) have yet to be fully elusive. Therefore, the present study was designed to determine the expression of lncRNA HNF1A-AS1 in patients with CRC, the role of lncRNA HNF1A-AS1 in CRC cells, as well as the underlying regulatory mechanisms. Our results indicated that the expression of lncRNA HNF1A-AS1 was significantly upregulated in both CRC tumor tissues and CRC cell lines in comparison with adjacent non-tumor tissues and the human normal colonic epithelial cell line (HcoEpiC). The Kaplan-Meier survival analysis further suggested that high expression of lncRNA HNF1A-AS1 might be an independent prognostic factor for disease-free survival (DFS) and overall survival (OS) in patients with CRC. Moreover, the area under the receiver operating characteristic (ROC) curve for HNF1A-AS1 was up to 0.8714, implying that HNF1A-AS1 had diagnostic significance as it could discriminate tumor tissues from nontumorous tissues. In addition, silencing of lncRNA HNF1A-AS1 abrogated the proliferation of CRC cells by MTS assay and clonogenic assay, arrested cell cycle at G0/G1 stage and reduced the migration and invasion in CRC cells. Finally, we found that decreased expression of lncRNA HNF1A-AS1 suppressed the Wnt/ß-catenin signaling pathway activity by downregulating the expression of ß-catenin,cyclinD1, and c-myc. In conclusion, these findings provide evidence that lncRNA HNF1A-AS1 may be considered as a new prognostic biomarker and therapeutic target in patients with CRC.


Subject(s)
Carcinogenesis/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , RNA, Long Noncoding/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease-Free Survival , Down-Regulation/genetics , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , Humans , Kaplan-Meier Estimate , Prognosis , Resting Phase, Cell Cycle/genetics , Up-Regulation/genetics
16.
Biotechnol J ; 12(12)2017 Dec.
Article in English | MEDLINE | ID: mdl-28846206

ABSTRACT

The C-terminal domain V of the extracellular matrix proteoglycan perlecan plays unique and often divergent roles in a number of biological processes, including angiogenesis, vascular cell interactions, wound healing, and autophagy. Recombinant forms of domain V have been proposed as therapeutic agents for the treatment of cancer, stroke, and the development of cardiovascular devices and bioartificial tissues. However, the effect of domain V appears to be related to the differences in domain V structure and function observed in different expression systems and environments and exactly how this occurs is not well understood. In this study, the sequence from amino acid 3626 to 4391 of the perlecan protein core, which includes domain V, is expressed in HEK-293 cells and purified as a secreted product from conditioned media. This recombinant domain V (rDV) is expressed as a proteoglycan decorated with heparan sulfate and chondroitin sulfate chains and supports endothelial cell interactions to the same extent as full-length perlecan. This expression system serves as an important model of recombinant proteoglycan expression, as well as a source of biologically active rDV for therapeutic applications.


Subject(s)
Endothelial Cells/drug effects , Heparan Sulfate Proteoglycans/pharmacology , Recombinant Fusion Proteins/pharmacology , Cell Adhesion/drug effects , Cell Line , Endothelial Cells/metabolism , HEK293 Cells , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Humans , Protein Domains/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
17.
Colloids Surf B Biointerfaces ; 148: 130-138, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27591944

ABSTRACT

Modulation of endothelial cell and platelet interactions is an essential feature of vascular materials. Silk biomaterials were functionalized with recombinantly expressed domain V of human perlecan, an essential vascular proteoglycan involved in vasculogenesis, angiogenesis and wound healing, using passive adsorption or covalent cross-linking via carbodiimide chemistry. The orientation of domain V on the surface of silk biomaterials was modulated by the immobilization technique and glycosaminoglycan chains played an essential role in the proteoglycan presentation on the material surface. Covalent immobilization supported improved integrin binding site presentation to endothelial cells compared to passive adsorption in the presence of glycosaminoglycan chains, but removal of glycosaminoglycan chains resulted in reduced integrin site availability and thus cell binding. Silk biomaterials covalently functionalized with domain V supported endothelial cell adhesion, spreading and proliferation and were anti-adhesive for platelets, making them promising surfaces for the development of the next-generation vascular grafts.


Subject(s)
Biocompatible Materials , Blood Platelets/cytology , Cell Adhesion , Endothelium, Vascular/cytology , Heparan Sulfate Proteoglycans/chemistry , Silk , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Recombinant Proteins/chemistry
18.
Metab Eng ; 38: 105-114, 2016 11.
Article in English | MEDLINE | ID: mdl-27445159

ABSTRACT

Heparin is a carbohydrate anticoagulant used clinically to prevent thrombosis, however impurities can limit its efficacy. Here we report the biosynthesis of heparin-like heparan sulfate via the recombinant expression of human serglycin in human cells. The expressed serglycin was also decorated with chondroitin/dermatan sulfate chains and the relative abundance of these glycosaminoglycan chains changed under different concentrations of glucose in the culture medium. The recombinantly expressed serglycin produced with 25mM glucose present in the culture medium was found to possess anticoagulant activity one-seventh of that of porcine unfractionated heparin, demonstrating that bioengineered human heparin-like heparan sulfate may be a safe next-generation pharmaceutical heparin.


Subject(s)
Blood Coagulation/drug effects , Genetic Engineering/methods , Heparin/analogs & derivatives , Proteoglycans/administration & dosage , Proteoglycans/biosynthesis , Vesicular Transport Proteins/administration & dosage , Vesicular Transport Proteins/biosynthesis , Anticoagulants/administration & dosage , Anticoagulants/metabolism , HEK293 Cells , Heparin/administration & dosage , Heparin/biosynthesis , Heparin/genetics , Humans , Metabolic Engineering , Proteoglycans/genetics , Vesicular Transport Proteins/genetics
19.
Ren Fail ; 38(2): 262-7, 2016.
Article in English | MEDLINE | ID: mdl-26627442

ABSTRACT

End-stage renal disease (ESRD) was defined as start of renal replacement therapy or death due to kidney disease. However, death due to acute kidney injury was not included. It typically occurs when chronic renal failure progresses to a point where the kidneys are permanently functioning at less than 10% of their capacity. Oxidative stress (OS) plays a crucial role in ESRD. Nicotinamide adenine dinucleotide phosphate (NADPH) is one of the most important enzymes during oxidative stress. Cytochrome b light chain (CYBA), encoded by a polymorphic gene, which is a critical component of the nicotinamide adenine dinucleotide (NADH)/NADPH oxidase system and plays an important role in electron transport and superoxide anion production, is located on chromosome band 16q24 and has six exons spanning almost 7.76 kb of genomic DNA. CYBA gene polymorphisms can influence the activity of NADPH oxidase. To evaluate the association between CYBA gene polymorphisms and ESRD, we genotyped five CYBA polymorphisms using TaqMan allelic discrimination assay on DNA samples from 306 healthy controls and 332 patients with ESRD. Our results suggested that rs1049255 polymorphism of CYBA modified the risk of ESRD (p = 0.019; OR = 0.625; 95%CI = 0.424-0.921). GG genotype and G allele might be a protective factor against the risk of ESRD, especially in patients with chronic glomerulonephritis.


Subject(s)
Kidney Failure, Chronic/genetics , NADPH Oxidases/genetics , Polymorphism, Single Nucleotide , Case-Control Studies , Chronic Disease , Female , Glomerulonephritis/complications , Humans , Kidney Failure, Chronic/epidemiology , Kidney Failure, Chronic/etiology , Male , Middle Aged , Risk Assessment
20.
Dis Markers ; 29(2): 89-93, 2010.
Article in English | MEDLINE | ID: mdl-21045268

ABSTRACT

BACKGROUND: The development of cardiovascular disease in ESRD patients is considered to be associated with oxidative stress. NAD(P)H oxidase has attracted attention as mechanisms of generating oxidative stress. We investigated the relation between the genotype of the C242T CYBA polymorphism of the NADPH oxidase and the development of cardiovascular disease in ESRD patients. METHODS: A total of 289 ESRD patients were recruited and allocated to one of the two groups: patients without cardiovascular disease (group N; n=192) and patients developing cardiovascular disease (group D; n=97). The C242T CYBA polymorphism was determined by RFLP-PCR methods. RESULTS: The frequency of the C242T CT+TT genotype was significantly lower in group D than in group N (9.1 vs. 20.2%). In multiple Logistic regression analysis, systolic blood pressure, smoking history and this gene polymorphism were shown to be independent variables for the development of cardiovascular disease in ESRD patients. CONCLUSIONS: These results suggest that assessment of the C242T CYBA polymorphism of the NADPH oxidase may be useful in identifying the risk for developing cardiovascular disease in ESRD patients.


Subject(s)
Cardiovascular Diseases/genetics , Kidney Failure, Chronic/genetics , NADPH Oxidases/genetics , Polymorphism, Genetic , Cardiovascular Diseases/etiology , Case-Control Studies , Genetic Association Studies , Genotype , Humans , Kidney Failure, Chronic/complications , Logistic Models , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...