Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 400
Filter
1.
PLoS One ; 15(11): e0242798, 2020.
Article in English | MEDLINE | ID: mdl-33253286

ABSTRACT

Dermatopontin (DPT) is an extracellular matrix (ECM) protein with diversified pharmaceutical applications. It plays important role in cell adhesion/migration, angiogenesis and ECM maintenance. The recombinant production of this protein will enable further exploration of its multifaceted functions. In this study, DPT protein has been expressed in Escherichia coli (E.coli) aiming at cost effective recombinant production. The E.coli GJ1158 expression system was transformed with constructed recombinant vector (pRSETA-DPT) and protein was expressed as inclusion bodies on induction with NaCl. The inclusion bodies were solubilised in urea and renaturation of protein was done by on-column refolding procedure in Nickel activated Sepharose column. The refolded Histidine-tagged DPT protein was purified and eluted from column using imidazole and its purity was confirmed by analytical techniques. The biological activity of the protein was confirmed by collagen fibril assay, wound healing assay and Chorioallantoic Membrane (CAM) angiogenesis assay on comparison with standard DPT. The purified DPT was found to enhance the collagen fibrillogenesis process and improved the migration of human endothelial cells. About 73% enhanced wound closure was observed in purified DPT treated endothelial cells as compared to control. The purified DPT also could induce neovascularisation in the CAM model. At this stage, scaling up the production process for DPT with appropriate purity and reproducibility will have a promising commercial edge.


Subject(s)
Chondroitin Sulfate Proteoglycans/genetics , Cloning, Molecular , Extracellular Matrix Proteins/genetics , Recombinant Proteins/genetics , Cell Movement/genetics , Chondroitin Sulfate Proteoglycans/biosynthesis , Endothelial Cells/metabolism , Escherichia coli/genetics , Extracellular Matrix Proteins/biosynthesis , Humans , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Protein Folding , Recombinant Proteins/biosynthesis , Wound Healing/genetics
2.
Glia ; 67(1): 125-145, 2019 01.
Article in English | MEDLINE | ID: mdl-30394599

ABSTRACT

Following spinal cord injury (SCI), the population of mature oligodendrocytes undergoes substantial cell death; promoting their preservation and replacement is a viable strategy for preserving axonal integrity and white matter repair in the injured spinal cord. Dramatic upregulation of matrix chondroitin sulfate proteoglycans (CSPGs) is shown to pose an obstacle to endogenous repair processes, and targeting CSPGs improves functional recovery after SCI. However, the cellular and molecular mechanisms underlying the inhibitory effects of CSPGs remain largely undefined. Modulation of CSPGs specific signaling receptors, leukocyte common antigen-related (LAR), and protein tyrosine phosphatase-sigma (PTPσ) allows us to uncover the role and mechanisms of CSPGs in regulating oligodendrocytes in SCI. Here, utilizing specific functionally blocking peptides in a clinically relevant model of contusive/compressive SCI in the rat, we demonstrate that inhibition of PTPσ and LAR receptors promotes oligodendrogenesis by endogenous precursor cells, attenuates caspase 3-mediated cell death in mature oligodendrocytes, and preserves myelin. In parallel in vitro systems, we have unraveled that CSPGs directly induce apoptosis in populations of neural precursor cells and oligodendrocyte progenitor cells and limit their ability for oligodendrocyte differentiation, maturation, and myelination. These negative effects of CSPGs are mediated through the activation of both LAR and PTPσ receptors and the downstream Rho/ROCK pathway. Thus, we have identified a novel inhibitory role for PTPσ and LAR in regulating oligodendrocyte differentiation and apoptosis in the injured adult spinal cord and a new feasible therapeutic strategy for optimizing endogenous cell replacement following SCI.


Subject(s)
Oligodendroglia/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Spinal Cord Injuries/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Cells, Cultured , Chondroitin Sulfate Proteoglycans/biosynthesis , Female , Mice , Mice, Inbred C57BL , Oligodendroglia/drug effects , Oligodendroglia/pathology , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology
3.
Glia ; 67(1): 68-77, 2019 01.
Article in English | MEDLINE | ID: mdl-30453391

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system (CNS), characterized by inflammation-mediated demyelination, axonal injury and neurodegeneration. The mechanisms underlying impaired neuronal function are not fully understood, but evidence is accumulating that the presence of the gliotic scar produced by reactive astrocytes play a critical role in these detrimental processes. Here, we identified astrocytic Transient Receptor Potential cation channel, subfamily M, member 7 (TRPM7), a Ca2+ -permeable nonselective cation channel, as a novel player in the formation of a gliotic scar. TRPM7 was found to be highly expressed in reactive astrocytes within well-characterized MS lesions and upregulated in primary astrocytes under chronic inflammatory conditions. TRPM7 overexpressing astrocytes impaired neuronal outgrowth in vitro by increasing the production of chondroitin sulfate proteoglycans, a key component of the gliotic scar. These findings indicate that astrocytic TRPM7 is a critical regulator of the formation of a gliotic scar and provide a novel mechanism by which reactive astrocytes affect neuronal outgrowth.


Subject(s)
Astrocytes/metabolism , Chondroitin Sulfate Proteoglycans/biosynthesis , Multiple Sclerosis/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/biosynthesis , TRPM Cation Channels/biosynthesis , Adult , Aged , Aged, 80 and over , Animals , Cells, Cultured , Chondroitin Sulfate Proteoglycans/genetics , Female , Humans , Male , Middle Aged , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Neurons/pathology , Protein Serine-Threonine Kinases/genetics , Rats , TRPM Cation Channels/genetics
4.
J Biomol Struct Dyn ; 36(3): 679-688, 2018 02.
Article in English | MEDLINE | ID: mdl-28278758

ABSTRACT

Chondroitin sulfate proteoglycans (CSPGs) are potent inhibitors of growth in the adult central nervous system. Use of the enzyme chondroitinase ABC I (ChABC I) as a strategy to reduce CSPG inhibition in experimental models of spinal cord injury has led to observations of its remarkable capacity for repair. More importantly, ChABC therapy has been demonstrated to promote significant recovery of function to spinal injured animals. Despite this incomparable function of ChABC I, its clinical application has been limited because of its thermal instability as reported in the literature. In a recent study by Nazari-Robati et al., thermal stability of ChABC I was improved by protein engineering using site-directed mutagenesis method. Here, in this study, molecular dynamics simulations were used to take a closer look into the phenomenon leading to the experimentally observed thermal stability improvement followed by the corresponding site-directed mutagenesis. We concluded that the mutations induce local flexibility along with a re-conformation into the native structure which consequently increase the protein thermal stability.


Subject(s)
Chondroitin ABC Lyase/chemistry , Chondroitin Sulfate Proteoglycans/chemistry , Enzyme Stability , Spinal Cord Injuries/enzymology , Animals , Central Nervous System/drug effects , Chondroitin ABC Lyase/genetics , Chondroitin Sulfate Proteoglycans/biosynthesis , Disease Models, Animal , Humans , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Temperature
5.
Tumour Biol ; 39(9): 1010428317724282, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28945172

ABSTRACT

Neuron-glial antigen 2 (NG2, also known as CSPG4) and hyaluronic acid receptor CD44 are chondroitin sulphate proteoglycans actively involved in brain development and its malignant transformation. Here, we aimed to compare prognostic significances of NG2, CD44 and Ki-67 expression in glioblastoma multiforme patients. Totally, 45 tissue samples and 83 paraffin-embedded tissues for 75 patients were analysed. The prognostic values of the genes were analysed using Kaplan-Meier survival curves. Grade III gliomas showed 2-fold difference in NG2 expression between anaplastic astrocytoma and oligoastrocytoma (10.1 ± 3.5 and 25.5 ± 14.5, respectively). For grade IV gliomas, upregulated NG2 expression (21.0 ± 6.8) was associated with poor glioblastoma multiforme prognosis (overall survival < 12 months) compared with glioblastoma multiforme patients with good prognosis (4.4 ± 3.2; overall survival > 12 months). Multivariate survival analysis using Cox proportional hazards model confirmed that high NG2 expression was associated with low survival of the patients (hazard ratio: 3.43; 95% confidence interval: 1.18-9.93; p = 0.02), whereas age (hazard ratio: 1.02; 95% confidence interval: 0.96-1.09; p = 0.42), tumour resection (hazard ratio: 1.03; 95% confidence interval: 0.98-1.08; p = 0.25) and sex (hazard ratio: 0.62; 95% confidence interval: 0.21-1.86; p = 0.40) did not show significant association with prognosis. Although the positive correlation was shown for NG2 and CD44 expression in the glioblastomas (Pearson coefficient = 0.954), Kaplan-Meier and multivariate survival analyses did not revealed a significant association of the increased CD44 expression (hazard ratio: 2.18; 95% confidence interval: 0.50-9.43; p = 0.30) or high Ki-67 proliferation index (hazard ratio: 1.10; 95% confidence interval: 1.02-1.20; p = 0.02) with the disease prognosis. The results suggest that upregulation of NG2/CSPG4 rather than changes in CD44 or Ki-67 expression is associated with low overall survival in glioblastoma multiforme patients, supporting NG2/CSPG4 as a potential prognostic marker in glioblastoma.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/pathology , Chondroitin Sulfate Proteoglycans/biosynthesis , Glioblastoma/pathology , Hyaluronan Receptors/biosynthesis , Ki-67 Antigen/biosynthesis , Membrane Proteins/biosynthesis , Adolescent , Adult , Aged , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Chondroitin Sulfate Proteoglycans/analysis , Female , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Hyaluronan Receptors/analysis , Immunohistochemistry , Kaplan-Meier Estimate , Ki-67 Antigen/analysis , Male , Membrane Proteins/analysis , Middle Aged , Polymerase Chain Reaction , Prognosis , Proportional Hazards Models , Up-Regulation , Young Adult
6.
Electron. j. biotechnol ; 28: 113-119, July. 2017. tab, ilus, graf
Article in English | LILACS | ID: biblio-1015986

ABSTRACT

Background: C4ST-1 catalyzes the transfer of sulfate groups in the sulfonation of chondroitin during chondroitin sulfate synthesis. Chondroitin sulfate consists of numerous copies of negatively charged sulfonic acid groups that participate in the nucleation process of biomineralization. In the present study, we obtained two CHST11 genes (PmCHST11a and PmCHST11b) which encoded the C4ST-1 and explored the functions of these genes in the synthesis of chondroitin sulfate and in the formation of the nacreous layer of shells. Results: Both PmCHST11a and PmCHST11b had a sulfotransferase-2 domain, a signal peptide and a transmembrane domain. These properties indicated that these genes localize in the Golgi apparatus. Real-time PCR revealed that both PmCHST11a and PmCHST11b were highly expressed in the central zone of the mantle tissue. Inhibiting PmCHST11a and PmCHST11b via RNA interference significantly decreased the expression levels of these genes in the central zone of the mantle tissue and the concentration of chondroitin sulfate in extrapallial fluid. Moreover, shell nacre crystallized irregularly with a rough surface after RNA interference. Conclusions: This study indicated that PmCHST11a and PmCHST11b are involved in the nacre formation of Pinctada fucata martensii through participating in the synthesis of chondroitin sulfate.


Subject(s)
Sulfotransferases/metabolism , Pinctada , Nacre/biosynthesis , Chondroitin Sulfate Proteoglycans/biosynthesis , Sulfotransferases/genetics , Nucleic Acid Amplification Techniques/methods , RNA Interference , Real-Time Polymerase Chain Reaction , Biomineralization
7.
BMC Complement Altern Med ; 17(1): 286, 2017 May 31.
Article in English | MEDLINE | ID: mdl-28569157

ABSTRACT

BACKGROUND: Numerous studies have reported on the health benefits of sesamin, a major lignin found in sesame (S. indicum) seeds. Recently, sesamin was shown to have the ability to promote chondroitin sulfate proteoglycan synthesis in normal human chondrocytes. This study assesses the anti-inflammatory effect of sesamin on proteoglycans production in 3D chondrocyte cultures. METHODS: To evaluate the effects of sesamin on IL-1ß-treated human articular chondrocytes (HAC) pellets, the pellets were pre-treated with IL-1ß then cultured in the presence of various concentrations of sesamin for 21 days. During that period, the expression of IL-1ß, glycosaminoglycans (GAGs) content and Chondroitin sulfate proteoglycans (CSPGs) synthesis genes (ACAN, XT-1, XT-2, CHSY1 and ChPF) was measured. The GAGs accumulation in the extracellular matrix was determined on day 21 by histological analysis. RESULTS: There was clear evidence that sesamin upregulated expression of all the CSPGs synthesis genes, in contrast to the down-regulation of IL-1ß expression both in genes and in protein levels. The level of release and matrix accumulation of GAGs in IL-1ß pre-treated HAC pellets in the presence of sesamin was recovered. These results correlate with the histological examination which showed that sesamin enhanced matrix CSPGs accumulation. CONCLUSIONS: Sesamin enhances CSPGs synthesis, suppresses IL-1ß expression and ameliorates IL-1ß induced inflammation in human chondrocytes. Sesamin could have therapeutic benefits for treating inflammation in osteoarthritis.


Subject(s)
Chondrocytes/drug effects , Chondroitin Sulfate Proteoglycans/biosynthesis , Dioxoles/pharmacology , Interleukin-1beta/metabolism , Lignans/pharmacology , Adult , Aggrecans/genetics , Aggrecans/metabolism , Cells, Cultured , Chondrocytes/metabolism , Female , Glucuronosyltransferase , Humans , Male , Middle Aged , Multifunctional Enzymes , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Young Adult
8.
Methods Cell Biol ; 134: 485-530, 2016.
Article in English | MEDLINE | ID: mdl-27312503

ABSTRACT

In the past 20years, appreciation for the varied roles of proteoglycans (PGs), which are specific types of sugar-coated proteins, has increased dramatically. PGs in the extracellular matrix were long known to impart structural functions to many tissues, especially articular cartilage, which cushions bones and allows mobility at skeletal joints. Indeed, osteoarthritis is a debilitating disease associated with loss of PGs in articular cartilage. Today, however, PGs have a demonstrated role in cell biological processes, such as growth factor signalling, prompting new perspectives on the etiology of PG-associated diseases. Here, we review diseases associated with defects in PG synthesis and sulfation, also highlighting current understanding of the underlying genetics, biochemistry, and cell biology. Since most research has analyzed a class of PGs called heparan sulfate PGs, more attention is paid here to studies of chondroitin sulfate PGs (CSPGs), which are abundant in cartilage. Interestingly, CSPG synthesis is tightly linked to the cell biological processes of secretion and lysosomal degradation, suggesting that these systems may be linked genetically. Animal models of loss of CSPG function have revealed CSPGs to impact skeletal development. Specifically, our work from a mutagenesis screen in zebrafish led to the hypothesis that cartilage PGs normally delay the timing of endochondral ossification. Finally, we outline emerging approaches in zebrafish that may revolutionize the study of cartilage PG function, including transgenic methods and novel imaging techniques. Our recent work with X-ray fluorescent imaging, for example, enables direct correlation of PG function with PG-dependent biological processes.


Subject(s)
Cartilage, Articular/metabolism , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Zebrafish/genetics , Animals , Cartilage, Articular/pathology , Chondroitin Sulfate Proteoglycans/biosynthesis , Disease Models, Animal , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Humans , Lysosomes/genetics , Lysosomes/metabolism , Mutagenesis/genetics , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Zebrafish/metabolism
9.
Nat Commun ; 7: 11312, 2016 04 26.
Article in English | MEDLINE | ID: mdl-27115988

ABSTRACT

Remyelination is the generation of new myelin sheaths after injury facilitated by processes of differentiating oligodendrocyte precursor cells (OPCs). Although this repair phenomenon occurs in lesions of multiple sclerosis patients, many lesions fail to completely remyelinate. A number of factors have been identified that contribute to remyelination failure, including the upregulated chondroitin sulfate proteoglycans (CSPGs) that comprise part of the astrogliotic scar. We show that in vitro, OPCs have dramatically reduced process outgrowth in the presence of CSPGs, and a medication library that includes a number of recently reported OPC differentiation drugs failed to rescue this inhibitory phenotype on CSPGs. We introduce a novel CSPG synthesis inhibitor to reduce CSPG content and find rescued process outgrowth from OPCs in vitro and accelerated remyelination following focal demyelination in mice. Preventing CSPG deposition into the lesion microenvironment may be a useful strategy to promote repair in multiple sclerosis and other neurological disorders.


Subject(s)
Central Nervous System/metabolism , Chondroitin Sulfate Proteoglycans/biosynthesis , Oligodendroglia/metabolism , Remyelination/physiology , Stem Cells/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Carbohydrate Sequence , Cells, Cultured , Central Nervous System/drug effects , Chondroitin Sulfate Proteoglycans/antagonists & inhibitors , Chondroitin Sulfate Proteoglycans/chemistry , Female , Glucosamine/chemistry , Glucosamine/pharmacology , Humans , Mice, Inbred C57BL , Molecular Structure , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Oligodendroglia/drug effects , Remyelination/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Stem Cells/drug effects , Uridine Diphosphate Sugars/chemistry , Uridine Diphosphate Sugars/pharmacology
10.
Nature ; 532(7598): 195-200, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27027288

ABSTRACT

Transected axons fail to regrow in the mature central nervous system. Astrocytic scars are widely regarded as causal in this failure. Here, using three genetically targeted loss-of-function manipulations in adult mice, we show that preventing astrocyte scar formation, attenuating scar-forming astrocytes, or ablating chronic astrocytic scars all failed to result in spontaneous regrowth of transected corticospinal, sensory or serotonergic axons through severe spinal cord injury (SCI) lesions. By contrast, sustained local delivery via hydrogel depots of required axon-specific growth factors not present in SCI lesions, plus growth-activating priming injuries, stimulated robust, laminin-dependent sensory axon regrowth past scar-forming astrocytes and inhibitory molecules in SCI lesions. Preventing astrocytic scar formation significantly reduced this stimulated axon regrowth. RNA sequencing revealed that astrocytes and non-astrocyte cells in SCI lesions express multiple axon-growth-supporting molecules. Our findings show that contrary to the prevailing dogma, astrocyte scar formation aids rather than prevents central nervous system axon regeneration.


Subject(s)
Astrocytes/pathology , Axons/physiology , Central Nervous System/pathology , Central Nervous System/physiology , Cicatrix/pathology , Models, Biological , Nerve Regeneration , Animals , Central Nervous System/cytology , Chondroitin Sulfate Proteoglycans/biosynthesis , Cicatrix/prevention & control , Female , Genomics , Mice , Reproducibility of Results , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology
11.
J Clin Invest ; 125(2): 636-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25574841

ABSTRACT

Cornelia de Lange syndrome (CdLS) is a genetically heterogeneous disorder that presents with extensive phenotypic variability, including facial dysmorphism, developmental delay/intellectual disability (DD/ID), abnormal extremities, and hirsutism. About 65% of patients harbor mutations in genes that encode subunits or regulators of the cohesin complex, including NIPBL, SMC1A, SMC3, RAD21, and HDAC8. Wiedemann-Steiner syndrome (WDSTS), which shares CdLS phenotypic features, is caused by mutations in lysine-specific methyltransferase 2A (KMT2A). Here, we performed whole-exome sequencing (WES) of 2 male siblings clinically diagnosed with WDSTS; this revealed a hemizygous, missense mutation in SMC1A that was predicted to be deleterious. Extensive clinical evaluation and WES of 32 Turkish patients clinically diagnosed with CdLS revealed the presence of a de novo heterozygous nonsense KMT2A mutation in 1 patient without characteristic WDSTS features. We also identified de novo heterozygous mutations in SMC3 or SMC1A that affected RNA splicing in 2 independent patients with combined CdLS and WDSTS features. Furthermore, in families from 2 separate world populations segregating an autosomal-recessive disorder with CdLS-like features, we identified homozygous mutations in TAF6, which encodes a core transcriptional regulatory pathway component. Together, our data, along with recent transcriptome studies, suggest that CdLS and related phenotypes may be "transcriptomopathies" rather than cohesinopathies.


Subject(s)
Codon, Nonsense , De Lange Syndrome , Exome , Gene Expression Regulation , Phenotype , Transcriptome , Adolescent , Adult , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Child , Child, Preschool , Chondroitin Sulfate Proteoglycans/biosynthesis , Chondroitin Sulfate Proteoglycans/genetics , Chromosomal Proteins, Non-Histone/biosynthesis , Chromosomal Proteins, Non-Histone/genetics , De Lange Syndrome/genetics , De Lange Syndrome/metabolism , De Lange Syndrome/pathology , Exonucleases , Gene Expression Profiling , Genome-Wide Association Study , Heterozygote , Histone Deacetylases/biosynthesis , Histone Deacetylases/genetics , Histone-Lysine N-Methyltransferase , Humans , Infant , Male , Myeloid-Lymphoid Leukemia Protein/biosynthesis , Myeloid-Lymphoid Leukemia Protein/genetics , Proteins/genetics , Proteins/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics
12.
Exp Neurol ; 263: 372-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25446723

ABSTRACT

The expression of chondroitin sulfate proteoglycans (CSPGs) by reactive astrocytes is a major factor contributing to glial scarring and regenerative failure after spinal cord injury, but the molecular mechanisms underlying CSPG expression remain largely undefined. One contributing factor is transforming growth factor ß (TGFß), which is upregulated after injury and has been shown to induce expression of CSPGs in vitro. TGFß typically mediates its effects through the Smad2/3 signaling pathway, and it has been suggested that this pathway is responsible for CSPG expression. However, there is evidence that TGFß can also activate non-Smad signaling pathways. In this study, we report that TGFß-induced expression of three different CSPGs--neurocan, brevican, and aggrecan--is mediated through non-Smad signaling pathways. We observed significant increases in TGFß-induced expression of neurocan, brevican, and aggrecan following siRNA knockdown of Smad2 or Smad4, which indicates that Smad signaling is not required for the expression of these CSPGs. In addition, we show that neurocan, aggrecan, and brevican levels are significantly reduced when TGFß is administered in the presence of either the PI3K inhibitor LY294002 or the mTOR inhibitor rapamycin, but not the MEK1/2 inhibitor U0126. This suggests that TGFß mediates this effect through non-Smad-dependent activation of the PI3K-Akt-mTOR signaling pathway, and targeting this pathway may therefore be an effective means of reducing CSPG expression in the injured CNS.


Subject(s)
Chondroitin Sulfate Proteoglycans/biosynthesis , Gliosis/metabolism , Signal Transduction/physiology , Spinal Cord Injuries/metabolism , Transforming Growth Factor beta/metabolism , Animals , Blotting, Western , Cells, Cultured , Female , Immunohistochemistry , Male , RNA, Small Interfering , Rats , Rats, Long-Evans , Smad Proteins/metabolism , Transfection
13.
Methods Mol Biol ; 1229: 69-78, 2015.
Article in English | MEDLINE | ID: mdl-25325945

ABSTRACT

Glycosaminoglycan (GAG) side chains of proteoglycans are involved in a wide variety of developmental and pathophysiological functions. Similar to a gene knockout, the ability to inhibit GAG biosynthesis would allow us to examine the function of endogenous GAG chains. However, ubiquitously and irreversibly knocking out all GAG biosynthesis would cause multiple effects making it difficult to attribute a specific biological role to a specific GAG structure in spatiotemporal manner. Reversible and selective inhibition of GAG biosynthesis would allow us to examine the importance of endogenous GAGs to specific cellular, tissue, or organ systems. In this chapter, we describe the chemical synthesis and biological evaluation of 4-deoxy-4-fluoro-xylosides as selective inhibitors of heparan sulfate and chondroitin/dermatan sulfate proteoglycan biosynthesis.


Subject(s)
Biochemistry/methods , Chondroitin Sulfate Proteoglycans/antagonists & inhibitors , Chondroitin Sulfate Proteoglycans/biosynthesis , Glycosides/chemical synthesis , Glycosides/pharmacology , Heparitin Sulfate/antagonists & inhibitors , Heparitin Sulfate/biosynthesis , Animals , CHO Cells , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , Glycosides/chemistry , Reproducibility of Results
14.
Virchows Arch ; 465(6): 703-13, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25304290

ABSTRACT

Giant cell tumor of bone (GCTB) displays worrisome clinical features such as local recurrence and occasionally metastatic disease which are unpredictable by morphology. Additional routinely usable biomarkers do not exist. Gene expression profiles of six clinically defined groups of GCTB and one group of aneurysmal bone cyst (ABC) were determined by microarray (n = 33). The most promising differentially expressed genes were validated by Q-PCR as potential biomarkers in a larger patient group (n = 41). Corresponding protein expression was confirmed by immunohistochemistry. Unsupervised hierarchical clustering reveals a metastatic GCTB cluster, a heterogeneous, non-metastatic GCTB cluster, and a primary ABC cluster. Balanced score testing indicates that lumican (LUM) and decorin (DCN) are the most promising biomarkers as they have lower level of expression in the metastatic group. Expression of dermatopontin (DPT) was significantly lower in recurrent tumors. Validation of the results was performed by paired and unpaired t test in primary GCTB and corresponding metastases, which proved that the differential expression of LUM and DCN is tumor specific rather than location specific. Our findings show that several genes related to extracellular matrix integrity (LUM, DCN, and DPT) are differentially expressed and may serve as biomarkers for metastatic and recurrent GCTB.


Subject(s)
Biomarkers, Tumor/analysis , Bone Neoplasms/genetics , Decorin/biosynthesis , Giant Cell Tumor of Bone/genetics , Lung Neoplasms/secondary , Adolescent , Adult , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Child , Chondroitin Sulfate Proteoglycans/biosynthesis , Chondroitin Sulfate Proteoglycans/genetics , Cluster Analysis , Decorin/genetics , Down-Regulation , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Female , Gene Expression Profiling , Giant Cell Tumor of Bone/metabolism , Giant Cell Tumor of Bone/pathology , Humans , Immunohistochemistry , Keratan Sulfate/biosynthesis , Keratan Sulfate/genetics , Lumican , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Transcriptome , Young Adult
15.
J Neurochem ; 130(5): 612-25, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24716865

ABSTRACT

Old astrocyte specifically induced substance (OASIS), a basic leucine zipper transcription factor of the cAMP response element binding/Activating transcription factor family, is induced in reactive astrocytes in vivo and has important roles in quality control of protein synthesis at the endoplasmic reticulum. Reactive astrocytes produce a non-permissive environment for regenerating axons by up-regulating chondroitin sulfate proteoglycans (CSPGs). In this study, we focus on the potential role of OASIS in CSPG production in the adult mouse cerebral cortex. CS-C immunoreactivity, which represents chondroitin sulfate moieties, was significantly attenuated in the stab-injured cortices of OASIS knockout mice compared to those of wild-type mice. We next examined expression of the CSPG-synthesizing enzymes and core proteins of CSPGs in the stab-injured cortices of OASIS knockout and wild-type mice. The levels of chondroitin 6-O-sulfotransferase 1 (C6ST1, one of the major enzymes involved in sulfation of CSPGs) mRNA and protein increased after cortical stab injury of wild-type, but not of OASIS knockout, mice. A C-terminal deletion mutant OASIS over-expressed in rat C6 glioma cells increased C6ST1 transcription by interacting with the first intron region. Neurite outgrowth of cultured hippocampal neurons was inhibited on culture dishes coated with membrane fractions of epidermal growth factor-treated astrocytes derived from wild type but not from OASIS knockout mice. These results suggest that OASIS regulates the transcription of C6ST1 and thereby promotes CSPG sulfation in astrocytes. Through these mechanisms, OASIS may modulate axonal regeneration in the injured cerebral cortex. OASIS, an ER stress-responsive CREB/ATF family member, is up-regulated in the reactive astrocytes of the injured brain. We found that the up-regulated OASIS is involved in the transcriptional regulation of C6ST1 gene, which promotes chondroitin sulfate proteoglycan (CSPG) sulfation. We conclude that OASIS functions as an anti-regenerative transcription factor by establishing a non-permissive microenvironment to regenerating axons.


Subject(s)
Brain Injuries/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation/physiology , Nerve Tissue Proteins/metabolism , Sulfotransferases/biosynthesis , Animals , Astrocytes/metabolism , Blotting, Western , Brain Injuries/genetics , Cerebral Cortex/metabolism , Chondroitin Sulfate Proteoglycans/biosynthesis , Chondroitin Sulfate Proteoglycans/genetics , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sulfotransferases/genetics , Transcription, Genetic , Carbohydrate Sulfotransferases
16.
Neuromolecular Med ; 16(2): 457-72, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24519742

ABSTRACT

To address the role of the transforming growth factor beta (TGFß)-Smad3 signaling pathway in dendrite growth and associated synaptogenesis, we used small inhibitory RNA to knockdown the Smad3 gene in either cultured neurons and or primary astrocytes. We found that TGFß1 treatment of primary neurons increased dendrite extensions and the number of synapsin-1-positive synapses. When Smad3 was knockdown in primary neurons, dendrite growth was inhibited and the number of synapsin-1-positive synapses reduced even with TGFß1 treatment. When astrocyte-conditioned medium (ACM), collected from TGFß1-treated astrocytes (TGFß1-stimulated ACM), was added to cultured neurons, dendritic growth was inhibited and the number of synapsin-1-positive puncta reduced. When TGFß1-stimulated ACM was collected from astrocytes with Smad3 knocked down, this conditioned media promoted the growth of dendrites and the number of synapsin-1-positive puncta in cultured neurons. We further found that TGFß1 signaling through Smad3 increased the expression of chondroitin sulfate proteoglycans, neurocan, and phosphacan in ACM. Application of chondroitinase ABC to the TGFß1-stimulated ACM reversed its inhibitory effects on the dendrite growth and the number of synapsin-1-positive puncta. On the other hand, we found that TGFß1 treatment caused a facilitation of Smad3 phosphorylation and translocation to the nucleus induced by status epilepticus (SE) in wild-type (Smad3(+/+)) mice, and this treatment also caused a promotion of γ-aminobutyric acid-ergic synaptogenesis impaired by SE in Smad3(+/+) as well as in Smad3(-/-) mice, but more dramatic promotion in Smad3(+/+) mice. Thus, we provide evidence for the first time that TGFß-Smad3 signaling pathways within neuron and astrocyte differentially regulate dendrite growth and synaptogenesis, and this pathway may be involved in the pathogenesis of some central nervous system diseases, such as epilepsy.


Subject(s)
Astrocytes/metabolism , Neurons/metabolism , Signal Transduction/physiology , Smad3 Protein/physiology , Synapses/ultrastructure , Transforming Growth Factor beta1/physiology , Active Transport, Cell Nucleus , Animals , Astrocytes/drug effects , Astrocytes/ultrastructure , Cells, Cultured , Chondroitin ABC Lyase/pharmacology , Chondroitin Sulfate Proteoglycans/biosynthesis , Chondroitin Sulfate Proteoglycans/genetics , Culture Media, Conditioned/pharmacology , Female , Gene Expression Regulation , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Neurocan/biosynthesis , Neurocan/genetics , Neurons/ultrastructure , Protein Processing, Post-Translational/drug effects , RNA Interference , RNA, Small Interfering/pharmacology , Receptor-Like Protein Tyrosine Phosphatases, Class 5/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Smad3 Protein/antagonists & inhibitors , Smad3 Protein/deficiency , Smad3 Protein/genetics , Status Epilepticus/metabolism , Synapsins/analysis , Transforming Growth Factor beta1/pharmacology
17.
Glycoconj J ; 31(3): 221-30, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24338203

ABSTRACT

Osteoarthritis (OA) is a degenerative joint disease that progressively causes a loss of joint functions and the impaired quality of life. The most significant event in OA is a high degree of degradation of articular cartilage accompanied by the loss of chondroitin sulfate-proteoglycans (CS-PGs). Recently, the chondroprotective effects of sesamin, the naturally occurring substance found in sesame seeds, have been proved in a rat model of papain-induced osteoarthritis. We hypothesized that sesamin may be associated with possible promotion of the biosynthesis of CS-PGs in human articular chondrocytes. The aim of the study was to investigate the effects of sesamin on the major CS-PG biosynthesis in primary human chondrocyte. The effects of sesamin on the gene expression of the PG core and the CS biosynthetic enzymes as well as on the secretion of glycosaminoglycans (GAGs) in monolayer and pellet culture systems of articular chondrocytes. Sesamin significantly increased the GAGs content both in culture medium and pellet matrix. Real-time-quantitative PCR showed that sesamin promoted the expression of the genes encoding the core protein (ACAN) of the major CS-PG aggrecan and the biosynthetic enzymes (XYLT1, XYLT2, CHSY1 and CHPF) required for the synthesis of CS-GAG side chains. Safranin-O staining of sesamin treated chondrocyte pellet section confirmed the high degree of GAG accumulation. These results were correlated with an increased level of secreted GAGs in the media of cultured articular chondrocytes in both culture systems. Thus, sesamin would provide a potential therapeutic strategy for treating OA patients.


Subject(s)
Chondrocytes/drug effects , Chondroitin Sulfate Proteoglycans/biosynthesis , Dioxoles/pharmacology , Lignans/pharmacology , Adolescent , Adult , Aggrecans/genetics , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/metabolism , Gene Expression Regulation/drug effects , Glucuronosyltransferase , Glycosaminoglycans/metabolism , Humans , Middle Aged , Multifunctional Enzymes , N-Acetylgalactosaminyltransferases/genetics , Pentosyltransferases/genetics , Young Adult , UDP Xylose-Protein Xylosyltransferase
18.
Cell Tissue Bank ; 15(1): 25-34, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23292197

ABSTRACT

The in vivo quiescent corneal stroma keratocytes need to be transformed to activated state in order to obtain sufficient number of cells either for monolayer evaluation or corneal stroma reconstruction. This study aimed to investigate the phenotypic characterization of corneal stromal cells during culture expansion from the limbal region of the cornea. Isolated corneal keratocytes from limbal tissue of New Zealand White Strain rabbits' corneas (n = 6) were culture expanded until three passages. Keratocytes morphology was examined daily with viability, growth rate, number of cell doubling and population doubling time were recorded at each passage. The expression of collagen type 1, aldehyde dehydrogenase (ALDH), lumican and alpha smooth muscle actin (α-SMA) were detected by RT-PCR. Immunocytochemistry was also used to detect ALDH, α-SMA, collagen type I and Cytokeratin-3 (CK3). Growth kinetic study revealed that the growth rate was low at the initial passage but increase to about two folds with concomitant reduction in population doubling time in later passages. Freshly isolated and cultured keratocytes expressed collagen type 1, ALDH and lumican but α-SMA expression was absent. However, α-SMA was expressed along with the other genes during culture expansion. Keratocytes at P1 expressed all the proteins except CK3. These results suggest that cultured keratocytes maintained most of the gene expression profile of native keratocytes while the emergence of α-SMA in serial passages showed a mix population of various phenotypes. The phenotypic characterization of monolayer keratocytes provides useful information before reconstruction of bioengineered tissue or in vitro pharmaceutical applications.


Subject(s)
Corneal Keratocytes/cytology , Corneal Stroma/cytology , Actins/biosynthesis , Aldehyde Dehydrogenase/biosynthesis , Animals , Bioengineering , Cell- and Tissue-Based Therapy , Cells, Cultured , Chondroitin Sulfate Proteoglycans/biosynthesis , Collagen Type I/biosynthesis , Corneal Keratocytes/transplantation , Fibroblasts , Gene Expression , Keratan Sulfate/biosynthesis , Keratin-3/biosynthesis , Lumican , Phenotype , Rabbits
19.
Biochem Biophys Res Commun ; 441(2): 514-8, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24177010

ABSTRACT

Chondroitin sulfate proteoglycan 4 (CSPG4), a transmembrane proteoglycan originally identified in melanoma cells, has been reported to be expressed in breast cancer cells. This study was performed to examine the expression and significance of CSPG4 in a cohort of breast cancer patients. Immunohistochemical analysis of CSPG4 was performed on tissue microarrays constructed from tissue specimens from 240 breast cancer patients. CSPG4 staining was correlated with clinical and pathological characteristics, overall survival (OS), and disease recurrence. Contradicting to a previous report, our results showed that high CSPG4 expression was not related to triple-negative status of breast cancer patients. The Kaplan-Meier method showed that high CSPG4 expression was significantly associated with shorter time to recurrence (TTR). Patients with high CSPG4 expression had poorer OS and shorter TTR in a multivariate survival analysis after adjustment for stage, tumor grade, expression of estrogen receptor and progesterone receptor, and HER2 overexpression. This study showed that high CSPG4 expression correlates with disease recurrence and OS in breast cancers.


Subject(s)
Breast Neoplasms/mortality , Chondroitin Sulfate Proteoglycans/biosynthesis , Membrane Proteins/biosynthesis , Neoplasm Recurrence, Local/mortality , Adult , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Recurrence, Local/metabolism , Prognosis , Time Factors , Tissue Array Analysis/statistics & numerical data
20.
Oncol Rep ; 30(4): 1609-21, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23846574

ABSTRACT

Lumican, a member of the class II small leucine-rich proteoglycan family, regulates the assembly and diameter of collagen fibers in the extracellular matrix of various tissues. We previously reported that lumican expression in the stromal tissues of pancreatic ductal adenocarcinoma (PDAC) correlates with tumor invasion, and tends to correlate with poor prognosis. Lumican stimulates growth and inhibits the invasion of a PDAC cell line. In the present study, we performed a global shotgun proteomic analysis using lumican-overexpressing PANC­1 cells and lumican downregulated PANC­1 cells to identify candidate proteins that are regulated by lumican and related to cell growth and invasion in PDAC cells. A total of 448 proteins were identified from lumican-overexpressing PANC­1 and control cells. Additionally, 451 proteins were identified from lumican-downregulated PANC­1 cells and control cells. As a result of semi-quantification based on spectral counting, 174 differentially expressed proteins were identified by lumican upregulation, and 143 differentially expressed proteins were identified by lumican downregulation. The expression levels of 24 proteins, including apoptosis- and invasion-related proteins correlated with lumican expression levels. It is likely that the expression of these proteins is regulated by lumican, and that they are involved in apoptosis and invasion in PDAC. These findings suggest that lumican may be involved in cell growth and invasion through the regulation of these 24 proteins expressed in PDAC.


Subject(s)
Apoptosis/genetics , Carcinoma, Pancreatic Ductal/pathology , Chondroitin Sulfate Proteoglycans/metabolism , Keratan Sulfate/metabolism , Neoplasm Invasiveness/genetics , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Proliferation , Chondroitin Sulfate Proteoglycans/biosynthesis , Down-Regulation , Gene Expression , Humans , Keratan Sulfate/biosynthesis , Lumican , Metallothionein/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Prognosis , Protein Isoforms/metabolism , Proteomics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...