Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Braz. j. med. biol. res ; 48(12): 1063-1070, Dec. 2015. tab, graf
Article in English | LILACS | ID: lil-762916

ABSTRACT

Damage to cartilage causes a loss of type II collagen (Col-II) and glycosaminoglycans (GAG). To restore the original cartilage architecture, cell factors that stimulate Col-II and GAG production are needed. Insulin-like growth factor I (IGF-I) and transcription factor SOX9are essential for the synthesis of cartilage matrix, chondrocyte proliferation, and phenotype maintenance. We evaluated the combined effect of IGF-I and SOX9 transgene expression on Col-II and GAG production by cultured human articular chondrocytes. Transient transfection and cotransfection were performed using two mammalian expression plasmids (pCMV-SPORT6), one for each transgene. At day 9 post-transfection, the chondrocytes that were over-expressing IGF-I/SOX9 showed 2-fold increased mRNA expression of the Col-II gene, as well as a 57% increase in Col-II protein, whereas type I collagen expression (Col-I) was decreased by 59.3% compared with controls. The production of GAG by these cells increased significantly compared with the controls at day 9 (3.3- vs 1.8-times, an increase of almost 83%). Thus, IGF-I/SOX9 cotransfected chondrocytes may be useful for cell-based articular cartilage therapies.


Subject(s)
Humans , Chondrocytes/metabolism , Collagen Type II/biosynthesis , Glycosaminoglycans/biosynthesis , Insulin-Like Growth Factor I/metabolism , Matrilin Proteins/biosynthesis , SOX9 Transcription Factor/metabolism , Transfection/methods , Cartilage, Articular/injuries , Cartilage, Articular/metabolism , Collagen Type II/analysis , Extracellular Matrix/chemistry , Gene Expression , Glycosaminoglycans/analysis , Insulin-Like Growth Factor I/genetics , Matrilin Proteins/genetics , Primary Cell Culture , Real-Time Polymerase Chain Reaction , RNA, Messenger/metabolism , SOX9 Transcription Factor/genetics , Spectrophotometry
2.
Yonsei Medical Journal ; : 277-286, 2015.
Article in English | WPRIM | ID: wpr-174623

ABSTRACT

PURPOSE: To investigate the molecular responses of various genes and proteins related to disc degeneration upon treatment with cytokines that affect disc-cell proliferation and phenotype in living human intervertebral discs (IVDs). Responsiveness to these cytokines according to the degree of disc degeneration was also evaluated. MATERIALS AND METHODS: The disc specimens were classified into two groups: group 1 (6 patients) showed mild degeneration of IVDs and group 2 (6 patients) exhibited severe degeneration of IVDs. Gene expression was analyzed after treatment with four cytokines: recombinant human bone morphogenic protein (rhBMP-2), transforming growth factor-beta (TGF-beta), interleukin-1beta (IL-1beta), and tumor necrosis factor-alpha (TNF-alpha). Molecular responses were assessed after exposure of cells from the IVD specimens to these cytokines via real-time polymerase chain reaction and immunofluorescence staining. RESULTS: mRNA gene expression was significantly greater for aggrecan, type I collagen, type II collagen, alkaline phosphatase, osteocalcin, and Sox9 in group 1 than mRNA gene expression in group 2, when the samples were not treated with cytokines. Analysis of mRNA levels for these molecules after morphogen treatment revealed significant increases in both groups, which were much higher in group 1 than in group 2. The average number of IVD cells that were immunofluorescence stained positive for alkaline phosphatase increased after treatment with rhBMP-2 and TGF-beta in group 1. CONCLUSION: The biologic responsiveness to treatment of rhBMP-2, TGF-beta, TNF-alpha, and IL-1beta in the degenerative living human IVD can be different according to the degree of degeneration of the IVD.


Subject(s)
Adult , Female , Humans , Male , Middle Aged , Aggrecans/genetics , Alkaline Phosphatase/genetics , Biological Products/pharmacology , Bone Morphogenetic Protein 2/pharmacology , Collagen Type I/genetics , Collagen Type II/genetics , Cytokines/pharmacology , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Interleukin-1/pharmacology , Intervertebral Disc/drug effects , Intervertebral Disc Degeneration/drug therapy , Osteocalcin/genetics , RNA, Messenger/genetics , Recombinant Proteins/pharmacology , SOX9 Transcription Factor/genetics , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
3.
Braz. j. med. biol. res ; 47(8): 637-645, 08/2014. tab, graf
Article in English | LILACS | ID: lil-716279

ABSTRACT

Tissue engineering encapsulated cells such as chondrocytes in the carrier matrix have been widely used to repair cartilage defects. However, chondrocyte phenotype is easily lost when chondrocytes are expanded in vitro by a process defined as “dedifferentiation”. To ensure successful therapy, an effective pro-chondrogenic agent is necessary to overcome the obstacle of limited cell numbers in the restoration process, and dedifferentiation is a prerequisite. Gallic acid (GA) has been used in the treatment of arthritis, but its biocompatibility is inferior to that of other compounds. In this study, we modified GA by incorporating sulfamonomethoxine sodium and synthesized a sulfonamido-based gallate, JJYMD-C, and evaluated its effect on chondrocyte metabolism. Our results showed that JJYMD-C could effectively increase the levels of the collagen II, Sox9, and aggrecan genes, promote chondrocyte growth, and enhance secretion and synthesis of cartilage extracellular matrix. On the other hand, expression of the collagen I gene was effectively down-regulated, demonstrating inhibition of chondrocyte dedifferentiation by JJYMD-C. Hypertrophy, as a characteristic of chondrocyte ossification, was undetectable in the JJYMD-C groups. We used JJYMD-C at doses of 0.125, 0.25, and 0.5 µg/mL, and the strongest response was observed with 0.25 µg/mL. This study provides a basis for further studies on a novel agent in the treatment of articular cartilage defects.


Subject(s)
Animals , Rabbits , Benzamides/chemical synthesis , Cell Dedifferentiation/drug effects , Cell Proliferation/drug effects , Chondrocytes/drug effects , Phenotype , Pyrimidines/chemical synthesis , Aggrecans/genetics , Aggrecans/metabolism , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Benzamides/pharmacology , Cell Survival , Cell Dedifferentiation/immunology , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type II/genetics , Collagen Type II/metabolism , Glycosaminoglycans/analysis , Immunohistochemistry , Laser Scanning Cytometry , Primary Cell Culture , Pyrimidines/pharmacology , Real-Time Polymerase Chain Reaction , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Tissue Engineering
4.
Braz. j. med. biol. res ; 47(4): 279-286, 8/4/2014. tab, graf
Article in English | LILACS | ID: lil-705770

ABSTRACT

SRY-related high-mobility-group box 9 (Sox9) gene is a cartilage-specific transcription factor that plays essential roles in chondrocyte differentiation and cartilage formation. The aim of this study was to investigate the feasibility of genetic delivery of Sox9 to enhance chondrogenic differentiation of human umbilical cord blood-derived mesenchymal stem cells (hUC-MSCs). After they were isolated from human umbilical cord blood within 24 h after delivery of neonates, hUC-MSCs were untreated or transfected with a human Sox9-expressing plasmid or an empty vector. The cells were assessed for morphology and chondrogenic differentiation. The isolated cells with a fibroblast-like morphology in monolayer culture were positive for the MSC markers CD44, CD105, CD73, and CD90, but negative for the differentiation markers CD34, CD45, CD19, CD14, or major histocompatibility complex class II. Sox9 overexpression induced accumulation of sulfated proteoglycans, without altering the cellular morphology. Immunocytochemistry demonstrated that genetic delivery of Sox9 markedly enhanced the expression of aggrecan and type II collagen in hUC-MSCs compared with empty vector-transfected counterparts. Reverse transcription-polymerase chain reaction analysis further confirmed the elevation of aggrecan and type II collagen at the mRNA level in Sox9-transfected cells. Taken together, short-term Sox9 overexpression facilitates chondrogenesis of hUC-MSCs and may thus have potential implications in cartilage tissue engineering.


Subject(s)
Humans , Cell Differentiation/genetics , Chondrogenesis/genetics , Fetal Blood/cytology , Mesenchymal Stem Cells/cytology , SOX9 Transcription Factor/genetics , Aggrecans/biosynthesis , Blotting, Western , Cartilage/metabolism , Cell Proliferation/genetics , Chondrocytes/metabolism , Collagen Type II/biosynthesis , Flow Cytometry , Green Fluorescent Proteins , Gene Expression Regulation/physiology , Human Umbilical Vein Endothelial Cells/cytology , Immunohistochemistry , Immunophenotyping , Primary Cell Culture , Reverse Transcriptase Polymerase Chain Reaction , Tissue Engineering , Transfection
5.
Gut and Liver ; : 508-518, 2014.
Article in English | WPRIM | ID: wpr-108130

ABSTRACT

BACKGROUND/AIMS: Doublecortin and CaM kinase-like-1 (DCAMKL1) is a marker of stem cells expressed predominantly in the crypt base in the intestine. However, DCAMKL1-positive cells have been shown to be differentiated tuft cells rather than quiescent progenitors. Tuft cells are the only epithelial cells that express cyclooxygenase 2 (COX-2) in the normal intestinal epithelium. We previously generated Cdx2-transgenic mice as model mice for intestinal metaplasia and gastric carcinoma. In the current study, we investigated the association between COX-2 and DCAMKL1 in gastric carcinoma. METHODS: We examined the association between COX-2 and DCAMKL1 expression in gastric carcinomas in clinical samples (early gastric well-differentiated adenocarcinoma) and Cdx2-transgenic mice; and the DCAMKL1-transgenic mouse stomach using immunohistochemistry and quantitative real-time polymerase chain reaction. RESULTS: The COX-2-expressing cells were scattered, not diffusely expressed, in gastric carcinomas from humans and Cdx2-transgenic mice. DCAMKL1-positive cells were also scattered in the gastric carcinomas, indicating that tuft cells could still be present in gastric carcinoma. COX-2 was expressed in DCAMKL1-positive tuft cells in Cdx2- and DCAMKL1-transgenic mouse stomachs, whereas the Sox9 transcription factor was ubiquitously expressed in gastric carcinomas, including COX-2-positive cells. CONCLUSIONS: COX-2 is expressed in DCAMKL1-expressing quiescent tuft cells in gastric carcinoma.


Subject(s)
Animals , Humans , Mice , Adenocarcinoma/metabolism , Cyclooxygenase 2/genetics , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Intestinal Mucosa/cytology , Intracellular Signaling Peptides and Proteins/genetics , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , SOX9 Transcription Factor/genetics , Stomach Neoplasms/enzymology
6.
Clinics ; 67(2): 99-106, 2012. ilus, graf, tab
Article in English | LILACS | ID: lil-614632

ABSTRACT

OBJECTIVES: Understanding the changes in chondrogenic gene expression that are involved in the differentiation of human adipose-derived stem cells to chondrogenic cells is important prior to using this approach for cartilage repair. The aims of the study were to characterize human adipose-derived stem cells and to examine chondrogenic gene expression after one, two, and three weeks of induction. MATERIALS AND METHODS: Human adipose-derived stem cells at passage 4 were evaluated by flow cytometry to examine the expression of surface markers. These adipose-derived stem cells were tested for adipogenic and osteogenic differentiation capacity. Ribonucleic acid was extracted from the cells for quantitative polymerase chain reaction analysis to determine the expression levels of chondrogenic genes after chondrogenic induction. RESULTS: Human adipose-derived stem cells were strongly positive for the mesenchymal markers CD90, CD73, CD44, CD9, and histocompatibility antigen and successfully differentiated into adipogenic and osteogenic lineages. The human adipose-derived stem cells aggregated and formed a dense matrix after chondrogenic induction. The expression of chondrogenic genes (collagen type II, aggrecan core protein, collagen type XI, COMP, and ELASTIN) was significantly higher after the first week of induction. However, a significantly elevated expression of collagen type X was observed after three weeks of chondrogenic induction. CONCLUSION: Human adipose-derived stem cells retain stem cell characteristics after expansion in culture to passage 4 and serve as a feasible source of cells for cartilage regeneration. Chondrogenesis in human adiposederived stem cells was most prominent after one week of chondrogenic induction.


Subject(s)
Humans , Adipose Tissue/cytology , Cartilage, Articular/cytology , Cell Differentiation/genetics , Chondrocytes/metabolism , Chondrogenesis/genetics , Collagen/metabolism , Mesenchymal Stem Cells , Adipogenesis/genetics , Biomarkers/metabolism , Cells, Cultured , Chondrocytes/cytology , Collagen/genetics , Elastin/genetics , Elastin/metabolism , Flow Cytometry , Gene Expression Regulation , Mesenchymal Stem Cells , Osteogenesis/genetics , RNA, Messenger/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Time Factors
7.
Journal of Korean Medical Science ; : 143-145, 2011.
Article in English | WPRIM | ID: wpr-211267

ABSTRACT

Campomelic dysplasia (CD; OMIM #114290), a rare form of congenital short-limbed dwarfism, is due to mutations in SOX9, a member of the SOX (SRY-related HMG box) gene family. Multiparous mother at 38 weeks' gestation delivered a 3,272 g baby boy with characteristic phenotypes including bowing of the lower limbs, a narrow thoracic cage, 11 pairs of ribs, hypoplastic scapulae, macrocephaly, flattened supraorbital ridges and nasal bridge, cleft palate, and micrognathia. He underwent a tracheostomy at the age of three months for severe laryngomalacia after a number of repeated hospitalizations due to respiratory problems and died at the age of four months from progressive respiratory failure. He was diagnosed as having CD based on a novel frameshift mutation (p.Gln458ArgfsX12) in the SOX9 gene, the mutation which has not yet been reported in Korea.


Subject(s)
Humans , Infant , Male , Campomelic Dysplasia/diagnosis , Disorders of Sex Development/genetics , Frameshift Mutation , Respiratory Insufficiency/complications , SOX9 Transcription Factor/genetics , Sequence Analysis, DNA
8.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 291-5, 2009.
Article in English | WPRIM | ID: wpr-634896

ABSTRACT

Sox9 gene was cloned from immortalized precartilaginous stem cells and its eukaryotic expression vector constructed in order to explore the possibility of bone marrow-derived stromal cells differentiation into precartilaginous stem cells induced by Sox9. A full-length fragment of Sox9 was obtained by RT-PCR, inserted into pGEM-T Easy clone vector, and ligated with pEGFP-IRES2 expression vector by double digestion after sequencing. The compound plasmid was transfected into born marrow-derived stromal cells by Lipofectamine 2000, and the transfection efficacy and the expression of Sox9 and FGFR-3 were observed. Flow cytometry was used to identify the cell phenotype, and MTT was employed to assay proliferative viability of cells. Sequencing, restrictive endonuclease identification and RT-PCR confirmed that the expansion of Sox9 and construction of Sox9 expression vector were successful. After transfection of the recombinant vector into bone marrow-derived stromal cells, the expression of Sox9 and FGFR-3 was detected, and proliferative viability was not different from that of precartilaginous stem cells. It was concluded that Sox9 gene eukaryotic expression vector was successfully constructed, and the transfected bone marrow-derived stromal cells differentiated into the precartilaginous stem cells.


Subject(s)
Base Sequence , Bone Marrow Cells/cytology , Cartilage/cytology , Cell Differentiation/genetics , Cells, Cultured , Cloning, Molecular , Genetic Vectors/genetics , Molecular Sequence Data , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , SOX9 Transcription Factor/biosynthesis , SOX9 Transcription Factor/genetics , Stem Cells/cytology , Stromal Cells/cytology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL