Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
JCI Insight ; 9(3)2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38127456

ABSTRACT

Despite clinical use of immunosuppressive agents, the immunopathogenesis of minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS) remains unclear. Src homology 3-binding protein 2 (SH3BP2), a scaffold protein, forms an immune signaling complex (signalosome) with 17 other proteins, including phospholipase Cγ2 (PLCγ2) and Rho-guanine nucleotide exchange factor VAV2 (VAV2). Bioinformatic analysis of human glomerular transcriptome (Nephrotic Syndrome Study Network cohort) revealed upregulated SH3BP2 in MCD and FSGS. The SH3BP2 signalosome score and downstream MyD88, TRIF, and NFATc1 were significantly upregulated in MCD and FSGS. Immune pathway activation scores for Toll-like receptors, cytokine-cytokine receptor, and NOD-like receptors were increased in FSGS. Lower SH3BP2 signalosome score was associated with MCD, higher estimated glomerular filtration rate, and remission. Further work using Sh3bp2KI/KI transgenic mice with a gain-in-function mutation showed ~6-fold and ~25-fold increases in albuminuria at 4 and 12 weeks, respectively. Decreased serum albumin and unchanged serum creatinine were observed at 12 weeks. Sh3bp2KI/KI kidney morphology appeared normal except for increased mesangial cellularity and patchy foot process fusion without electron-dense deposits. SH3BP2 co-immunoprecipitated with PLCγ2 and VAV2 in human podocytes, underscoring the importance of SH3BP2 in immune activation. SH3BP2 and its binding partners may determine the immune activation pathways resulting in podocyte injury leading to loss of the glomerular filtration barrier.


Subject(s)
Glomerulosclerosis, Focal Segmental , Nephrosis, Lipoid , Nephrotic Syndrome , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Kidney/pathology , Kidney Glomerulus/pathology , Mice, Transgenic , Nephrosis, Lipoid/pathology , Nephrotic Syndrome/metabolism , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism
2.
Prog Rehabil Med ; 7: 20220026, 2022.
Article in English | MEDLINE | ID: mdl-35633759

ABSTRACT

Objectives: Hip fracture is a common injury occurring in elderly people and often impairs their activities of daily living (ADL). This study aimed to identify and analyze factors associated with ADL following hip fracture treatment. Methods: A total of 371 consecutive patients with hip fractures who were surgically treated in our hospital were enrolled. Among these, 103 patients who underwent acute- to recovery-phase postoperative rehabilitation at our hospital and whose motor scale of the functional independence measure (mFIM) score was ≥70 before the fracture were finally included in this study. Single and multiple regression analyses were performed to identify the factors correlated with ADL. The mFIM at hospital discharge was set as the outcome variable, and various clinical factors, such as fracture type, surgical technique, serum and biological data, mini-mental state examination (MMSE) score, and serial mFIM scores, were used as explanatory variables. Results: Only MMSE and preinjury mFIM scores were significantly correlated with mFIM at discharge, and MMSE had the larger effect on the outcome. Receiver operating characteristic curve analysis revealed an MMSE cutoff value of 20/21. Patients with an MMSE score of ≤20 showed a relatively poor recovery of mFIM from 2-3 weeks postoperatively compared with those with an MMSE score of ≥21. Conclusion: Cognitive impairment and the preinjury ADL level were correlated with short-term ADL outcomes following hip fracture. Cognitive impairment was the most important factor affecting ADL; treatment and postoperative rehabilitation should be carefully considered for cognitively disturbed patients from the acute phase after hip fracture.

3.
J Bone Miner Res ; 33(1): 167-181, 2018 01.
Article in English | MEDLINE | ID: mdl-28914985

ABSTRACT

Currently, it is believed that osteoclasts positive for tartrate-resistant acid phosphatase (TRAP+) are the exclusive bone-resorbing cells responsible for focal bone destruction in inflammatory arthritis. Recently, a mouse model of cherubism (Sh3bp2KI/KI ) with a homozygous gain-of-function mutation in the SH3-domain binding protein 2 (SH3BP2) was shown to develop auto-inflammatory joint destruction. Here, we demonstrate that Sh3bp2KI/KI mice also deficient in the FBJ osteosarcoma oncogene (c-Fos) still exhibit noticeable bone erosion at the distal tibia even in the absence of osteoclasts at 12 weeks old. Levels of serum collagen I C-terminal telopeptide (ICTP), a marker of bone resorption generated by matrix metalloproteinases (MMPs), were elevated, whereas levels of serum cross-linked C-telopeptide (CTX), another resorption marker produced by cathepsin K, were not increased. Collagenolytic MMP levels were increased in the inflamed joints of the Sh3bp2KI/KI mice deficient in c-Fos. Resorption pits contained a large number of F4/80+ macrophages and genetic depletion of macrophages rescued these erosive changes. Importantly, administration of NSC405020, an MMP14 inhibitor targeted to the hemopexin (PEX) domain, suppressed bone erosion in c-Fos-deficient Sh3bp2KI/KI mice. After activation of the NF-κB pathway, macrophage colony-stimulating factor (M-CSF)-dependent macrophages from c-Fos-deficient Sh3bp2KI/KI mice expressed increased amounts of MMP14 compared with wild-type macrophages. Interestingly, receptor activator of NF-κB ligand (RANKL)-deficient Sh3bp2KI/KI mice failed to show notable bone erosion, whereas c-Fos deletion did restore bone erosion to the RANKL-deficient Sh3bp2KI/KI mice, suggesting that osteolytic transformation of macrophages requires both loss-of-function of c-Fos and gain-of-function of SH3BP2 in this model. These data provide the first genetic evidence that cells other than osteoclasts can cause focal bone destruction in inflammatory bone disease and suggest that MMP14 is a key mediator conferring pathological bone-resorbing capacity on c-Fos-deficient Sh3bp2KI/KI macrophages. In summary, the paradigm that osteoclasts are the exclusive cells executing inflammatory bone destruction may need to be reevaluated based on our findings with c-Fos-deficient cherubism mice lacking osteoclasts. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Bone and Bones/pathology , Cherubism/pathology , Inflammation/pathology , Macrophages/pathology , Matrix Metalloproteinase 14/metabolism , Osteoclasts/metabolism , Proto-Oncogene Proteins c-fos/deficiency , Tartrate-Resistant Acid Phosphatase/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Joints/drug effects , Joints/pathology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Macrophages/enzymology , Matrix Metalloproteinase 2/metabolism , Mice , NF-kappa B/metabolism , Osteoclasts/drug effects , Proto-Oncogene Proteins c-fos/metabolism , RANK Ligand/deficiency , RANK Ligand/metabolism , Signal Transduction/drug effects
4.
Arthritis Rheumatol ; 67(3): 656-67, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25470448

ABSTRACT

OBJECTIVE: SH3 domain-binding protein 2 (SH3BP2) is a signaling adapter protein that regulates the immune and skeletal systems. The present study was undertaken to investigate the role of SH3BP2 in arthritis using 2 experimental mouse models, i.e., human tumor necrosis factor α-transgenic (hTNF-Tg) mice and mice with collagen-induced arthritis (CIA). METHODS: First, Sh3bp2(-/-) and wild-type (Sh3bp2(+/+) ) mice were crossed with hTNF-Tg mice. Inflammation and bone loss were examined by clinical inspection and histologic and micro-computed tomography analysis, and osteoclastogenesis was evaluated using primary bone marrow-derived macrophage colony-stimulating factor-dependent macrophages (BMMs). Second, CIA was induced in Sh3bp2(-/-) and Sh3bp2(+/+) mice, and the incidence and severity of arthritis were evaluated. Anti-mouse type II collagen (CII) antibody levels were measured by enzyme-linked immunosorbent assay, and lymph node cell responses to CII were determined. RESULTS: SH3BP2 deficiency did not alter the severity of joint swelling but did suppress bone erosion in the hTNF-Tg mouse model. Bone loss at the talus and tibia was prevented in Sh3bp2(-/-) /hTNF-Tg mice compared to Sh3bp2(+/+) /hTNF-Tg mice. RANKL- and TNFα-induced osteoclastogenesis was suppressed in Sh3bp2(-/-) mouse BMM cultures. NF-ATc1 nuclear localization in response to TNFα was decreased in Sh3bp2(-/-) mouse BMMs compared to Sh3bp2(+/+) mouse BMMs. In the CIA model, SH3BP2 deficiency suppressed the incidence of arthritis and this was associated with decreased anti-CII antibody production, while antigen-specific T cell responses in lymph nodes were not significantly different between Sh3bp2(+/+) and Sh3bp2(-/-) mice. CONCLUSION: SH3BP2 deficiency prevents loss of bone via impaired osteoclastogenesis in the hTNF-Tg mouse model and suppresses the induction of arthritis via decreased autoantibody production in the CIA model. Therefore, SH3BP2 could potentially be a therapeutic target in rheumatoid arthritis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Arthritis, Experimental/prevention & control , Bone Resorption/prevention & control , Tumor Necrosis Factor-alpha/genetics , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Autoantibodies/blood , Blotting, Western , Bone Resorption/metabolism , Bone Resorption/pathology , Collagen Type II/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , X-Ray Microtomography
5.
Bone ; 71: 201-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25445458

ABSTRACT

Cherubism (OMIM#118400) is a genetic disorder in children characterized by excessive jawbone destruction with proliferation of fibro-osseous lesions containing a large number of osteoclasts. Mutations in the SH3-domain binding protein 2 (SH3BP2) are responsible for cherubism. Analysis of the knock-in (KI) mouse model of cherubism showed that homozygous cherubism mice (Sh3bp2(KI/KI)) spontaneously develop systemic autoinflammation and inflammatory bone loss and that cherubism is a TNF-α-dependent hematopoietic disorder. In this study, we investigated whether bone marrow transplantation (BMT) is effective for the treatment of inflammation and bone loss in Sh3bp2(KI/KI) mice. Bone marrow (BM) cells from wild-type (Sh3bp2(+/+)) mice were transplanted to 6-week-old Sh3bp2(KI/KI) mice with developing inflammation and to 10-week-old Sh3bp2(KI/KI) mice with established inflammation. Six-week-old Sh3bp2(KI/KI) mice transplanted with Sh3bp2(+/+) BM cells exhibited improved body weight loss, facial swelling, and survival rate. Inflammatory lesions in the liver and lung as well as bone loss in calvaria and mandibula were ameliorated at 10weeks after BMT compared to Sh3bp2(KI/KI) mice transplanted with Sh3bp2(KI/KI) BM cells. Elevation of serum TNF-α levels was not detected after BMT. BMT was effective for up to 20weeks in 6-week-old Sh3bp2(KI/KI) mice transplanted with Sh3bp2(+/+) BM cells. BMT also ameliorated the inflammation and bone loss in 10-week-old Sh3bp2(KI/KI) mice. Thus our study demonstrates that BMT improves the inflammation and bone loss in cherubism mice. BMT may be effective for the treatment of cherubism patients.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Marrow Transplantation , Bone Resorption/pathology , Cherubism/pathology , Gene Knock-In Techniques , Inflammation/pathology , Inflammation/therapy , Adaptor Proteins, Signal Transducing/deficiency , Animals , Body Weight , Bone Marrow Cells/metabolism , Female , Inflammation/blood , Liver/pathology , Lung/pathology , Male , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/blood
6.
Cell Rep ; 8(6): 1752-1766, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25220465

ABSTRACT

Cherubism is caused by mutations in SH3BP2. Studies of cherubism mice showed that tumor necrosis factor α (TNF-α)-dependent autoinflammation is a major cause of the disorder but failed to explain why human cherubism lesions are restricted to jaws and regress after puberty. We demonstrate that the inflammation in cherubism mice is MYD88 dependent and is rescued in the absence of TLR2 and TLR4. However, germ-free cherubism mice also develop inflammation. Mutant macrophages are hyperresponsive to PAMPs (pathogen-associated molecular patterns) and DAMPs (damage-associated molecular patterns) that activate Toll-like receptors (TLRs), resulting in TNF-α overproduction. Phosphorylation of SH3BP2 at Y183 is critical for the TNF-α production. Finally, SYK depletion in macrophages prevents the inflammation. These data suggest that the presence of a large amount of TLR ligands, presumably oral bacteria and DAMPs during jawbone remodeling, may cause the jaw-specific development of human cherubism lesions. Reduced levels of DAMPs after stabilization of jaw remodeling may contribute to the age-dependent regression.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cherubism/etiology , Inflammation , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Jaw/diagnostic imaging , Liver/pathology , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , NF-kappa B/metabolism , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/metabolism , Radiography , Syk Kinase , Toll-Like Receptor 2/chemistry , Toll-Like Receptor 4/chemistry , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
PLoS One ; 9(8): e105518, 2014.
Article in English | MEDLINE | ID: mdl-25144740

ABSTRACT

OBJECTIVE: SH3BP2 is a signaling adapter protein which regulates immune and skeletal systems. Gain-of-function mutations in SH3BP2 cause cherubism, characterized by jawbone destruction. This study was aimed to examine the role of SH3BP2 in inflammatory bone loss using a collagen-induced arthritis (CIA) model. METHODS: CIA was induced in wild-type (Sh3bp2(+/+)) and heterozygous P416R SH3BP2 cherubism mutant knock-in (Sh3bp2(KI/+)) mice, an SH3BP2 gain-of-function model. Severity of the arthritis was determined by assessing the paw swelling and histological analyses of the joints. Micro-CT analysis was used to determine the levels of bone loss. Inflammation and osteoclastogenesis in the joints were evaluated by quantitating the gene expression of inflammatory cytokines and osteoclast markers. Furthermore, involvement of the T- and B-cell responses was determined by draining lymph node cell culture and measurement of the serum anti-mouse type II collagen antibody levels, respectively. Finally, roles of the SH3BP2 mutation in macrophage activation and osteoclastogenesis were determined by evaluating the TNF-α production levels and osteoclast formation in bone marrow-derived M-CSF-dependent macrophage (BMM) cultures. RESULTS: Sh3bp2(KI/+) mice exhibited more severe inflammation and bone loss, accompanying an increased number of osteoclasts. The mRNA levels for TNF-α and osteoclast marker genes were higher in the joints of Sh3bp2(KI/+) mice. Lymph node cell culture showed that lymphocyte proliferation and IFN-γ and IL-17 production were comparable between Sh3bp2(+/+) and Sh3bp2(KI/+) cells. Serum anti-type II collagen antibody levels were comparable between Sh3bp2(+/+) and Sh3bp2(KI/+) mice. In vitro experiments showed that TNF-α production in Sh3bp2(KI/+) BMMs is elevated compared with Sh3bp2(+/+) BMMs and that RANKL-induced osteoclastogenesis is enhanced in Sh3bp2(KI/+) BMMs associated with increased NFATc1 nuclear localization. CONCLUSION: Gain-of-function of SH3BP2 augments inflammation and bone loss in the CIA model through increased macrophage activation and osteoclast formation. Therefore, modulation of the SH3BP2 expression may have therapeutic potential for the treatment of rheumatoid arthritis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Bone Resorption/genetics , Mutation , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Substitution , Animals , Arthritis, Experimental/diagnosis , Arthritis, Experimental/immunology , Cartilage/pathology , Cell Proliferation , Cytokines/metabolism , Gene Expression , Lymphoid Tissue/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Transgenic , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , RANK Ligand/metabolism , RANK Ligand/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , X-Ray Microtomography
8.
J Bone Miner Res ; 29(5): 1170-82, 2014.
Article in English | MEDLINE | ID: mdl-24978678

ABSTRACT

Cherubism is a genetic disorder of the craniofacial skeleton caused by gain-of-function mutations in the signaling adaptor protein, SH3-domain binding protein 2 (SH3BP2). In a knock-in mouse model for cherubism, we previously demonstrated that homozygous mutant mice develop T/B cell-independent systemic macrophage inflammation leading to bone erosion and joint destruction. Homozygous mice develop multiostotic bone lesions whereas cherubism lesions in humans are limited to jawbones. We identified a critical role of tumor necrosis factor α (TNF-α) in the development of autoinflammation by creating homozygous TNF-α-deficient cherubism mutants, in which systemic inflammation and bone destruction were rescued. In this study, we examined whether postnatal administration of an anti-TNF-α antagonist can prevent or ameliorate the disease progression in cherubism mice. Neonatal homozygous mutants, in which active inflammation has not yet developed, were treated with a high dose of etanercept (25 mg/kg, twice/week) for 7 weeks. Etanercept-treated neonatal mice showed strong rescue of facial swelling and bone loss in jaws and calvariae. Destruction of joints was fully rescued in the high-dose group. Moreover, the high-dose treatment group showed a significant decrease in lung and liver inflammatory lesions. However, inflammation and bone loss, which were successfully treated by etanercept administration, recurred after etanercept discontinuation. No significant effect was observed in low-dose-treated (0.5 mg/kg, twice/week) and vehicle-treated groups. In contrast, when 10-week-old cherubism mice with fully active inflammation were treated with etanercept for 7 weeks, even the high-dose administration did not decrease bone loss or lung or liver inflammation. Taken together, the results suggest that anti-TNF-α therapy may be effective in young cherubism patients, if treated before the inflammatory phase or bone resorption occurs. Therefore, early genetic diagnosis and early treatment with anti-TNF-α antagonists may be able to prevent or ameliorate cherubism, especially in patients with a mutation in SH3BP2.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/metabolism , Cherubism/drug therapy , Cherubism/metabolism , Immunoglobulin G/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/pathology , Cherubism/genetics , Cherubism/pathology , Etanercept , Gene Knock-In Techniques , Humans , Inflammation , Mice , Mice, Mutant Strains , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics
9.
J Bone Miner Res ; 29(12): 2618-35, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24916406

ABSTRACT

Cherubism (OMIM# 118400) is a genetic disorder with excessive jawbone resorption caused by mutations in SH3 domain binding protein 2 (SH3BP2), a signaling adaptor protein. Studies on the mouse model for cherubism carrying a P416R knock-in (KI) mutation have revealed that mutant SH3BP2 enhances tumor necrosis factor (TNF)-α production and receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation in myeloid cells. TNF-α is expressed in human cherubism lesions, which contain a large number of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells, and TNF-α plays a critical role in inflammatory bone destruction in homozygous cherubism mice (Sh3bp2(KI/KI) ). The data suggest a pathophysiological relationship between mutant SH3BP2 and TNF-α-mediated bone loss by osteoclasts. Therefore, we investigated whether P416R mutant SH3BP2 is involved in TNF-α-mediated osteoclast formation and bone loss. Here, we show that bone marrow-derived M-CSF-dependent macrophages (BMMs) from the heterozygous cherubism mutant (Sh3bp2(KI/+) ) mice are highly responsive to TNF-α and can differentiate into osteoclasts independently of RANKL in vitro by a mechanism that involves spleen tyrosine kinase (SYK) and phospholipase Cγ2 (PLCγ2) phosphorylation, leading to increased nuclear translocation of NFATc1. The heterozygous cherubism mutation exacerbates bone loss with increased osteoclast formation in a mouse calvarial TNF-α injection model as well as in a human TNF-α transgenic mouse model (hTNFtg). SH3BP2 knockdown in RAW264.7 cells results in decreased TRAP-positive multinucleated cell formation. These findings suggest that the SH3BP2 cherubism mutation can cause jawbone destruction by promoting osteoclast formation in response to TNF-α expressed in cherubism lesions and that SH3BP2 is a key regulator for TNF-α-induced osteoclastogenesis. Inhibition of SH3BP2 expression in osteoclast progenitors could be a potential strategy for the treatment of bone loss in cherubism as well as in other inflammatory bone disorders.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Resorption/metabolism , Cherubism/metabolism , Mutation , NFATC Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Resorption/genetics , Bone Resorption/pathology , Cherubism/genetics , Cherubism/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , NFATC Transcription Factors/genetics , Osteoclasts/metabolism , Osteoclasts/pathology , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Syk Kinase , Tumor Necrosis Factor-alpha/genetics
10.
J Orthop Res ; 31(12): 1992-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23940002

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs capable of inhibiting gene expression post-transcriptionally and expression profiling can provide therapeutic targets and tools for cancer diagnosis. Chondrosarcoma is a mesenchymal tumor with unknown cause and differentiation status. Here, we profiled miRNA expression of chondrosarcoma, namely clinical samples from human conventional chondrosarcoma tissue, established chondrosarcoma cell lines, and primary non-tumorous adult articular chondrocytes, by miRNA array and quantitative real-time PCR. A wide variety of miRNAs were differently downregulated in chondrosarcoma compared to non-tumorous articular chondrocytes; 27 miRNAs: miR-10b, 23b, 24-1*, 27b, 100, 134, 136, 136*, 138, 181d, 186, 193b, 221*, 222, 335, 337-5p, 376a, 376a*, 376b, 376c, 377, 454, 495, 497, 505, 574-3p, and 660, were significantly downregulated in chondrosarcoma and only 2: miR-96 and 183, were upregulated. We further validated the expression levels of miRNAs by quantitative real-time PCR for miR-181a, let-7a, 100, 222, 136, 376a, and 335 in extended number of chondrosarcoma clinical samples. Among them, all except miR-181a were found to be significantly downregulated in chondrosarcoma derived samples. The findings provide potential diagnostic value and new molecular understanding of chondrosarcoma.


Subject(s)
Bone Neoplasms/genetics , Chondrosarcoma/genetics , MicroRNAs/analysis , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Middle Aged
11.
Proc Natl Acad Sci U S A ; 107(23): 10538-42, 2010 Jun 08.
Article in English | MEDLINE | ID: mdl-20498044

ABSTRACT

Mohawk (Mkx) is a member of the Three Amino acid Loop Extension superclass of atypical homeobox genes that is expressed in developing tendons. To investigate the in vivo functions of Mkx, we generated Mkx(-/-) mice. These mice had hypoplastic tendons throughout the body. Despite the reduction in tendon mass, the cell number in tail tendon fiber bundles was similar between wild-type and Mkx(-/-) mice. We also observed small collagen fibril diameters and a down-regulation of type I collagen in Mkx(-/-) tendons. These data indicate that Mkx plays a critical role in tendon differentiation by regulating type I collagen production in tendon cells.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Tendons/growth & development , Tendons/metabolism , Animals , Collagen Type I/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Tendons/cytology , Tendons/embryology , Tensile Strength
12.
Mod Rheumatol ; 20(1): 11-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19784544

ABSTRACT

Recent studies suggest that histone deacetylase (HDAC) inhibitors may therapeutically prevent cartilage degradation in osteoarthritis (OA). Matrix metalloproteinase-13 (MMP-13) plays an important role in the pathogenesis of this disease and in the present study we investigated the correlation between HDACs and MMP-13. Comparing the expression of different HDACs in cartilage from OA patients and healthy donors, HDAC7 showed a significant elevation in cartilage from OA patients. High level of HDAC7 expression in OA cartilage was also confirmed by immunohistochemistry. Knockdown of HDAC7 by small interference RNA (siRNA) in SW1353 human chondrosarcoma cells strongly suppressed interleukin (IL)-1-dependent and independent induction of MMP-13 gene expression. In conclusion, elevated HDAC7 expression in human OA may contribute to cartilage degradation via promoting MMP-13 gene expression, suggesting the critical role of MMP-13 in OA pathogenesis.


Subject(s)
Gene Expression Regulation, Enzymologic , Histone Deacetylases/metabolism , Matrix Metalloproteinase 13/metabolism , Osteoarthritis, Knee/metabolism , Adult , Biomarkers/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Line, Tumor , Chondrocytes/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockdown Techniques , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Humans , Hydroxamic Acids/pharmacology , Immunohistochemistry , Matrix Metalloproteinase 13/genetics , Middle Aged , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , RNA, Small Interfering/genetics , Young Adult
13.
J Neurotrauma ; 27(4): 739-51, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20038195

ABSTRACT

Spinal cord injury results in disruption of the cord microstructure, which is followed by inflammation leading to additional deterioration. Perivascular basement membranes are a component of the spinal cord microstructure that lies between blood vessels and astrocytes. The impact of disrupting the basement membrane structure on the expansion of inflammation has not been fully examined. The objective of this study was to clarify the relationship between damage to basement membranes and inflammation after spinal cord injury. Immunohistochemical analyses of the perivascular extracellular matrix were performed in a mouse spinal cord injury model. In normal tissue, the perivascular basement membrane was a single-layer structure produced by both endothelial cells and surrounding astrocytes. After spinal cord injury, however, the perivascular basement membrane often separated into an inner endothelial basement membrane and an outer parenchymal basement membrane. The altered basement membranes formed during the acute phase (within 7 days after spinal cord injury). During the subacute phase of injury, numerous monocytes and macrophages accumulated in the space between the separated basement membranes and infiltrated into the parenchyma where astrocytic endfeet were displaced. Infiltration of inflammatory cells from the injury core was attenuated coincident with the appearance of the glia limitans and glial scar. Furthermore, the outer parenchymal basement membrane was connected to the basement membrane of the glia limitans surrounding the injury core. Our data suggest that structurally altered basement membranes facilitate expansion of secondary inflammation during the subacute phase of spinal cord injury.


Subject(s)
Basement Membrane/physiopathology , Blood Vessels/physiopathology , Chemotaxis, Leukocyte/physiology , Myelitis/physiopathology , Spinal Cord Injuries/physiopathology , Spinal Cord/blood supply , Spinal Cord/physiopathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Basement Membrane/pathology , Blood Vessels/pathology , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Gliosis/etiology , Gliosis/pathology , Gliosis/physiopathology , Immunohistochemistry , Macrophages/cytology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/physiology , Myelitis/etiology , Myelitis/pathology , Spinal Cord/pathology , Spinal Cord Injuries/complications , Spinal Cord Injuries/pathology
14.
Mod Rheumatol ; 18(4): 359-65, 2008.
Article in English | MEDLINE | ID: mdl-18461274

ABSTRACT

We prospectively evaluated the disease-specific features of the early postoperative plasma D: -dimer value and the relationship with deep venous thrombosis and/or pulmonary thromboembolism (DVT/PE) in 95 patients following total knee arthroplasty. Patients in whom DVT/PE was highly suspected were diagnosed by high-resolution multi-detector row computed tomography scanning (MDCT). Forty-nine knees in 46 patients with rheumatoid arthritis (RA, 24 knees) or osteoarthritis (OA, 25 knees) were finally recruited. DVT/PE was detected in 28 (57.1%) of the 49 cases examined by diagnostic MDCT: 12 (50.0%) of the 24 cases of RA, and 16 (64.0%) of the 25 cases of OA. Of these, PE was found in 11 cases (39.2%), but none of them showed clinical symptomatic signs of dyspnea or chest pain. In both RA and OA cases, there were statistically significant differences in the D: -dimer value on postoperative day 3 (P = 0.027) and after day 28 (P = 0.037) between the groups with and without DVT/PE. In OA cases, there were significant differences between the two groups on postoperative days 1 (P = 0.034), 3 (P = 0.020), 5 (P = 0.005), and 7 (P = 0.045), respectively. At the baseline, perioperative D: -dimer levels in the RA group without DVT/PE were higher than in the OA group. However, multivariate logistic regression analysis showed that RA was not a significant risk factor of DVT/PE in comparison with OA. In conclusion, individual evaluation of the D: -dimer level between RA and OA should provide a more precise predictive indicator of early postoperative DVT/PE.


Subject(s)
Arthroplasty, Replacement, Knee/adverse effects , Fibrin Fibrinogen Degradation Products/analysis , Pulmonary Embolism/diagnosis , Venous Thrombosis/diagnosis , Adult , Aged , Aged, 80 and over , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/surgery , Female , Humans , Male , Middle Aged , Osteoarthritis, Knee/complications , Osteoarthritis, Knee/surgery , Prospective Studies , Pulmonary Embolism/etiology , ROC Curve , Venous Thrombosis/etiology
15.
Glia ; 44(3): 190-204, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14603461

ABSTRACT

We report the molecular cloning of a new member of the transmembrane-type immunoglobulin superfamily and designate the encoded protein as limitrin, since it localized selectively to glia limitans in mouse brain. Limitrin cDNA was obtained using a subtractive hybridization procedure designed to identify molecules responsible for blood-brain barrier function. Western blots using a limitrin-specific antibody demonstrated that the gene product is expressed significantly in mouse brain and primary murine astrocytes and is distributed in the plasma membrane. Immunohistochemical studies using confocal and electron microscopy clearly demonstrated highly polarized localization in astroglial endfeet in the perivascular region and under the pia mater in vivo. Limitrin is expressed in the spinal cord and in many areas of the brain, but not in the median eminence or subfornical organ (the circumventricular organs), where the blood-brain barrier is lacking. Disruption of the blood-brain barrier by cold injury resulted in a drastic reduction in limitrin expression. Furthermore, during retrieval from cold injury, the increased expression of limitrin in perivascular endfeet correlated with the recovery of angiogenesis in capillaries within the lesion margins. Our results suggest that limitrin is physically and functionally associated with the blood-brain barrier, implying that this protein may be useful as a diagnostic tool of barrier integrity.


Subject(s)
Astrocytes/physiology , Blood-Brain Barrier/physiology , Immunoglobulins/genetics , Membrane Proteins/genetics , Pseudopodia/physiology , Amino Acid Sequence , Animals , Antibody Specificity , Astrocytes/chemistry , Base Sequence , Cloning, Molecular , Female , Gene Expression , Immunoglobulins/analysis , Immunoglobulins/immunology , Median Eminence/physiology , Membrane Proteins/analysis , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Pseudopodia/chemistry , Rabbits , Subfornical Organ/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...