Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Am J Pathol ; 190(10): 2123-2135, 2020 10.
Article in English | MEDLINE | ID: mdl-32650003

ABSTRACT

Tenascin-C (TNC) is strongly expressed by fibroblasts and cancer cells in breast cancer. To assess the effects of TNC on stromal formation, we examined phenotypic changes in human mammary fibroblasts treated with TNC. The addition of TNC significantly up-regulated α-smooth muscle actin (α-SMA) and calponin. TNC increased the number of α-SMA- and/or calponin-positive cells with well-developed stress fibers in immunofluorescence, which enhanced contractile ability in collagen gel contraction. The treatment with TNC also significantly up-regulated its own synthesis. Double immunofluorescence of human breast cancer tissues showed α-SMA- and/or calponin-positive myofibroblasts in the TNC-deposited stroma. Among several receptors for TNC, the protein levels of the αv and ß1 integrin subunits were significantly increased after the treatment. Immunofluorescence showed the augmented colocalization of αv and ß1 at focal adhesions. Immunoprecipitation using an anti-αv antibody revealed a significant increase in coprecipitated ß1 with TNC in lysates. The knockdown of αv and ß1 suppressed the up-regulation of α-SMA and calponin. The addition of TNC induced the phosphorylation of SMAD2/3, whereas SB-505124 and SIS3 blocked myofibroblast differentiation. Therefore, TNC enhances its own synthesis by forming a positive feedback loop and increases integrin αvß1 heterodimer levels to activate transforming growth factor-ß signaling, which is followed by a change to highly contractile myofibroblasts. TNC may essentially contribute to the stiffer stromal formation characteristic of breast cancer tissues.


Subject(s)
Breast Neoplasms/pathology , Fibroblasts/pathology , Myofibroblasts/pathology , Tenascin/pharmacology , Breast Neoplasms/metabolism , Cell Differentiation/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/pharmacology , Fibroblasts/metabolism , Humans , Myofibroblasts/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Phosphorylation/drug effects , Receptors, Vitronectin/metabolism , Signal Transduction/physiology , Tenascin/metabolism , Transforming Growth Factor beta/metabolism
2.
Am J Physiol Cell Physiol ; 319(5): C781-C796, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32845719

ABSTRACT

Tenascin-C (TNC) is a large extracellular matrix glycoprotein classified as a matricellular protein that is generally upregulated at high levels during physiological and pathological tissue remodeling and is involved in important biological signaling pathways. In the heart, TNC is transiently expressed at several important steps during embryonic development and is sparsely detected in normal adult heart but is re-expressed in a spatiotemporally restricted manner under pathological conditions associated with inflammation, such as myocardial infarction, hypertensive cardiac fibrosis, myocarditis, dilated cardiomyopathy, and Kawasaki disease. Despite its characteristic and spatiotemporally restricted expression, TNC knockout mice develop a grossly normal phenotype. However, various disease models using TNC null mice combined with in vitro experiments have revealed many important functions for TNC and multiple molecular cascades that control cellular responses in inflammation, tissue repair, and even myocardial regeneration. TNC has context-dependent diverse functions and, thus, may exert both harmful and beneficial effects in damaged hearts. However, TNC appears to deteriorate adverse ventricular remodeling by proinflammatory and profibrotic effects in most cases. Its specific expression also makes TNC a feasible diagnostic biomarker and target for molecular imaging to assess inflammation in the heart. Several preclinical studies have shown the utility of TNC as a biomarker for assessing the prognosis of patients and selecting appropriate therapy, particularly for inflammatory heart diseases.


Subject(s)
Cardiomyopathy, Dilated/genetics , Endomyocardial Fibrosis/genetics , Mucocutaneous Lymph Node Syndrome/genetics , Myocardial Infarction/genetics , Myocarditis/genetics , Tenascin/genetics , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Disease Models, Animal , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Gene Expression Regulation , Humans , Inflammation , Mice , Mice, Knockout , Mucocutaneous Lymph Node Syndrome/metabolism , Mucocutaneous Lymph Node Syndrome/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocarditis/metabolism , Myocarditis/pathology , Myocardium/metabolism , Myocardium/pathology , Tenascin/metabolism , Ventricular Remodeling/genetics , Wound Healing/genetics
3.
J Neurosci Res ; 98(1): 42-56, 2020 01.
Article in English | MEDLINE | ID: mdl-30242870

ABSTRACT

Subarachnoid hemorrhage (SAH) by a rupture of cerebral aneurysms remains the most devastating cerebrovascular disease. Early brain injury (EBI) is increasingly recognized to be the primary determinant for poor outcomes, and also considered to cause delayed cerebral ischemia (DCI) after SAH. Both clinical and experimental literatures emphasize the impact of global cerebral edema in EBI as negative prognostic and direct pathological factors. The nature of the global cerebral edema is a mixture of cytotoxic and vasogenic edema, both of which may be caused by post-SAH induction of tenascin-C (TNC) that is an inducible, non-structural, secreted and multifunctional matricellular protein. Experimental SAH induces TNC in brain parenchyma in rats and mice. TNC knockout suppressed EBI in terms of brain edema, blood-brain barrier disruption, neuronal apoptosis and neuroinflammation, associated with the inhibition of post-SAH activation of mitogen-activated protein kinases and nuclear factor-kappa B in mice. In a clinical setting, more severe SAH increases more TNC in cerebrospinal fluid and peripheral blood, which could be a surrogate marker of EBI and predict DCI development and outcomes. In addition, cilostazol, a selective inhibitor of phosphodiesterase type III that is a clinically available anti-platelet agent and is known to suppress TNC induction, dose-dependently inhibited delayed cerebral infarction and improved outcomes in a pilot clinical study. Thus, further studies may facilitate application of TNC as biomarkers for non-invasive diagnosis or assessment of EBI and DCI, and lead to development of a molecular target drug against TNC, contributing to the improvement of post-SAH outcomes.


Subject(s)
Brain Edema/metabolism , Brain Injuries/metabolism , Subarachnoid Hemorrhage/metabolism , Tenascin/metabolism , Animals , Brain Edema/etiology , Extracellular Matrix/metabolism , Humans , Subarachnoid Hemorrhage/complications
4.
Acta Neurochir Suppl ; 127: 91-96, 2020.
Article in English | MEDLINE | ID: mdl-31407069

ABSTRACT

Toll-like receptor 4 (TLR4) is expressed in various cell types in the central nervous system and exerts maximal inflammatory responses among the TLR family members. TLR4 can be activated by many endogenous ligands having damage-associated molecular patterns including heme and fibrinogen at the rupture of a cerebral aneurysm, and therefore its activation is reasonable as an initial step of cascades to brain injuries after aneurysmal subarachnoid hemorrhage (SAH). TLR4 activation induces tenascin-C (TNC), a representative of matricellular proteins that are a class of inducible, nonstructural, secreted, and multifunctional extracellular matrix glycoproteins. TNC is also an endogenous activator and inducer of TLR4, forming positive feedback mechanisms leading to more activation of the signaling transduction. Our studies have demonstrated that TLR4 as well as TNC are involved in inflammatory reactions, blood-brain barrier disruption, neuronal apoptosis, and cerebral vasospasm after experimental SAH. This article reviews recent understanding of TLR4 and TNC in SAH to suggest that the TLR4-TNC signaling may be an important therapeutic target for post-SAH brain injuries.


Subject(s)
Brain Injuries , Subarachnoid Hemorrhage , Tenascin , Toll-Like Receptor 4 , Vasospasm, Intracranial , Brain Injuries/metabolism , Extracellular Matrix , Humans , Subarachnoid Hemorrhage/metabolism , Tenascin/metabolism , Toll-Like Receptor 4/metabolism , Vasospasm, Intracranial/metabolism
5.
J Orthop Sci ; 25(2): 324-330, 2020 Mar.
Article in English | MEDLINE | ID: mdl-30975540

ABSTRACT

BACKGROUND: The effects of tenascin-C (TNC) on cartilage repair were examined in cartilage defect model mice. An in vitro study was also performed to determine the mechanism of cartilage repair with TNC. METHODS: Full-thickness osteochondral defects were filled with TNC (group A: 100 µg/ml, group B: 10 µg/ml, group C: empty). Mice were sacrificed at 1, 2, 3, and 6 weeks postoperatively. Cartilage repair was histologically evaluated using the modified WAKITANI score. Chondrocytes were isolated and cultured, and they were treated with TNC. The expressions of various mRNAs including TNC, inflammatory cytokines, and anabolic and catabolic factors for cartilage were compared by real-time polymerase chain reaction. RESULTS: The defects in group A were covered with hyaline-like cartilage after 3 weeks. Average modified WAKITANI scores were significantly better in group A than in groups B and C at 3 and 6 weeks. TNC upregulated the expressions of endogenous TNC, inflammatory cytokines, and anabolic and catabolic factors for cartilage. TNC downregulated the expression of a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) 5. CONCLUSIONS: Intra-articular injection of full-length TNC repaired cartilage in murine models of full-thickness osteochondral defects. TNC upregulated the expression of ADAMTS4, but downregulated the expression of ADAMTS5 that contributed to cartilage degradation.


Subject(s)
Cartilage Diseases/drug therapy , Cartilage, Articular/drug effects , Knee Joint/drug effects , Tenascin/administration & dosage , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Humans , Injections, Intra-Articular , Mice , Mice, Inbred BALB C , Middle Aged
6.
Mod Rheumatol ; 30(4): 765-772, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31339809

ABSTRACT

Objectives: Rebamipide is a protective drug used for gastric mucosal injuries, and it also exerts protective effects for a variety of other tissues. In this study, murine post-traumatic (PT) osteoarthritis (OA) models in vivo and human OA chondrocytes in vitro were used to examine the effects of rebamipide on articular cartilage degeneration.Methods: Male BALB/c mice were used. The knee ligaments were transected in both knees. Mice were injected with rebamipide into the knee joint every week. Human chondrocytes were stimulated with IL-1ß, then treated with or without rebamipide. The levels of mRNA expression of COL2A, IL-1ß, TNFα, NF-κB, MMP3, MMP13, ADAMTS5, TIMP3, FGF2, and TGFß were estimated using real-time PCR.Results: Histological scores were significantly better in the rebamipide 1 mg/mL and 10 mg/mL groups than in the control group. Rebamipide up-regulated the mRNA expressions of COL2A, TIMP3, TGFß, and FGF2 in chondrocytes and down-regulated IL-1ß, TNFα, NF-κB, MMP3, MMP13, and ADAMTS5.Conclusion: Intra-articular injection of rebamipide prevents articular cartilage degeneration for 6 weeks in murine models of OA in vivo. Rebamipide down-regulates inflammatory cytokines and catabolic factors and up-regulates anabolic factors in human chondrocytes in vitro. Rebamipide could be an important treatment for prevention of articular cartilage degeneration.


Subject(s)
Alanine/analogs & derivatives , Antioxidants/therapeutic use , Cartilage, Articular/drug effects , Osteoarthritis/drug therapy , Quinolones/therapeutic use , Alanine/administration & dosage , Alanine/pharmacology , Alanine/therapeutic use , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacology , Cartilage, Articular/metabolism , Injections, Intra-Articular , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Quinolones/administration & dosage , Quinolones/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Int Heart J ; 60(1): 151-158, 2019 Jan 25.
Article in English | MEDLINE | ID: mdl-30464121

ABSTRACT

Inflammation after myocardial infarction (MI) may be a major factor influencing ventricular remodeling, leading to congestive heart failure and arrhythmia. Therefore, inflammation in the heart needs to be monitored. Tenascin-C (TNC) is an extracellular matrix molecule not normally expressed, but it is strongly upregulated when associated with active inflammation. Based on this characteristic, we successfully imaged in vivo inflammatory lesions in rat models using 111Indium (111In)-labeled anti-TNC antibodies. The aim of the present study was to further assess the applicability of this molecular imaging probe to detect inflammatory activity in primate hearts.We generated an MI model of cynomolgus monkeys (Macaca fascicularis) by coronary artery ligation and performed dual-isotope single-photon emission computed tomography (SPECT) imaging with an 111In-labeled anti-TNC antibody Fab' fragment (111In-TNC Fab') and 99mtechnetium methoxy-isobutyl isonitrile (99mTc-MIBI). Dual autoradiography was used to compare the uptake of 111In-TNC Fab' with histology and immunostaining for TNC. Dual-isotope SPECT showed the regional myocardial uptake of 111In-TNC Fab' complementary to a defect in the perfusion image by 99mTc-MIBI. The high radioactivity of 111In-TNC Fab' by autoradiography corresponded to immunostaining for TNC, which was observed in inflammatory lesions at the border zone between the infarcted and non-infarcted areas of the left ventricle and at the epi/pericarditis lesions of the right ventricle. These results demonstrate the potential of 111In-TNC-Fab' imaging to monitor myocardial injury and inflammation and suggest the feasibility of the non-invasive detection of cardiac inflammation following acute MI in a preclinical stage before testing in humans.


Subject(s)
Inflammation/pathology , Molecular Imaging/methods , Myocardial Infarction/pathology , Tenascin/immunology , Animals , Coronary Vessels/surgery , Environmental Biomarkers , Extracellular Matrix/pathology , Heart , Indium , Inflammation/diagnostic imaging , Inflammation/veterinary , Ligation , Macaca fascicularis , Myocardial Infarction/complications , Myocardial Infarction/diagnostic imaging , Primates , Rats , Tomography, Emission-Computed, Single-Photon/methods , Ventricular Remodeling
8.
Artif Organs ; 42(6): E90-E101, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29473183

ABSTRACT

In vitro biocompatibility assessments that consider physiologically appropriate conditions of cell exposure to peritoneal dialysis fluids (PDFs) are still awaited. In this study, we found that fragmentation of Golgi apparatus occurred in a pH-dependent manner within 30-min exposure to five distinct commercially available PDFs, which showed no marked difference in their effects on cell viability in the conventional MTT assay. Fluorescence microscopy analysis of labeling antibody against cis-Golgi protein GM130 indicated that the stacked cisternal structure was maintained in the perinuclear area of both M199 culture medium and a neutral-pH PDF groups. However, this specific structure became partially disassembled over time even in a neutral-pH PDF, and fragmentation was markedly enhanced in cells exposed to neutralized-pH PDFs in correspondence with their intracellular pH; moreover, in acidic PDFs, Golgi staining was diffuse and scattered in the entire cytoplasm and showed partial aggregation. The Golgi fragmentation markedly observed with the neutralized PDFs could be reversed by replacing either the media with a neutral-pH medium or a mixture of PDF and PD effluent (PDF) in a gradient manner mimicking clinical conditions. Furthermore, although weaker than pH effect, notable effects of other PDF-related factors were also observed after 30-min exposure to pH-adjusted PDFs. Lastly, the results of studies conducted using MAPK/SAPK inhibitors indicated that the mechanism underlying the Golgi fragmentation described here differs from that associated with the fragmentation that occurs at the G2/M checkpoint in the cell cycle. We conclude that Golgi fragmentation is suitable for rapid biocompatibility assessment of PDF not only because of its strong pH dependence but also because the fragmentation is recognizably affected by PDF constituents.


Subject(s)
Dialysis Solutions/adverse effects , Golgi Apparatus/pathology , Peritoneal Dialysis/adverse effects , Cell Line , Cell Survival , Dialysis Solutions/chemistry , G2 Phase Cell Cycle Checkpoints , Golgi Apparatus/ultrastructure , Humans , Hydrogen-Ion Concentration , M Phase Cell Cycle Checkpoints , Osmolar Concentration
9.
Mod Rheumatol ; 28(2): 215-220, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28722504

ABSTRACT

Tenascin-C (TN-C) is a glycoprotein component of the extracellular matrix (ECM). TN-C consists of four distinct domains, including the tenascin assembly domain, epidermal growth factor-like repeats, fibronectin type III-like repeats, and the fibrinogen-like globe (FBG) domain. This review summarizes the role of TN-C in articular cartilage. Expression of TN-C is associated with the development of articular cartilage but markedly decreases during maturation of chondrocytes and disappears almost completely in adult articular cartilage. Increased expression of TN-C has been found at diseased cartilage and synovial sites in osteoarthritis (OA) and rheumatoid arthritis (RA). TN-C is increased in the synovial fluid in patients with OA and RA. In addition, serum TN-C is elevated in RA patients. TN-C could be a useful biochemical marker for joint disease. The addition of TN-C results in different effects among TN-C domains. TN-C fragments might be endogenous inducers of cartilage matrix degradation; however, full-length TN-C could promote cartilage repair and prevent cartilage degeneration. The deficiency of TN-C enhanced cartilage degeneration in the spontaneous OA in aged joints and surgical OA model. The clinical significance of TN-C effects on cartilage is not straightforward.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cartilage, Articular/metabolism , Osteoarthritis/metabolism , Tenascin/metabolism , Animals , Arthritis, Rheumatoid/etiology , Cartilage, Articular/growth & development , Cartilage, Articular/pathology , Humans , Osteoarthritis/etiology , Tenascin/genetics
10.
Stroke ; 48(4): 1108-1111, 2017 04.
Article in English | MEDLINE | ID: mdl-28242775

ABSTRACT

BACKGROUND AND PURPOSE: A matricellular protein tenascin-C is implicated in early brain injury after experimental subarachnoid hemorrhage (SAH). This study first evaluated the role of another matricellular protein periostin and the relationships with tenascin-C in post-SAH early brain injury. METHODS: Wild-type (n=226) and tenascin-C knockout (n=9) C57BL/6 male adult mice underwent sham or filament perforation SAH modeling. Vehicle, anti-periostin antibody, or recombinant periostin was randomly administrated by an intracerebroventricular injection at 30 minutes post-modeling. Neuroscores, SAH grading, brain water content, immunostaining, and Western blotting were blindly evaluated at 24 to 48 hours post-SAH. RESULTS: Periostin was induced in brain capillary endothelial cells and neurons at 24 hours post-SAH. Anti-periostin antibody improved post-SAH neurobehavior, brain edema, and blood-brain barrier disruption associated with downregulation of tenascin-C, inactivation of p38, extracellular signal-related kinase 1/2 and matrix metalloproteinase-9, and subsequent preservation of zona occludens-1. Recombinant periostin aggravated post-SAH brain edema and tenascin-C induction. Tenascin-C knockout prevented post-SAH neurobehavioral impairments and periostin induction. CONCLUSIONS: Periostin may cause post-SAH early brain injury through activating downstream signaling pathways and interacting with tenascin-C, providing a novel approach for the treatment of early brain injury.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Injuries/metabolism , Cell Adhesion Molecules/metabolism , Subarachnoid Hemorrhage/metabolism , Tenascin/metabolism , Animals , Antibodies , Cell Adhesion Molecules/immunology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins , Subarachnoid Hemorrhage/complications
11.
Circ J ; 80(11): 2376-2381, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27746411

ABSTRACT

BACKGROUND: Tenascin-C (TN-C) is an extracellular matrix glycoprotein that is heavily upregulated at sites of inflammation. We conducted a retrospective study to assess the utility of TN-C as a novel biomarker to predict the risk of developing coronary artery lesions (CAL) and resistance to intravenous immunoglobulin (IVIG) in patients with Kawasaki disease (KD).Methods and Results:We collected blood samples of 111 KD patients (IVIG-responder: 89, IVIG-resistant: 22; CAL: 8) and 23 healthy controls, and measured the serum levels of TN-C. TN-C levels on admission were significantly higher in patients than in healthy controls and in patients during convalescence after IVIG administration (69.6 vs. 20.4 vs. 39.7 ng/ml, respectively; P<0.001), and correlated positively with C-reactive protein (P<0.001), neutrophil (percentage; P=0.005), and ALT (P<0.001), and negatively with platelet count (P=0.023) and sodium level (P=0.025). On admission, TN-C levels in patients who later developed CAL were significantly higher than in those without CAL (P=0.010), and significantly higher in IVIG-resistant subjects than in IVIG-responders (P=0.003). The accuracy of TN-C testing for the prediction of IVIG resistance was comparable to that of the Kobayashi score. CONCLUSIONS: Serum TN-C could be a biomarker for predicting the risk of developing CAL and IVIG resistance during the acute phase of KD. (Circ J 2016; 80: 2376-2381).


Subject(s)
Coronary Vessels/metabolism , Drug Resistance , Immunoglobulins, Intravenous/administration & dosage , Mucocutaneous Lymph Node Syndrome/blood , Tenascin/blood , Biomarkers/blood , Child , Child, Preschool , Female , Humans , Infant , Male , Mucocutaneous Lymph Node Syndrome/drug therapy , Retrospective Studies , Risk Factors
12.
Acta Neurochir Suppl ; 121: 151-6, 2016.
Article in English | MEDLINE | ID: mdl-26463940

ABSTRACT

Accumulated evidence suggests that blood-brain barrier disruption or brain edema is an important pathologic manifestation for poor outcome after aneurysmal subarachnoid hemorrhage. Many molecules may be involved, acting simultaneously or at different stages during blood-brain barrier disruption via multiple independent or interconnected signaling pathways. Matricellular protein is a class of nonstructural, secreted, and multifunctional extracellular matrix proteins, which potentially mediates brain edema formation. This study reviews the role of osteopontin and tenascin-C, representatives of matricellular proteins, in the context of brain edema formation after subarachnoid hemorrhage in both clinical and experimental settings.


Subject(s)
Brain Edema/metabolism , Brain/metabolism , Subarachnoid Hemorrhage/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain Edema/etiology , Cell Adhesion Molecules/metabolism , Galectins/metabolism , Humans , Osteonectin/metabolism , Osteopontin/metabolism , Subarachnoid Hemorrhage/complications , Tenascin/metabolism , Thrombospondin 1/metabolism , Thrombospondins/metabolism
13.
J Stroke Cerebrovasc Dis ; 25(4): 771-80, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26856458

ABSTRACT

BACKGROUND: This study aimed to deliver gellan sulfate core platinum coil with tenascin-C (GSCC-TNC) into rabbit side-wall aneurysms endovascularly and to evaluate the organization effects in a simulated clinical setting. METHODS: Elastase-induced rabbit side-wall aneurysms were randomly coiled via a transfemoral route like clinical settings with platinum coils (PCs), gellan sulfate core platinum coils (GSCCs), or GSCC-TNCs (n = 5, respectively). Aneurysm-occlusion status was evaluated angiographically and histologically at 2 weeks post coiling. As each rabbit coiled aneurysm provided only 2-3 tissue slices due to technical limitations and prevented immunohistochemical evaluations, a PC, GSCC, or GSCC-TNC was randomly implanted in a rat blind-ended model (n = 3, respectively) and the organization effects were immunohistochemically evaluated for expressions of tenascin-C (TNC), transforming growth factor-beta (TGF-ß), and matrix metalloproteinase-9 (MMP-9) 2 weeks later. RESULTS: Coil handling was similar among the 3 kinds of coils. GSCCs showed a significantly higher ratio of organized area to the aneurysmal cavity than PCs, but GSCC-TNCs had the greatest organization-promoting effects on aneurysms (the ratio of organized area/aneurysmal luminal area: PC, 17.9 ± 7.1%; GSCC, 54.2 ± 18.3%; GSCC-TNC, 82.5 ± 5.8%). GSCC-TNCs had intense immunoreactivities for TNC, TGF-ß, and MMP-9 in the organized thrombosis and tunica media. GSCCs also showed intense immunoreactivities for TNC, TGF-ß, and MMP-9, although the extent was less than GSCC-TNCs. The immunoreactivities were hardly found in unorganized thrombus and the tunica media of aneurysm wall in the PC group. CONCLUSIONS: This study first showed that GSCC-TNCs promote intra-aneurysmal clot organization in simulated clinical settings using rabbits possibly through the TGF-ß and MMP-9 upregulation.


Subject(s)
Embolization, Therapeutic/methods , Intracranial Aneurysm/pathology , Intracranial Aneurysm/therapy , Platinum , Polysaccharides/therapeutic use , Sulfuric Acid Esters/therapeutic use , Tenascin/metabolism , Angiography , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Glioma/pathology , Male , Matrix Metalloproteinase 9/metabolism , Rabbits , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/metabolism
14.
Acta Neurochir Suppl ; 120: 69-73, 2015.
Article in English | MEDLINE | ID: mdl-25366602

ABSTRACT

BACKGROUND AND PURPOSE: We previously reported that tenascin-C (TNC), a matricellular protein, was involved in the pathogenesis of cerebral vasospasm after subarachnoid hemorrhage (SAH), but the role of TNC in early brain injury (EBI) is unknown. This study assessed whether inhibition of TNC upregulation in brain by imatinib mesylate (imatinib), an inhibitor of the tyrosine kinases of platelet-derived growth factor receptors, prevents EBI after experimental SAH. METHODS: Rats were assigned to sham, SAH plus vehicle, and SAH plus imatinib groups (n = 4 per group). Imatinib (50 mg/kg body weight) was administered intraperitoneally to rats undergoing SAH by endovascular perforation, and EBI was evaluated using terminal deoxynucleotidyl transferase-mediated uridine 5-triphosphate-biotin nick end-labeling staining at 24 h after SAH. Imatinib-treated SAH rats were also treated by a cisternal injection of recombinant TNC. RESULTS: SAH upregulated TNC and caused EBI. Imatinib treatment suppressed both TNC upregulation and EBI at 24 h. Recombinant TNC reinduced EBI in imatinib-treated SAH rats. CONCLUSIONS: TNC may be involved in the pathogenesis of EBI after SAH.


Subject(s)
Benzamides/pharmacology , Brain Injuries/drug therapy , Piperazines/pharmacology , Pyrimidines/pharmacology , Subarachnoid Hemorrhage/drug therapy , Tenascin/antagonists & inhibitors , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Disease Models, Animal , Imatinib Mesylate , In Situ Nick-End Labeling , Male , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology , Tenascin/metabolism , Up-Regulation/drug effects
15.
Acta Neurochir Suppl ; 120: 117-21, 2015.
Article in English | MEDLINE | ID: mdl-25366610

ABSTRACT

INTRODUCTION: Tenascin-C (TNC), a matricellular protein, exerts diverse functions, including tissue remodeling and apoptosis, and is induced in cerebrospinal fluid (CSF) after aneurysmal subarachnoid hemorrhage (SAH). The purpose of this study was to examine the relationships among CSF TNC levels, initial brain injury, delayed cerebral ischemia (DCI), and vasospasm after SAH. METHODS: CSF TNC levels were measured in 30 patients with aneurysmal SAH of Fisher computed tomography (CT) group III who were treated microsurgically or endovascularly with CSF drainage within 24 h of SAH. Admission World Federation of Neurosurgical Societies (WFNS) grade was supposed to indicate the severity of initial brain injury. Cerebral vasospasm was defined as narrowed (≥ 25 %) cerebral arteries demonstrated by angiography. DCI was defined as any neurological deterioration presumed related to ischemia that persisted for ≥ 1 h. RESULTS: Higher CSF TNC levels were correlated with worse admission WFNS grades. Vasospasm was aggravated with higher TNC levels. DCI occurred regardless of the degree of vasospasm but was associated with TNC induction. Multivariate analyses showed that higher TNC levels and vasospasm were independent predictors of DCI occurrence. CONCLUSIONS: SAH (initial brain injury) that is more severe induces more TNC, which may cause the subsequent development of both vasospasm and vasospasm-unrelated secondary brain injury, leading to DCI.


Subject(s)
Brain Injuries/cerebrospinal fluid , Brain Ischemia/cerebrospinal fluid , Subarachnoid Hemorrhage/cerebrospinal fluid , Tenascin/cerebrospinal fluid , Vasospasm, Intracranial/cerebrospinal fluid , Adult , Aged , Aged, 80 and over , Brain Injuries/etiology , Brain Ischemia/etiology , Extracellular Matrix/metabolism , Female , Humans , Male , Middle Aged , Severity of Illness Index , Subarachnoid Hemorrhage/complications , Vasospasm, Intracranial/etiology
16.
Acta Neurochir Suppl ; 120: 99-103, 2015.
Article in English | MEDLINE | ID: mdl-25366607

ABSTRACT

BACKGROUND AND PURPOSE: The authors have reported that tenascin-C (TNC), a matricellular protein, is induced after subarachnoid hemorrhage (SAH), associated with cerebral vasospasm. In this study, we examined whether TNC alone causes cerebral vasospasm-like constriction of the intracranial internal carotid arteries (ICAs) in rats, focusing on the p38 mitogen-activated protein kinase (MAPK)-mediated mechanisms. METHODS: First, we injected 10 µg of TNC into the cisterna magna of healthy rats and studied morphologically whether TNC caused constriction of the left ICA at 24-72 h after administration. Second, we examined the effect of SB203580 (a p38 MAPK inhibitor) on the vessel diameter of the left ICA in healthy rats at 24 h. Third, we evaluated the effect of SB203580 on TNC-induced constriction of the left ICA in healthy rats at 24 h. RESULTS: TNC significantly induced cerebral vasospasm-like angiographic constriction of the left ICAs, which continued at least for 72 h. SB203580 itself had no effect on the diameter of normal ICAs, but abolished the TNC-induced vasoconstrictive effect on the left ICA. CONCLUSION: These findings show that TNC causes left ICA constriction via activation of p38 MAPK, resembling post-SAH vasospasm, and suggest the possible involvement of TNC in the pathogenesis of cerebral vasospasm.


Subject(s)
Carotid Artery, Internal/drug effects , Subarachnoid Hemorrhage/drug therapy , Tenascin/pharmacology , Vasoconstriction/drug effects , Vasospasm, Intracranial/drug therapy , Animals , Carotid Artery, Internal/physiology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Male , Pyridines/pharmacology , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/physiopathology , Tenascin/physiology , Vasoconstriction/physiology , Vasospasm, Intracranial/physiopathology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Neurobiol Dis ; 55: 104-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23333289

ABSTRACT

Tenascin-C (TNC), a matricellular protein, is induced in association with cerebral vasospasm after subarachnoid hemorrhage. The aim of this study was to assess the vasoconstrictive effects of TNC and its mechanisms of action on cerebral arteries in vivo. Two dosages (1 and 10µg) of TNC were administered intracisternally to healthy rats, and the effects were evaluated by neurobehavioral tests and India-ink angiography at 24, 48, and 72h after the administration. Western blotting and immunohistochemistry were performed to explore the underlying mechanisms on constricted cerebral arteries after 24h. The effects of toll-like receptor 4 (TLR4) antagonists (LPS-RS), c-Jun N-terminal kinase (JNK), and p38 inhibitors (SP600125 and SB203580) on TNC-induced vasoconstriction were evaluated at 24h. Higher dosages of TNC induced more severe cerebral arterial constriction, which continued for more than 72h. TNC administration also upregulated TLR4, and activated JNK and p38 in the smooth muscle cell layer of the constricted cerebral artery. LPS-RS blocked TNC-induced TLR4 upregulation, JNK and p38 activation, and vasoconstrictive effects. SP600125 and SB203580 abolished TNC-induced TLR4 upregulation and vasoconstrictive effects. TNC may cause prolonged cerebral arterial constriction via TLR4 and activation of JNK and p38, which may upregulate TLR4. These findings suggest that TNC causes cerebral vasospasm and provides a novel therapeutic approach against it.


Subject(s)
Cerebral Arteries/drug effects , Tenascin/toxicity , Vasospasm, Intracranial/chemically induced , Angiography , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , MAP Kinase Kinase 4/metabolism , Male , Nervous System Diseases/etiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Statistics, Nonparametric , Time Factors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Up-Regulation/drug effects , Vasospasm, Intracranial/complications , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Immunol ; 187(11): 5851-64, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22039306

ABSTRACT

The interaction between matricellular proteins such as tenascin-C (TN-C) and osteopontin (OPN) and integrins has been implicated in the pathology of rheumatoid arthritis in which Th17 cells are recognized as primary pathogenic cells. The differentiation of Th17 cells is tightly regulated by cytokines derived from APCs, receiving various signals including TLR stimuli. In this study, we used a collagen-induced arthritis model and found that increased numbers of α(9) integrin-positive conventional dendritic cells and macrophage were detectable in the draining lymph node (dLN) shortly following first immunization, and these cells produced both TN-C and OPN, ligands for α(9) integrin. α(9) integrin-mediated signaling, induced by TN-C and OPN, promoted the production of Th17-related cytokines by conventional dendritic cells and macrophages in synergy with TLR2 and 4 signaling. This led to the Th17 cell differentiation and arthritis development. Moreover, Th17 cells generated under blocking of α(9) integrin-mediated signaling showed low level of CCR6 expression and impaired migration ability toward CCL20. Thus, we have identified α(9) integrin-mediated signaling by TN-C and OPN as a novel intrinsic regulator of pathogenic Th17 cell generation that contributes to the development of rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/immunology , Glycoproteins/immunology , Integrins/immunology , Signal Transduction/immunology , Tenascin/immunology , Th17 Cells/cytology , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Blotting, Western , Cell Differentiation/immunology , Cell Separation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Glycoproteins/biosynthesis , Humans , Integrins/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred DBA , Real-Time Polymerase Chain Reaction , Tenascin/biosynthesis , Th17 Cells/immunology
19.
Nat Med ; 12(1): 128-32, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16327803

ABSTRACT

Many heat-shock proteins (Hsp) are members of evolutionarily conserved families of chaperone proteins that inhibit the aggregation of unfolded polypeptides and refold denatured proteins, thereby remedying phenotypic effects that may result from protein aggregation or protein instability. Here we report that the mitochondrial chaperone Hsp40, also known as Dnaja3 or Tid1, is differentially expressed during cardiac development and pathological hypertrophy. Mice deficient in Dnaja3 developed dilated cardiomyopathy (DCM) and died before 10 weeks of age. Progressive respiratory chain deficiency and decreased copy number of mitochondrial DNA were evident in cardiomyocytes lacking Dnaja3. Profiling of Dnaja3-interacting proteins identified the alpha-subunit of DNA polymerase gamma (Polga) as a client protein. These findings suggest that Dnaja3 is crucial for mitochondrial biogenesis, at least in part, through its chaperone activity on Polga and provide genetic evidence of the necessity for mitochondrial Hsp40 in preventing DCM.


Subject(s)
Cardiomyopathy, Dilated/pathology , DNA-Directed DNA Polymerase/metabolism , HSP40 Heat-Shock Proteins/chemistry , HSP40 Heat-Shock Proteins/physiology , Animals , Aorta/pathology , Cardiomyopathy, Dilated/prevention & control , Cell Line , DNA/metabolism , DNA Polymerase gamma , DNA, Mitochondrial/metabolism , Electron Transport , Electron Transport Complex IV/metabolism , Evolution, Molecular , Green Fluorescent Proteins/metabolism , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Protein Binding , Protein Denaturation , Protein Folding , Protein Renaturation , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection
20.
Heart Vessels ; 28(5): 646-57, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23277455

ABSTRACT

We aimed to investigate whether atrial natriuretic peptide (ANP) attenuates angiotensin II (Ang II)-induced myocardial remodeling and to clarify the possible molecular mechanisms involved. Thirty-five 8-week-old male Wistar-Kyoto rats were divided into control, Ang II, Ang II + ANP, and ANP groups. The Ang II and Ang II + ANP rats received 1 µg/kg/min Ang II for 14 days. The Ang II + ANP and ANP rats also received 0.1 µg/kg/min ANP intravenously. The Ang II and Ang II + ANP rats showed comparable blood pressure. Left ventricular fractional shortening and ejection fraction were lower in the Ang II rats than in controls; these indices were higher (P < 0.001) in the Ang II + ANP rats than in the Ang II rats. In the Ang II rats, the peak velocity of mitral early inflow and its ratio to atrial contraction-related peak flow velocity were lower, and the deceleration time of mitral early inflow was significantly prolonged; these changes were decreased by ANP. Percent fibrosis was higher (P < 0.001) and average myocyte diameters greater (P < 0.01) in the Ang II rats than in controls. ANP decreased both myocardial fibrosis (P < 0.01) and myocyte hypertrophy (P < 0.01). Macrophage infiltration, expression of mRNA levels of collagen types I and III, monocyte chemotactic protein-1, and a profibrotic/proinflammatory molecule, tenascin-C (TN-C) were increased in the Ang II rats; ANP significantly decreased these changes. In vitro, Ang II increased expression of TN-C and endothelin-1 (ET-1) in cardiac fibroblasts, which were reduced by ANP. ET-1 upregulated TN-C expression via endothelin type A receptor. These results suggest that ANP may protect the heart from Ang II-induced remodeling by attenuating inflammation, at least partly through endothelin 1/endothelin receptor A cascade.


Subject(s)
Angiotensin II , Anti-Inflammatory Agents/pharmacology , Atrial Natriuretic Factor/pharmacology , Endothelin-1/metabolism , Heart Diseases/prevention & control , Inflammation/prevention & control , Myocardium/metabolism , Receptor, Endothelin A/metabolism , Signal Transduction/drug effects , Ventricular Remodeling/drug effects , Animals , Anti-Inflammatory Agents/administration & dosage , Atrial Natriuretic Factor/administration & dosage , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Cells, Cultured , Disease Models, Animal , Fibrillar Collagens/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Inflammation Mediators/metabolism , Infusions, Intravenous , Macrophages/drug effects , Macrophages/metabolism , Male , Mitral Valve/drug effects , Mitral Valve/physiopathology , Myocardial Contraction/drug effects , Myocardium/pathology , Rats , Rats, Inbred WKY , Stroke Volume/drug effects , Time Factors , Ventricular Function, Left/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL