Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 128
1.
J Plast Reconstr Aesthet Surg ; 83: 69-76, 2023 08.
Article En | MEDLINE | ID: mdl-37270997

BACKGROUND: Although autologous fat grafting is considered a successful method for the management of contour deformities, the fat graft could potentially induce cancer reappearance by fueling dormant breast cancer cells. Our aim was to characterize the role of adipose-derived stem cells on active and dormant breast cancer cell growth. METHODS: Cobalt chloride was used to induce dormancy in MCF-7 cancer cells. Proliferation of active and dormant cancer cells was determined in the presence of adipose-derived stem cells. A proteome array was used to detect cancer-related protein expression in the cell-conditioned medium. The migration of cancer cells was measured in response to conditioned medium from the adipose-derived stem cells. RESULTS: The adipose-derived stem cells showed variable effects on active MCF-7 cells growth and inhibited MCF-7 proliferation after the withdrawal of cobalt chloride. Of the 84 different proteins measured in the conditioned medium, only tenascin-C was differentially expressed in the co-cultures. MCF-7 cells alone did not express tenascin-C, whereas co-cultures between MCF-7 and adipose-derived stem cells expressed more tenascin-C versus adipose-derived stem cells alone. The conditioned medium from co-cultures significantly increased the migration of the cancer cells. CONCLUSIONS: Adipose-derived stem cells themselves neither increased the growth or migration of cancer cells, suggesting that autologous fat grafting may be oncologically safe if reconstruction is postponed until there is no evidence of active disease. However, interactions between adipose-derived stem cells and MCF-7 cancer cells could potentially lead to the production of factors, which further promote cancer cell migration.


Adipose Tissue , Breast Neoplasms , Humans , Female , Adipose Tissue/transplantation , Culture Media, Conditioned/pharmacology , Culture Media, Conditioned/metabolism , Tenascin/metabolism , Tenascin/pharmacology , Stem Cells , Cell Proliferation
2.
Adv Mater ; 35(33): e2301493, 2023 Aug.
Article En | MEDLINE | ID: mdl-37227134

The lung extracellular matrix (ECM) maintains the structural integrity of the tissue and regulates the phenotype and functions of resident fibroblasts. Lung-metastatic breast cancer alters these cell-ECM interactions, promoting fibroblast activation. There is a need for bio-instructive ECM models that match the ECM composition and biomechanics of the lung to study these cell-matrix interactions in vitro. Here, a synthetic, bioactive hydrogel is synthesized that mimics the native lung modulus and includes a representative distribution of the most abundant ECM peptide motifs responsible for integrin-binding and matrix metalloproteinase (MMP)-mediated degradation in the lung, which enables quiescent culture of human lung fibroblasts (HLFs). Stimulation with transforming growth factor ß1 (TGF-ß1), metastatic breast cancer conditioned media (CM), or tenascin-C-derived integrin-binding peptide activated hydrogel-encapsulated HLFs demonstrates multiple environmental methods to activate HLFs in a lung ECM-mimicking hydrogel. This lung hydrogel platform is a tunable, synthetic approach to studying the independent and combinatorial effects of ECM in regulating fibroblast quiescence and activation.


Breast Neoplasms , Tenascin , Humans , Female , Tenascin/metabolism , Tenascin/pharmacology , Extracellular Matrix/metabolism , Fibroblasts , Hydrogels/chemistry , Peptides/chemistry , Lung , Integrins/metabolism
3.
Nephrology (Carlton) ; 27(5): 458-466, 2022 May.
Article En | MEDLINE | ID: mdl-35213087

AIM: Tenascin-C (TNC), a non-structural extracellular matrix glycoprotein, is transiently expressed during development or after injury, playing an important role in injury and repair process. The potential role of TNC in the pathogenesis of IgA nephropathy (IgAN) remains to be clarified. METHODS: Immunohistochemistry staining for TNC was conducted on paraffin-embedded slices from renal biopsies of 107 IgAN patients, and correlation analysis was made between mesangial TNC expression and clinic-pathological parameters. In situ hybridization for TNC mRNA was further performed to figure out the cells that express TNC within glomeruli. In vitro experiments were also carried out on mouse mesangial cells (SV40 MES13) to elucidate the effect of TNC on mesangial cells. RESULTS: TNC was expressed in the mesangial area of IgAN, as well as in fibrotic regions. Correlation analysis showed that higher mesangial TNC was associated with higher level of proteinuria, lower estimated glomerular filtration rate and more serious pathological lesions (MEST score). In situ hybridization revealed that abundant TNC mRNA expression was observed in the affected glomeruli of IgAN, but not in minimal change disease. Moreover, TNC mRNA co-localized with PDGFRß mRNA, but not with PODXL mRNA, suggesting that TNC mRNA was expressed in the mesangial cells within glomeruli in IgAN. In vitro experiments showed that exogenous TNC promoted matrix protein production and mesangial cell proliferation, which was attenuated by an epidermal growth factor receptor inhibitor. CONCLUSION: Taken together, these results suggest that mesangial cell-derived TNC contributes to mesangial matrix expansion and mesangial cell proliferation, which is a potential therapeutic target in IgAN.


Glomerulonephritis, IGA , Mesangial Cells , Animals , Cell Proliferation , Extracellular Matrix/metabolism , Glomerulonephritis, IGA/pathology , Humans , Mesangial Cells/pathology , Mice , Tenascin/genetics , Tenascin/pharmacology
4.
Transl Stroke Res ; 13(2): 326-337, 2022 04.
Article En | MEDLINE | ID: mdl-34342874

Activation of α-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor (AMPAR) is thought to cause acute brain injury, but the role remains poorly understood in subarachnoid hemorrhage (SAH). This study was conducted to evaluate if AMPAR activation induces acute blood-brain barrier (BBB) disruption after SAH. C57BL/6 male adult mice (n = 117) underwent sham or filament perforation SAH modeling, followed by a random intraperitoneal injection of vehicle or two dosages (1 mg/kg or 3 mg/kg) of a selective noncompetitive AMPAR antagonist perampanel (PER) at 30 min post-modeling. The effects were evaluated by mortality, neurological scores, and brain water content at 24-48 h and video electroencephalogram monitoring, immunostaining, and Western blotting at 24 h post-SAH. PER significantly suppressed post-SAH neurological impairments, brain edema, and BBB disruption. SAH developed epileptiform spikes without obvious convulsion, which were also inhibited by PER. Western blotting showed that the expression of AMPAR subunits GluA1 and GluA2 was unchanged after SAH, but they were significantly activated after SAH. PER prevented post-SAH activation of GluA1/2, associated with the suppression of post-SAH induction of tenascin-C, a causative mediator of post-SAH BBB disruption. Meanwhile, an intracerebroventricular injection of a subtype-selective GluA1/2 agonist augmented the activation of GluA1/2 and the induction of tenascin-C in brain capillary endothelial cells and aggravated post-SAH BBB disruption without increases in epileptiform spikes. Neurological impairments and brain edema were not correlated with the occurrence of epileptiform spikes. This study first showed that AMPAR plays an important role in the development of post-SAH BBB disruption and can be a novel therapeutic target against it.


Brain Edema , Subarachnoid Hemorrhage , Animals , Blood-Brain Barrier/metabolism , Brain Edema/drug therapy , Brain Edema/etiology , Brain Edema/prevention & control , Endothelial Cells/metabolism , Female , Isoxazoles/metabolism , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Male , Mice , Mice, Inbred C57BL , Propionates/metabolism , Propionates/pharmacology , Propionates/therapeutic use , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Tenascin/metabolism , Tenascin/pharmacology , Tenascin/therapeutic use , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/therapeutic use
5.
Am J Pathol ; 190(10): 2123-2135, 2020 10.
Article En | MEDLINE | ID: mdl-32650003

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.


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
6.
Cancer Immunol Res ; 8(9): 1122-1138, 2020 09.
Article En | MEDLINE | ID: mdl-32665262

Inherent immune suppression represents a major challenge in the treatment of human cancer. The extracellular matrix molecule tenascin-C promotes cancer by multiple mechanisms, yet the roles of tenascin-C in tumor immunity are incompletely understood. Using a 4NQO-induced oral squamous cell carcinoma (OSCC) model with abundant and absent tenascin-C, we demonstrated that tenascin-C enforced an immune-suppressive lymphoid stroma via CCL21/CCR7 signaling, leading to increased metastatic tumors. Through TLR4, tenascin-C increased expression of CCR7 in CD11c+ myeloid cells. By inducing CCL21 in lymphatic endothelial cells via integrin α9ß1 and binding to CCL21, tenascin-C immobilized CD11c+ cells in the stroma. Inversion of the lymph node-to-tumor CCL21 gradient, recruitment of T regulatory cells, high expression of anti-inflammatory cytokines, and matrisomal components were hallmarks of the tenascin-C-instructed lymphoid stroma. Ablation of tenascin-C or CCR7 blockade inhibited the lymphoid immune-suppressive stromal properties, reducing tumor growth, progression, and metastasis. Thus, targeting CCR7 could be relevant in human head and neck tumors, as high tenascin-C expression and an immune-suppressive stroma correlate to poor patient survival.


Mouth Neoplasms/immunology , Squamous Cell Carcinoma of Head and Neck/immunology , Tenascin/immunology , Animals , Chemokine CCL21/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mouth Neoplasms/pathology , Receptors, CCR7/immunology , Recombinant Proteins/pharmacology , T-Lymphocytes, Regulatory/immunology , Tenascin/pharmacology , Tumor Microenvironment/immunology
7.
Cell Death Dis ; 11(4): 249, 2020 04 20.
Article En | MEDLINE | ID: mdl-32312959

A central and unique aspect of high-grade serous ovarian carcinoma (HGSC) is the extensive transcoelomic spreading of tumor cell via the peritoneal fluid or malignant ascites. We and others identified tumor-associated macrophages (TAM) in the ascites as promoters of metastasis-associated processes like extracellular matrix (ECM) remodeling, tumor cell migration, adhesion, and invasion. The precise mechanisms and mediators involved in these functions of TAM are, however, largely unknown. We observed that HGSC migration is promoted by soluble mediators from ascites-derived TAM, which can be emulated by conditioned medium from monocyte-derived macrophages (MDM) differentiated in ascites to TAM-like asc-MDM. A similar effect was observed with IL-10-induced alternatively activated m2c-MDM but not with LPS/IFNγ-induced inflammatory m1-MDM. These observations provided the basis for deconvolution of the complex TAM secretome by performing comparative secretome analysis of matched triplets of different MDM phenotypes with different pro-migratory properties (asc-MDM, m2c-MDM, m1-MDM). Mass spectrometric analysis identified an overlapping set of nine proteins secreted by both asc-MDM and m2c-MDM, but not by m1-MDM. Of these, three proteins, i.e., transforming growth factor beta-induced (TGFBI) protein, tenascin C (TNC), and fibronectin (FN1), have been associated with migration-related functions. Intriguingly, increased ascites concentrations of TGFBI, TNC, and fibronectin were associated with short progression-free survival. Furthermore, transcriptome and secretome analyses point to TAM as major producers of these proteins, further supporting an essential role for TAM in promoting HGSC progression. Consistent with this hypothesis, we were able to demonstrate that the migration-inducing potential of asc-MDM and m2c-MDM secretomes is inhibited, at least partially, by neutralizing antibodies against TGFBI and TNC or siRNA-mediated silencing of TGFBI expression. In conclusion, the present study provides the first experimental evidence that TAM-derived TGFBI and TNC in ascites promote HGSC progression.


Cell Movement/drug effects , Extracellular Matrix Proteins/antagonists & inhibitors , Macrophages/drug effects , Tenascin/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Tumor-Associated Macrophages/drug effects , Carcinoma, Ovarian Epithelial/pathology , Cell Differentiation/drug effects , Cell Movement/physiology , Culture Media, Conditioned/pharmacology , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Female , Humans , Macrophages/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Tenascin/metabolism , Transforming Growth Factor beta/metabolism , Tumor-Associated Macrophages/metabolism
8.
Folia Histochem Cytobiol ; 58(1): 1-8, 2020.
Article En | MEDLINE | ID: mdl-32003441

INTRODUCTION: Osteoarthritis (OA) is the most common degenerative disease in middle-aged and elderly individuals that causes joint deformity and limb disability. Accumulating evidence has suggested that the pathogenesis of OA has been related to various mechanisms such as apoptosis, inflammation, oxidative stress and metabolic disorders. The aim of this study is to clarify the role of Foxo3a in the progress of OA in an in vitro model. MATERIALS AND METHODS: The chondrocytes were derived from rabbit, and treated with IL-1ß, which was used as an in vitro OA model. The over-expression and down-regulation of Foxo3a were achieved by transfection with overexpression vector or shRNA, respectively. The mRNA level of iNOS in chondrocytes was quantified by qPCR. Tenascin-c (Tnc) production was measured by ELISA and apoptosis-associated proteins were analyzed by Western blotting. The MTT assay was used to assess the viability of chondrocytes. RESULTS: Foxo3a and iNOS expression were upregulated in IL-1ß-treated chondrocytes. Foxo3a silencing decreased iNOS expression, and inhibited apoptosis of IL-1ß-treated chondrocytes. The production of Tnc was significantly increased in IL-1b-treated chondrocytes and was positively regulated by Foxo3. Importantly, extracellular addition of Tnc abrogated the protective effects of Foxo3a knockdown on IL-1ß - treated chondrocytes. CONCLUSION: The present study indicated that down-regulation of Foxo3a protected IL-1ß-treated chondrocytes by decreasing iNOS expression and suppressing chondrocytes' apoptosis via modulating tenascin-c, which could be regarded as a potent therapeutic target for the treatment of OA.


Apoptosis/physiology , Chondrocytes/drug effects , Forkhead Box Protein O3/metabolism , Inflammation/metabolism , Tenascin/metabolism , Animals , Chondrocytes/metabolism , Down-Regulation , Forkhead Box Protein O3/genetics , Gene Knockdown Techniques , Interleukin-1beta , Nitric Oxide Synthase Type II/metabolism , Osteoarthritis/chemically induced , Osteoarthritis/metabolism , Rabbits , Tenascin/pharmacology , Up-Regulation
9.
Anticancer Res ; 39(7): 3487-3492, 2019 Jul.
Article En | MEDLINE | ID: mdl-31262872

BACKGROUND/AIM: Despite intensive chemotherapy, the survival rates for high-risk neuroblastoma, most of which have MYCN amplification, remain low. Overexpression of N-myc oncoprotein promotes expression of cancer-associated properties. We recently found that combination of all-trans retinoic acid (ATRA) with the ß1-integrin-activating peptide TNIIIA2 attenuated cancer-associated properties of neuroblastoma cells through N-Myc degradation. However, ATRA has serious side-effects and there are concerns about late adverse effects. The aim of this study was to examine the effects of the combination of acyclic retinoid (ACR) with TNIIIA2 on neuroblastoma. MATERIALS AND METHODS: The effects of ACR and TNIIIA2 were examined by neuroblastoma cell proliferation and survival assays as well as by using a neuroblastoma xenograft model. The levels of N-Myc and cancer-associated malignant properties were assayed by western blot and colony formation assay, respectively. RESULTS: Combining ACR, which is clinically safe, with TNIIIA2 induced proteasomal degradation of N-Myc and reduction of neuroblastoma cell malignant properties. An in vivo experiment showed therapeutic potential. CONCLUSION: ACR-TNIIIA2 combination treatment may be efficacious and clinical safe chemotherapy for high-risk neuroblastoma.


Antineoplastic Agents/therapeutic use , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/drug therapy , Peptides/therapeutic use , Tenascin/therapeutic use , Tretinoin/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Mice, Inbred BALB C , Mice, Nude , Neuroblastoma/metabolism , Neuroblastoma/pathology , Peptides/pharmacology , Phenotype , Tenascin/pharmacology , Tretinoin/pharmacology , Tretinoin/therapeutic use , Tumor Burden/drug effects
10.
Cytotherapy ; 20(8): 1061-1076, 2018 08.
Article En | MEDLINE | ID: mdl-30087008

BACKGROUND: Impaired cutaneous wound healing is common in humans, and treatments are often ineffective. Based on the significant emotional and economic burden of impaired wound healing, innovative therapies are needed. The potential of mesenchymal stromal cell (MSC)-secreted factors to treat cutaneous wounds is an active area of research that is in need of refinement before effective clinical trials can be initiated. The aims of the present study were to (i) study which MSC-secreted factors stimulate dermal fibroblast (DF) migration in vitro and (ii) evaluate the potential of these factors to promote wound healing in vivo. METHODS: To this end, MSCs were isolated from the peripheral blood of healthy horses, a physiologically relevant large animal model appropriate for translational wound-healing studies. Conditioned medium (CM) from cultured equine MSCs was analyzed using liquid chromatography-mass spectrophotometry (LC-MS/MS) to identify secreted proteins of interest. Double-stranded RNA-mediated interference (RNAi) was used to silence the genes encoding selected proteins, and the effects of CM from these transfected MSCs on migration of cultured equine DF cells in vitro and full-thickness wounds in mice were evaluated. RESULTS: We found that MSC-derived plasminogen activator inhibitor-1 (PAI-1) and tenascin-C significantly increased DF migration in vitro and improved wound healing in vivo by decreasing time to wound closure. DISCUSSION: These results suggest that in a complex wound environment, MSC-secreted factors PAI-1 and tenascin-C contribute to the positive effect of therapeutically applied MSC CM on wound healing.


Dermis/drug effects , Fibroblasts/drug effects , Mesenchymal Stem Cells/metabolism , Plasminogen Activator Inhibitor 1 , Tenascin , Wound Healing/drug effects , Animals , Cell Line , Cell Movement/drug effects , Coculture Techniques , Culture Media, Conditioned/pharmacology , Dermis/cytology , Extracellular Vesicles/metabolism , Extracellular Vesicles/physiology , Female , Fibroblasts/physiology , Horses , Mesenchymal Stem Cells/cytology , Mice , NIH 3T3 Cells , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Tenascin/metabolism , Tenascin/pharmacology , Wound Healing/physiology
11.
Biomater Sci ; 6(7): 1859-1868, 2018 Jun 25.
Article En | MEDLINE | ID: mdl-29799029

The development of new biomaterials mimicking the neuronal extracellular matrix (ECM) requires signals for the induction of neuronal differentiation and regeneration. In addition to the biological and chemical cues, the physical properties of the ECM should also be considered while designing regenerative materials for nervous tissue. In this study, we investigated the influence of the microenvironment on tenascin-C signaling using 2D surfaces and 3D scaffolds generated by a peptide amphiphile nanofiber gel with a tenascin-C derived peptide epitope (VFDNFVLK). While tenascin-C mimetic PA nanofibers significantly increased the length and number of neurites produced by PC12 cells on 2D cell culture, more extensive neurite outgrowth was observed in the 3D gel environment. PC12 cells encapsulated within the 3D tenascin-C mimetic peptide nanofiber gel also exhibited significantly increased expression of neural markers compared to the cells on 2D surfaces. Our results emphasize the synergistic effects of the 3D conformation of peptide nanofibers along with the tenascin-C signaling and growth factors on the neuronal differentiation of PC12 cells, which may further provide more tissue-like morphology for therapeutic applications.


Biomimetic Materials/pharmacology , Nanofibers/chemistry , Neurons/drug effects , Peptides/pharmacology , Signal Transduction , Tissue Scaffolds , Animals , Biomarkers/metabolism , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Cell Culture Techniques , Cell Differentiation/drug effects , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Gels , Gene Expression , Intercellular Signaling Peptides and Proteins/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuronal Outgrowth/drug effects , Neuronal Outgrowth/physiology , Neurons/cytology , Neurons/metabolism , PC12 Cells , Peptides/chemical synthesis , Rats , Tenascin/metabolism , Tenascin/pharmacology
12.
Mol Med Rep ; 17(6): 7603-7610, 2018 Jun.
Article En | MEDLINE | ID: mdl-29620204

There are currently limitations in stem cell therapy due to the low rate of homing and proliferation of cells following transplantation. The present study was designed to investigate the effects of Tenascin­C (TN­C) on bone marrow mesenchymal stem cells (BMSCs) and its underlying mechanisms. BMSCs were obtained from C57BL/6 mice. The survival and proliferation of BMSCs was analyzed by Cell Counting Kit­8 assay, migration was evaluated using the Transwell method, and differentiation was assessed by immunocytochemistry and immunofluorescence. In addition, the levels of proteins were detected by western blotting. High concentrations of TN­C promoted the migration of BMSCs. H2O2 at concentrations of 60­90 µmol/ml induced cell death in BMSCs, and thus, it was used to simulate oxidative stress in the microenvironment of acute myocardial infarction (AMI). High concentrations of TN­C were able to protect BMSCs from cell death, and promoted the migration of BMSCs (P<0.05). However, TAK­242 [the inhibitor of Toll­like receptor 4, (TLR4)] reduced the promoting effect of TN­C (P<0.05). By contrast, TN­C had no effect on the proliferation and differentiation of BMSCs. TN­C reduced the phosphorylation levels of p38 mitogen­activated protein kinase (MAPK), and increased the phosphorylation levels of Ser473 protein kinase B (AKT) and ß­catenin, all of which were inhibited by TAK­242 (P<0.05). In the simulated AMI microenvironment, TN­C promoted the migration of BMSCs via TLR4­mediated signaling pathways, including MAPK, AKT and Wnt.


Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Tenascin/pharmacology , Toll-Like Receptor 4/metabolism , Animals , Biomarkers , Bone Marrow Cells/cytology , Cell Differentiation , Cell Movement , Cell Proliferation/drug effects , Cell Survival/drug effects , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Stem Cells/cytology , Wnt Proteins/metabolism
13.
J Tissue Eng Regen Med ; 12(4): e2123-e2133, 2018 04.
Article En | MEDLINE | ID: mdl-29327429

Biomaterials hold great promise in helping the adult brain regenerate and rebuild after trauma. Peptide amphiphiles (PAs) are highly versatile biomaterials, gelling and forming macromolecular structures when exposed to physiological levels of electrolytes. We are here reporting on the first ever in vivo use of self-assembling PA carrying a Tenascin-C signal (E2 Ten-C PA) for the redirection of endogenous neuroblasts in the rodent brain. The PA forms highly aligned nanofibers, displaying the migratory sequence of Tenascin-C glycoprotein as epitope. In this in vivo work, we have formed in situ a gel of aligned PA nanofibers presenting a migratory Tenascin-C signal sequence in the ventral horn of the rostral migratory stream, creating a track reaching the neocortex. Seven days posttransplant, doublecortin positive cells were observed migrating inside and alongside the injected biomaterial, reaching the cortex. We observed a 24-fold increase in number of redirected neuroblasts for the E2 Ten-C PA-injected animals compared to control. We also found injecting the E2 Ten-C PA to cause minimal neuroinflammatory response. Analysing GFAP+ astrocytes and Iba1+ microglia activation, the PA does not elicit a stronger neuroinflammatory response than would be expected from a small needle stab wound. Redirecting endogenous neuroblasts and increasing the number of cells reaching a site of injury using PAs may open up new avenues for utilizing the pool of neuroblasts and neural stem cells within the adult brain for regenerating damaged brain tissue and replacing neurons lost to injury.


Biocompatible Materials/pharmacology , Brain Injuries/therapy , Cell Movement/drug effects , Nanofibers/therapeutic use , Neocortex/metabolism , Neural Stem Cells/metabolism , Peptides/pharmacology , Tenascin/pharmacology , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Doublecortin Protein , Male , Microglia/metabolism , Microglia/pathology , Neocortex/pathology , Neural Stem Cells/pathology , Rats , Rats, Sprague-Dawley
14.
Cartilage ; 9(1): 80-88, 2018 01.
Article En | MEDLINE | ID: mdl-29219023

Objective The objective of this study was to determine whether intra-articular injections of tenascin-C (TNC) could prevent cartilage damage in murine models of osteoarthritis (OA). Design Fluorescently labeled TNC was injected into knee joints and its distribution was examined at 1 day, 4 days, 1 week, 2 weeks, and 4 weeks postinjection. To investigate the effects of TNC on cartilage degeneration after surgery to knee joints, articular spaces were filled with 100 µg/mL (group I), 10 µg/mL (group II) of TNC solution, or control (group III). TNC solution of 10 µg/mL was additionally injected twice after 3 weeks (group IV) or weekly after 1 week, 2 weeks, and 3 weeks (group V). Joint tissues were histologically assessed using the Mankin score and the modified Chambers system at 2 to 8 weeks after surgery. Results Exogenous TNC was maintained in the cartilage and synovium for 1 week after administration. Histological scores in groups I and II were better than scores in group III at 4 and 6 weeks, but progressive cartilage damage was seen in all groups 8 weeks postoperatively. Sequential TNC injections (groups IV and V) showed significantly better Mankin score than single injection (group II) at 8 weeks. Conclusion TNC administered exogenously remained in the cartilage of knee joints for 1 week, and could decelerate articular cartilage degeneration in murine models of OA. We also showed that sequential administration of TNC was more effective than a single injection. TNC could be an important molecule for prevention of articular cartilage damage.


Cartilage Diseases/pathology , Cartilage, Articular/drug effects , Knee Joint/pathology , Osteoarthritis, Knee/drug therapy , Synovial Membrane/drug effects , Tenascin/pharmacology , Animals , Cartilage Diseases/prevention & control , Cartilage, Articular/pathology , Cartilage, Articular/ultrastructure , Collagen Type II/metabolism , Disease Models, Animal , Injections, Intra-Articular , Knee Joint/ultrastructure , Male , Mice , Mice, Inbred BALB C/metabolism , Osteoarthritis, Knee/prevention & control , Synovial Membrane/pathology , Tenascin/administration & dosage
15.
Cancer Res ; 77(12): 3231-3243, 2017 06 15.
Article En | MEDLINE | ID: mdl-28416488

Oncogenic signaling by NOTCH is elevated in brain tumor-initiating cells (BTIC) in malignant glioma, but the mechanism of its activation is unknown. Here we provide evidence that tenascin-C (TNC), an extracellular matrix protein prominent in malignant glioma, increases NOTCH activity in BTIC to promote their growth. We demonstrate the proximal localization of TNC and BTIC in human glioblastoma specimens and in orthotopic murine xenografts of human BTIC implanted intracranially. In tissue culture, TNC was superior amongst several extracellular matrix proteins in enhancing the sphere-forming capacity of glioma patient-derived BTIC. Exogenously applied or autocrine TNC increased BTIC growth through an α2ß1 integrin-mediated mechanism that elevated NOTCH ligand Jagged1 (JAG1). Microarray analyses and confirmatory PCR and Western analyses in BTIC determined that NOTCH signaling components including JAG1, ADAMTS15, and NICD1/2 were elevated in BITC after TNC exposure. Inhibition of γ-secretase and metalloproteinase proteolysis in the NOTCH pathway, or silencing of α2ß1 integrin or JAG1, reduced the proliferative effect of TNC on BTIC. Collectively, our findings identified TNC as a pivotal initiator of elevated NOTCH signaling in BTIC and define the establishment of a TN-α2ß1-JAG1-NOTCH signaling axis as a candidate therapeutic target in glioma patients. Cancer Res; 77(12); 3231-43. ©2017 AACR.


Brain Neoplasms/pathology , Glioma/pathology , Neoplastic Stem Cells/pathology , Receptors, Notch/metabolism , Tenascin/metabolism , Animals , Blotting, Western , Brain Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Glioma/metabolism , Heterografts , Humans , Immunoprecipitation , Mice , Mice, SCID , Neoplastic Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Tenascin/pharmacology
16.
Int J Mol Sci ; 18(1)2017 Jan 17.
Article En | MEDLINE | ID: mdl-28106752

The extracellular matrix (ECM) molecule tenascin C (TNC) is known to be highly expressed under various pathological conditions such as inflammation and cancer. It has been reported that the expression of TNC is correlated with the malignant potential of cancer. In our laboratory, it was found that the peptide derived from the alternative splicing domain A2 in TNC, termed TNIIIA2, has been shown to influence a variety of cellular processes, such as survival, proliferation, migration, and differentiation. In this study, we investigated the effect of TNC/TNIIIA2 on the invasion and metastasis of colon cancer cells, Colon26-M3.1, or PMF-Ko14, using an in vitro and in vivo experimental system. The degree of cell invasion was increased by the addition of TNC and TNIIIA2 in a dose-dependent manner. The invasion by TNC and TNIIIA2 were suppressed by an MMP inhibitor or TNIIIA2-blocking antibody. In an in vivo experiment, pulmonary metastasis was promoted conspicuously by the addition of TNIIIA2. In this study, we found that colon cancer cell invasion and metastasis was accelerated by TNC/TNIIIA2 via MMP induction. This result suggests the possibility of a new strategy targeting TNC/TNIIIA2 for colon cancer.


Cell Movement/drug effects , Colonic Neoplasms/enzymology , Matrix Metalloproteinases/metabolism , Peptides/pharmacology , Tenascin/pharmacology , Amino Acid Sequence , Animals , Cell Line, Tumor , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Extracellular Matrix Proteins/chemistry , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinases/genetics , Mice, Inbred BALB C , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , Reverse Transcriptase Polymerase Chain Reaction , Tenascin/chemistry
17.
Cell Transplant ; 26(1): 103-113, 2017 01 24.
Article En | MEDLINE | ID: mdl-27452449

Mesenchymal stem cells (MSCs) remain of great interest in regenerative medicine because of their ability to home to sites of injury, differentiate into a variety of relevant lineages, and modulate inflammation and angiogenesis through paracrine activity. Many studies have found that despite the promise of MSC therapy, cell survival upon implant is highly limited and greatly reduces the therapeutic utility of MSCs. The matrikine tenascin C, a protein expressed often at the edges of a healing wound, contains unique EGF-like repeats that are able to bind EGFR at low affinities and induce downstream prosurvival signaling without inducing receptor internalization. In this study, we utilized tenascin C in a collagen/GAG-based polymer (TPolymer) that has been shown to be beneficial for skin wound healing, incorporating human MSCs into the polymer prior to application to mouse punch biopsy wound beds. We found that the TPolymer was able to promote MSC survival for 21 days in vivo, leading to associated improvements in wound healing such as dermal maturation and collagen content. This was most marked in a model of hypertrophic scarring, in which the scar formation was limited. This approach also reduced the inflammatory response in the wound bed, limiting CD3e+ cell invasion by approximately 50% in the early wound-healing process, while increasing the numbers of endothelial cells during the first week of wound healing as well. Ultimately, this matrikine-based approach to improving MSC survival may be of great use across a variety of cell therapies utilizing matrices as delivery vehicles for cells.


Mesenchymal Stem Cell Transplantation/methods , Polymers/chemistry , Tenascin/chemistry , Animals , Cell Survival/drug effects , Cells, Cultured , Collagen/metabolism , Extracellular Matrix/metabolism , Female , Flow Cytometry , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Polyethylene Glycols/chemistry , Skin/metabolism , Skin/pathology , Skin Diseases/metabolism , Skin Diseases/pathology , Tenascin/pharmacology , Wound Healing/drug effects
18.
Nat Commun ; 7: 11703, 2016 06 03.
Article En | MEDLINE | ID: mdl-27256716

The factors responsible for maintaining persistent organ fibrosis in systemic sclerosis (SSc) are not known but emerging evidence implicates toll-like receptors (TLRs) in the pathogenesis of SSc. Here we show the expression, mechanism of action and pathogenic role of endogenous TLR activators in skin from patients with SSc, skin fibroblasts, and in mouse models of organ fibrosis. Levels of tenascin-C are elevated in SSc skin biopsy samples, and serum and SSc fibroblasts, and in fibrotic skin tissues from mice. Exogenous tenascin-C stimulates collagen gene expression and myofibroblast transformation via TLR4 signalling. Mice lacking tenascin-C show attenuation of skin and lung fibrosis, and accelerated fibrosis resolution. These results identify tenascin-C as an endogenous danger signal that is upregulated in SSc and drives TLR4-dependent fibroblast activation, and by its persistence impedes fibrosis resolution. Disrupting this fibrosis amplification loop might be a viable strategy for the treatment of SSc.


Collagen/genetics , Fibroblasts/metabolism , Lung/pathology , Scleroderma, Systemic/genetics , Skin/metabolism , Tenascin/genetics , Adult , Aged , Animals , Case-Control Studies , Cell Differentiation , Cells, Cultured , Collagen/drug effects , Disease Models, Animal , Female , Fibrosis/genetics , Fibrosis/metabolism , Gene Expression Regulation , Humans , Lung/drug effects , Male , Mice , Middle Aged , Myofibroblasts/drug effects , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Signal Transduction , Skin/pathology , Tenascin/metabolism , Tenascin/pharmacology , Toll-Like Receptor 4/metabolism , Up-Regulation
19.
PLoS One ; 11(5): e0155261, 2016.
Article En | MEDLINE | ID: mdl-27182834

Tenascin-C (TNC) is a newly identified innate HIV-1-neutralizing protein present in breast milk, yet its presence and potential HIV-inhibitory function in other mucosal fluids is unknown. In this study, we identified TNC as a component of semen and cervical fluid of HIV-1-infected and uninfected individuals, although it is present at a significantly lower concentration and frequency compared to that of colostrum and mature breast milk, potentially due to genital fluid protease degradation. However, TNC was able to neutralize HIV-1 after exposure to low pH, suggesting that TNC could be active at low pH in the vaginal compartment. As mucosal fluids are complex and contain a number of proteins known to interact with the HIV-1 envelope, we further studied the relationship between the concentration of TNC and neutralizing activity in breast milk. The amount of TNC correlated only weakly with the overall innate HIV-1-neutralizing activity of breast milk of uninfected women and negatively correlated with neutralizing activity in milk of HIV-1 infected women, indicating that the amount of TNC in mucosal fluids is not adequate to impede HIV-1 transmission. Moreover, the presence of polyclonal IgG from milk of HIV-1 infected women, but not other HIV-1 envelope-binding milk proteins or monoclonal antibodies, blocked the neutralizing activity of TNC. Finally, as exogenous administration of TNC would be necessary for it to mediate measurable HIV-1 neutralizing activity in mucosal compartments, we established that recombinantly produced TNC has neutralizing activity against transmitted/founder HIV-1 strains that mimic that of purified TNC. Thus, we conclude that endogenous TNC concentration in mucosal fluids is likely inadequate to block HIV-1 transmission to uninfected individuals.


Extracellular Fluid/immunology , Genitalia , HIV Infections/immunology , HIV-1/immunology , Milk Proteins/immunology , Milk, Human/immunology , Tenascin/immunology , Antibodies, Neutralizing/immunology , Cervix Uteri/immunology , Female , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/metabolism , HIV Infections/virology , HIV-1/drug effects , Humans , Hydrogen-Ion Concentration , Immunity, Innate , Immunoglobulin G/immunology , Male , Milk Proteins/pharmacology , Mucous Membrane/immunology , Mucous Membrane/metabolism , Neutralization Tests , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Binding , Recombinant Proteins , Semen/immunology , Tenascin/pharmacology
20.
Mol Vis ; 22: 436-45, 2016.
Article En | MEDLINE | ID: mdl-27186070

PURPOSE: We previously demonstrated that tenascin-C was highly expressed in the fibrovascular membranes (FVMs) of patients with proliferative diabetic retinopathy (PDR). However, its role in the pathogenesis of FVMs has not been determined. The purpose of this study was to investigate what role tenascin-C plays in the formation and angiogenesis of FVMs. METHODS: The level of tenascin-C was determined by sandwich enzyme-linked immunosorbent assay in the vitreous samples collected from patients with PDR and with a macular hole as control. The locations of tenascin-C, α- smooth muscle actin (SMA), CD34, glial fibrillary acidic protein (GFAP), and integrin αV in the FVMs from PDR patients were determined by immunohistochemistry. We also measured the in vitro expression of the mRNA and protein of tenascin-C in vascular smooth muscle cells (VSMCs) stimulated by interleukin (IL)-13. The effects of tenascin-C on cell proliferation, migration, and tube formation were determined in human retinal endothelial cells (HRECs) in culture. RESULTS: The mean vitreous levels of tenascin-C were significantly higher in patients with PDR than in patients with a macular hole (p<0.001). Double immunofluorescence analyses of FVMs from PDR patients showed that tenascin-C co-stained FVMs with α-SMA, CD34, and integrin αV but not with GFAP. In addition, IL-13 treatment increased both the expression and secretion of tenascin-C by VSMCs in a dose-dependent manner. Tenascin-C exposure promoted proliferation, migration, and tube formation in HRECs. Tenascin-C neutralizing antibody significantly blocked the tube formation by HRECs exposed to VSMC-IL-13-conditioned medium. CONCLUSIONS: Our findings suggest that tenascin-C is secreted from VSMCs and promotes angiogenesis in the FVMs associated with PDR.


Diabetic Retinopathy/metabolism , Epiretinal Membrane/metabolism , Retinal Neovascularization/metabolism , Tenascin/physiology , Vitreous Body/metabolism , Actins/metabolism , Aged , Antigens, CD34/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Diabetic Retinopathy/pathology , Endothelium, Vascular/cytology , Enzyme-Linked Immunosorbent Assay , Epiretinal Membrane/pathology , Female , Fluorescent Antibody Technique, Indirect , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/genetics , Retinal Neovascularization/pathology , Retinal Perforations/metabolism , Retinal Perforations/pathology , Retinal Vessels/cytology , Tenascin/pharmacology
...