Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Neuroimmunol ; 361: 577749, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34688067

ABSTRACT

We examined the mechanism how 2-carba-cyclic phosphatidic acid (2ccPA), a lipid mediator, regulates neuronal apoptosis in traumatic brain injury (TBI). First, we found 2ccPA suppressed neuronal apoptosis after the injury, and increased the immunoreactivity of tenascin-C (TN-C), an extracellular matrix protein by 2ccPA in the vicinity of the wound region. 2ccPA increased the mRNA expression levels of Tnc in primary cultured astrocytes, and the conditioned medium of 2ccPA-treated astrocytes suppressed the apoptosis of cortical neurons. The neuroprotective effect of TN-C was abolished by knockdown of TN-C. These results indicate that 2ccPA contributes to neuroprotection via TN-C from astrocytes in TBI.


Subject(s)
Astrocytes/metabolism , Brain Injuries, Traumatic/metabolism , Neuroprotective Agents/therapeutic use , Phosphatidic Acids/physiology , Tenascin/metabolism , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Brain Injuries, Traumatic/drug therapy , Cells, Cultured , Cerebral Cortex/cytology , Culture Media, Conditioned/pharmacology , Female , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Injections, Intraperitoneal , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred ICR , Neurons/drug effects , Neurons/pathology , Phosphatidic Acids/pharmacology , Phosphatidic Acids/therapeutic use , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tenascin/antagonists & inhibitors , Tenascin/genetics , Wounds, Stab/drug therapy , Wounds, Stab/metabolism
2.
Front Immunol ; 12: 635166, 2021.
Article in English | MEDLINE | ID: mdl-33790905

ABSTRACT

The extracellular matrix (ECM) molecule Tenascin-C (TNC) is well-known to promote tumor progression by multiple mechanisms. However, reliable TNC detection in tissues of tumor banks remains limited. Therefore, we generated dromedary single-domain nanobodies Nb3 and Nb4 highly specific for human TNC (hTNC) and characterized the interaction with TNC by several approaches including ELISA, western blot, isothermal fluorescence titration and negative electron microscopic imaging. Our results revealed binding of both nanobodies to distinct sequences within fibronectin type III repeats of hTNC. By immunofluroescence and immunohistochemical imaging we observed that both nanobodies detected TNC expression in PFA and paraffin embedded human tissue from ulcerative colitis, solid tumors and liver metastasis. As TNC impairs cell adhesion to fibronectin we determined whether the nanobodies abolished this TNC function. Indeed, Nb3 and Nb4 restored adhesion of tumor and mesangial cells on a fibronectin/TNC substratum. We recently showed that TNC orchestrates the immune-suppressive tumor microenvironment involving chemoretention, causing tethering of CD11c+ myeloid/dendritic cells in the stroma. Here, we document that immobilization of DC2.4 dendritic cells by a CCL21 adsorbed TNC substratum was blocked by both nanobodies. Altogether, our novel TNC specific nanobodies could offer valuable tools for detection of TNC in the clinical practice and may be useful to inhibit the immune-suppressive and other functions of TNC in cancer and other diseases.


Subject(s)
Antibodies, Neutralizing/immunology , Camelus/immunology , Single-Domain Antibodies/immunology , Tenascin/antagonists & inhibitors , Animals , Antibodies, Neutralizing/pharmacology , Antibody Specificity , Binding Sites, Antibody , Cell Adhesion/drug effects , Cell Line, Tumor , Colitis, Ulcerative/immunology , Colon/immunology , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Immunohistochemistry , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Protein Binding , Single-Domain Antibodies/pharmacology , Tenascin/administration & dosage , Tenascin/immunology
3.
MAbs ; 12(1): 1836713, 2020.
Article in English | MEDLINE | ID: mdl-33136526

ABSTRACT

Antibody-based delivery of bioactive molecules represents a promising strategy for the improvement of cancer immunotherapy. Here, we describe the generation and characterization of R6N, a novel fully human antibody specific to the alternatively spliced domain D of Tenascin C, which is highly expressed in the stroma of primary tumors and metastasis. The R6N antibody recognized its cognate tumor-associated antigen with identical specificity in mouse and human specimens. Moreover, the antibody was able to selectively localize to solid tumors in vivo as evidenced by immunofluorescence-based biodistribution analysis. Encouraged by these results, we developed a novel fusion protein (termed mIL12-R6N) consisting of the murine interleukin 12 fused to the R6N antibody in homodimeric tandem single-chain variable fragment arrangement. mIL12-R6N exhibited potent antitumor activity in immunodeficient mice bearing SKRC52 renal cell carcinoma, as well as in immunocompetent mice bearing SMA-497 glioma. The experiments presented in this work provide a rationale for possible future applications for the R6N antibody for the treatment of cancer patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Interleukin-12/administration & dosage , Neoplasms, Experimental , Tenascin/antagonists & inhibitors , Alternative Splicing , Animals , Humans , Mice , Molecular Targeted Therapy/methods , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies
4.
PLoS One ; 15(8): e0237889, 2020.
Article in English | MEDLINE | ID: mdl-32817625

ABSTRACT

Tenascin-C (TNC) is an extracellular matrix (ECM) glycoprotein that plays an important role in cell proliferation, migration, and tumour invasion in various cancers. TNC is one of the main protein overexpressed in breast cancer, indicating a role for this ECM molecule in cancer pathology. In this study we have evaluated the TNC loss-off-function in breast cancer cells. In our approach, we used dsRNA sharing sequence homology with TNC mRNA, called ATN-RNA. We present the data showing the effects of ATN-RNA in MDA-MB-231 cells both in monolayer and three-dimensional culture. Cells treated with ATN-RNA were analyzed for phenotypic alterations in proliferation, migration, adhesion, cell cycle, multi-caspase activation and the involvement in epithelial to mesenchymal transition (EMT) processes. As complementary analysis the oncogenomic portals were used to assess the clinical implication of TNC expression on breast cancer patient's survival, showing the TNC overexpression associated with a poor survival outcome. Our approach applied first in brain tumors and then in breast cancer cell lines reveals that ATN-RNA significantly diminishes the cell proliferation, migration and additionally, reverses the mesenchymal cells phenotype to the epithelial one. Thus, TNC could be considered as the universal target in different types of tumors, where TNC overexpression is associated with poor prognosis.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , RNA, Double-Stranded/genetics , Tenascin/genetics , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Disease-Free Survival , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Prognosis , RNA, Double-Stranded/pharmacology , Tenascin/antagonists & inhibitors
5.
Bioorg Med Chem ; 27(15): 3248-3253, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31208798

ABSTRACT

Tenatumomab is an anti-tenascin murine monoclonal antibody previously used in clinical trials for delivering radionuclides to tumors by both pre-targeting (biotinylated Tenatumomab within PAGRIT) and direct 131Iodine labeling approaches. Here we present the synthesis and in vitro characterization of three Tenatumomab conjugates to bifunctional chelating agents (NHS-DOTA, NCS-DOTA and NCS-DTPA). Results indicate ST8198AA1 (Tenatumomab-DOTAMA, derived by conjugation of NHS-DOTA), as the most promising candidate in terms of conjugation rate and yield, stability, antigen immunoreactivity and affinity. Labeling efficiency of the different chelators was investigated with a panel of cold metals indicating DOTAMA as the best chelator. Labeling of Tenatumomab-DOTAMA was then optimized with several metals and stability performed confirms suitability of this conjugate for further development. ST8198AA1 represents an improvement of the previous antibody forms because the labeling with radionuclides like 177Lu or 64Cu would allow theranostic applications in patients bearing tenascin expressing tumors.


Subject(s)
Heterocyclic Compounds, 1-Ring/pharmacology , Neoplasms/drug therapy , Tenascin/antagonists & inhibitors , Theranostic Nanomedicine , Dose-Response Relationship, Drug , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Molecular Structure , Neoplasms/genetics , Structure-Activity Relationship , Tenascin/genetics
6.
Carcinogenesis ; 40(7): 871-882, 2019 07 20.
Article in English | MEDLINE | ID: mdl-30668646

ABSTRACT

Altered flux through major metabolic pathways is a hallmark of cancer cells and provides opportunities for therapy. Stem cell-like cancer (SCLC) cells can cause metastasis and therapy resistance. They possess metabolic plasticity, theoretically enabling resistance to therapies targeting a specific metabolic state. The C-terminal binding protein (CtBP) transcriptional regulators are potential therapeutic targets in highly glycolytic cancer cells, as they are activated by the glycolytic coenzyme nicotinamide adenine dinucleotide (NADH). However, SCLC cells commonly exist in an oxidative state with low rates of glycolysis. Metformin inhibits complex I of the mitochondrial electron transport chain; it can kill oxidative SCLC cells and has anti-cancer activity in patients. SCLC cells can acquire resistance to metformin through increased glycolysis. Given the potential for long-term metformin therapy, we have studied acquired metformin resistance in cells from the claudin-low subtype of breast cancer. Cells cultured for 8 weeks in sub-IC50 metformin concentration proliferated comparably to untreated cells and exhibited higher rates of glucose uptake. SCLC cells were enriched in metformin-adapted cultures. These SCLC cells acquired sensitivity to multiple methods of inhibition of CtBP function, including a cyclic peptide inhibitor of NADH-induced CtBP dimerization. Single-cell mRNA sequencing identified a reprogramming of epithelial-mesenchymal and stem cell gene expression in the metformin-adapted SCLC cells. These SCLC cells demonstrated an acquired dependency on one of these genes, Tenascin C. Thus, in addition to acquisition of sensitivity to glycolysis-targeting therapeutic strategies, the reprograming of gene expression in the metformin-adapted SCLC cells renders them sensitive to potential therapeutic approaches not directly linked to cell metabolism.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Antineoplastic Agents, Alkylating/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Metformin/pharmacology , Neoplastic Stem Cells/drug effects , Protein Multimerization/drug effects , Triple Negative Breast Neoplasms/drug therapy , Alcohol Oxidoreductases/metabolism , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Glycolysis , Humans , Inhibitory Concentration 50 , Metabolic Networks and Pathways/drug effects , Metformin/therapeutic use , Mice , NAD/metabolism , Neoplastic Stem Cells/pathology , Sequence Analysis, RNA , Single-Cell Analysis , Spheroids, Cellular , Tenascin/antagonists & inhibitors , Tenascin/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
7.
Biochem Biophys Res Commun ; 495(1): 104-109, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29101036

ABSTRACT

Extensive changes of neuronal transcriptome occur post ischemic stroke and during the following reperfusion. Although numerous studies focused on transcriptome changes of mRNAs associated with ischemic stroke, little is known about whether and how long non-coding RNAs (lncRNAs), which play critical roles in cellular homeostasis, are involved in this process. In this study, we performed high throughput screening to analyze expression changes of lncRNAs in primarily cultured hippocampal neurons under an oxygen-glucose deprivation/reperfusion (OGD/R) condition at 0 h, 6 h, 12 h, and 18 h, respectively. Knock down of one validated lncRNAs (Tnxa-ps1) promoted neuronal survival by inhibiting apoptosis. Coding non-coding co-expression network analysis revealed that the expression of Tnxa-ps1 was highly correlated with changes of a particular group of genes, many of which are associated with neural protection. Finally, we showed that down-regulation of Tnxa-ps1 reversed the expression changes of four mRNAs post OGD/R, revealing a regulatory effect between Tnxa-ps1 and selected genes. Together, our data revealed possible participation of lncRNAs in the pathophysiology of OGD/R and thereby provided new insights into the studies of potential therapeutic targets for ischemic stroke.


Subject(s)
Neurons/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Tenascin/genetics , Tenascin/metabolism , Animals , Apoptosis , Cell Survival , Cells, Cultured , Gene Expression Profiling , Gene Knockdown Techniques , Hippocampus/metabolism , Hippocampus/pathology , Neurons/pathology , RNA, Long Noncoding/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Reperfusion Injury/pathology , Tenascin/antagonists & inhibitors
8.
Biomol Concepts ; 7(2): 117-32, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27135623

ABSTRACT

Matricellular proteins influence wide-ranging fundamental cellular processes including cell adhesion, migration, growth and differentiation. They achieve this both through interactions with cell surface receptors and regulation of the matrix environment. Many matricellular proteins are also associated with diverse clinical disorders including cancer and diabetes. Alternative splicing is a precisely regulated process that can produce multiple isoforms with variable functions from a single gene. To date, the expression of alternate transcripts for the matricellular family has been reported for only a handful of genes. Here we analyse the evidence for alternative splicing across the matricellular family including the secreted protein acidic and rich in cysteine (SPARC), thrombospondin, tenascin and CCN families. We find that matricellular proteins have double the average number of splice variants per gene, and discuss the types of domain affected by splicing in matricellular proteins. We also review the clinical significance of alternative splicing for three specific matricellular proteins that have been relatively well characterised: osteopontin (OPN), tenascin-C (TNC) and periostin. Embracing the complexity of matricellular splice variants will be important for understanding the sometimes contradictory function of these powerful regulatory proteins, and for their effective clinical application as biomarkers and therapeutic targets.


Subject(s)
Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Alternative Splicing , Animals , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/chemistry , Gene Expression Regulation , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Molecular Targeted Therapy , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Osteopontin/genetics , Osteopontin/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Isoforms , Protein Processing, Post-Translational , Tenascin/antagonists & inhibitors , Tenascin/genetics , Tenascin/metabolism
9.
Mediators Inflamm ; 2015: 613249, 2015.
Article in English | MEDLINE | ID: mdl-26557739

ABSTRACT

Tenascin-C (TN-C), an extracellular matrix (ECM) glycoprotein, is specifically induced upon tissue injury and infection and during septic conditions. Carbon monoxide (CO) gas is known to exert various anti-inflammatory effects in various inflammatory diseases. However, the mechanisms underlying the effect of CO on TN-C-mediated inflammation are unknown. In the present study, we found that treatment with LPS significantly enhanced TN-C expression in macrophages. CO gas, or treatment with the CO-donor compound, CORM-2, dramatically reduced LPS-induced expression of TN-C and proinflammatory cytokines while significantly increased the expression of IL-10. Treatment with TN-C siRNA significantly suppressed the effects of LPS on proinflammatory cytokines production. TN-C siRNA did not affect the CORM-2-dependent increase of IL-10 expression. In cells transfected with IL-10 siRNA, CORM-2 had no effect on the LPS-induced expression of TN-C and its downstream cytokines. These data suggest that IL-10 mediates the inhibitory effect of CO on TN-C and the downstream production of proinflammatory cytokines. Additionally, administration of CORM-2 dramatically reduced LPS-induced TN-C and proinflammatory cytokines production while expression of IL-10 was significantly increased. In conclusion, CO regulated IL-10 expression and thus inhibited TN-C-mediated inflammation in vitro and in vivo.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Carbon Monoxide/pharmacology , Interleukin-10/physiology , Sepsis/drug therapy , Tenascin/antagonists & inhibitors , Animals , Cells, Cultured , Cytokines/biosynthesis , Disease Models, Animal , Dose-Response Relationship, Drug , Heme Oxygenase-1/physiology , Inflammation/drug therapy , Inflammation/immunology , Lipopolysaccharides/pharmacology , Male , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Tenascin/physiology
10.
Cent Nerv Syst Agents Med Chem ; 15(2): 126-37, 2015.
Article in English | MEDLINE | ID: mdl-25924917

ABSTRACT

Malignant glioma is the most prevalent and lethal primary brain tumor. Due to molecular heterogeneity and organ-specific clinical manifestations, it is essential to improve glioma treatment by shifting from conventional cytotoxic chemotherapy to more targeted therapies. Hence, innovative approaches based on ligands able to specifically detect and measure mutated proteins associated to a specific tumor phenotype are needed in order to refine diagnosis and therapy of glioma. To date, antibody- based approaches have been developed for in vivo applications but, in most cases, they show toxicity, do not reach adequate sensitivity and have a low permeability across the blood-brain-barrier. Single-stranded nucleic acid aptamers, generated by the SELEX (Systematic Evolution of Ligands by EXponential enrichment) process, have been shown as a valuable alternative to protein antibodies because may couple the advantages of their chemical nature to the high specific binding of antibodies to their proper targets. The simplicity of aptamers selection and derivatization with other molecules (nanocarriers, tracers for imaging, drugs) combined to low immunogenicity and toxicity, render them a versatile tool for identification of new biomarkers, in vitro diagnosis, in vivo imaging and targeted therapy. Aim of this review article is to discuss contemporary applications of aptamers as innovative tools for glioma diagnosis and therapy. We will describe promising new approaches for the identification of aptamers targeting proteins with a crucial role in glioma, including SELEX protocols against living glioma cells and brain tumor-initiating cells. Additionally, we will review recently proposed aptamer-based strategies for site-targeted controlled delivery of therapeutics and imaging agents to the brain. Summarizing, we hope that this article will provide an updated overview of perspectives and challenges for aptamer-based glioma treatment in the near future.


Subject(s)
Antineoplastic Agents/therapeutic use , Aptamers, Nucleotide/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Molecular Targeted Therapy , Animals , Antineoplastic Agents/pharmacokinetics , Aptamers, Nucleotide/pharmacokinetics , Blood-Brain Barrier , Brain Neoplasms/diagnosis , Clinical Trials as Topic , Drug Carriers , Glioma/diagnosis , Humans , Mice , Neoplasm Proteins/antagonists & inhibitors , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , SELEX Aptamer Technique , Tenascin/antagonists & inhibitors , Xenograft Model Antitumor Assays
11.
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
12.
Anticancer Res ; 33(5): 1927-34, 2013 May.
Article in English | MEDLINE | ID: mdl-23645740

ABSTRACT

BACKGROUND: Tenascin-C (TNC) is an extracellular matrix glycoprotein, usually derived from myofibroblasts in the cancer microenvironment. Recently, however, the significance of tumor-derived TNC in initiation of cancer metastasis was disclosed. We investigated the clinical significance of cancer-derived TNC in colorectal cancer (CRC) cases. MATERIALS AND METHODS: TNC expression in 170 cases of CRC was analyzed by quantitative real-time polymerase chain reaction (PCR). In addition, gene expression arrays using purely-separated cancer tissues of another 86 cases was performed and the functional implications of cancer-specific TNC were investigated. RESULTS: The expression of TNC mRNA was significantly higher in CRC tissues than in the corresponding normal tissues. Cancer cell-specific TNC expression was a significant prognostic factor in CRC cases. Moreover, cancer cell-derived TNC was associated with the epithelial-mesenchymal transition (EMT) signature. CONCLUSION: Cancer cell-derived TNC promotes cancer invasiveness via EMT regulation, and not cancer tissue TNC but cancer cell-specific TNC is a novel indicator of poor prognosis.


Subject(s)
Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Liver Neoplasms/secondary , Peritoneal Neoplasms/secondary , Tenascin/genetics , Aged , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Adhesion , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Laser Capture Microdissection , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lymphatic Metastasis , Male , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism , Stromal Cells/pathology , Tenascin/antagonists & inhibitors , Tenascin/metabolism , Tumor Cells, Cultured
13.
Neurosci Lett ; 525(2): 129-34, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22902990

ABSTRACT

Tenascin-R (TN-R) is a neural specific protein and an important molecule involved in inhibition of axonal regeneration after spinal cord injury (SCI). Here we report on rabbit-derived TN-R polyclonal antibody, which acts as a TN-R antagonist with high titer and high specificity, promoted neurite outgrowth and sprouting of rat cortical neurons cultured on the inhibitory TN-R substrate in vitro. When locally administered into the lesion sites of rats received spinal cord dorsal hemisection, these TN-R antibodies could significantly decrease RhoA activation and improve functional recovery from corticospinal tract (CST) transection. Thus, passive immunotherapy with specific TN-R antagonist may represent a promising repair strategy following acute SCI.


Subject(s)
Antibodies/pharmacology , Axons/drug effects , Spinal Cord Injuries/therapy , Tenascin/antagonists & inhibitors , Animals , Animals, Newborn , Antibodies/therapeutic use , Axons/physiology , Cells, Cultured , Female , Hindlimb/physiopathology , Immunization, Passive , Motor Activity , Nerve Regeneration , Neurites/drug effects , Neurites/physiology , Rabbits , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/immunology , Spinal Cord Injuries/physiopathology , Tenascin/immunology , rhoA GTP-Binding Protein/metabolism
14.
Curr Pharm Biotechnol ; 12(11): 1805-22, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21902632

ABSTRACT

Malignant gliomas are the deadliest brain tumors, which are characterized by highly invasive growth, a rampant genetic instability and intense resistance to apoptosis. Such an aggressive behavior of malignant gliomas is reflected in the resistance to chemo- and radiotherapy and weak prognosis in spite of cytoreduction through surgery. Brain tumors preferentially express a number of specific protein and RNA markers, that may be exploited as potential therapeutic targets in design of the new treatment modalities based on nucleic acids. For almost three decades, a possibility to apply DNA and RNA molecules as anticancer therapeutics have been studied. A variety of antisense oligonucleotides, ribozymes, DNAzymes, and aptamers can be designed to trigger the sequence-specific inhibition of particular mRNA of interest. RNA interference (RNAi) is the latest and the most promising technique in the long line of nucleic acid-based therapeutic technologies. Recently, we designed and implemented the experimental therapy of patients suffering from malignant brain tumors based on application of double-stranded RNA (dsRNA) specific for tenascin-C (TN-C) mRNA. That therapeutic agent, called ATN-RNA, induces RNAi pathway to inhibit the synthesis of TN-C, the extracellular matrix protein which is highly overexpressed in brain tumor tissue. In the chapter specific problems of application of nucleic acid-based technologies in glioma tumors treatment will be discussed.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Oligonucleotides, Antisense/therapeutic use , RNA Interference , RNA, Small Interfering/therapeutic use , Tenascin/antagonists & inhibitors , Animals , Apoptosis/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/adverse effects , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacokinetics , RNA, Small Interfering/genetics , Tenascin/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Exp Cell Res ; 317(12): 1714-25, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21586285

ABSTRACT

Activation and migration of resident stellate cells (HSCs) within the hepatic space of Disse play an important role in hepatic fibrosis, which accounts for the increased numbers of activated HSCs in areas of inflammation during hepatic fibrosis. Currently, microRNAs have been found to play essential roles in HSC differentiation, proliferation, apoptosis, fat accumulation and collagen production. However, little is known about microRNA mediated HSC activation and migration. In this study, the miRNA expression profiles of quiescent HSCs, partially activated HSCs and fully activated HSCs were compared in pairs. Gene ontology (GO) and GO-Map network analysis indicated that the activation of HSCs was regulated by microRNAs. Among them miR-335 was confirmed to be significantly reduced during HSC activation by qRT-PCR, and restoring expression of miR-335 inhibited HSC migration and reduced α-SMA and collagen type I. Previous study revealed that tenascin-C (TNC), an extracellular matrix glycoprotein involved in cell migration, might be a target of miR-335. Therefore, we further studied the TNC expression in miR-335 over-expressed HSCs. Our data showed that exogenous TNC could enhance HSC migration in vitro and miR-335 restoration resulted in a significant inhibition of TNC expression. These results demonstrated that miR-335 restoration inhibited HSC migration, at least in part, via downregulating the TNC expression.


Subject(s)
Cell Movement , Cell Proliferation , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , MicroRNAs/metabolism , Tenascin/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Blotting, Western , Cells, Cultured , Down-Regulation , Gene Expression Profiling , Male , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tenascin/antagonists & inhibitors , Tenascin/genetics , Wound Healing
16.
Plant Biotechnol J ; 7(1): 59-72, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18793269

ABSTRACT

The aims of this work were to obtain a human antibody against the tumour-associated antigen tenascin-C (TNC) and to compare the yield and quality of plant-produced antibody in either stable transgenics or using a transient expression system. To this end, the characterization of a full-sized human immunoglobulin G (IgG) [monoclonal antibody H10 (mAb H10)], derived from a selected single-chain variable fragment (scFv) and produced in plants, is presented. The human mAb gene was engineered for plant expression, and Nicotiana tabacum transgenic lines expressing both heavy (HC) and light (LC) chain were obtained and evaluated for antibody expression levels, in vivo assembly and functionality. Affinity-purified H10 from transgenics (yield, 0.6-1.1 mg/kg fresh weight) revealed that more than 90% of HC was specifically degraded, leading to the formation of functional antigen-binding fragments (Fab). Consequently, H10 was transiently expressed in Nicotiana benthamiana plants through an Agrobacterium-mediated gene-transfer system. Moreover, the use of the p19 silencing suppressor gene from artichoke mottled crinkle virus raised antibody expression levels by an order of magnitude (yields of purified H10, 50-100 mg/kg fresh weight). Approximately 75% of purified protein consisted of full-sized antibody functionally binding to TNC (K(D) = 14 nm), and immunohistochemical analysis on tumour tissues revealed specific accumulation around tumour blood vessels. The data indicate that the purification yields of mAb H10, using a transient expression system boosted by the p19 silencing suppressor, are exceptionally high when compared with the results reported previously, providing a technique for the over-expression of anticancer mAbs by a rapid, cost-effective, molecular farming approach.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Neoplasm/biosynthesis , Plants, Genetically Modified/metabolism , Tenascin/antagonists & inhibitors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Gene Expression , Humans , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Mice , Molecular Sequence Data , Neoplasms, Experimental/immunology , Plants, Genetically Modified/genetics , Plants, Genetically Modified/immunology , Protein Engineering , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Nicotiana/genetics , Nicotiana/metabolism , Transformation, Genetic
17.
Oncogene ; 27(40): 5373-84, 2008 Sep 11.
Article in English | MEDLINE | ID: mdl-18504437

ABSTRACT

Metastasis is the primary cause of death in patients with breast cancer. Overexpression of c-myc in humans correlates with metastases, but transgenic mice only show low rates of micrometastases. We have generated transgenic mice that overexpress both c-myc and vascular endothelial growth factor (VEGF) (Myc/VEGF) in the mammary gland, which develop high rates of pulmonary macrometastases. Gene expression profiling revealed a set of deregulated genes in Myc/VEGF tumors compared to Myc tumors associated with the increased metastatic phenotype. Cross-comparisons between this set of genes with a human breast cancer lung metastasis gene signature identified five common targets: tenascin-C(TNC), matrix metalloprotease-2, collagen-6-A1, mannosidase-alpha-1A and HLA-DPA1. Signaling blockade or knockdown of TNC in MDA-MB-435 cells resulted in a significant impairment of cell migration and anchorage-independent cell proliferation. Mice injected with clonal MDA-MB-435 cells with reduced expression of TNC demonstrated a significant decrease (P<0.05) in (1) primary tumor growth; (2) tumor relapse after surgical removal of the primary tumor and (3) incidence of lung metastasis. Our results demonstrate that VEGF induces complex alterations in tissue architecture and gene expression. The TNC signaling pathway plays an important role in mammary tumor growth and metastases, suggesting that TNC may be a relevant target for therapy against metastatic breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-myc/physiology , Tenascin/pharmacology , Vascular Endothelial Growth Factor A/physiology , Animals , Biomarkers, Tumor/genetics , Blotting, Northern , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Lung Neoplasms/genetics , Lung Neoplasms/prevention & control , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tenascin/antagonists & inhibitors , Tenascin/genetics , Tumor Cells, Cultured , Tumor Stem Cell Assay
18.
Circ Res ; 99(8): 837-44, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16990566

ABSTRACT

Tenascin-C (TN-C) is an extracellular matrix (ECM) protein expressed within remodeling systemic and pulmonary arteries (PAs), where it supports vascular smooth muscle cell (SMC) proliferation. Previously, we showed that A10 SMCs cultivated on native type I collagen possess a spindle-shaped morphology and do not express TN-C, whereas those on denatured collagen possess a well-defined F-actin stress fiber network, a spread morphology, and they do express TN-C. To determine whether changes in cytoskeletal architecture control TN-C, SMCs on denatured collagen were treated with cytochalasin D, which decreased SMC spreading and activation of extracellular signal-regulated kinase 1/2 (ERK1/2), signaling effectors required for TN-C transcription. Next, to determine whether cell shape, dictated by the F-actin cytoskeleton, regulates TN-C, different geometries of SMCs (ranging from spread to round) were engineered on denatured collagen: as SMCs progressively rounded, ERK1/2 activity and TN-C transcription declined. Because RhoA and Rho kinase (ROCK) regulate cell morphology by controlling cytoskeletal architecture, we reasoned that these factors might also regulate TN-C. Indeed, SMCs on denatured collagen possessed higher levels of RhoA activity than those on native collagen, and blocking RhoA or ROCK activities attenuated SMC spreading, ERK1/2 activity, and TN-C expression in SMCs on denatured collagen. Thus, ROCK controls the configuration of the F-actin cytoskeleton and SMC shape in a manner that is permissive for ERK1/2-dependent production of TN-C. Finally, we showed that inhibition of ROCK activity suppresses SMC TN-C expression and disease progression in hypertensive rat PAs. Thus, in addition to its role in regulating vasoconstriction, ROCK also controls matrix production.


Subject(s)
Extracellular Matrix/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Serine-Threonine Kinases/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Actins/physiology , Animals , Blood Vessels/physiology , Cell Adhesion/physiology , Cell Shape/physiology , Cells, Cultured , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Disease Progression , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypertension/chemically induced , Hypertension/metabolism , Hypertension/physiopathology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Monocrotaline , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats , Stress, Mechanical , Tenascin/antagonists & inhibitors , Tenascin/biosynthesis , Tenascin/genetics , Tenascin/metabolism , Transcription, Genetic/physiology , Vasoconstriction/physiology , rho-Associated Kinases , rhoA GTP-Binding Protein/physiology
19.
J Thorac Cardiovasc Surg ; 128(3): 357-63, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15354092

ABSTRACT

OBJECTIVE: Accumulation of smooth muscle cells and extracellular matrix in the intima of artery bypass grafts induces neointimal hyperplasia, resulting in graft failure. We investigated the inhibitory effect of locally applied cilostazol, an inhibitor of cyclic adenosine monophosphate phosphodiesterase III, on neointimal hyperplasia and the role of tenascin-C synthesis and smooth muscle cell proliferation in free artery grafts. Methods and results We established a distal anastomotic stricture model of free artery graft stenosis using rat abdominal aorta. In this model, neointimal hyperplasia was observed not only in the distal anastomotic site but also in the graft body at postoperative day 14 and was markedly progressed at day 28. Strong expression of tenascin-C was found in the media and neointima of the graft body. When cilostazol was locally administered around the graft using Pluronic gel, neointimal hyperplasia of the graft was significantly suppressed in comparison with gel-treated control graft. The mean neointima/media area ratio was reduced by 86.6% for the graft body and by 75.8% for the distal anastomotic site versus the control. Cilostazol treatment decreased cell proliferation and tenascin-C expression in the neointima. In an in vitro experiment using cultured smooth muscle cells isolated from rat aorta, cilostazol completely suppressed the tenascin-C mRNA expression induced by platelet-derived growth factor-BB. CONCLUSION: A single topical administration of cilostazol may suppress neointimal hyperplasia by inhibiting cell proliferation and tenascin-C synthesis in free artery grafts, presenting the potential for clinical use in vascular surgery.


Subject(s)
Arteries/drug effects , Phosphodiesterase Inhibitors/pharmacology , Tenascin/antagonists & inhibitors , Tenascin/biosynthesis , Tetrazoles/pharmacology , Tunica Intima/drug effects , Tunica Intima/pathology , Administration, Topical , Animals , Arteries/cytology , Cell Division , Cells, Cultured , Cilostazol , Hyperplasia , In Vitro Techniques , Male , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Rats , Rats, Inbred Lew
20.
Cancer Control ; 11(3): 192-207, 2004.
Article in English | MEDLINE | ID: mdl-15153843

ABSTRACT

BACKGROUND: Despite advances in surgical and adjuvant radiation therapy and chemotherapy strategies, malignant gliomas continue to be associated with poor prognoses. METHODS: We review immune-mediated treatment approaches for malignant glioma and the relevance of recent clinical trials and their outcomes. We specifically address the increasing evidence implicating the role of cytotoxic T cells in ensuring adequate immune-mediated clearance of neoplastic cells and the need for the optimization of therapies that can elicit and support such antitumor T-cell activity. RESULTS: The poor outcome of this disease has spurred the search for novel experimental therapies that can address and overcome the root biological phenomena associated with the lethality of this disease. The use of immunotherapy to bolster the otherwise impaired antitumor immune responses in glioma patients has received increasing attention. CONCLUSIONS: An effective treatment paradigm for malignant gliomas may eventually require a multifaceted approach combining two or more different immunotherapeutic strategies. Such scenarios may involve the use of local cytokine gene therapy to enhance glioma-cell immunogenicity in conjunction with dendritic cell-based active vaccination to stimulate systemic tumoricidal T-cell immunity. Given the heterogeneity of this disease process and the potential risk of immunoediting out a selected, treatment-refractory tumor cell population, the concurrent use of multiple modalities that target disparate tumor characteristics may be of greatest therapeutic relevance.


Subject(s)
Glioma/immunology , Glioma/therapy , Immunotherapy/methods , Adult , Aged , Clinical Trials as Topic , Cytokines/therapeutic use , ErbB Receptors/antagonists & inhibitors , Humans , Immunization, Passive/methods , Immunotherapy, Active/methods , Immunotherapy, Adoptive/methods , Middle Aged , Tenascin/antagonists & inhibitors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...