Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732135

ABSTRACT

Glioblastoma (GBM) is the most lethal and common malignant primary brain tumor in adults. An important feature that supports GBM aggressiveness is the unique composition of its extracellular matrix (ECM). Particularly, fibronectin plays an important role in cancer cell adhesion, differentiation, proliferation, and chemoresistance. Thus, herein, a hydrogel with mechanical properties compatible with the brain and the ability to disrupt the dynamic and reciprocal interaction between fibronectin and tumor cells was produced. High-molecular-weight hyaluronic acid (HMW-HA) functionalized with the inhibitory fibronectin peptide Arg-Gly-Asp-Ser (RGDS) was used to produce the polymeric matrix. Liposomes encapsulating doxorubicin (DOX) were also included in the hydrogel to kill GBM cells. The resulting hydrogel containing liposomes with therapeutic DOX concentrations presented rheological properties like a healthy brain. In vitro assays demonstrated that unmodified HMW-HA hydrogels only caused GBM cell killing after DOX incorporation. Conversely, RGDS-functionalized hydrogels displayed per se cytotoxicity. As GBM cells produce several proteolytic enzymes capable of disrupting the peptide-HA bond, we selected MMP-2 to illustrate this phenomenon. Therefore, RGDS internalization can induce GBM cell apoptosis. Importantly, RGDS-functionalized hydrogel incorporating DOX efficiently damaged GBM cells without affecting astrocyte viability, proving its safety. Overall, the results demonstrate the potential of the RGDS-functionalized hydrogel to develop safe and effective GBM treatments.


Subject(s)
Doxorubicin , Fibronectins , Glioblastoma , Hyaluronic Acid , Hydrogels , Oligopeptides , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , Oligopeptides/chemistry , Oligopeptides/pharmacology , Fibronectins/metabolism , Fibronectins/antagonists & inhibitors , Hydrogels/chemistry , Cell Line, Tumor , Hyaluronic Acid/chemistry , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Liposomes/chemistry , Apoptosis/drug effects , Matrix Metalloproteinase 2/metabolism
2.
Arch Toxicol ; 96(10): 2767-2783, 2022 10.
Article in English | MEDLINE | ID: mdl-35864199

ABSTRACT

Sustained exposure of the lung to various environmental or occupational toxins may eventually lead to pulmonary fibrosis, a devastating disease with no cure. Pulmonary fibrosis is characterized by excessive deposition of extracellular matrix (ECM) proteins such as fibronectin and collagens. The peptidase plasmin degrades the ECM, but protein levels of the plasmin activator inhibitor-1 (PAI-1) are increased in fibrotic lung tissue, thereby dampening plasmin activity. Transforming growth factor-ß1 (TGF-ß1)-induced activation of SMAD transcription factors promotes ECM deposition by enhancing collagen, fibronectin and PAI-1 levels in pulmonary fibroblasts. Hence, counteracting TGF-ß1-induced signaling is a promising approach for the therapy of pulmonary fibrosis. Transient receptor potential cation channel subfamily M Member 7 (TRPM7) supports TGF-ß1-promoted SMAD signaling in T-lymphocytes and the progression of fibrosis in kidney and heart. Thus, we investigated possible effects of TRPM7 on plasmin activity, ECM levels and TGF-ß1 signaling in primary human pulmonary fibroblasts (pHPF). We found that two structurally unrelated TRPM7 blockers enhanced plasmin activity and reduced fibronectin or PAI-1 protein levels in pHPF under basal conditions. Further, TRPM7 blockade strongly inhibited fibronectin and collagen deposition induced by sustained TGF-ß1 stimulation. In line with these data, inhibition of TRPM7 activity diminished TGF-ß1-triggered phosphorylation of SMAD-2, SMAD-3/4-dependent reporter activation and PAI-1 mRNA levels. Overall, we uncover TRPM7 as a novel supporter of TGF-ß1 signaling in pHPF and propose TRPM7 blockers as new candidates to control excessive ECM levels under pathophysiological conditions conducive to pulmonary fibrosis.


Subject(s)
Pulmonary Fibrosis , TRPM Cation Channels , Collagen/antagonists & inhibitors , Collagen/metabolism , Fibrinolysin/metabolism , Fibroblasts , Fibronectins/adverse effects , Fibronectins/antagonists & inhibitors , Fibronectins/metabolism , Fibrosis , Humans , Lung/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Protein Serine-Threonine Kinases , Pulmonary Fibrosis/chemically induced , TRPM Cation Channels/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism
3.
Theranostics ; 12(2): 929-943, 2022.
Article in English | MEDLINE | ID: mdl-34976221

ABSTRACT

Background: Bone is a frequent site of metastases from breast cancer, but existing therapeutic options are not satisfactory. Although osteoblasts have active roles in cancer progression by assisting the vicious bone-destructive cycle, we employed a counterintuitive approach of activating pro-tumorigenic Wnt signaling and examined the paradoxical possibility of developing osteoblast-derived tumor-suppressive, bone-protective secretomes. Methods: Wnt signaling was activated by the overexpression of Lrp5 and ß-catenin in osteoblasts as well as a pharmacological agent (BML284), and the therapeutic effects of their conditioned medium (CM) were evaluated using in vitro cell cultures, ex vivo breast cancer tissues, and a mouse model of osteolysis. To explore the unconventional regulatory mechanism of the action of Wnt-activated osteoblasts, whole-genome proteomics analysis was conducted, followed by immunoprecipitation and gain- and loss-of-function assays. Results: While osteoblasts did not present any innate tumor-suppressing ability, we observed that the overexpression of Lrp5 and ß-catenin in Wnt signaling made their CM tumor-suppressive and bone-protective. The growth of breast cancer cells and tissues was inhibited by Lrp5-overexpressing CM (Lrp5 CM), which suppressed mammary tumors and tumor-driven bone destruction in a mouse model. Lrp5 CM also inhibited the differentiation and maturation of bone-resorbing osteoclasts by downregulating NFATc1 and cathepsin K. The overexpression of Lrp5 upregulated osteopontin that enriched Hsp90ab1 (Hsp90 beta) and moesin (MSN) in Lrp5 CM. Hsp90ab1 and MSN are atypical tumor-suppressing proteins since they are multi-tasking, moonlighting proteins that promote tumorigenesis in tumor cells. Importantly, Hsp90ab1 immuno-precipitated latent TGFß and inactivated TGFß, whereas MSN interacted with CD44, a cancer stem-cell marker, as well as fibronectin 1, an ECM protein. Furthermore, Hsp90ab1 and MSN downregulated KDM3A that demethylated histones, together with PDL1 that inhibited immune responses. Conclusion: In contrast to inducing tumor-enhancing secretomes and chemoresistance in general by inhibiting varying oncogenic pathways in chemotherapy, this study presented the unexpected outcome of generation tumor-suppressive secretomes by activating the pro-tumorigenic Wnt pathway. The results shed light on the contrasting role of oncogenic signaling in tumor cells and osteoblast-derived secretomes, suggesting a counterintuitive option for the treatment of breast cancer-associated bone metastasis.


Subject(s)
Breast Neoplasms/complications , HSP90 Heat-Shock Proteins/metabolism , Microfilament Proteins/metabolism , Osteoblasts/metabolism , Osteolysis/prevention & control , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Fibronectins/antagonists & inhibitors , Fibronectins/metabolism , Humans , Hyaluronan Receptors/antagonists & inhibitors , Hyaluronan Receptors/metabolism , Mammary Neoplasms, Experimental/complications , Mammary Neoplasms, Experimental/therapy , Mice , Osteoclasts/metabolism , Osteogenesis , Osteolysis/metabolism , Proteome/metabolism , Secretome , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
4.
Oxid Med Cell Longev ; 2021: 5391706, 2021.
Article in English | MEDLINE | ID: mdl-34745418

ABSTRACT

Ischemia-reperfusion injury (IRI) is defined as the total combined damage that occurs during a period of ischemia and following the recovery of blood flow. Oxidative stress, mitochondrial dysfunction, and an inflammatory response are factors contributing to IRI-related damage that can each result in cell death. Irisin is a polypeptide that is proteolytically cleaved from the extracellular domain of fibronectin type III domain-containing protein 5 (FNDC5). Irisin acts as a myokine that potentially mediates beneficial effects of exercise by reducing oxidative stress, improving mitochondrial fitness, and suppressing inflammation. The existing literature also suggests a possible link between irisin and IRI, involving mechanisms similar to those associated with exercise. This article will review the pathogenesis of IRI and the potential benefits and current limitations of irisin as a therapeutic strategy for IRI, while highlighting the mechanistic correlations between irisin and IRI.


Subject(s)
Fibronectins/antagonists & inhibitors , Oxidative Stress , Reperfusion Injury/pathology , Reperfusion Injury/therapy , Animals , Fibronectins/metabolism , Humans , Reperfusion Injury/metabolism
5.
Biochem Biophys Res Commun ; 582: 77-85, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34695754

ABSTRACT

PURPOSE: The purpose of this study was to investigate the effect of FNDC5 expression levels in hepatocellular carcinoma on the phenotypic changes of macrophages in tumor tissues. METHODS: In this study, we established an in vitro co-culture system of hepatocellular carcinoma cells and macrophages. Then we performed overexpression or knockdown of FNDC5 gene in hepatocellular carcinoma cells to observe the effect of changes in FNDC5 expression level on the phenotypic changes of THP-1 macrophages. And the conclusions obtained in the in vitro assay were further validated by a subcutaneous tumorigenic nude mice model. RESULTS: Our findings suggest that elevated FNDC5 expression in hepatocellular carcinoma cells lead to an increased M2 phenotype and decreased M1 phenotype in macrophages. This effect may be achieved by elevating PPARγ levels in macrophages while decreasing NF-κB and NLRP3 levels. These changes could be reversed by using PPARγ inhibitors. CONCLUSION: We preliminarily demonstrated that FNDC5 in hepatocellular carcinoma cells promotes the polarization of M2 macrophages by affecting the PPARγ/NF-κB/NLRP3 pathway.


Subject(s)
Carcinoma, Hepatocellular/genetics , Fibronectins/genetics , Liver Neoplasms/genetics , NF-kappa B/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , PPAR gamma/genetics , Anilides/pharmacology , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Differentiation , Cell Line, Tumor , Coculture Techniques , Fibronectins/antagonists & inhibitors , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Mice , Mice, Nude , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , PPAR gamma/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , THP-1 Cells , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Neoplasia ; 23(9): 837-850, 2021 09.
Article in English | MEDLINE | ID: mdl-34298233

ABSTRACT

Understanding how the extracellular matrix affects cancer development constitutes an emerging research field. Fibronectin and collagen are two intriguing matrix components found in cancer. Large concentrations of fibronectin or collagen type I have been implicated in poor prognosis in patients. In a mouse model, we had shown that genetically decreasing circulating fibronectin resulted in smaller tumors. We therefore aimed to manipulate fibronectin pharmacologically and determine how cancer development is affected. Deletion of fibronectin in human breast cancer cells (MDA-MB-231) using shRNA (knockdown: Kd) improved survival and diminished tumor burden in a model of metastatic lesions and in a model of local growth. Based on these findings, it seemed reasonable to attempt to prevent fibronectin accumulation using a bacterial derived peptide called pUR4. Treatment with this peptide for 10 days in the breast cancer local growth model or for 5 days in a melanoma skin cancer model (B16) was associated with a significant suppression of cancer growth. Treatment aimed at inhibiting collagen type I accumulation without interfering with fibronectin could not affect any changes in vivo. In the absence of fibronectin, diminished cancer progression was due to inhibition of proliferation, even though changes in blood vessels were also detected. Decreased proliferation could be attributed to decreased ERK phosphorylation and diminished YAP expression. In summary, manipulating fibronectin diminishes cancer progression, mostly by suppressing cell proliferation. This suggests that matrix modulation could be used as an adjuvant to conventional therapy as long as a decrease in fibronectin is obtained.


Subject(s)
Fibronectins/antagonists & inhibitors , Fibronectins/metabolism , Neoplasms/metabolism , Neoplasms/prevention & control , Amino Acid Sequence , Animals , Fibronectins/genetics , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Nude , NIH 3T3 Cells , Neoplasms/genetics , Neoplasms/pathology , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Tumor Burden/drug effects , Tumor Burden/physiology , Xenograft Model Antitumor Assays/methods
7.
Nanomedicine ; 36: 102424, 2021 08.
Article in English | MEDLINE | ID: mdl-34174417

ABSTRACT

In order to generate an antibody directed enzyme prodrug therapy, here we designed a chimeric protein by fusing the F8 antibody that recognizes the EDA of fibronectin (expressed on the tumor neovasculature) and an evolved variant of the ROS-generating enzyme D-amino acid oxidase (DAAO). The F8(scFv)-DAAO-Q144R recombinant protein is expressed by both CHO-S and E. coli cells. The F8(scFv)-DAAO-Q144R from E. coli cells is fully soluble, shows a high specific activity, is more thermostable in blood than the native DAAO, possesses a binding affinity for EDA well suited for efficient tumor accumulation, and localizes in tumor tissues. Notably, the F8(scFv)-DAAO-Q144R conjugate generates a stronger cytotoxicity to tumor cells than the native enzyme, especially when an inhibitor of heme oxygenase-1 (HO-1) is used, making it a promising candidate for a selective antitumor oxidative therapy controlled by the substrate addition, in the so called "activity on demand", thus sparing normal tissue from damage.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents , Cytotoxins , D-Amino-Acid Oxidase , Fibronectins , Neoplasm Proteins , Neoplasms/drug therapy , Recombinant Fusion Proteins , Single-Chain Antibodies , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetulus , Cytotoxins/chemistry , Cytotoxins/pharmacology , D-Amino-Acid Oxidase/chemistry , D-Amino-Acid Oxidase/genetics , D-Amino-Acid Oxidase/pharmacology , Fibronectins/antagonists & inhibitors , Fibronectins/genetics , Fibronectins/metabolism , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacology
8.
J Clin Invest ; 131(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33529175

ABSTRACT

Intratumor heterogeneity is an important mediator of poor outcomes in many cancers, including breast cancer. Genetic subclones frequently contribute to this heterogeneity; however, their growth dynamics and interactions remain poorly understood. PIK3CA and HER2 alterations are known to coexist in breast and other cancers. Herein, we present data that describe the ability of oncogenic PIK3CA mutant cells to induce the proliferation of quiescent HER2 mutant cells through a cell contact-mediated mechanism. Interestingly, the HER2 cells proliferated to become the major subclone over PIK3CA counterparts both in vitro and in vivo. Furthermore, this phenotype was observed in both hormone receptor-positive and -negative cell lines, and was dependent on the expression of fibronectin from mutant PIK3CA cells. Analysis of human tumors demonstrated similar HER2:PIK3CA clonal dynamics and fibronectin expression. Our study provides insight into nonrandom subclonal architecture of heterogenous tumors, which may aid the understanding of tumor evolution and inform future strategies for personalized medicine.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Cell Communication/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Coculture Techniques , Female , Fibronectins/antagonists & inhibitors , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic , Gene Frequency , Gene Knockout Techniques , Humans , Immunohistochemistry , MCF-7 Cells , Mutation , Phenotype , Receptor, ErbB-2/genetics
9.
Cancer Immunol Res ; 9(3): 279-290, 2021 03.
Article in English | MEDLINE | ID: mdl-33355188

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy has had limited success in early-phase clinical studies for solid tumors. Lack of efficacy is most likely multifactorial, including a limited array of targetable antigens. We reasoned that targeting the cancer-specific extra domain B (EDB) splice variant of fibronectin might overcome this limitation because it is abundantly secreted by cancer cells and adheres to their cell surface. In vitro, EDB-CAR T cells recognized and killed EDB-positive tumor cells. In vivo, 1 × 106 EDB-CAR T cells had potent antitumor activity in both subcutaneous and systemic tumor xenograft models, resulting in a significant survival advantage in comparison with control mice. EDB-CAR T cells also targeted the tumor vasculature, as judged by IHC and imaging, and their antivascular activity was dependent on the secretion of EDB by tumor cells. Thus, targeting tumor-specific splice variants such as EDB with CAR T cells is feasible and has the potential to improve the efficacy of CAR T-cell therapy.


Subject(s)
Fibronectins/antagonists & inhibitors , Immunotherapy, Adoptive/methods , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/therapy , T-Lymphocytes/transplantation , Animals , Antigens, Neoplasm , Cell Line, Tumor , Coculture Techniques , Feasibility Studies , Fibronectins/genetics , Fibronectins/immunology , Fibronectins/metabolism , Healthy Volunteers , Human Umbilical Vein Endothelial Cells , Humans , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Neoplasms/immunology , Neoplasms/pathology , Primary Cell Culture , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , RNA Splicing , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
10.
Biosci Biotechnol Biochem ; 84(12): 2415-2424, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32942951

ABSTRACT

This study aims to discover the effects of ursolic acid (UA) on papillary thyroid carcinoma (PTC). Human PTC cells were under UA treatment, and cell viability, clone formation, and apoptosis were measured by MTT assay, clone formation assay, and flow cytometry, respectively. Expressions of apoptosis- and epithelial-mesenchymal transition (EMT)-related markers were determined via qRT-PCR and western blot. Fibronectin-1 (FN1) expression in thyroid carcinoma was analyzed by GEPIA2 and qRT-PCR. The effects of overexpressed FN1 on UA-treated cells were detected following the previous procedures. Cell viability, proliferation, and EMT-related marker expressions were inhibited, while cell apoptosis and apoptosis-related marker expressions were promoted by UA. FN1 was higher expressed in thyroid carcinoma and downregulated by UA. Effects of FN1 on cell viability, proliferation, and apoptosis- and EMT-related marker expressions were partially reversed by UA. UA inhibited human PTC cell viability, proliferation, and EMT but promoted apoptosis via suppressing FN1.


Subject(s)
Antineoplastic Agents/pharmacology , Fibronectins/antagonists & inhibitors , Thyroid Cancer, Papillary/pathology , Triterpenes/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ursolic Acid
11.
Exp Cell Res ; 396(1): 112289, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32950474

ABSTRACT

Lesion healing without treatment is a unique clinical characteristic of the early stages of syphilis infection. Angiogenesis, which involves endothelial cell migration, is an important process in wound healing. Tp0136, an outer membrane protein of T. pallidum, has the ability to bind host fibronectin-producing cells, which plays a crucial role in the pathogenesis of syphilis. In this research, we purposed to analyze the role of Tp0136 in the migration of human microvascular endothelial (HMEC-1) cells and to explore the related mechanism. First, Tp0136 significantly promoted HMEC-1 cell migration. Furthermore, the levels of C-C motif ligand 2 (CCL2) mRNA and protein expression rose with the concentration and time increasing of Tp0136. The migration of HMEC-1 cells was significantly suppressed by an anti-CCL2 antibody and a CCR2 (the CCL2 receptor) inhibitor. Further study revealed that, in cells pretreated with anti-fibronectin antibody, anti-integrin ß1 antibody or RGD (Arg-Gly-Asp), the expression levels of CCL2 induced by Tp0136 were notably decreased. Additionally, after pretreatment with an anti-fibronectin antibody, an anti-integrin ß1 antibody or RGD, the migration of HMEC-1 cells treated with Tp0136 was obviously suppressed. These results show that Tp0136 promots the migration of HMEC-1 cells by inducing CCL2 expression via the interaction of the fibronectin RGD domain with integrin ß1 and the CCL2/CCR2 signaling pathway, and these interactions may contribute to the mechanisms that increase the capacity for self-healing syphilis infection.


Subject(s)
Bacterial Proteins/pharmacology , Cell Movement/drug effects , Fibronectins/genetics , Integrin beta1/genetics , Treponema pallidum/metabolism , Antibodies, Neutralizing/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cloning, Molecular , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Escherichia coli/genetics , Escherichia coli/metabolism , Fibronectins/antagonists & inhibitors , Fibronectins/metabolism , Gene Expression , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Host-Pathogen Interactions/genetics , Humans , Integrin beta1/metabolism , Oligopeptides/pharmacology , Protein Binding , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , Treponema pallidum/chemistry
12.
Respir Res ; 21(1): 108, 2020 May 07.
Article in English | MEDLINE | ID: mdl-32381012

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a rapidly progressing disease with challenging management. To find novel effective therapies, better preclinical models are needed for the screening of anti-fibrotic compounds. Activated fibroblasts drive fibrogenesis and are the main cells responsible for the accumulation of extracellular matrix (ECM). Here, a prolonged Scar-in-a-Jar assay was combined with clinically validated biochemical markers of ECM synthesis to evaluate ECM synthesis over time. To validate the model as a drug screening tool for novel anti-fibrotic compounds, two approved compounds for IPF, nintedanib and pirfenidone, and a compound in development, omipalisib, were tested. METHODS: Primary human lung fibroblasts from healthy donors were cultured for 12 days in the presence of ficoll and were stimulated with TGF-ß1 with or without treatment with an ALK5/TGF-ß1 receptor kinase inhibitor (ALK5i), nintedanib, pirfenidone or the mTOR/PI3K inhibitor omipalisib (GSK2126458). Biomarkers of ECM synthesis were evaluated over time in cell supernatants using ELISAs to assess type I, III, IV, V and VI collagen formation (PRO-C1, PRO-C3, PRO-C4, PRO-C5, PRO-C6), fibronectin (FBN-C) deposition and α-smooth muscle actin (α-SMA) expression. RESULTS: TGF-ß1 induced synthesis of PRO-C1, PRO-C6 and FBN-C as compared with unstimulated fibroblasts at all timepoints, while PRO-C3 and α-SMA levels were not elevated until day 8. Elevated biomarkers were reduced by suppressing TGF-ß1 signalling with ALK5i. Nintedanib and omipalisib were able to reduce all biomarkers induced by TGF-ß1 in a concentration dependent manner, while pirfenidone had no effect on α-SMA. CONCLUSIONS: TGF-ß1 stimulated synthesis of type I, III and VI collagen, fibronectin and α-SMA but not type IV or V collagen. Synthesis was increased over time, although temporal profiles differed, and was modulated pharmacologically by ALK5i, nintedanib, pirfenidone and omipalisib. This prolonged 12-day Scar-in-a-Jar assay utilising biochemical markers of ECM synthesis provides a useful screening tool for novel anti-fibrotic compounds.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cicatrix/chemically induced , Cicatrix/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Protein Kinase Inhibitors/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/metabolism , Cells, Cultured , Cicatrix/drug therapy , Collagen/antagonists & inhibitors , Collagen/metabolism , Drug Evaluation, Preclinical/methods , Extracellular Matrix/drug effects , Fibroblasts/drug effects , Fibronectins/antagonists & inhibitors , Fibronectins/metabolism , Fibrosis/chemically induced , Fibrosis/drug therapy , Fibrosis/metabolism , Humans , Indoles/antagonists & inhibitors , Indoles/metabolism , Protein Kinase Inhibitors/therapeutic use , Pyridones/antagonists & inhibitors , Pyridones/metabolism , Transforming Growth Factor beta1/toxicity
13.
Signal Transduct Target Ther ; 5(1): 12, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32296050

ABSTRACT

Stage IV breast cancer, which has a high risk of invasion, often develops into metastases in distant organs, especially in the lung, and this could threaten the lives of women. Thus, the development of more advanced therapeutics that can efficiently target metastatic foci is crucial. In this study, we built an dual-acting therapeutic strategy using micelles with high stability functionalized with fibronectin-targeting CREKA peptides encapsulating two slightly soluble chemotherapy agents in water, doxorubicin (D) and vinorelbine (V), which we termed C-DVM. We found that small C-DVM micelles could efficiently codeliver drugs into 4T1 cells and disrupt microtubule structures. C-DVM also exhibited a powerful ability to eradicate and inhibit invasion of 4T1 cells. Moreover, an in vivo pharmacokinetics study showed that C-DVM increased the drug circulation half-life and led to increased enrichment of drugs in lung metastatic foci after 24 h. Moreover, dual-acting C-DVM treatment led to 90% inhibition of metastatic foci development and reduced invasion of metastases. C-DVM could potentially be used as a targeted treatment for metastasis and represents a new approach with higher therapeutic efficacy than conventional chemotherapy for stage IV breast cancer that could be used in the future.


Subject(s)
Breast Neoplasms/drug therapy , Fibronectins/antagonists & inhibitors , Molecular Targeted Therapy , Oligopeptides/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Combined Modality Therapy , Doxorubicin/chemistry , Doxorubicin/pharmacology , Female , Fibronectins/genetics , Humans , Micelles , Nanoparticles/chemistry , Neoplasm Metastasis , Neoplasm Staging , Oligopeptides/chemistry , Vinorelbine/chemistry , Vinorelbine/pharmacology
14.
Curr Mol Med ; 20(9): 741-750, 2020.
Article in English | MEDLINE | ID: mdl-32031067

ABSTRACT

BACKGROUND: Autophagy is deeply associated with aging, but little is known about its association with the extracellular matrix (ECM). 3-methyladenine (3-MA) is a commonly used autophagy inhibitor. OBJECTIVE: We used this compound to investigate the role of autophagy in dermal ECM protein synthesis. METHODS: Normal human dermal fibroblasts (NHDFs) were treated with 3-MA for 24 h, and mRNA encoding several ECM proteins was analyzed in addition to the protein expression of procollagen-1 and fibronectin. Several phosphoinositide 3-kinase (PI3K) inhibitors, an additional autophagy inhibitor, and small interfering RNA (siRNA) targeting autophagy-related genes were additionally used to confirm the role of autophagy in ECM synthesis. RESULTS: Only 3-MA, but not other chemical compounds or autophagy-related genetargeting siRNA, inhibited the transcription of procollagen-1 and fibronectin-encoding genes. Further, 3-MA did not affect the activation of regulatory Smads, but inhibited the interaction between Smad3 with p300. Moreover, 3-MA treatment increased the phosphorylation of cAMP response element-binding protein (CREB); however, CREB knock-down did not recover 3-MA-induced procollagen-1 and fibronectin downregulation. CONCLUSION: We revealed that 3-MA might inhibit procollagen-1 and fibronectin synthesis in an autophagy-independent manner by interfering with the binding between Smad3 and p300. Therefore, 3-MA could be a candidate for the treatment of diseases associated with the accumulation of ECM proteins.


Subject(s)
Adenine/analogs & derivatives , Autophagy , Dermis/drug effects , Extracellular Matrix/drug effects , Fibroblasts/drug effects , Fibronectins/antagonists & inhibitors , Procollagen/antagonists & inhibitors , Adenine/pharmacology , Cells, Cultured , Dermis/metabolism , Dermis/pathology , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibronectins/genetics , Fibronectins/metabolism , Humans , Phosphorylation , Procollagen/genetics , Procollagen/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism
15.
Biochem Biophys Res Commun ; 520(3): 619-626, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31623832

ABSTRACT

Elevated plasma free fatty acid (FFA) levels are associated with insulin resistance and can cause lipotoxicity in skeletal muscles. In response to FFAs, skeletal muscle can secrete a variety of cytokines. Irisin, one such muscle-secreted cytokine, can improve glucose tolerance, glucose uptake, and lipid metabolism. It is produced by the transmembrane protein fibronectin type Ⅲ domain containing 5 (FNDC5) by specific proteases. The purpose of this study was to investigate the regulatory mechanisms of the FNDC5 response to palmitate and their relationships with insulin resistance in C2C12 myotubes. RNA sequencing analysis results from C2C12 myotubes treated with palmitate showed that palmitate could activate the TGF-ß signaling pathway. Palmitate directly affected the expression of Smad3, but not its phosphorylation level, in C2C12 myotubes. Furthermore, knockdown and knockout of Smad3 alleviated the inhibitory effect of palmitate on the expression of FNDC5. In contrast, overexpression of Smad3 aggravated the inhibition of FNDC5 expression. There is a Smad3 binding motif in the -660 bp to -649 bp region of the Fndc5 promoter. CRISPR/Cas9 knockout of this region also alleviated the inhibition of FNDC5 expression in response to palmitate. More importantly, inhibition of FNDC5 expression mediated by Smad3 led to a decrease in insulin sensitivity in C2C12 myotubes. Collectively, these findings suggest that palmitate could induce insulin resistance through Smad3-mediated down-regulation of the Fndc5 gene.


Subject(s)
Fibronectins/metabolism , Insulin Resistance/physiology , Muscle Fibers, Skeletal/metabolism , Palmitic Acid/metabolism , Smad3 Protein/metabolism , Animals , Binding Sites/genetics , Cell Line , Down-Regulation/drug effects , Fibronectins/antagonists & inhibitors , Fibronectins/genetics , Gene Knockdown Techniques , Gene Knockout Techniques , Mice , Muscle Fibers, Skeletal/drug effects , Palmitic Acid/pharmacology , Promoter Regions, Genetic , Signal Transduction/drug effects , Smad3 Protein/antagonists & inhibitors , Smad3 Protein/genetics , Transforming Growth Factor beta/metabolism
16.
Am J Physiol Renal Physiol ; 317(6): F1430-F1438, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31545926

ABSTRACT

Renal fibrosis is a key pathological phenomenon of chronic kidney disease (CKD) contributing to the progressive loss of renal function. UK383,367 is a procollagen C proteinase inhibitor that has been selected as a candidate for dermal antiscarring agents, whereas its role in renal fibrosis is unclear. In the present study, UK383,367 was applied to a CKD mouse model of unilateral ureteral obstruction (UUO) and cell lines of renal tubular epithelial cells (mouse proximal tubular cells) and renal fibroblast cells (NRK-49F cells) challenged by transforming growth factor-ß1. In vivo, bone morphogenetic protein 1, the target of UK383,367, was significantly enhanced in UUO mouse kidneys and renal biopsies from patients with CKD. Strikingly, UK383,367 administration ameliorated tubulointerstitial fibrosis as shown by Masson's trichrome staining in line with the blocked expression of collagen type I/III, fibronectin, and α-smooth muscle actin in the kidneys from UUO mice. Similarly, the enhanced inflammatory factors in obstructed kidneys were also blunted. In vitro, UK383,367 pretreatment inhibited the induction of collagen type I/III, fibronectin, and α-smooth muscle actin in both mouse proximal tubular cells and NRK-49F cells treated with transforming growth factor-ß1. Taken together, these findings indicate that the bone morphogenetic protein 1 inhibitor UK383,367 could serve as a potential drug in antagonizing CKD renal fibrosis by acting on the maturation and deposition of collagen and the subsequent profibrotic response and inflammation.


Subject(s)
Bone Morphogenetic Protein 1/antagonists & inhibitors , Oxadiazoles/therapeutic use , Renal Agents/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Animals , Cell Line , Child , Child, Preschool , Collagen Type I/antagonists & inhibitors , Collagen Type I/biosynthesis , Collagen Type III/antagonists & inhibitors , Collagen Type III/biosynthesis , Female , Fibronectins/antagonists & inhibitors , Fibronectins/biosynthesis , Fibrosis/drug therapy , Humans , Inflammation/pathology , Inflammation/prevention & control , Kidney/pathology , Kidney Function Tests , Male , Mice , Mice, Inbred C57BL , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/pathology , Ureteral Obstruction/complications
17.
Oxid Med Cell Longev ; 2019: 7857082, 2019.
Article in English | MEDLINE | ID: mdl-31191804

ABSTRACT

Intestinal ischemia/reperfusion (I/R), which is associated with high morbidity and mortality, is also accompanied with abnormal energy metabolism and liver injury. Irisin, a novel exercise-induced hormone, can regulate adipose browning and thermogenesis. The following study investigated the potential role of dexmedetomidine in liver injury during intestinal I/R in rats. Adult male Sprague-Dawley rats underwent occlusion of the superior mesenteric artery for 90 min followed by 2 h of reperfusion. Dexmedetomidine or irisin-neutralizing antibody was intravenously administered for 1 h before surgery. The results demonstrated that severe intestine and liver injuries occurred during intestinal I/R as evidenced by pathological scores and an apparent increase in serum diamine oxidase (DAO), aspartate aminotransferase (AST), alanine aminotransferase (ALT), and lactate dehydrogenase (LDH) levels. In addition, the hepatic irisin, cleaved caspase-3, Bax, and NLRP3 inflammasome components (including NLRP3, ASC, and caspase-1), protein expressions, apoptotic index, reactive oxygen species (ROS), malondialdehyde (MDA), myeloperoxidase (MPO), tumor necrosis factor- (TNF-) α, and interleukin- (IL-) 6 levels increased; however, the serum irisin level and hepatic Bcl-2 protein expression and superoxide dismutase (SOD) activity decreased after intestinal I/R. Interestingly, dexmedetomidine could reduce the above listed changes and increase the irisin levels in plasma and the liver in I/R rats. Dexmedetomidine-mediated protective effects on liver injury and NLRP3 inflammasome activation during intestinal I/R were partially abrogated via irisin-neutralizing antibody treatment. The results suggest that irisin might contribute to the hepatoprotection of dexmedetomidine during intestinal ischemia/reperfusion.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Dexmedetomidine/therapeutic use , Fibronectins/metabolism , Intestines/drug effects , Intestines/pathology , Ischemia/drug therapy , Reperfusion Injury/drug therapy , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , Fibronectins/antagonists & inhibitors , Fibronectins/blood , Ischemia/blood , Liver/drug effects , Liver/metabolism , Male , Malondialdehyde/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/blood , Tumor Necrosis Factor-alpha/metabolism
18.
Kidney Blood Press Res ; 44(4): 479-495, 2019.
Article in English | MEDLINE | ID: mdl-31238319

ABSTRACT

BACKGROUND/AIMS: Skeletal muscle atrophy is one of the main manifestations of protein energy wasting. We hypothesized that urotensin II (UII) can lead to skeletal muscle atrophy through upregulating autophagy and affecting Irisin precursor fibronectin type III domain containing 5 (FNDC5) expressions. METHODS: Three animal models (the sham operation, wild-type C57BL/6 mice with 5/6 nephrectomy, UII receptor (UT) gene knockout (UTKO) mice with 5/6 nephrectomy) were designed. Skeletal muscle weight, cross-sectional area (CSA) along with UII, FNDC5, LC3, and p62 expression were investigated. C2C12 cells were differentiated for up to 4 days into myotubes. These cells were then exposed to different UII concentrations (10-5 to 10-7 M) for 6-12 h and analyzed for the expressions of autophagic markers. These cells were also exposed to the same predetermined UII concentrations for 48-72 h and analyzed for the FNDC5 expression. Myotube diameter was measured. RESULTS: Upregulation of UII expression in skeletal muscle tissue was accompanied by reduced muscle weight and skeletal muscle CSA in the 2 posterior limbs, upregulated autophagy markers expression, and downregulated FNDC5 expression in 5/6 nephrectomy mice. The decrease of skeletal muscle weight, skeletal muscle CSA, downregulation of FNDC5 expression, and the upregulation of autophagy markers were inhibited in UTKO with 5/6 nephrectomy mice. Our in vitrostudy showed that UII could directly decrease myotube diameter, induce autophagy markers upregulation, and inhibit expression of FNDC5. When UII receptor gene was interfered by UT-specific siRNA, UII induced autophagy markers upregulation and FNDC5 downregulation were inhibited. CONCLUSION: We are the first to verify UII induces mice skeletal muscle atrophy associated with enhanced skeletal muscle autophagy and inhibited FNDC5 expression in chronic renal failure.


Subject(s)
Atrophy/chemically induced , Autophagy/drug effects , Fibronectins/antagonists & inhibitors , Kidney Failure, Chronic/metabolism , Muscle, Skeletal/pathology , Urotensins/pharmacology , Animals , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Fibronectin Type III Domain , Fibronectins/metabolism , Kidney Failure, Chronic/pathology , Mice
19.
BMC Cancer ; 19(1): 596, 2019 Jun 17.
Article in English | MEDLINE | ID: mdl-31208372

ABSTRACT

BACKGROUND: Gemcitabine remains a cornerstone in chemotherapy of pancreatic ductal adenocarcinoma (PDAC) despite suboptimal clinical effects that are partly due to the development of chemoresistance. Pancreatic stellate cells (PSCs) of the tumor stroma are known to interact with pancreatic cancer cells (PCCs) and influence the progression of PDAC through a complex network of signaling molecules that involve extracellular matrix (ECM) proteins. To understand tumor-stroma interactions regulating chemosensitivity, the role of PSC-secreted fibronectin (FN) in the development of gemcitabine resistance in PDAC was examined. METHODS: PSC cultures obtained from ten different human PDAC tumors were co-cultured with PCC lines (AsPC-1, BxPC-3, Capan-2, HPAF-II, MIA PaCa-2, PANC-1 and SW-1990) either directly, or indirectly via incubation with PSC-conditioned medium (PSC-CM). Gemcitabine dose response cytotoxicity was determined using MTT based cell viability assays. Protein expression was assessed by western blotting and immunofluorescence. PSC-CM secretome analysis was performed by proteomics-based LC-MS/MS, and FN content in PSC-CM was determined with ELISA. Radiolabeled gemcitabine was used to determine the capacity of PCCs to uptake the drug. RESULTS: In both direct and indirect co-culture, PSCs induced varying degrees of resistance to the cytotoxic effects of gemcitabine among all cancer cell lines examined. A variable degree of increased phosphorylation of ERK1/2 was observed across all PCC lines upon incubation with PSC-CM, while activation of AKT was not detected. Secretome analysis of PSC-CM identified 796 different proteins, including several ECM-related proteins such as FN and collagens. Soluble FN content in PSC-CM was detected in the range 175-350 ng/ml. Neither FN nor PSC-CM showed any effect on PCC uptake capacity of gemcitabine. PCCs grown on FN-coated surface displayed higher resistance to gemcitabine compared to cells grown on non-coated surface. Furthermore, a FN inhibitor, synthetic Arg-Gly-Asp-Ser (RGDS) peptide significantly inhibited PSC-CM-induced chemoresistance in PCCs via downregulation of ERK1/2 phosphorylation. CONCLUSIONS: The findings of this study suggest that FN secreted by PSCs in the ECM plays a key role in the development of resistance to gemcitabine via activation of ERK1/2. FN-blocking agents added to gemcitabine-based chemotherapy might counteract chemoresistance in PDAC and provide better clinical outcomes.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Fibronectins/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Extracellular Matrix/metabolism , Fibronectins/antagonists & inhibitors , Flavonoids/pharmacology , Humans , MAP Kinase Signaling System , Oligopeptides/pharmacology , Phosphorylation , Proteome/analysis , Proto-Oncogene Proteins c-akt/metabolism , Gemcitabine
20.
J Biomed Sci ; 26(1): 37, 2019 May 16.
Article in English | MEDLINE | ID: mdl-31096970

ABSTRACT

BACKGROUND: The blood-spinal cord barrier (BSCB) is composed of a monolayer of endothelium linked with tight junctions and extracellular matrix (ECM)-rich basement membranes and is surrounded by astrocyte foot processes. Endothelial permeability is regulated by interaction between endothelial cells and ECM proteins. Fibronectin (FN) is a principal ECM component of microvessels. Excessive FN deposition disrupts cell-cell adhesion in fibroblasts through ß1 integrin ligation. To determine whether excessive FN deposition contributes to the disruption of endothelial integrity, we used an in vitro model of the endothelial monolayer to investigate whether the FN inhibitor pUR4 prevents FN deposition into the subendothelial matrix and attenuates endothelial leakage. METHODS: To correlate the effects of excessive FN accumulation in microvessels on BSCB disruption, spinal nerve ligation-which induces BSCB leakage-was applied, and FN expression in the spinal cord was evaluated through immunohistochemistry and immunoblotting. To elucidate the effects by which pUR4 modulates endothelial permeability, brain-derived endothelial (bEND.3) cells treated with tumor necrosis factor (TNF)-α were used to mimic a leaky BSCB. A bEND.3 monolayer was preincubated with pUR4 before TNF-α treatment. The transendothelial electrical resistance (TEER) measurement and transendothelial permeability assay were applied to assess the endothelial integrity of the bEND.3 monolayer. Immunofluorescence analysis and immunoblotting were performed to evaluate the inhibitory effects of pUR4 on TNF-α-induced FN deposition. To determine the mechanisms underlying pUR4-mediated endothelial permeability, cell morphology, stress fiber formation, myosin light chain (MLC) phosphorylation, and ß1 integrin-mediated signaling were evaluated through immunofluorescence analysis and immunoblotting. RESULTS: Excessive FN was accumulated in the microvessels of the spinal cord after spinal nerve ligation; moreover, pUR4 inhibited TNF-α-induced FN deposition in the bEND.3 monolayer and maintained intact TEER and endothelial permeability. Furthermore, pUR4 reduced cell morphology alteration, actin stress fiber formation, and MLC phosphorylation, thereby attenuating paracellular gap formation. Moreover, pUR4 reduced ß1 integrin activation and downstream signaling. CONCLUSIONS: pUR4 reduces TNF-α-induced ß1 integrin activation by depleting ECM FN, leading to a decrease in endothelial hyperpermeability and maintenance of monolayer integrity. These findings suggest therapeutic benefits of pUR4 in pathological vascular leakage treatment.


Subject(s)
Fibronectins/antagonists & inhibitors , Integrin beta1/metabolism , Peptide Fragments/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Adhesion/drug effects , Cell Line , Cell Membrane Permeability/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Humans , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL