Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Mol Ther ; 29(1): 347-364, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33007201

ABSTRACT

Follistatin-like 1 (FSTL1) is a matricellular protein that is upregulated during development and disease, including idiopathic pulmonary fibrosis (IPF), keloid, and arthritis. The profibrotic and pro-inflammatory roles of FSTL1 have been intensively studied during the last several years, as well as in this report. We screened and identified epitope-specific monoclonal neutralizing antibodies (nAbs) to functionally block FSTL1. FSTL1 nAbs attenuated bleomycin-induced pulmonary and dermal fibrosis in vivo and transforming growth factor (TGF)-ß1-induced dermal fibrosis ex vivo in human skin. In addition, FSTL1 nAbs significantly reduced existing lung fibrosis and skin fibrosis in experimental models. FSTL1 nAbs exerted their potent antifibrotic effects via reduced TGF-ß1 responsiveness and subsequent myofibroblast activation and extracellular matrix production. We also observed that FSTL1 nAbs attenuated the severity of collagen-induced arthritis in mice, which was accompanied by reduced inflammatory responses in vitro. Our findings suggest that FSTL1 nAbs are a promising new therapeutic strategy for the treatment of multiple organ fibrosis and systemic autoimmune diseases.


Subject(s)
Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Biomarkers , Disease Susceptibility , Follistatin-Related Proteins/metabolism , Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/metabolism , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Disease Models, Animal , Drug Discovery , Fibrosis , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/genetics , Gene Expression , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Mice , Molecular Targeted Therapy , Transforming Growth Factor beta1/metabolism
2.
Cell Commun Signal ; 18(1): 151, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32933544

ABSTRACT

BACKGROUND: Hepatic fibrosis is a pathological response of the liver to a variety of chronic stimuli. Hepatic stellate cells (HSCs) are the major source of myofibroblasts in the liver. Follistatin like 1 (Fstl1) is a secreted glycoprotein induced by transforming growth factor-ß1 (TGF-ß1). However, the precise functions and regulation mechanisms of Fstl1 in liver fibrogenesis remains unclear. METHODS: Hepatic stellate cell (HSC) line LX-2 stimulated by TGF-ß1, primary culture of mouse HSCs and a model of liver fibrosis induced by CCl4 in mice was used to assess the effect of Fstl1 in vitro and in vivo. RESULTS: Here, we found that Fstl1 was significantly up regulated in human and mouse fibrotic livers, as well as activated HSCs. Haplodeficiency of Fstl1 or blockage of Fstl1 with a neutralizing antibody 22B6 attenuated CCl4-induced liver fibrosis in vivo. Fstl1 modulates TGF-ß1 classic Samd2 and non-classic JNK signaling pathways. Knockdown of Fstl1 in HSCs significantly ameliorated cell activation, cell migration, chemokines C-C Motif Chemokine Ligand 2 (CCL2) and C-X-C Motif Chemokine Ligand 8 (CXCL8) secretion and extracellular matrix (ECM) production, and also modulated microRNA-29a (miR29a) expression. Furthermore, we identified that Fstl1 was a target gene of miR29a. And TGF-ß1 induction of Fstl1 expression was partially through down regulation of miR29a in HSCs. CONCLUSIONS: Our data suggests TGF-ß1-miR29a-Fstl1 regulatory circuit plays a key role in regulation the HSC activation and ECM production, and targeting Fstl1 may be a strategy for the treatment of liver fibrosis. Video Abstract.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Follistatin-Related Proteins/antagonists & inhibitors , Liver Cirrhosis/therapy , MicroRNAs/genetics , Transforming Growth Factor beta1/metabolism , Animals , Carbon Tetrachloride , Cells, Cultured , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Gene Expression Regulation/drug effects , Haploidy , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/metabolism
3.
Mol Pharmacol ; 97(2): 90-101, 2020 02.
Article in English | MEDLINE | ID: mdl-31757861

ABSTRACT

Myocardial infarction is a frequent cardiovascular event and a major cause for cardiomyocyte loss. In adult mammals, cardiomyocytes are traditionally considered to be terminally differentiated cells, unable to proliferate. Therefore, the wound-healing response in the infarct area typically yields scar tissue rather than newly formed cardiomyocytes. In the last decade, several lines of evidence have challenged the lack of proliferative capacity of the differentiated cardiomyocyte: studies in zebrafish and neonatal mammals have convincingly demonstrated the regenerative capacity of cardiomyocytes. Moreover, multiple signaling pathways have been identified in these models that-when activated in adult mammalian cardiomyocytes-can reactivate the cell cycle in these cells. However, cardiomyocytes frequently exit the cell cycle before symmetric division into daughter cells, leading to polyploidy and multinucleation. Now that there is more insight into the reactivation of the cell cycle machinery, other prerequisites for successful symmetric division of cardiomyocytes, such as the control of sarcomere disassembly to allow cytokinesis, require more investigation. This review aims to discuss the signaling pathways involved in cardiomyocyte proliferation, with a specific focus on wingless/int-1 protein signaling. Comparing the conflicting results from in vitro and in vivo studies on this pathway illustrates that the interaction with other cells and structures around the infarct is likely to be essential to determine the outcome of these interventions. The extensive crosstalk with other pathways implicated in cardiomyocyte proliferation calls for the identification of nodal points in the cell signaling before cardiomyocyte proliferation can be moved forward toward clinical application as a cure of cardiac disease. SIGNIFICANCE STATEMENT: Evidence is mounting that proliferation of pre-existing cardiomyocytes can be stimulated to repair injury of the heart. In this review article, an overview is provided of the different signaling pathways implicated in cardiomyocyte proliferation with emphasis on wingless/int-1 protein signaling, crosstalk between the pathways, and controversial results obtained in vitro and in vivo.


Subject(s)
Cardiovascular Agents/pharmacology , Cell Proliferation/radiation effects , Cicatrix/prevention & control , Myocardial Infarction/drug therapy , Myocytes, Cardiac/drug effects , Wnt Signaling Pathway/drug effects , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Newborn , Cardiovascular Agents/therapeutic use , Cell Cycle/drug effects , Cell Differentiation , Cicatrix/pathology , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/metabolism , Hippo Signaling Pathway , Humans , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Neuregulins/antagonists & inhibitors , Neuregulins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , Sarcomeres/drug effects , Sarcomeres/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Wnt Signaling Pathway/physiology , YAP-Signaling Proteins , Zebrafish
4.
Oncol Rep ; 42(6): 2806-2813, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31578589

ABSTRACT

Gastric cancer is an aggressive disease and a common cause of cancer­associated mortality worldwide. Recent studies have indicated that follistatin­like protein 1 (FSTL­1) is expressed and serves essential roles in tumorigenesis; however, the specific functional mechanism of FSTL­1 in gastric cancer progression remains ambiguous. CellTiter­Glo Luminescent Cell Viability and lactate dehydrogenase assays were used to measure cell survival and cell cytotoxicity, respectively. Cell apoptosis was ascertained using the Cell Death Detection ELISA assay and caspase­3/9 activity kits. Reverse transcription­quantitative polymerase chain reaction and western blotting were used to detect the expression levels of FSTL­1. The present study confirmed that FSTL­1 was highly expressed in gastric cancer cells compared with in control cells. Subsequently, FSTL­1 inhibition by small interfering RNA significantly reduced cancer cell survival and induced cytotoxic effects. In addition, knockdown of FSTL­1 in gastric cancer cells promoted apoptosis by increasing caspase­3 and caspase­9 expression. A decrease in signal transducer and activator of transcription 6 (STAT6) phosphorylation was observed in FSTL­1 knockdown cells, and the results confirmed that STAT6 phosphorylation was essential for FSTL­1 knockdown­induced cell apoptosis of cancer cells. Taken together, these results demonstrated that FSTL­1 knockdown may promote cell apoptosis via the STAT6 signaling pathway; therefore, FSTL1 may be considered a novel diagnostic and therapeutic target for gastric cancer.


Subject(s)
Apoptosis/genetics , Follistatin-Related Proteins/genetics , STAT6 Transcription Factor/genetics , Stomach Neoplasms/genetics , Carcinogenesis/genetics , Caspase 3/genetics , Caspase 9/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Enzyme-Linked Immunosorbent Assay , Follistatin-Related Proteins/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , RNA, Small Interfering/genetics , Signal Transduction/genetics , Stomach Neoplasms/pathology
5.
Cell Rep ; 24(7): 1790-1801, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30110636

ABSTRACT

Immune dysfunction is a strong factor in the resistance of cancer to treatment. Blocking immune checkpoint pathways is a promising approach to improve anti-tumor immunity, but the clinical efficacies are still limited. We previously identified follistatin-like 1 (FSTL1) as a determinant of immune dysfunction mediated by mesenchymal stromal/stem cells (MSCs) and immunoregulatory cells. Here, we demonstrate that blocking FSTL1 but not immune checkpoint pathways significantly suppresses cancer progression and metastasis in several mouse tumor models with increased MSCs. Expression of DIP2A (the receptor of FSTL1) in tumor cells is critical for FSTL1-induced immunoresistance. FSTL1/DIP2A co-positivity in tumor tissues correlates with poor prognosis in NSCLC patients. Thus, breaking the FSTL1-DIP2A axis may be a useful strategy for successfully inducing anti-tumor immunity.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carrier Proteins/genetics , Follistatin-Related Proteins/genetics , Gene Expression Regulation, Neoplastic , Immunity, Innate , Lung Neoplasms/genetics , Nuclear Proteins/genetics , Animals , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/mortality , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/immunology , Case-Control Studies , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/immunology , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Male , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/immunology , Prognosis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Tumor Burden , Xenograft Model Antitumor Assays
6.
Cell Physiol Biochem ; 44(4): 1616-1628, 2017.
Article in English | MEDLINE | ID: mdl-29212066

ABSTRACT

BACKGROUND: Gliomas result in the highest morbidity and mortality rates of intracranial primary central nervous system tumors because of their aggressive growth characteristics and high postoperative recurrence. They are characterized by genetic instability, intratumoral histopathological variability and unpredictable clinical behavior in patients. Proliferation is a key aspect of the clinical progression of malignant gliomas, complicating complete surgical resection and enabling tumor regrowth and further proliferation of the surviving tumor cells. METHODS: The expression of Fstl1 was detected by western blotting and qRT-PCR. We used cell proliferation and colony formation assays to measure proliferation. Then, flow cytometry was used to analyze cell cycle progression. The expression of Fstl1, p-Smad1/5/8 and p21 in GBM tissue sections was evaluated using immunohistochemical staining. Furthermore, we used coimmunoprecipitation (Co-IP) and immunoprecipitation to validate the relationship between Fstl1, BMP4 and BMPR2. Finally, we used orthotopic xenograft studies to measure the growth of tumors in vivo. RESULTS: We found that follistatin-like 1 (Fstl1) was upregulated in high-grade glioma specimens and that its levels correlated with poor prognosis. Fstl1 upregulation increased cell proliferation, colony formation and cell cycle progression, while its knockdown inhibited these processes. Moreover, Fstl1 interacted with bone morphogenetic protein (BMP) 4, but not BMP receptor (BMPR) 2, and competitively inhibited their association. Furthermore, Fstl1 overexpression suppressed the activation of the BMP4/Smad1/5/8 signaling pathway, while BMP4 overexpression reversed this effect. CONCLUSION: Our study demonstrated that Fstl1 promoted glioma growth through the BMP4/Smad1/5/8 signaling pathway, and these findings suggest potential new glioblastoma treatment strategies.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Brain Neoplasms/pathology , Follistatin-Related Proteins/metabolism , Glioma/pathology , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type II/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Cell Line, Tumor , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/genetics , Glioma/metabolism , Glioma/mortality , Humans , Immunoprecipitation , Kaplan-Meier Estimate , Mice , Phosphorylation , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , S Phase Cell Cycle Checkpoints , Signal Transduction , Transplantation, Heterologous
7.
Mol Med Rep ; 16(5): 7119-7123, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28901425

ABSTRACT

Follistatin­like 1 (Fstl1) is a secreted glycoprotein that belongs to the follistatin and SPARC (secreted protein, acidic and rich in cysteine) families and was identified to serve a critical role in lung fibrosis. However, the role of Fstl1 in liver fibrosis remains undefined. Therefore, the aim of the present study was to investigate the role of Fstl1 in liver fibrosis. The results indicated that Fstl1 was highly expressed in human hepatic fibrosis tissues and activated hepatic stellate cells (HSCs). Furthermore, knockdown of Fstl1effectively suppressed HSC proliferation and the protein expression levels of α­SMA and collagen I in transforming growth factor (TGF)­ß1­treated HSCs. Mechanistically, knockdown of Fstl1 remarkably decreased the phosphorylation level of Smad3 in TGF­ß1­induced HSCs. Taken together, the present study demonstrated that Fstl1serves an important role in liver fibrosis and target deletion of Fstl1 attenuated HSCs activation through suppressing TGF­ß1/Smad3 signaling pathway. Therefore, Fstl1 may be a potential therapeutic target for the treatment of liver fibrosis.


Subject(s)
Follistatin-Related Proteins/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Actins/metabolism , Cell Proliferation , Cells, Cultured , Collagen Type I/metabolism , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/genetics , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects
8.
Sci Rep ; 7: 45820, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28361925

ABSTRACT

Pulmonary hypertension (PH) remains a life-limiting disease characterized by pulmonary vascular remodelling due to aberrant proliferation and migration of pulmonary artery smooth muscle cells (PASMCs), thus leading to raised pulmonary arterial pressure and right ventricular hypertrophy. Secreted glycoprotein follistatin-like 1 (FSTL1) has been reported to ameliorate tissue remodelling in cardiovascular injuries. However, the role of FSTL1 in deranged pulmonary arteries remains elusive. We found that there were higher serum levels of FSTL1 in patients with PH related to chronic obstructive pulmonary diseases (COPD) and in mice model of hypoxia-induced PH (HPH). Haploinsufficiency of Fstl1 in mice contributed to an exacerbated HPH, as demonstrated by increased right ventricular systolic pressure, pulmonary arterial muscularization and right ventricular hypertrophy index. Conversely, FSTL1 administration attenuated HPH. In cultured human PASMCs, hypoxia-promoted cellular viability, DNA synthesis and migration were suppressed by exogenous FSTL1 but enhanced by small interfering RNA targeting FSTL1. Additionally, FSTL1 inhibited the proliferation and migration of PASMCs via extracellular regulated kinase (ERK) signal pathway. All these findings indicate that FSTL1 imposed a protective modulation on pulmonary vascular remodelling, thereby suggesting its role in the regulation of HPH.


Subject(s)
Follistatin-Related Proteins/genetics , Hypertension, Pulmonary/drug therapy , Hypoxia/drug therapy , Pulmonary Disease, Chronic Obstructive/genetics , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Follistatin-Related Proteins/administration & dosage , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/blood , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Hypoxia/blood , Hypoxia/complications , Hypoxia/pathology , Mice , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/drug therapy , RNA, Small Interfering/administration & dosage
9.
J Exp Med ; 212(2): 235-52, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25584011

ABSTRACT

Progressive tissue fibrosis is a cause of major morbidity and mortality. Pulmonary fibrosis is an epithelial-mesenchymal disorder in which TGF-ß1 plays a central role in pathogenesis. Here we show that follistatin-like 1 (FSTL1) differentially regulates TGF-ß and bone morphogenetic protein signaling, leading to epithelial injury and fibroblast activation. Haplodeletion of Fstl1 in mice or blockage of FSTL1 with a neutralizing antibody in mice reduced bleomycin-induced fibrosis in vivo. Fstl1 is induced in response to lung injury and promotes the accumulation of myofibroblasts and subsequent fibrosis. These data suggest that Fstl1 may serve as a novel therapeutic target for treatment of progressive lung fibrosis.


Subject(s)
Follistatin-Related Proteins/genetics , Pulmonary Fibrosis/genetics , Aged , Animals , Antibiotics, Antineoplastic/adverse effects , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bleomycin/adverse effects , Bone Morphogenetic Protein 4/metabolism , Cell Communication , Cell Line , Disease Models, Animal , Female , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/deficiency , Follistatin-Related Proteins/metabolism , Gene Expression , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lung Injury/chemically induced , Lung Injury/genetics , Lung Injury/metabolism , Lung Injury/pathology , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Middle Aged , Myofibroblasts/immunology , Myofibroblasts/metabolism , Phenotype , Protein Binding , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism
10.
Cardiovasc Res ; 103(1): 111-20, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24743592

ABSTRACT

AIMS: It is well-established that exercise diminishes cardiovascular risk, but whether humoral factors secreted by muscle confer these benefits has not been conclusively shown. We have shown that the secreted protein follistatin-like 1 (Fstl1) has beneficial actions on cardiac and endothelial function. However, the role of muscle-derived Fstl1 in proliferative vascular disease remains largely unknown. Here, we investigated whether muscle-derived Fstl1 modulates vascular remodelling in response to injury. METHODS AND RESULTS: The targeted ablation of Fstl1 in muscle led to an increase in neointimal formation following wire-induced arterial injury compared with control mice. Conversely, muscle-specific Fstl1 transgenic (TG) mice displayed a decrease in the neointimal thickening following arterial injury. Muscle-specific Fstl1 ablation and overexpression increased and decreased, respectively, the frequency of BrdU-positive proliferating cells in injured vessels. In cultured human aortic smooth muscle cells (HASMCs), treatment with human FSTL1 protein decreased proliferation and migration induced by stimulation with PDGF-BB. Treatment with FSTL1 enhanced AMPK phosphorylation, and inhibition of AMPK abrogated the inhibitory actions of FSTL1 on HASMC responses to PDGF-BB. The injured arteries of Fstl1-TG mice exhibited an increase in AMPK phosphorylation, and administration of AMPK inhibitor reversed the anti-proliferative actions of Fstl1 on the vessel wall. CONCLUSION: Our findings indicate that muscle-derived Fstl1 attenuates neointimal formation in response to arterial injury by suppressing SMC proliferation through an AMPK-dependent mechanism. Thus, the release of protein factors from muscle, such as Fstl1, may partly explain why the maintenance of muscle function can have a therapeutic effect on the cardiovascular system.


Subject(s)
Femoral Artery/injuries , Follistatin-Related Proteins/physiology , Neointima/pathology , Neointima/physiopathology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Becaplermin , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Femoral Artery/pathology , Femoral Artery/physiopathology , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/genetics , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/physiology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/physiology , Neointima/prevention & control , Proto-Oncogene Proteins c-sis/metabolism
11.
Cancer Res ; 73(20): 6185-93, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23966294

ABSTRACT

Bone metastasis greatly deteriorates the quality of life in patients with cancer. Although mechanisms have been widely investigated, the relationship between cancer bone metastasis and antitumor immunity in the host has been much less studied. Here, we report a novel mechanism of bone metastasis mediated by FSTL1, a follistatin-like glycoprotein secreted by Snail(+) tumor cells, which metastasize frequently to bone. We found that FSTL1 plays a dual role in bone metastasis-in one way by mediating tumor cell invasion and bone tropism but also in a second way by expanding a population of pluripotent mesenchymal stem-like CD45(-)ALCAM(+) cells derived from bone marrow. CD45(-)ALCAM(+) cells induced bone metastasis de novo, but they also generated CD8(low) T cells with weak CTL activity in the periphery, which also promoted bone metastasis in an indirect manner. RNA interference-mediated attenuation of FSTL1 in tumor cells prevented bone metastasis along with the parallel increase in ALCAM(+) cells and CD8(low) T cells. These effects were accompanied by heightened antitumor immune responses in vitro and in vivo. In clinical specimens of advanced breast cancer, ALCAM(+) cells increased with FSTL1 positivity in tumor tissues, but not in adjacent normal tissues, consistent with a causal connection between these molecules. Our findings define FSTL1 as an attractive candidate therapeutic target to prevent or treat bone metastasis, which remains a major challenge in patients with cancer.


Subject(s)
Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/immunology , Melanoma, Experimental/prevention & control , Animals , Bone Marrow Cells/pathology , Bone Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Flow Cytometry , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transfection
12.
Int J Cardiol ; 167(3): 698-703, 2013 Aug 10.
Article in English | MEDLINE | ID: mdl-22464481

ABSTRACT

BACKGROUND: Although skeletal muscle atrophy and changes in myosin heavy chain (MyHC) isoforms have often been observed during heart failure, their pathophysiological mechanisms are not completely defined. In this study we tested the hypothesis that skeletal muscle phenotype changes are related to myogenic regulatory factors and myostatin/follistatin expression in spontaneously hypertensive rats (SHR) with heart failure. METHODS: After developing tachypnea, SHR were subjected to transthoracic echocardiogram. Pathological evidence of heart failure was assessed during euthanasia. Age-matched Wistar-Kyoto (WKY) rats were used as controls. Soleus muscle morphometry was analyzed in histological sections, and MyHC isoforms evaluated by electrophoresis. Protein levels were assessed by Western blotting. STATISTICAL ANALYSIS: Student'st test and Pearson correlation. RESULTS: All SHR presented right ventricular hypertrophy and seven had pleuropericardial effusion. Echocardiographic evaluation showed dilation in the left chambers and left ventricular hypertrophy with systolic and diastolic dysfunction in SHR. Soleus weight and fiber cross sectional areas were lower (WKY 3615 ± 412; SHR 2035 ± 224 µm(2); P<0.001), and collagen fractional volume was higher in SHR. The relative amount of type I MyHC isoform was increased in SHR. Myogenin, myostatin, and follistatin expression was lower and MRF4 levels higher in SHR. Myogenin and follistatin expression positively correlated with fiber cross sectional areas and MRF4 levels positively correlated with I MyHC isoform. CONCLUSION: Reduced myogenin and follistatin expression seems to participate in muscle atrophy while increased MRF4 protein levels can modulate myosin heavy chain isoform shift in skeletal muscle of spontaneously hypertensive rats with heart failure.


Subject(s)
Heart Failure/pathology , Muscle, Skeletal/pathology , Muscular Diseases/metabolism , Myogenic Regulatory Factors/antagonists & inhibitors , Myogenic Regulatory Factors/biosynthesis , Animals , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/biosynthesis , Heart Failure/genetics , Heart Failure/metabolism , Male , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Diseases/genetics , Muscular Diseases/pathology , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Species Specificity
13.
FEBS Lett ; 586(4): 319-24, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22265692

ABSTRACT

Follistatin-related protein (FRP)/follistatin-like 1 (FSTL1) has multi-specific binding nature especially with TGF-ß superfamily proteins, and thereby modulates organ development. However, its function in immune systems remains unclear. Previously, we reported FRP interacts with CD14, which is known to mediate toll-like receptor 4 (TLR4) signaling. Here, we investigated whether FRP activates TLR4 signaling. Recombinant FRP induced interleukin 6 or interleukin 8 production from target cells in a CD14- and TLR4-dependent manner. Moreover, similar to lipopolysaccharide (LPS), FRP induced tolerance to the second LPS stimulation. FRP has the function of evoking innate immune responses as one of the endogenous TLR4 agonists.


Subject(s)
Follistatin-Related Proteins/immunology , Immunity, Innate , Lipopolysaccharide Receptors/metabolism , Toll-Like Receptor 4/metabolism , Animals , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/pharmacology , Gene Knockdown Techniques , HEK293 Cells , Humans , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , NIH 3T3 Cells , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Signal Transduction , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...