Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Int J Mol Sci ; 21(21)2020 10 22.
Article in English | MEDLINE | ID: mdl-33105556

ABSTRACT

CCN1 (cysteine-rich 61, connective tissue growth factor, and nephroblastoma-1), previously named CYR61 (cysteine-rich angiogenic inducer 61) belongs to the CCN family of matricellular proteins. CCN1 plays critical roles in the regulation of proliferation, differentiation, apoptosis, angiogenesis, and fibrosis. Recent studies have extensively characterized the important physiological and pathological roles of CCN1 in various tissues and organs. In this review, we summarize both basic and clinical aspects of CCN1 in pulmonary diseases, including acute lung injury (ALI), chronic obstructive pulmonary disease (COPD), lung fibrosis, pulmonary arterial hypertension (PAH), lung infection, and lung cancer. We also emphasize the important challenges for future investigations to better understand the CCN1 and its role in physiology and pathology, as well as the questions that need to be addressed for the therapeutic development of CCN1 antagonists in various lung diseases.


Subject(s)
Cysteine-Rich Protein 61/physiology , Lung Diseases/etiology , Acute Lung Injury/etiology , Bronchopulmonary Dysplasia/etiology , Humans , Lung Neoplasms/etiology , Pulmonary Disease, Chronic Obstructive/etiology
2.
J Bone Miner Res ; 35(11): 2289-2300, 2020 11.
Article in English | MEDLINE | ID: mdl-32634285

ABSTRACT

CCN1/Cyr61 is a dynamically expressed matricellular protein that serves regulatory functions in multiple tissues. Previous studies from our laboratory demonstrated that CCN1 regulates bone maintenance. Using an osteoblast and osteocyte conditional knockout mouse model (Ccn1OCN ), we found a significant decrease in trabecular and cortical bone mass in vivo, in part through suppression of Wnt signaling since the expression of the Wnt antagonist sclerostin (SOST) is increased in osteoblasts lacking CCN1. It has been established that parathyroid hormone (PTH) signaling also suppresses SOST expression in bone. We therefore investigated the interaction between CCN1 and PTH-mediated responses in this study. We find that loss of Ccn1 in osteoblasts leads to impaired responsiveness to anabolic intermittent PTH treatment in Ccn1OCN mice in vivo and in osteoblasts from these mice in vitro. Analysis of Ccn1OCN mice demonstrated a significant decrease in parathyroid hormone receptor-1 (PTH1R) expression in osteoblasts in vivo and in vitro. We investigated the regulatory role of a non-canonical integrin-binding domain of CCN1 because several studies indicate that specific integrins are critical to mechanotransduction, a PTH-dependent response, in bone. These data suggest that CCN1 regulates the expression of PTH1R through interaction with the αvß3 and/or αvß5 integrin complexes. Osteoblasts that express a mutant form of CCN1 that cannot interact with αvß3/ß5 integrin demonstrate a significant decrease in mRNA and protein expression of both PTH1R and αv integrin. Overall, these data suggest that the αvß3/ß5-binding domain of CCN1 is required to endow PTH signaling with anabolic activity in bone cells. © 2020 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Cysteine-Rich Protein 61/physiology , Mechanotransduction, Cellular , Osteoblasts/cytology , Parathyroid Hormone , Animals , Mice , Osteoblasts/drug effects , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1 , Wnt Signaling Pathway
3.
Proc Natl Acad Sci U S A ; 113(33): E4847-56, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27482114

ABSTRACT

Inflammation is characterized by the recruitment of leukocytes from the bloodstream. The rapid arrival of neutrophils is followed by a wave of inflammatory lymphocyte antigen 6 complex (Ly6C)-positive monocytes. In contrast Ly6C(low) monocytes survey the endothelium in the steady state, but their role in inflammation is still unclear. Here, using confocal intravital microscopy, we show that upon Toll-like receptor 7/8 (TLR7/8)-mediated inflammation of mesenteric veins, platelet activation drives the rapid mobilization of Ly6C(low) monocytes to the luminal side of the endothelium. After repeatedly interacting with platelets, Ly6C(low) monocytes commit to a meticulous patrolling of the endothelial wall and orchestrate the subsequent arrival and extravasation of neutrophils through the production of proinflammatory cytokines and chemokines. At a molecular level, we show that cysteine-rich protein 61 (CYR61)/CYR61 connective tissue growth factor nephroblastoma overexpressed 1 (CCN1) protein is released by activated platelets and enables the recruitment of Ly6C(low) monocytes upon vascular inflammation. In addition endothelium-bound CCN1 sustains the adequate patrolling of Ly6C(low) monocytes both in the steady state and under inflammatory conditions. Blocking CCN1 or platelets with specific antibodies impaired the early arrival of Ly6C(low) monocytes and abolished the recruitment of neutrophils. These results refine the leukocyte recruitment cascade model by introducing endothelium-bound CCN1 as an inflammation mediator and by demonstrating a role for platelets and patrolling Ly6C(low) monocytes in acute vascular inflammation.


Subject(s)
Antigens, Ly/analysis , Cysteine-Rich Protein 61/physiology , Monocytes/physiology , Vasculitis/etiology , Animals , Blood Platelets/physiology , Cell Movement , Mice , Mice, Inbred C57BL , Neutrophils/physiology , Toll-Like Receptor 7/physiology , Toll-Like Receptor 8/physiology
5.
Brain Tumor Pathol ; 32(4): 245-52, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26201842

ABSTRACT

Recently, research efforts in identifying prognostic molecular biomarkers for malignant glioma have intensified. Cysteine-rich protein 61 (CCN1) is one of the CCN family of matricellular proteins that promotes cell growth and angiogenesis in cancers through its interaction with several integrins. In this study, we investigated the relationships among CCN1, O(6)-methylguanine-DNA methyltransferase expression, the tumor removal rate, and prognosis in 46 glioblastoma patients treated at the Okayama University Hospital. CCN1 expression was high in 31 (67 %) of these patients. The median progression-free survival (PFS) and overall survival (OS) times of patients with high CCN1 expression was significantly shorter than those of patients with low CCN1 expression (p < 0.005). In a multivariate Cox analysis, CCN1 proved to be an independent prognostic factor for patient survival [PFS, hazard ratio (HR) = 3.53 (1.55-8.01), p = 0.003 and OS, HR = 3.05 (1.35-6.87), p = 0.007]. Moreover, in the 31 patients who underwent gross total resection, the PFS and OS times of those with high CCN1 expression were significantly shorter than those with low CCN1 expression. It was concluded that CCN1 might emerge as a significant prognostic factor regarding the prognosis of glioblastoma patients.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Cysteine-Rich Protein 61/analysis , Glioblastoma/diagnosis , Glioblastoma/genetics , Adult , Aged , Aged, 80 and over , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/physiology , Female , Gene Expression , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Immunohistochemistry , Male , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/analysis , O(6)-Methylguanine-DNA Methyltransferase/genetics , Prognosis , Proportional Hazards Models , Survival Rate , Young Adult
6.
J Clin Invest ; 125(5): 1886-900, 2015 May.
Article in English | MEDLINE | ID: mdl-25822023

ABSTRACT

Liver cholestatic diseases, which stem from diverse etiologies, result in liver toxicity and fibrosis and may progress to cirrhosis and liver failure. We show that CCN1 (also known as CYR61), a matricellular protein that dampens and resolves liver fibrosis, also mediates cholangiocyte proliferation and ductular reaction, which are repair responses to cholestatic injury. In cholangiocytes, CCN1 activated NF-κB through integrin αvß5/αvß3, leading to Jag1 expression, JAG1/NOTCH signaling, and cholangiocyte proliferation. CCN1 also induced Jag1 expression in hepatic stellate cells, whereupon they interacted with hepatic progenitor cells to promote their differentiation into cholangiocytes. Administration of CCN1 protein or soluble JAG1 induced cholangiocyte proliferation in mice, which was blocked by inhibitors of NF-κB or NOTCH signaling. Knock-in mice expressing a CCN1 mutant that is unable to bind αvß5/αvß3 were impaired in ductular reaction, leading to massive hepatic necrosis and mortality after bile duct ligation (BDL), whereas treatment of these mice with soluble JAG1 rescued ductular reaction and reduced hepatic necrosis and mortality. Blockade of integrin αvß5/αvß3, NF-κB, or NOTCH signaling in WT mice also resulted in defective ductular reaction after BDL. These findings demonstrate that CCN1 induces cholangiocyte proliferation and ductular reaction and identify CCN1/αvß5/NF-κB/JAG1 as a critical axis for biliary injury repair.


Subject(s)
Bile Ducts/metabolism , Cysteine-Rich Protein 61/physiology , Liver/metabolism , NF-kappa B/metabolism , Receptors, Vitronectin/physiology , Animals , Bile Ducts/physiology , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/pharmacology , Calcium-Binding Proteins/therapeutic use , Cell Division , Cells, Cultured , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/metabolism , Cholestasis, Extrahepatic/pathology , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/pharmacology , Gene Expression Regulation , Gene Knock-In Techniques , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Humans , Integrin alphaVbeta3 , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/therapeutic use , Jagged-1 Protein , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Membrane Proteins/therapeutic use , Mice , Mice, Inbred C57BL , RNA Interference , Receptors, Notch/physiology , Recombinant Fusion Proteins/metabolism , Regeneration , Serrate-Jagged Proteins
7.
J Biol Chem ; 290(13): 8232-42, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25623072

ABSTRACT

Platelet-derived growth factor (PDGF), a potent chemoattractant, induces cell migration via the MAPK and PI3K/Akt pathways. However, the downstream mediators are still elusive. In particular, the role of extracellular mediators is largely unknown. In this study, we identified the matricellular protein Cyr61, which is de novo synthesized in response to PDGF stimulation, as the key downstream mediator of the ERK and JNK pathways, independent of the p38 MAPK and AKT pathways, and, thereby, it mediates PDGF-induced smooth muscle cell migration but not proliferation. Our results revealed that, when Cyr61 was newly synthesized by PDGF, it was promptly translocated to the extracellular matrix and physically interacted with the plasma membrane integrins α6ß1 and αvß3. We further demonstrate that Cyr61 and integrins are integral components of the PDGF signaling pathway via an "outside-in" signaling route to activate intracellular focal adhesion kinase (FAK), leading to cell migration. Therefore, this study provides the first evidence that the PDGF-induced endogenous extracellular matrix component Cyr61 is a key mediator in modulating cell migration by connecting intracellular PDGF-ERK and JNK signals with integrin/FAK signaling. Therefore, extracellular Cyr61 convergence with growth factor signaling and integrin/FAK signaling is a new concept of growth factor-induced cell migration. The discovered signaling pathway may represent an important therapeutic target in growth factor-mediated cell migration/invasion-related vascular diseases and tumorigenesis.


Subject(s)
Cell Movement , Cysteine-Rich Protein 61/physiology , Proto-Oncogene Proteins c-sis/physiology , Animals , Becaplermin , Cell Proliferation , Cells, Cultured , Enzyme Activation , Extracellular Matrix/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrins/metabolism , MAP Kinase Signaling System , Mice , Myocytes, Smooth Muscle/physiology , Transcriptional Activation
8.
Cell Signal ; 26(6): 1326-34, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24631528

ABSTRACT

Cell proliferation from pre-existing cardiomyocytes is a major source of cells for normal mammalian myocardial renewal or for regeneration after myocardial injury. These proliferative cardiomyocytes may act differently from the postmitotic cardiomyocytes in a stressed heart. Extracellular matrix molecule CCN1 is produced to promote Fas ligand (FasL)-induced cardiomyocyte apoptosis in mice with stress-induced cardiac injury. We aimed to investigate the effect of CCN1 on the proliferative cardiomyocytes. We used rat embryonic cardiomyoblast H9c2 cells to study the cardiotoxicity of CCN1. We found that FasL dose-dependently increased the X-linked inhibitor of apoptosis protein (XIAP) levels to prevent the progression of apoptosis in H9c2 cells. CCN1, though it did not induce apoptosis by itself, sensitized H9c2 cells to FasL-induced apoptosis. CCN1 functions by engaging its cell-surface receptor integrin α6ß1 and elevating reactive oxygen species levels, which leads to mitogen-activated protein kinase p38 activation, cytosolic Bax translocation to mitochondria, and the release of mitochondrial Smac and HtrA2 to cytosol. These elevated cytosolic Smac and HtrA2 dismantle the inhibition of XIAP, thereby facilitating the activation of caspase-3 and the apoptosis-induced by FasL. In summary, we demonstrated a novel mechanism underlying the resistance of cardiomyoblasts to FasL-induced apoptosis, and the pro-apoptotic function of CCN1 by disrupting this resistance.


Subject(s)
Apoptosis , Cysteine-Rich Protein 61/physiology , Fas Ligand Protein/physiology , Myoblasts, Cardiac/physiology , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Apoptosis Regulatory Proteins , Carrier Proteins/metabolism , Cell Line , Integrin alpha6beta1/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Transport , RNA-Binding Proteins/metabolism , Rats , Reactive Oxygen Species/metabolism , Serine-Arginine Splicing Factors , bcl-2-Associated X Protein/metabolism , fas Receptor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Biochim Biophys Acta ; 1843(5): 902-14, 2014 May.
Article in English | MEDLINE | ID: mdl-24487063

ABSTRACT

UNLABELLED: Cysteine-rich protein 61 (CCN1/CYR61) is a CCN (CYR61, CTGF (connective tissue growth factor), and NOV (Nephroblastoma overexpressed gene)) family matricellular protein comprising six secreted CCN proteins in mammals. CCN1/CYR61 expression is associated with inflammation and injury repair. Recent studies show that CCN1/CYR61 limits fibrosis in models of cutaneous wound healing by inducing cellular senescence in myofibroblasts of the granulation tissue which thereby transforms into an extracellular matrix-degrading phenotype. We here investigate CCN1/CYR61 expression in primary profibrogenic liver cells (i.e., hepatic stellate cells and periportal myofibroblasts) and found an increase of CCN1/CYR61 expression during early activation of hepatic stellate cells that declines in fully transdifferentiated myofibroblasts. By contrast, CCN1/CYR61 levels found in primary parenchymal liver cells (i.e., hepatocytes) were relatively low compared to the levels exhibited in hepatic stellate cells and portal myofibroblasts. In models of ongoing liver fibrogenesis, elevated levels of CCN1/CYR61 were particularly noticed during early periods of insult, while expression declined during prolonged phases of fibrogenesis. We generated an adenovirus type 5 encoding CCN1/CYR61 (i.e., Ad5-CMV-CCN1/CYR61) and overexpressed CCN1/CYR61 in primary portal myofibroblasts. Interestingly, overexpressed CCN1/CYR61 significantly inhibited production of collagen type I at both mRNA and protein levels as evidenced by quantitative real-time polymerase chain reaction, Western blot and immunocytochemistry. CCN1/CYR61 further induces production of reactive oxygen species (ROS) leading to dose-dependent cellular senescence and apoptosis. Additionally, we demonstrate that CCN1/CYR61 attenuates TGF-ß signaling by scavenging TGF-ß thereby mitigating in vivo liver fibrogenesis in a bile duct ligation model. CONCLUSION: In line with dermal fibrosis and scar formation, CCN1/CYR61 is involved in liver injury repair and tissue remodeling. CCN1/CYR61 gene transfer into extracellular matrix-producing liver cells is therefore potentially beneficial in liver fibrotic therapy.


Subject(s)
Apoptosis , Cellular Senescence , Cysteine-Rich Protein 61/physiology , Myofibroblasts/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley
10.
Arthritis Rheumatol ; 66(1): 49-59, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24449575

ABSTRACT

OBJECTIVE: We previously showed that Cyr61 acts to promote fibroblast-like synoviocyte (FLS) proliferation and Th17 cell differentiation, suggesting that Cyr61 plays an important role in mediating the joint inflammation and damage in rheumatoid arthritis (RA). The aim of this study was to investigate whether Cyr61 expression is regulated at the posttranscription level, and if so, how this regulation connects to other etiologic factors in RA. METHODS: Expression of microRNA-22 (miR-22) in synovial tissue was detected by real-time polymerase chain reaction (PCR) using miRNA-specific TaqMan MGB probes. MicroRNA-22 promoter activity was analyzed using a Dual-Luciferase Reporter Assay. Cytokine expression was measured by enzyme-linked immunosorbent assay, and the expression of other factors was measured by real-time PCR or Western blotting. RESULTS: MicroRNA-22 directly targeted the 3'-untranslated region of Cyr61 messenger RNA and inhibited Cyr61 expression. Expression of miR-22 was down-regulated and was negatively correlated with Cyr61 expression in RA synovial tissue. Furthermore, wild-type p53 activated miR-22 transcription by binding to the promoter region of the miR-22 gene, while the mutant forms of p53 frequently found in RA synovial tissue were shown to have lost the ability to activate miR-22 expression. As a result, miR-22 was down-regulated, contributing to the overexpression of Cyr61 in RA FLS. CONCLUSION: Our results not only reveal a novel mechanism whereby p53 is involved in the posttranscriptional regulation of Cyr61 expression via miRNA-22, but also provide a molecular explanation for the role of somatic mutations of p53, which are frequently observed in RA synovial tissue, in the etiology of this autoimmune disease.


Subject(s)
Arthritis, Rheumatoid/genetics , Cysteine-Rich Protein 61/genetics , Gene Expression Regulation/physiology , MicroRNAs/genetics , RNA, Messenger/physiology , Signal Transduction/physiology , Synovial Membrane/metabolism , Tumor Suppressor Protein p53/physiology , 3' Untranslated Regions/genetics , Adult , Aged , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/physiopathology , Cell Line , Cysteine-Rich Protein 61/physiology , Cytokines/metabolism , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts , HCT116 Cells , HeLa Cells , Humans , Male , MicroRNAs/physiology , Middle Aged , Real-Time Polymerase Chain Reaction
11.
Age (Dordr) ; 36(1): 353-64, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23881607

ABSTRACT

Chronic exposure of human skin to solar ultraviolet (UV) irradiation causes premature skin aging, which is characterized by reduced type I collagen production and increased fragmentation of the dermal collagenous extracellular matrix. This imbalance of collagen homeostasis is mediated, in part, by elevated expression of the matricellular protein cysteine-rich protein 61 (CCN1), in dermal fibroblasts, the primary collagen producing cell type in human skin. Here, we report that the actions of CCN1 are mediated by induction of interleukin 1ß (IL-1ß). CCN1 and IL-1ß are strikingly induced by acute UV irradiation, and constitutively elevated in sun-exposed prematurely aged human skin. Elevated CCN1 rapidly induces IL-1ß, inhibits type I collagen production, and upregulates matrix metalloproteinase-1, which degrades collagen fibrils. Blockade of IL-1ß actions by IL-1 receptor antagonist largely prevents the deleterious effects of CCN1 on collagen homeostasis. Furthermore, knockdown of CCN1 significantly reduces induction of IL-1ß by UV irradiation, and thereby partially prevents collagen loss. These data demonstrate that elevated CCN1promotes inflammaging and collagen loss via induction of IL-1ß and thereby contributes to the pathophysiology of premature aging in chronically sun-exposed human skin.


Subject(s)
Cysteine-Rich Protein 61/physiology , Interleukin-1beta/metabolism , Skin Aging/radiation effects , Sunlight/adverse effects , Adult , Blotting, Western , Cells, Cultured , Collagen Type I/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Matrix Metalloproteinase 1/metabolism , Microscopy, Atomic Force , Middle Aged , Real-Time Polymerase Chain Reaction , Transfection , Up-Regulation
12.
Mol Cell Biol ; 33(10): 2078-90, 2013 May.
Article in English | MEDLINE | ID: mdl-23508104

ABSTRACT

Liver fibrosis occurs as a wound-healing response to chronic hepatic injuries irrespective of the underlying etiology and may progress to life-threatening cirrhosis. Here we show that CCN1, a matricellular protein of the CCN (CYR61/CTGF/NOV) family, is accumulated in hepatocytes of human cirrhotic livers. CCN1 is not required for liver development or regeneration, since these processes are normal in mice with hepatocyte-specific Ccn1 deletion. However, Ccn1 expression is upregulated upon liver injuries and functions to inhibit liver fibrogenesis induced by either carbon tetrachloride intoxication or bile duct ligation and promote fibrosis regression. CCN1 acts by triggering cellular senescence in activated hepatic stellate cells and portal fibroblasts by engaging integrin α6ß1 to induce reactive oxygen species accumulation through the RAC1-NADPH oxidase 1 enzyme complex, whereupon the senescent cells express an antifibrosis genetic program. Mice with hepatocyte-specific Ccn1 deletion suffer exacerbated fibrosis with a concomitant deficit in cellular senescence, whereas overexpression of hepatic Ccn1 reduces liver fibrosis with enhanced senescence. Furthermore, tail vein delivery of purified CCN1 protein accelerates fibrosis regression in mice with established fibrosis. These findings reveal a novel integrin-dependent mechanism of fibrosis resolution in chronic liver injury and identify the CCN1 signaling pathway as a potential target for therapeutic intervention.


Subject(s)
Cellular Senescence , Cysteine-Rich Protein 61/metabolism , Liver Cirrhosis/metabolism , Myofibroblasts/metabolism , Animals , Binding Sites , Carbon Tetrachloride , Cells, Cultured , Cholestasis/complications , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/physiology , Female , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/physiology , Humans , Integrin alpha6beta1/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Myofibroblasts/physiology , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , Neuropeptides/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tissue Array Analysis , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
14.
J Biol Chem ; 288(12): 8544-8559, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23362279

ABSTRACT

The shape of the dendritic arbor is one of the criteria of neuron classification and reflects functional specialization of particular classes of neurons. The development of a proper dendritic branching pattern strongly relies on interactions between the extracellular environment and intracellular processes responsible for dendrite growth and stability. We previously showed that mammalian target of rapamycin (mTOR) kinase is crucial for this process. In this work, we performed a screen for modifiers of dendritic growth in hippocampal neurons, the expression of which is potentially regulated by mTOR. As a result, we identified Cyr61, an angiogenic factor with unknown neuronal function, as a novel regulator of dendritic growth, which controls dendritic growth in a ß1-integrin-dependent manner.


Subject(s)
Cysteine-Rich Protein 61/physiology , Dendrites/physiology , Extracellular Matrix/metabolism , Hippocampus/cytology , Neurons/physiology , Animals , Cell Shape , Cells, Cultured , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Gene Knockdown Techniques , Genes, Immediate-Early , Hippocampus/metabolism , Insulin/physiology , Integrin beta1/metabolism , Integrin beta1/physiology , Phosphatidylinositol 3-Kinases/metabolism , RNA, Small Interfering/genetics , Rats , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , ras Proteins/metabolism , ras Proteins/physiology
15.
Cardiovasc Res ; 98(1): 64-72, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23329650

ABSTRACT

AIMS: Expression of extracellular matrix protein CCN1 is induced in end-stage ischaemic cardiomyopathy in humans, and after cardiac ischaemia and reperfusion in experimental animal models. Despite its well-documented angiogenic activities, CCN1 increases the cytotoxicities of the tumour necrosis factor family cytokines, which promotes apoptosis in fibroblasts. We aimed to determine the physiological function of CCN1 in an injured heart. METHODS AND RESULTS: To assess the function of CCN1 in vivo, knock-in mice carrying the apoptosis-defective mutant allele Ccn1-dm were tested in an isoproterenol (ISO)-induced myocardial injury model (100 mg/kg/day of sc injected ISO for 5 days). Compared with wild-type mice, Ccn1(dm/dm) mice were remarkably resistant to ISO-induced cardiac injury; they showed no post-treatment cardiomyocyte apoptosis or myocardial tissue damage. ISO cardiotoxicity was dependent on Fas ligand (FasL) and its downstream signalling. Using primary cultures of cardiomyocytes isolated from rats, we demonstrated that CCN1 sensitized FasL-mediated apoptosis by engaging its cell-surface receptor integrin α6ß1 and up-regulating intracellular reactive oxygen species (ROS), which activated mitogen-activated protein kinase p38, and increased cell-surface Fas expression. CONCLUSION: CCN1 is a critical pathophysiological regulator that mediates cardiomyocyte apoptosis during work-overload-induced cardiac injury. CCN1 increases cellular susceptibility to Fas-induced apoptosis by increasing ROS and cell-surface Fas expression.


Subject(s)
Apoptosis , Cysteine-Rich Protein 61/physiology , Isoproterenol/toxicity , Myocytes, Cardiac/pathology , Animals , Fas Ligand Protein/physiology , Integrin alpha6beta1/physiology , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/physiology
16.
Am J Physiol Lung Cell Mol Physiol ; 304(6): L415-27, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23316072

ABSTRACT

Repair of the lung epithelium after injury is integral to the pathogenesis and outcomes of diverse inflammatory lung diseases. We previously reported that ß-catenin signaling promotes epithelial repair after inflammatory injury, but the ß-catenin target genes that mediate this effect are unknown. Herein, we examined which ß-catenin transcriptional coactivators and target genes promote epithelial repair after inflammatory injury. Transmigration of human neutrophils across cultured monolayers of human lung epithelial cells resulted in a fall in transepithelial resistance and the formation of discrete areas of epithelial denudation ("microinjury"), which repaired via cell spreading by 96 h. In mice treated with intratracheal (i.t.) LPS or keratinocyte chemokine, neutrophil emigration was associated with increased permeability of the lung epithelium, as determined by increased bronchoalveolar lavage (BAL) fluid albumin concentration, which decreased over 3-6 days. Activation of ß-catenin/p300-dependent gene expression using the compound ICG-001 accelerated epithelial repair in vitro and in murine models. Neutrophil transmigration induced epithelial expression of the ß-catenin/p300 target genes Wnt-induced secreted protein (WISP) 1 and cysteine-rich (Cyr) 61, as determined by real-time PCR (qPCR) and immunostaining. Purified neutrophil elastase induced WISP1 upregulation in lung epithelial cells, as determined by qPCR. WISP1 expression increased in murine lungs after i.t. LPS, as determined by ELISA of the BAL fluid and qPCR of whole lung extracts. Finally, recombinant WISP1 and Cyr61 accelerated repair, and Cyr61-neutralizing antibodies delayed repair of the injured epithelium in vitro. We conclude that ß-catenin/p300-dependent expression of WISP1 and Cyr61 is critical for epithelial repair and represents a potential therapeutic target to promote epithelial repair after inflammatory injury.


Subject(s)
Acute Lung Injury/metabolism , CCN Intercellular Signaling Proteins/physiology , Cysteine-Rich Protein 61/physiology , Proto-Oncogene Proteins/physiology , Respiratory Mucosa/metabolism , Transendothelial and Transepithelial Migration , beta Catenin/physiology , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , CCN Intercellular Signaling Proteins/genetics , CCN Intercellular Signaling Proteins/metabolism , Cells, Cultured , Coculture Techniques , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/physiology , Female , Gene Expression , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Neutrophils/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Respiratory Mucosa/immunology , Signal Transduction , beta Catenin/metabolism
17.
J Biol Chem ; 287(46): 38569-79, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23027863

ABSTRACT

CCN1 is a matricellular protein and a member of the CCN family of growth factors. CCN1 is associated with the development of various cancers including pancreatic ductal adenocarcinoma (PDAC). Our recent studies found that CCN1 plays a critical role in pancreatic carcinogenesis through the induction of EMT and stemness. CCN1 mRNA and protein were detected in the early precursor lesions, and their expression intensified with disease progression. However, biochemical activity and the molecular targets of CCN1 in pancreatic cancer cells are unknown. Here we show that CCN1 regulates the Sonic Hedgehog (SHh) signaling pathway, which is associated with the PDAC progression and poor prognosis. SHh regulation by CCN1 in pancreatic cancer cells is mediated through the active Notch-1. Notably, active Notch-1is recruited by CCN1 in these cells via the inhibition of proteasomal degradation results in stabilization of the receptor. We find that CCN1-induced activation of SHh signaling might be necessary for CCN1-dependent in vitro pancreatic cancer cell migration and tumorigenicity of the side population of pancreatic cancer cells (cancer stem cells) in a xenograft in nude mice. Moreover, the functional role of CCN1 could be mediated through the interaction with the αvß3 integrin receptor. These extensive studies propose that targeting CCN1 can provide a new treatment option for patients with pancreatic cancer since blocking CCN1 simultaneously blocks two critical pathways (i.e. SHh and Notch1) associated with the development of the disease as well as drug resistance.


Subject(s)
Carcinoma/metabolism , Cysteine-Rich Protein 61/physiology , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Pancreatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cysteine-Rich Protein 61/chemistry , Disease Progression , Drug Resistance, Neoplasm , Humans , Integrins/metabolism , Male , Mice , Mice, Nude , Models, Biological , Neoplasm Transplantation , Proteasome Endopeptidase Complex/metabolism , Receptors, Notch/metabolism , Signal Transduction
18.
Int J Gynecol Pathol ; 31(5): 405-15, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22833079

ABSTRACT

Members of the CCN [cystein-rich 61 (Cyr61)/connective tissue growth factor (CTGF)/nephroblastoma (NOV)] protein family are involved in the regulation of cellular proliferation, apoptosis, and migration and are also assumed to play a role in carcinogenesis. Therefore, we performed a retrospective study to investigate the immunohistochemical expression of both Cyr61 and CTGF in 92 borderline tumors (BOTs) and 107 invasive carcinomas of the ovary (IOCs). To determine their diagnostic and prognostic value, we correlated protein expression with clinicopathologic factors including overall and disease-free survival. Cyr61 and CTGF were found to be inversely expressed in both BOTs and IOCs, with a stronger expression of Cyr61 in IOCs. Moreover, Cyr61 was found to be preferentially expressed in high-grade serous carcinomas, whereas CTGF was found more frequently in low-grade serous carcinomas. Weak Cyr61 levels correlated with both low estrogen receptor and p53 expression (P=0.038, P=0.04, respectively). However, no association was observed between CTGF, estrogen receptor, and p53 expression levels in IOCs. Regarding prognosis, Cyr61 was found to be of no value, but the loss of CTGF was found to be associated with a poor prognosis in multivariate analysis of overall (relative risk 2.8; P=0.050) and disease-free (relative risk 2.3; P=0.031) survival. Cyr61 and CTGF are inversely expressed in BOTs and IOCs, and loss of CTGF independently indicates poor prognosis in IOCs.


Subject(s)
Connective Tissue Growth Factor/analysis , Cysteine-Rich Protein 61/analysis , Neoplasms, Glandular and Epithelial/chemistry , Ovarian Neoplasms/chemistry , Adult , Aged , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Connective Tissue Growth Factor/physiology , Cysteine-Rich Protein 61/physiology , Fallopian Tubes/chemistry , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasms, Glandular and Epithelial/mortality , Ovarian Neoplasms/mortality , Ovary/chemistry , Prognosis , Retrospective Studies
19.
Placenta ; 33(7): 561-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22551568

ABSTRACT

OBJECTIVE: To investigate the function and mechanism of CYR61 on the migration and invasion of the trophoblast cell line, HTR-8/SVneo cells. STUDY DESIGN: The mRNA and protein levels of NUR77 in the placentas of normal and preeclampsia (PE) women were evaluated using real-time PCR and Western blot, respectively. Paraffin-embedded tissues were processed for localization of NUR77 protein in placental villus by immunohistochemistry. HTR-8/SVneo cells were cultured in the presence of CYR61, Ad-NUR77 or a small interfering RNA for NUR77 (Ad-sinur77). The expression of NUR77 in the HTR-8/SVneo cells was detected and the effects of CYR61 on the migration and invasion of HTR-8/SVneo cells were assessed in wound-healing and transwell experiments, respectively. Gelatin zymography was used to measure the MMP2 release in HTR-8/SVneo cells. RESULTS: NUR77 is significantly decreased in the placenta of women with PE compared with the levels during a normal pregnancy. CYR61 can significantly increase the expression of NUR77 in HTR-8/SVneo cells. CYR61, as well as NUR77, can promote HTR-8/SVneo cells migration and invasion, which can be blocked by Ad-sinur77. Both CYR61 and Ad-nur77 reduced the mRNA expression of TIMP2 in HTR-8/SVneo cells. CONCLUSIONS: CYR61 may promote HTR-8/SVneo cells migration and invasion through the upregulation of NUR77, leading to the increase of MMP2 release and the downregulation of TIMP2 expression.


Subject(s)
Cell Movement/physiology , Cysteine-Rich Protein 61/physiology , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Tissue Inhibitor of Metalloproteinase-2/genetics , Trophoblasts/cytology , Cell Line , Cysteine-Rich Protein 61/analysis , Cysteine-Rich Protein 61/pharmacology , Down-Regulation , Female , Gene Expression Regulation/physiology , Humans , Matrix Metalloproteinase 2/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/analysis , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Placenta/chemistry , Placenta/cytology , Pre-Eclampsia/etiology , Pre-Eclampsia/metabolism , Pregnancy , RNA, Messenger/analysis , RNA, Small Interfering/pharmacology , Trophoblasts/drug effects , Trophoblasts/physiology
20.
J Immunol ; 188(11): 5776-84, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22547695

ABSTRACT

Cysteine-rich protein 61 (Cyr61)/CCN1 is a product of an immediate early gene and functions in mediating cell adhesion and inducing cell migration. We previously showed that increased production of Cyr61 by fibroblast-like synoviocytes (FLS) in rheumatoid arthritis (RA) promotes FLS proliferation and participates in RA pathogenesis with the IL-17-dependent pathway. However, whether Cyr61 in turn regulates Th17 cell differentiation and further enhances inflammation of RA remained unknown. In the current study, we explored the potential role of Cyr61 as a proinflammatory factor in RA pathogenesis. We found that Cyr61 treatment dramatically induced IL-6 production in FLS isolated from RA patients. Moreover, IL-6 production was attenuated by Cyr61 knockdown in FLS. Mechanistically, we showed that Cyr61 activated IL-6 production via the αvß5/Akt/NF-κB signaling pathway. Further, using a coculture system consisting of purified CD4(+) T cells and RA FLS, we found that RA FLS stimulated Th17 differentiation, and the pro-Th17 differentiation effect of RA FLS can be attenuated or stimulated by Cyr61 RNA interference or addition of exogenous Cyr61, respectively. Finally, using the collagen-induced arthritis animal model, we showed that treatment with the anti-Cyr61 mAb led to reduction of IL-6 levels, decrease of Th17 response, and attenuation of inflammation and disease progression in vivo. Taken together, our results reveal a novel role of Cyr61 in promoting Th17 development in RA via upregulation of IL-6 production by FLS, thus adding a new layer into the functional interplay between FLS and Th17 in RA pathogenesis. Our study also suggests that targeting of Cyr61 may represent a novel strategy in RA treatment.


Subject(s)
Arthritis, Rheumatoid/immunology , Cell Differentiation/immunology , Cysteine-Rich Protein 61/physiology , Fibroblasts/immunology , Interleukin-6/biosynthesis , Synovial Membrane/immunology , Th17 Cells/immunology , Adult , Aged , Animals , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cells, Cultured , Coculture Techniques , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Interleukin-6/physiology , Male , Mice , Mice, Inbred DBA , Middle Aged , Receptors, Vitronectin/physiology , Signal Transduction/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Th17 Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...