Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Int J Mol Sci ; 22(12)2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34205668

ABSTRACT

Matricellular proteins (MCPs) are defined as extracellular matrix (ECM) associated proteins that are important regulators and integrators of microenvironmental signals, contributing to the dynamic nature of ECM signalling. There is a growing understanding of the role of matricellular proteins in cellular processes governing tissue development as well as in disease pathogenesis. In this review, the expression and functions of different MP family members (periostin, CCNs, TSPs, SIBLINGs and others) are presented, specifically in relation to craniofacial development and the maintenance of orofacial tissues, including bone, gingiva, oral mucosa, palate and the dental pulp. As will be discussed, each MP family member has been shown to have non-redundant roles in development, tissue homeostasis, wound healing, pathology and tumorigenesis of orofacial and dental tissues.


Subject(s)
Cell Adhesion Molecules/physiology , Extracellular Matrix Proteins/physiology , Mouth/growth & development , Osteonectin/physiology , Thrombospondins/physiology , Animals , CCN Intercellular Signaling Proteins/physiology , Head and Neck Neoplasms/etiology , Humans , Mouth/embryology , Tenascin/physiology , Wound Healing
2.
DNA Cell Biol ; 40(4): 629-637, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33646053

ABSTRACT

Our study aimed at validating the effect of WISP1 on osteoarthritis (OA) and the pathway involved in the WISP1-induced protection against OA. The expression of WISP1 was measured by immunohistochemical analyses. We found that WISP1 expression was shown to be upregulated within human OA cartilage compared with controls. WISP1 expression was related to knee OA severity. rhWISP1 inhibited OA chondrocyte senescence and apoptosis in vitro, which was reversed by the αvß3 antibody and PI3K/Akt inhibitor LY294002. WISP1 overexpression induced by knee injection of LiCI could also prevent the senescence and apoptosis of rat chondrocytes. Safranin-O staining and Mankin score revealed that WISP1 overexpression can protect rat chondrocytes from degeneration. Nearly opposite results were obtained in the treatment of ICG-001 and siRNA-WISP1 in vivo. These data strongly suggest that WISP1 can protect against the senescence and apoptosis of chondrocytes via modulating the αvß3 receptor and PI3K/Akt signaling pathway within OA. Therefore, the development of specific activators of WISP1 may present the value of an underlying OA treatment.


Subject(s)
CCN Intercellular Signaling Proteins/metabolism , Chondrocytes/metabolism , Osteoarthritis, Knee/metabolism , Proto-Oncogene Proteins/metabolism , Adult , Aging/physiology , Animals , Apoptosis/physiology , CCN Intercellular Signaling Proteins/physiology , Cartilage, Articular/metabolism , Female , Humans , Integrin alphaVbeta3/metabolism , Interleukin-1beta/metabolism , Male , MicroRNAs/metabolism , Middle Aged , Osteoarthritis/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
3.
Shock ; 56(3): 461-472, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33394970

ABSTRACT

ABSTRACT: IL-33 and WNT1-inducible secreted protein (WISP1) play central roles in acute lung injury (ALI) induced by mechanical ventilation with moderate tidal volume (MTV) in the setting of sepsis. Here, we sought to determine the inter-relationship between IL-33 and WISP1 and the associated signaling pathways in this process.We used a two-hit model of cecal ligation puncture (CLP) followed by MTV ventilation (4 h 10 mL/kg) in wild-type, IL-33-/- or ST2-/- mice or wild-type mice treated with intratracheal antibodies to WISP1. Macrophages (Raw 264.7 and alveolar macrophages from wild-type or ST2-/- mice) were used to identify specific signaling components.CLP + MTV resulted in ALI that was partially sensitive to genetic ablation of IL-33 or ST2 or antibody neutralization of WISP1. Genetic ablation of IL-33 or ST2 significantly prevented ALI after CLP + MTV and reduced levels of WISP1 in the circulation and bronchoalveolar lung fluid. rIL-33 increased WISP1 in alveolar macrophages in an ST2, PI3K/AKT, and ERK dependent manner. This WISP1 upregulation and WNT ß-catenin activation were sensitive to inhibition of the ß-catenin/TCF/CBP/P300 nuclear pathway.We show that IL-33 drives WISP1 upregulation and ALI during MTV in CLP sepsis. The identification of this relationship and the associated signaling pathways reveals a number of possible therapeutic targets to prevent ALI in ventilated sepsis patients.


Subject(s)
CCN Intercellular Signaling Proteins/physiology , Interleukin-33/physiology , Proto-Oncogene Proteins/physiology , Respiration, Artificial/adverse effects , Sepsis/complications , Tidal Volume/physiology , Ventilator-Induced Lung Injury/etiology , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Sepsis/therapy , Signal Transduction/physiology , Ventilator-Induced Lung Injury/metabolism
4.
Anim Reprod Sci ; 207: 44-51, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31208847

ABSTRACT

The number of Sertoli cells has a major effect on adult testis size and sperm production capacity. Mechanisms that regulate the number of Sertoli cells in livestock are at best nebulously understood; however, with lesser testicular estrogen production, proliferation of Sertoli cells is prolonged compared with vehicle-treated littermates. Decreased WISP2 gene expression in testes as a result of less endogenous estrogen is similar to altered WISP2 gene expression following corticosteroid treatment of some cultured cells. Taken together, these findings indicate decreased testicular cortisol might be in the signaling pathway between reduced endogenous estrogens and the prolonged interval of Sertoli cell proliferation. Hence, in these studies, potential actions of testicular corticosteroid on Sertoli cell numbers were evaluated. Testicular cortisol concentrations were reduced at 6.5 weeks of age (P < 0.05) in littermates treated with the aromatase inhibitor, letrozole, compared with littermates treated with vehicle. Letrozole treatment leads to reduced testicular estradiol and greater Sertoli cell numbers during the early juvenile interval in pigs. The inverse relationship between testicular glucocorticoid and Sertoli cell proliferation was also tested by increasing local testicular glucocorticoids using the synthetic compound, dexamethasone. Local administration beginning at 1.5 weeks of age (osmotic pump and catheter (n = 3) or a silastic implant (n = 5)) reduced Sertoli cell numbers at 6.5 weeks of age compared with littermates that received the vehicle treatment (P< 0.05). In summary, testicular glucocorticoid concentration was inversely correlated with Sertoli cell numbers during the first wave of Sertoli cell proliferation.


Subject(s)
CCN Intercellular Signaling Proteins/physiology , Cell Proliferation/drug effects , Estrogens/pharmacology , Hydrocortisone/physiology , Sertoli Cells/drug effects , Testis/metabolism , Animals , CCN Intercellular Signaling Proteins/genetics , Dexamethasone/pharmacology , Fulvestrant/pharmacology , Gene Expression/drug effects , Hydrocortisone/metabolism , Letrozole/pharmacology , Male , Sertoli Cells/physiology , Sex Differentiation/drug effects , Sex Differentiation/genetics , Spermatozoa/drug effects , Spermatozoa/physiology , Swine
5.
Breast Cancer Res Treat ; 172(3): 577-586, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30220054

ABSTRACT

PURPOSE: Metaplastic breast carcinomas are an aggressive subtype of triple-negative breast cancer (TNBC) in which part or all of the adenocarcinoma transforms into a non-glandular component (e.g., spindled, squamous, or heterologous). We discovered that mammary-specific Ccn6/Wisp3 knockout mice develop mammary carcinomas with spindle and squamous differentiation that share upregulation of the oncofetal proteins IGF2BP2 (IMP2) and HMGA2 with human metaplastic carcinomas. Here, we investigated the functional relationship between CCN6, IGF2BP2, and HMGA2 proteins in vitro and in vivo, and their expression in human tissue samples. METHODS: MMTV-cre;Ccn6fl/fl tumors and spindle TNBC cell lines were treated with recombinant CCN6 protein or vehicle. IGF2BP2 was downregulated using shRNAs in HME cells with stable CCN6 shRNA knockdown, and subjected to invasion and adhesion assays. Thirty-one human metaplastic carcinomas were arrayed in a tissue microarray (TMA) and immunostained for CCN6, IGF2BP2, and HMGA2. RESULTS: CCN6 regulates IGF2BP2 and HMGA2 protein expression in MMTV-cre;Ccn6fl/fl tumors, in MDA-MB-231 and - 468, and in HME cells. CCN6 recombinant protein reduced IGF2BP2 and HMGA2 protein expression, and decreased growth of MMTV-cre;Ccn6fl/fl tumors in vivo. IGF2BP2 shRNA knockdown was sufficient to reverse the invasive abilities conferred by CCN6 knockdown in HME cells. Analyses of the TCGA Breast Cancer Cohort (n = 1238) showed that IGF2BP2 and HMGA2 are significantly upregulated in metaplastic carcinoma compared to other breast cancer subtypes. In clinical samples, low CCN6 is frequent in tumors with high IGF2BP2/HMGA2 with spindle and squamous differentiation. CONCLUSIONS: These data shed light into the pathogenesis of metaplastic carcinoma and demonstrate a novel CCN6/IGF2BP2/HMGA2 oncogenic pathway with biomarker and therapeutic implications.


Subject(s)
CCN Intercellular Signaling Proteins/physiology , HMGA2 Protein/physiology , RNA-Binding Proteins/physiology , Signal Transduction/physiology , Triple Negative Breast Neoplasms/pathology , Animals , Cell Adhesion , Cell Line, Tumor , Female , HMGA2 Protein/analysis , Humans , Mice , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , RNA-Binding Proteins/analysis , Triple Negative Breast Neoplasms/therapy
6.
Cell Signal ; 51: 72-85, 2018 11.
Article in English | MEDLINE | ID: mdl-30044965

ABSTRACT

Portal fibroblasts are mesenchyme-derived fibroblasts surrounding the bile ducts, and activated into portal myofibroblasts (pMF) during cholestatic liver injury. pMF express α-smooth muscle actin (α-SMA) and produce the fibrogenic extracellular matrix (ECM) collagen type I and fibronectin, playing important roles in portal fibrosis. A cholestatic bile duct-ligated (BDL) model is characterized by impaired hepatobiliary excretion of bile, leading to increased bile acid accumulation. Accumulation of bile acids is known to induce endoplasmic reticulum (ER) stress leading to liver damage and cell death. Additionally, a BDL fibrotic model is also associated with upregulation of CCN (CYR61, CTGF and NOV) matricellular proteins and reported to induce ER stress both in vitro and in vivo. To explore the effects of CCN proteins, we used adenovirus-mediated CCN1-4 (Ad-CCN1-4) gene transfers into cultured pMF. Overexpression of CCN proteins leads to protein accumulation in the ER lumen, causing ER stress and unfolded protein response (UPR). We further found ER stress and UPR to mitigate fibrogenesis in pMF by decreased cellular production of fibronectin, collagen type 1 and α-SMA. In this scenario, Tauroursodeoxycholic acid, a pharmaceutical chaperone and ER stress inhibitor, attenuated Ad-CCN1-4 induced pMF apoptosis and restored collagen and fibronectin levels. Since hepatic fibrogenesis is accompanied by ER stress and upregulation of CCN proteins in a BDL, we further evaluated ER stress responses after Ad-CCN1 gene transfer in such a model and found overexpressed CCN1 to enhance the ER stress-associated proteins BiP and CHOP with positive cleaved caspase 3 and 9 staining in periportal nonparenchymal cells. This indicates that these nonparenchymal cells, most likely pMF, have the tendency to undergo apoptosis during later stages of BDL. Ad-CCN1 transduction furthermore sensitized pMF for ER stress and apoptosis. We suggest that CCN proteins are key factors in the fibrotic microenvironment impacting pMF survival during fibrogenesis and pMF apoptosis during fibrosis resolution.


Subject(s)
Bile Ducts, Intrahepatic/metabolism , CCN Intercellular Signaling Proteins/physiology , Cholestasis/metabolism , Endoplasmic Reticulum/metabolism , Myofibroblasts/metabolism , Actins/metabolism , Animals , Apoptosis , Bile Ducts, Intrahepatic/pathology , Caspase 3/metabolism , Caspase 9/metabolism , Cells, Cultured , Cholestasis/pathology , Collagen Type I/metabolism , Endoplasmic Reticulum Stress , Fibronectins/metabolism , Fibrosis , Heat-Shock Proteins/metabolism , Male , Mice , Myofibroblasts/cytology , Rats , Rats, Sprague-Dawley , Signal Transduction , Transcription Factor CHOP/metabolism , Unfolded Protein Response
7.
J Matern Fetal Neonatal Med ; 30(8): 942-946, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27267804

ABSTRACT

OBJECTIVE: To investigate Wnt1-inducible signaling pathway protein-1 (WISP1) levels and their correlation with metabolic parameters in pregnant women with gestational diabetes mellitus (GDM) and non-GDM healthy pregnant women. MATERIALS AND METHODS: In this prospective cross-sectional study, the study group was composed of 62 women with GDM and 73 healthy pregnant women matched for age, body mass index (BMI) and gestational age. Blood samples were collected at 25-29th gestational week. Serum WISP1, betatrophin, glucose, fasting insulin, glycosylated hemoglobin A1c, total cholesterol, triglyceride, high density lipoprotein cholesterol, low density lipoprotein cholesterol, C reactive protein, alanine aminotransferase and creatinine levels were measured. Homeostasis model assessment of insulin resistance (HOMA-IR) values was calculated. The level of significance was accepted as p < 0.05. RESULTS: Circulating WISP1 in the GDM group was significantly higher than the control group (p <0.001). Further, WISP1 was positively correlated with BMI, HOMA-IR values and fasting glucose, fasting insulin, triglyceride, betatrophin levels. BMI, HOMA-IR and betatrophin independently and positively predicted WISP1 levels. CONCLUSION: These results demonstrate a relationship between WISP1 and the metabolic parameters of GDM. And, WISP1 might be involved in the pathophysiology of GDM. As a part of this pathophysiological mechanism, the activation of WISP1 and betatrophin might take place through several ways; WISP1 and betatrophin might either use same signaling pathways and potentiate each other or they might also constitute the sequential steps of a common pathway.


Subject(s)
Adipokines/physiology , CCN Intercellular Signaling Proteins/physiology , Diabetes, Gestational/metabolism , Proto-Oncogene Proteins/physiology , Adipokines/blood , Adult , Biomarkers/blood , Blood Glucose/metabolism , C-Reactive Protein/analysis , CCN Intercellular Signaling Proteins/blood , Case-Control Studies , Cholesterol/blood , Cross-Sectional Studies , Female , Glucose Tolerance Test , Humans , Insulin/blood , Pregnancy , Proto-Oncogene Proteins/blood , Triglycerides/blood
8.
Metabolism ; 65(2): 30-40, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26773927

ABSTRACT

OBJECTIVE: Wnt signaling pathway significantly participates in cardiac fibrosis and CFs activation. Therefore, we reviewed current evidence on the new perspectives and biological association between Wnt signaling pathway and cardiac fibrosis. DESIGN AND METHODS: A PubMed database search was performed for studies of Wnt signaling pathway in cardiac fibrosis and CFs activation. RESULTS: Numerous studies have shown that the Wnt signaling pathway significantly participates in cardiac fibrosis pathogenesis. The aim of this review is to describe the present knowledge about the Wnt signaling pathway significantly participating in cardiac fibrosis and CFs activation, and look ahead on new perspectives of Wnt signaling pathway research. Moreover, we will discuss the different insights that interact with the Wnt signaling pathway-regulated cardiac fibrosis. The Wnt proteins are glycoproteins that bind to the Fz receptors on the cell surface, which lead to several important biological functions, such as cell differentiation and proliferation. There are several signals among the characterized pathways of cardiac fibrosis, including Wnt/ß-catenin signaling. In this review, new insight into the Wnt signaling pathway in cardiac fibrosis pathogenesis is discussed, with special emphasis on Wnt/ß-catenin. CONCLUSION: It seems reasonable to suggest the potential targets of Wnt signaling pathway and it can be developed as a therapeutic target for cardiac fibrosis.


Subject(s)
Myocardium/pathology , Wnt Signaling Pathway/physiology , CCN Intercellular Signaling Proteins/physiology , Calcium Signaling , Fibrosis , Glycoproteins/physiology , Humans , Intracellular Signaling Peptides and Proteins , Proto-Oncogene Proteins/physiology , beta Catenin/antagonists & inhibitors , beta Catenin/physiology
9.
Rheumatology (Oxford) ; 55(9): 1536-47, 2016 09.
Article in English | MEDLINE | ID: mdl-26667213

ABSTRACT

The Wnt signalling pathway is gaining increasing attention in the field of joint pathologies, attributable to its role in the development and homeostasis of the tissues found in the joint, including bone and cartilage. Imbalance in this pathway has been implicated in the development and progression of OA, and interference with the pathway might therefore depict an effective treatment strategy. Though offering multiple opportunities, it is yet to be decided which starting point will bring forth the most promising results. The complexity of the pathway and its interaction with other pathways (such as the TGF-ß signalling pathway, which also has a central role in the maintenance of joint homeostasis) means that acting directly on proteins in this signalling cascade entails a high risk of undesired side effects. Therefore, interference with Wnt-induced proteins, such as WISP1, might be an overall more effective and safer therapeutic approach to inhibit the pathological events that take place during OA.


Subject(s)
CCN Intercellular Signaling Proteins/physiology , Osteoarthritis/etiology , Proto-Oncogene Proteins/physiology , Transforming Growth Factor beta/physiology , Wnt Proteins/physiology , Wnt Signaling Pathway/physiology , Cartilage Diseases/etiology , Cartilage Diseases/physiopathology , Cell Communication/physiology , Cell Nucleus/physiology , Chondrocytes/physiology , Cytoplasm/physiology , Homeostasis/physiology , Humans , Osteoarthritis/physiopathology , Receptor Cross-Talk/physiology
10.
Cancer ; 121(22): 4088-96, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26349011

ABSTRACT

BACKGROUND: Desmoid tumors (DTs) are rare mesenchymal lesions that can recur repeatedly. When it is feasible, DTs are surgically resected; however, this often results in high recurrence rates. Recently, treatment with PF-03084014, a potent γ-secretase inhibitor, has been shown to have antitumor activity in several tumor types by affecting the WNT/ß-catenin pathway. Consequently, Notch pathway inhibition by PF-03084014 might be a promising approach for DT treatment. METHODS: The expression of Notch pathway components was analyzed in DT tissues and cell strains with immunohistochemistry and Western blotting, respectively. A panel of DT cell strains was exposed to PF-03084014 and evaluated for cell proliferation. Antitumor effects were assessed via cell cycle, apoptosis, and migration and invasion analysis. Cells treated with PF-03084014 were characterized with a gene array analysis combined with Ingenuity Pathway Analysis. RESULTS: The results showed that Notch pathway components were expressed at different levels in DTs. Hes1 (Hes Family BHLH Transcription Factor 1) was overexpressed in DT tumors versus dermal scar tissue, and PF-03084014 caused significant decreases in Notch intracellular domain and Hes1 expression in DT cell strains. PF-03084014 decreased DT cell migration and invasion and also caused cell growth inhibition in DT cell strains, most likely through cell cycle arrest. Gene array analysis combined with Ingenuity Pathway Analysis showed that Wnt1-inducible signaling pathway protein 2 possibly regulated Notch and WNT pathways after treatment with PF-03084014 through integrin. CONCLUSION: Our findings suggest that the Notch pathway is an important DT therapeutic target. Furthermore, PF-03084014 has significant antitumor activity against DTs, and it may be an alternative strategy for DT treatment.


Subject(s)
Fibromatosis, Aggressive/drug therapy , Receptors, Notch/antagonists & inhibitors , Signal Transduction/physiology , CCN Intercellular Signaling Proteins/physiology , Cell Cycle Checkpoints/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Fibromatosis, Aggressive/etiology , Fibromatosis, Aggressive/pathology , Humans , Neoplasm Invasiveness , Receptors, Notch/physiology , Repressor Proteins/physiology , Tetrahydronaphthalenes/pharmacology , Valine/analogs & derivatives , Valine/pharmacology
11.
Hum Genomics ; 9: 24, 2015 Sep 23.
Article in English | MEDLINE | ID: mdl-26395334

ABSTRACT

"CCN" is an acronym referring to the first letter of each of the first three members of this original group of mammalian functionally and phylogenetically distinct extracellular matrix (ECM) proteins [i.e., cysteine-rich 61 (CYR61), connective tissue growth factor (CTGF), and nephroblastoma-overexpressed (NOV)]. Although "CCN" genes are unlikely to have arisen from a common ancestral gene, their encoded proteins share multimodular structures in which most cysteine residues are strictly conserved in their positions within several structural motifs. The CCN genes can be subdivided into members developmentally indispensable for embryonic viability (e.g., CCN1, 2 and 5), each assuming unique tissue-specific functions, and members not essential for embryonic development (e.g., CCN3, 4 and 6), probably due to a balance of functional redundancy and specialization during evolution. The temporo-spatial regulation of the CCN genes and the structural information contained within the sequences of their encoded proteins reflect diversity in their context and tissue-specific functions. Genetic association studies and experimental anomalies, replicated in various animal models, have shown that altered CCN gene structure or expression is associated with "injury" stimuli--whether mechanical (e.g., trauma, shear stress) or chemical (e.g., ischemia, hyperglycemia, hyperlipidemia, inflammation). Consequently, increased organ-specific susceptibility to structural damages ensues. These data underscore the critical functions of CCN proteins in the dynamics of tissue repair and regeneration and in the compensatory responses preceding organ failure. A better understanding of the regulation and mode of action of each CCN member will be useful in developing specific gain- or loss-of-function strategies for therapeutic purposes.


Subject(s)
CCN Intercellular Signaling Proteins/genetics , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Amino Acid Sequence , Animals , CCN Intercellular Signaling Proteins/classification , CCN Intercellular Signaling Proteins/physiology , Disease/etiology , Disease/genetics , Exons , Humans , Introns , Molecular Sequence Data
12.
BMC Musculoskelet Disord ; 16: 218, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26293784

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is a multifactorial degenerative condition of the whole joint with a complex pathogenesis whose development and progression is significantly mediated by interactions between the joint cartilage and articular tissues, particularly, proinflammatory mediators and oxidative stress, which results in cartilage deterioration and subchondral bone destruction. HIF-1 alpha regulates oxygen homeostasis in hypoxic tissues such as joint cartilage; efficiency of transcriptional activity of the HIF1A gene is strongly influenced by the presence of polymorphic variants. Given the loss of articular cartilage and with intention to restore damaged tissue, WISP-1 participates in the development of subchondral bone; further, its expression is highly increased in chondrocytes of OA patients. The aim of this study was to evaluate gene frequencies of HIF1A and WISP1 polymorphisms in Mexican patients suffering from knee OA. METHODS: We determined HIF1A rs11549465 (P582S), rs11549467 (A588T), and rs2057482 (C191T), and WISP1 rs2929970 (A2364G) polymorphisms in 70 Mexican patients with knee OA and compare them to those present in 66 ethnically matched healthy controls. Genotyping for these polymorphisms was performed by Real-Time PCR using TaqMan probes. RESULTS: Gene frequencies exhibited a significant increase of the CC genotype of rs11549465 polymorphism in knee OA patients as compared with those present in controls (P = 0.003 OR = 5.7, 95% CI = 1.7-21.6); CT genotype and T allele showed decreased frequency in the knee OA group vs. the controls (P = 0.003 OR = 0.2, CI = 0.05-0.6; and P = 0.004 OR = 0.2, CI = 0.05-0.65, respectively). Allele frequencies of the other polymorphic variants were similar in both patients and controls. CONCLUSIONS: These results suggest that the presence of the rs11549465 SNP (HIF1A) plays a role protective in the loss of articular cartilage in our population, and offers the possibility to further study the molecular mechanisms within cartilage and subchondral bone.


Subject(s)
Cartilage/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Osteoarthritis, Knee/genetics , Polymorphism, Single Nucleotide , Adult , Aged , Alleles , CCN Intercellular Signaling Proteins/genetics , CCN Intercellular Signaling Proteins/physiology , Case-Control Studies , Female , Gene Frequency , Genetic Predisposition to Disease , Genotype , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mexico/epidemiology , Middle Aged , Osteoarthritis, Knee/ethnology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Single-Blind Method
13.
DNA Cell Biol ; 34(8): 524-33, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26061139

ABSTRACT

The mTOR/S6K1 signaling pathway is the primary regulator of milk protein synthesis. While mTOR is known to be regulated at the translational level by amino acids, the mechanism by which mTOR accepts the amino acid signal is not yet clear. In this study, we describe the discovery of WISP3 as a potentially novel signaling factor that connects mTOR and amino acids. Treatment of dairy cow mammary epithelial cells with amino acids (lysine or methionine) increased both cell growth and the expression of ß-casein (CSN2), WISP3, mTOR, and phospho-mTOR (p-mTOR). Notably, overexpressing WISP3 in these cells also increased both cell growth and the expression of CSN2, mTOR, and p-mTOR and decreased the expression of glycogen synthase kinase 3ß (GSK3ß), while repressing WISP3 had the opposite effect. The increase of the expression of CSN2, mTOR, and p-mTOR mediated by amino acid could be inhibited by repressing WISP3. The increase of the expression of CSN2, mTOR, and p-mTOR mediated by WISP3 overexpression could be inhibited by overexpressing GSK3ß, and vice versa. Taken together, these results reveal that through its amino acid-mediated regulation of the mTOR pathway, WISP3 is an important regulatory factor involved in the amino acid-mediated regulation of milk protein synthesis and cell growth.


Subject(s)
CCN Intercellular Signaling Proteins/physiology , Cell Proliferation/genetics , Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , Milk Proteins/biosynthesis , TOR Serine-Threonine Kinases/physiology , Animals , CCN Intercellular Signaling Proteins/genetics , Cattle , Dairying , Female , Lactation/physiology , Protein Biosynthesis , Signal Transduction
14.
Int J Biochem Cell Biol ; 62: 142-6, 2015 May.
Article in English | MEDLINE | ID: mdl-25794425

ABSTRACT

The matricellular protein WISP1 is a member of the CCN protein family. It is induced by WNT1 and is a downstream target of ß-catenin. WISP1 is expressed during embryonic development, wound healing and tissue repair. Aberrant WISP1 expression is associated with various pathologies including osteoarthritis, fibrosis and cancer. Its role in tumor progression and clinical outcome makes WISP1 an emerging candidate for the detection and treatment of tumors.


Subject(s)
CCN Intercellular Signaling Proteins/physiology , Neoplasms/genetics , Proto-Oncogene Proteins/physiology , Animals , CCN Intercellular Signaling Proteins/chemistry , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/diagnosis , Neoplasms/pathology , Neoplasms/therapy , Prognosis , Proto-Oncogene Proteins/chemistry , Signal Transduction
15.
Oncogene ; 34(24): 3152-63, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25132260

ABSTRACT

The matricellular protein CCN5/WISP-2 represents a promising target in triple-negative breast cancer (TNBC) because treatment or induced activation of CCN5 in TNBC cells promotes cell growth arrest at the G0/G1 phase, reduces cell proliferation and delays tumor growth in the xenograft model. Our studies found that the p27(Kip1) tumor suppressor protein is upregulated and relocalized to the nucleus from cytoplasm by CCN5 in these cells and that these two events (upregulation and relocalization of p27(Kip1)) are critical for CCN5-induced growth inhibition of TNBC cells. In the absence of CCN5, p27(Kip1) resides mostly in the cytoplasm, which is associated with the aggressive nature of cancer cells. Mechanistically, CCN5 inhibits Skp2 expression, which seems to stabilize the p27(Kip1) protein in these cells. On the other hand, CCN5 also recruits FOXO3a to mediate the transcriptional regulation of p27(Kip1). The recruitment of FOXO3a is achieved by the induction of its expression and activity through shifting from cytoplasm to the nucleus. Our data indicate that CCN5 blocks PI3K/AKT signaling to dephosphorylate at S318, S253 and Thr32 in FOXO3a for nuclear relocalization and activation of FOXO3a. Moreover, inhibition of α6ß1 receptors diminishes CCN5 action on p27(Kip1) in TNBC cells. Collectively, these data suggest that CCN5 effectively inhibits TNBC growth through the accumulation and trafficking of p27(Kip1) via Skp2 and FOXO3a regulation, and thus, activation of CCN5 may have the therapeutic potential to kill TNBC.


Subject(s)
CCN Intercellular Signaling Proteins/physiology , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p27 , Forkhead Transcription Factors/physiology , Repressor Proteins/physiology , S-Phase Kinase-Associated Proteins/physiology , Triple Negative Breast Neoplasms/pathology , Animals , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Forkhead Box Protein O3 , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Mice, Nude , Protein Stability , Protein Transport/drug effects , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/genetics , Tumor Cells, Cultured
16.
Arterioscler Thromb Vasc Biol ; 34(11): 2449-56, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25212236

ABSTRACT

OBJECTIVE: Apoptosis of vascular smooth muscle cells (VSMCs) contributes to thinning and rupture of the atherosclerotic plaque fibrous cap and is thereby associated with myocardial infarction. Wnt protein activation of ß-catenin regulates numerous genes that are associated with cell survival. We therefore investigated Wnt/ß-catenin survival signaling in VSMCs and assessed the presence of this pathway in human atherosclerotic plaques at various stages of the disease process. APPROACH AND RESULTS: Wnt5a induced ß-catenin/T-cell factor signaling and retarded oxidative stress (H2O2)-induced apoptosis in mouse aortic VSMCs. Quantification of mRNA levels revealed a >4-fold (P<0.05; n=9) increase in the expression of the Wnt/ß-catenin responsive gene, Wnt1-inducible secreted protein-1 (WISP-1), which was dependent on cAMP response element-binding protein and sustained in the presence of H2O2. Exogenous WISP-1 significantly reduced H2O2-induced apoptosis by 43% (P<0.05; n=3) and was shown using silencing small interfering RNA, to be important for Wnt5a-dependent survival responses to H2O2 (P<0.05; n=3). WISP-1 protein levels were significantly lower (≈50%) in unstable atherosclerosis compared with stable plaques (n=11 and n=14). CONCLUSIONS: These results indicate for the first time that Wnt5a induces ß-catenin survival signaling in VSMCs via WISP-1. The deficiency of the novel survival factor, WISP-1 in intimal VSMCs of unstable coronary plaques, suggests that there is altered Wnt/ß-catenin/ T-cell factor signaling with progressive atherosclerosis, and restoration of WISP-1 protein might be an effective stabilization factor for vulnerable atherosclerotic plaques.


Subject(s)
Apoptosis/drug effects , CCN Intercellular Signaling Proteins/physiology , Muscle, Smooth, Vascular/pathology , Oxidative Stress/physiology , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins/physiology , Wnt Proteins/pharmacology , Animals , Apoptosis/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Humans , Hydrogen Peroxide/pharmacology , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Signal Transduction/drug effects , Signal Transduction/physiology , TCF Transcription Factors/physiology , Wnt Proteins/physiology , Wnt-5a Protein , beta Catenin/physiology
17.
Curr Neurovasc Res ; 11(4): 378-89, 2014.
Article in English | MEDLINE | ID: mdl-25219658

ABSTRACT

As a proliferative and restorative entity, Wnt1 inducible signaling pathway protein 1 (WISP1) is emerging as a novel target for a number of therapeutic strategies that are relevant for disorders such as traumatic injury, neurodegeneration, musculoskeletal disorders, cardiovascular disease, pulmonary compromise, and control of tumor growth as well as distant metastases. WISP1, a target of the wingless pathway Wnt1, oversees cellular mechanisms that include apoptosis, autophagy, cellular migration, stem cell proliferation, angiogenesis, immune cell modulation, and tumorigenesis. The signal transduction pathways of WISP1 are broad and involve phosphoinositide 3-kinase (PI 3-K), protein kinase B (Akt), mitogen activated protein (MAP) kinase, c-Jun N-terminal kinase (JNK), caspases, forkhead transcription factors, sirtuins, c-myc, glycogen synthase kinase -3ß (GSK-3ß), ß-catenin, miRNAs, and the mechanistic target of rapamaycin (mTOR). Ultimately, these signal transduction pathways of WISP1 can result in varied and sometimes unpredictable outcomes especially for cell survival, tissue repair, and tumorigenesis that demand increased insight into the critical role WISP1 holds for cellular biology and clinical medicine.


Subject(s)
CCN Intercellular Signaling Proteins/physiology , Cell Proliferation/physiology , Proto-Oncogene Proteins/metabolism , Recovery of Function/physiology , Animals , Apoptosis/physiology , CCN Intercellular Signaling Proteins/metabolism , Humans , Signal Transduction/physiology
18.
Neural Plast ; 2014: 321209, 2014.
Article in English | MEDLINE | ID: mdl-24551460

ABSTRACT

Matricellular proteins are secreted, nonstructural proteins that regulate the extracellular matrix (ECM) and interactions between cells through modulation of growth factor signaling, cell adhesion, migration, and proliferation. Despite being well described in the context of nonneuronal tissues, recent studies have revealed that these molecules may also play instrumental roles in central nervous system (CNS) development and diseases. In this minireview, we discuss the matricellular protein families SPARC (secreted protein acidic and rich in cysteine), Hevin/SC1 (SPARC-like 1), TN-C (Tenascin C), TSP (Thrombospondin), and CCN (CYR61/CTGF/NOV), which are secreted by astrocytes during development. These proteins exhibit a reduced expression in adult CNS but are upregulated in reactive astrocytes following injury or disease, where they are well placed to modulate the repair processes such as tissue remodeling, axon regeneration, glial scar formation, angiogenesis, and rewiring of neural circuitry. Conversely, their reexpression in reactive astrocytes may also lead to detrimental effects and promote the progression of neurodegenerative diseases.


Subject(s)
Astrocytes/metabolism , Central Nervous System Diseases/pathology , Central Nervous System/growth & development , Central Nervous System/pathology , Extracellular Matrix Proteins/metabolism , Animals , CCN Intercellular Signaling Proteins/metabolism , CCN Intercellular Signaling Proteins/physiology , Calcium-Binding Proteins/metabolism , Central Nervous System/metabolism , Central Nervous System Diseases/metabolism , Extracellular Matrix/physiology , Humans , Osteonectin/metabolism , Tenascin/metabolism , Thrombospondins/metabolism
19.
Cell Mol Life Sci ; 71(12): 2299-311, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24448904

ABSTRACT

The CCN family of proteins consists of six high homologous matricellular proteins which act predominantly by binding to heparin sulphate proteoglycan and a variety of integrins. Interestingly, CCN proteins are regulated by ovarian steroid hormones and are able to adapt to changes in oxygen concentration, which is a necessary condition for successful implantation. CCN1 is involved in processes of angiogenesis within reproductive systems, thereby potentially contributing to diseases such as endometriosis and disturbed angiogenesis in the placenta and fetus. In the ovary, CCN2 is the key factor for follicular development, ovulation and corpora luteal luteolysis, and its deletion leads to fertility defects. CCN1, CCN2 and CCN3 seem to be regulators for human trophoblast proliferation and migration, but with CCN2 acting as a counterweight. Alterations in the expression of these three proteins could contribute to the shallow invasion properties observed in preeclampsia. Little is known about the role of CCN4-6 in the reproductive organs. The ability of CCN1, CCN2 and CCN3 to interact with numerous receptors enables them to adapt their biological function rapidly to the continuous remodelling of the reproductive organs and in the development of the placenta. The CCN proteins mediate their specific cell physiological function through the receptor type of their binding partner followed by a defined signalling cascade. Because of their partly overlapping expression patterns, they could act in a concert synergistically or in an opposite way within the reproductive organs. Imbalances in their expression levels are correlated to different human reproductive diseases, such as endometriosis and preeclampsia.


Subject(s)
CCN Intercellular Signaling Proteins/physiology , Reproduction/genetics , Animals , Embryo Implantation/genetics , Endometriosis/genetics , Endometriosis/metabolism , Female , Humans , Ovary/physiology , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , Sex Factors , Uterine Diseases/genetics , Uterine Diseases/metabolism
20.
Int J Clin Oncol ; 19(2): 325-35, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23592278

ABSTRACT

BACKGROUND: The differences in clinical, pathological, and biological characteristics between adenocarcinoma (AC) and squamous cell/adenosquamous carcinoma (SC/ASC) of gallbladder cancer have not been well documented. This study is to compare the clinicopathological characteristics and FGFBP1 and WISP-2 expression between AC and SC/ASC patients. METHODS: We examined FGFBP1 and WISP-2 expression in 46 SC/ASC and 80 AC samples using immunohistochemistry and analyzed their correlations with clinicopathological characteristics. RESULTS: SC/ASCs occur more frequently in older patients and often correspond to larger tumor masses than ACs. Positive FGFBP1 and negative WISP-2 expression were significantly associated with lymph node metastasis and invasion of SC/ASCs and ACs. In addition, positive FGFBP1 and negative WISP-2 expression were significantly associated with differentiation and TMN stage in ACs. Univariate Kaplan-Meier analysis showed that either elevated FGFBP1 (p < 0.001) or lowered WISP-2 (p < 0.001) expression was closely associated with decreased overall survival in both SC/ASC and AC patients. Multivariate Cox regression analysis showed that positive FGFBP1 expression (p = 0.001) or negative WISP-2 expression (p = 0.035 for SC/ASC and p = 0.009 for AC) is an independent predictor of poor prognosis in both SC/ASC and AC patients. We also revealed that differentiation, tumor size, TNM stage, lymph node metastasis, invasion, and surgical procedure were associated with survival of both SC/ASC and AC patients. CONCLUSION: Our study suggested that the overexpression of FGFBP1 or loss of WISP-2 expression is closely related to the metastasis, invasion and poor prognosis of gallbladder cancer.


Subject(s)
Adenocarcinoma/pathology , CCN Intercellular Signaling Proteins/analysis , Carcinoma, Adenosquamous/pathology , Carcinoma, Squamous Cell/pathology , Carrier Proteins/analysis , Gallbladder Neoplasms/pathology , Intercellular Signaling Peptides and Proteins/analysis , Repressor Proteins/analysis , Adenocarcinoma/chemistry , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , CCN Intercellular Signaling Proteins/physiology , Carcinoma, Adenosquamous/chemistry , Carcinoma, Adenosquamous/mortality , Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/mortality , Carrier Proteins/physiology , Female , Gallbladder Neoplasms/chemistry , Gallbladder Neoplasms/mortality , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/physiology , Male , Middle Aged , Neoplasm Staging , Proportional Hazards Models , Repressor Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...