Metalloproteinase Meprin Α Regulates Migration and Invasion of Human Hepatocarcinoma Cells and Is a Mediator of the Oncoprotein Reptin

Total Page:16

File Type:pdf, Size:1020Kb

Metalloproteinase Meprin Α Regulates Migration and Invasion of Human Hepatocarcinoma Cells and Is a Mediator of the Oncoprotein Reptin www.impactjournals.com/oncotarget/ Oncotarget, 2017, Vol. 8, (No. 5), pp: 7839-7851 Research Paper Metalloproteinase meprin α regulates migration and invasion of human hepatocarcinoma cells and is a mediator of the oncoprotein Reptin Osman Breig1, Maïlyn Yates1, Véronique Neaud1, Gabrielle Couchy2, Aude Grigoletto1, Carlo Lucchesi3, Johannes Prox4, Jessica Zucman-Rossi2, Christoph Becker-Pauly4, Jean Rosenbaum1 1University Bordeaux, INSERM, U1053, BordeAux Research in Translational Oncology, BaRITOn, Bordeaux, France 2Inserm, U1162, Génomique Fonctionnelle des Tumeurs Solides, Université Paris Diderot, Université Paris Descartes, Université Paris 13, Paris, France 3SIRIC BRIO, Bordeaux, France 4Unit for Degradomics of the Protease Web, University of Kiel, Germany Correspondence to: Jean Rosenbaum, email: [email protected] Keywords: RUVBL2, proteolysis, prognosis Received: August 23, 2016 Accepted: November 30, 2016 Published: December 16, 2016 ABSTRACT Hepatocellular carcinoma is associated with a high rate of intra-hepatic invasion that carries a poor prognosis. Meprin alpha (Mep1A) is a secreted metalloproteinase with many substrates relevant to cancer invasion. We found that Mep1A was a target of Reptin, a protein that is oncogenic in HCC. We studied Mep1A regulation by Reptin, its role in HCC, and whether it mediates Reptin oncogenic effects. MepA and Reptin expression was measured in human HCC by qRT-PCR and in cultured cells by PCR, western blot and enzymatic activity measurements. Cell growth was assessed by counting and MTS assay. Cell migration was measured in Boyden chambers and wound healing assays, and cell invasion in Boyden chambers. Silencing Reptin decreased Mep1A expression and activity, without affecting meprin β. Mep1A, but not meprin β, was overexpressed in a series of 242 human HCC (2.04 fold, p < 0.0001), and a high expression correlated with a poor prognosis. Mep1A and Reptin expressions were positively correlated (r = 0.39, p < 0.0001). Silencing Mep1A had little effect on cell proliferation, but decreased cell migration and invasion of HuH7 and Hep3B cells. Conversely, overexpression of Mep1A or addition of recombinant Mep1A increased migration and invasion. Finally, overexpression of Mep1A restored a normal cell migration in cells where Reptin was depleted. Mep1A is overexpressed in most HCC and induces HCC cell migration and invasion. Mep1A expression is regulated by Reptin, and Mep1A mediates Reptin-induced migration. Overall, we suggest that Mep1A may be a useful target in HCC. INTRODUCTION responsible for HCC cells migration and invasion have been identified, this knowledge has not so far impacted Hepatocellular carcinoma (HCC) remains one of the handling of patients. There is thus a great need for the deadliest cancers worldwide. The prognosis of HCC discovering new actionable targets. is dictated both by the status of the host liver, very often In the last few years, we have discovered the cirrhotic, and by the tumor extension. HCC is characterized overexpression in HCC of Reptin, or RUVBL2, a by a very frequent intra-hepatic invasion, notably of portal member of the AAA+ ATPase family [2]. Reptin, and its veins, and by microvascular invasion, which carries a very homolog and partner protein, Pontin/RUVBL1 (also found poor prognosis [1]. Although a number of mechanisms overexpressed in HCC [3]) are part of several protein www.impactjournals.com/oncotarget 7839 Oncotarget complexes that have pleiotropic functions (for review, meprin α proteolytic activity in the conditioned medium see [4]). Briefly, they are involved in the remodeling of was decreased upon Reptin silencing. As a control, chromatin, the regulation of gene transcription through we confirmed that the substrate cleaving activity was various mechanisms, and they also act as chaperones abrogated by the meprin inhibitor actinonin (Figure 1F). for proteins of the Phosphatidylinositol Kinase-like Kinases family such as mTOR, ATM or DNA-PKcs. We Meprin α is overexpressed in human HCC and found that both proteins were required for the growth its expression correlates with that of Reptin and viability of HCC cells [2, 3, 5] and that silencing Reptin within established HCC xenografts induced tumor Expression of meprin α, meprin β and Reptin regression, thus establishing Reptin as a potential target mRNAs was assessed using RT-PCR on a previously in HCC [6]. There is also evidence showing that Reptin described series of 242 HCC from the French Biological silencing reduces invasion of cancer cells from kidney Resource Center on HCC [22]. Meprin α mRNA level [7] and prostate [8]. The downstream mediators of Reptin was significantly higher in HCC samples as compared to underlying its oncogenic effects remain however to be non-tumor liver (2.04 fold, p < 0.0001; Figure 2A). On found. the other hand, meprin β expression was not increased, Meprin α is a secreted metalloproteinase that can and was even significantly decreased (p < 0.0001) when cleave a potentially large number of protein substrates, comparing HCC to non-tumor liver (Figure 2A). many of them being highly relevant for cancer [9]. Immunohistochemistry experiments in 3 HCC Those include extracellular matrix and related proteins tumors confirmed in all cases an overexpression of Meprin (pro-collagen I, fibronectin, SPARC, osteopontin), α in HCC as compared to the non-tumor liver in the same cytokines (pro-IL1β, IL6), growth factors (VEGF-A, patient. Furthermore, these experiments demonstrated CTGF), membrane proteins (occludin), and other unambiguously that Meprin α overexpression takes place proteinases (MMP1, ADAM10) [10]. Meprin α had so far in tumor cells and not stromal cells (Figure 2B). received little attention in cancer, with limited evidence Analysis of HCC subgroups revealed that meprin showing that it could stimulate cancer cell proliferation α expression was especially high in transcriptomic [11], migration [12] and invasion [13]. While this work groups G1, G2 and G3 (Figure 2C). These groups are was in progress, however, OuYang et al. showed that it characterized by a higher rate of cell proliferation, could induce HCC cell migration and invasion [14]. and accordingly meprin α expression was significantly Interestingly, there are already available good meprin correlated with that of Ki67 mRNA (Spearman r = 0.41, α inhibitors like actinonin [15] that can be used in vivo p < 0.0001). Groups G1-G3 are also enriched in patients [16–18], and more specific inhibitors are currently under with TP53 mutations [23] and we found indeed that development [19], raising the hope that meprin α may meprin α expression was significantly higher in become a clinically useful target. patients with TP53 mutation, and conversely lower in Knowing that Reptin regulates the expression those with CTNNB1 mutations, that are enriched in of many genes [20, 21], we hypothesized that it G4-G6 patients (Figure 2D). There was no significant might regulate meprin α. Thus, in this study, we have difference in meprin α expression whether the cause investigated the regulation of the expression of meprin α of liver disease was excess alcohol consumption, B or by Reptin in HCC, the effects of meprin α on HCC cell C viral hepatitis, hemochromatosis or metabolic liver phenotype, and whether meprin α mediated the oncogenic disease (not shown). effects of Reptin. Additionally, several data pointed to a significant correlation of high meprin α expression with a poor RESULTS prognosis. First, meprin α expression was significantly higher in tumors from Edmonson grades II-IV, as Reptin silencing decreases the expression of compared to grades I-II (Figure 2E). We previously found meprin α that a score based on the expression of the 5 genes HN1, RAN, RAMP3, KRT19, and TAF9, was associated with Reptin expression was efficiently silenced in the survival of HCC patients following surgery [22]. We HuH7 cells using two different siRNAs, or with a found here that meprin α expression was significantly doxycycline-inducible shRNA, as described previously higher in tumors from patients with high 5-gene prognosis [2, 6]. This resulted in a significantly decreased expression score (Figure 2E). Finally, we performed a Kaplan-Meier of meprin α mRNA, whereas the expression of meprin β analysis by first dichotomizing the patients as meprin was not decreased (Figure 1A–1C). Similar results were low or high according to the median of expression in all obtained in another HCC cell line (Hep 3B (Supplementary samples. This analysis showed that the overall survival of Figure S1A). Furthermore, meprin α, but not meprin β patients was significantly worse in patients with a high protein expression was also reduced as shown by Western expression of meprin α, whereas meprin β expression blot (Figures 1D–1E, Supplementary Figure S1D). Finally, carried no prognostic value (Figure 2F). www.impactjournals.com/oncotarget 7840 Oncotarget Very interestingly, and in line with our results strategies. First, we constructed HuH7 and Hep3B cell showing the regulation of meprin α expression by Reptin, lines constitutively overexpressing meprin α with a V5 we found that meprin α expression was significantly C-terminal tag. Because the C-terminus of meprin α is and positively correlated with that of Reptin in HCC removed during the maturation and secretion process, (Spearman r = 0.23, p = 0.0003), whereas no correlation the tag is no more present in the secreted protein and was found for meprin β (Spearman r = 0.083, p = 0.20). does not interfere with the proteolytic activity. As shown on Figure 3D, the V5-tagged protein was found in Generation of tools for loss and gain of function large amounts in cell extracts where it can be detected of meprin α with both a meprin α and a V5 antibody. On the other hand, the overexpressed protein is detectable only with For loss of function, we designed two siRNAs the meprin α antibody in the supernatant of the cells. targeting meprin α. Both siRNAs efficiently depleted Immunofluorescence with both anti-meprin α and anti-V5 meprin α mRNA and protein (Figure 3A–3C).
Recommended publications
  • Discovery and Optimization of Selective Inhibitors of Meprin Α (Part II)
    pharmaceuticals Article Discovery and Optimization of Selective Inhibitors of Meprin α (Part II) Chao Wang 1,2, Juan Diez 3, Hajeung Park 1, Christoph Becker-Pauly 4 , Gregg B. Fields 5 , Timothy P. Spicer 1,6, Louis D. Scampavia 1,6, Dmitriy Minond 2,7 and Thomas D. Bannister 1,2,* 1 Department of Molecular Medicine, Scripps Research, Jupiter, FL 33458, USA; [email protected] (C.W.); [email protected] (H.P.); [email protected] (T.P.S.); [email protected] (L.D.S.) 2 Department of Chemistry, Scripps Research, Jupiter, FL 33458, USA; [email protected] 3 Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, 3321 College Avenue, CCR r.605, Fort Lauderdale, FL 33314, USA; [email protected] 4 The Scripps Research Molecular Screening Center, Scripps Research, Jupiter, FL 33458, USA; [email protected] 5 Unit for Degradomics of the Protease Web, Institute of Biochemistry, University of Kiel, Rudolf-Höber-Str.1, 24118 Kiel, Germany; fi[email protected] 6 Department of Chemistry & Biochemistry and I-HEALTH, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA 7 Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, FL 33314, USA * Correspondence: [email protected] Abstract: Meprin α is a zinc metalloproteinase (metzincin) that has been implicated in multiple diseases, including fibrosis and cancers. It has proven difficult to find small molecules that are capable Citation: Wang, C.; Diez, J.; Park, H.; of selectively inhibiting meprin α, or its close relative meprin β, over numerous other metzincins Becker-Pauly, C.; Fields, G.B.; Spicer, which, if inhibited, would elicit unwanted effects.
    [Show full text]
  • On and Polymorphisms in Q Fever
    Matrix metalloproteinase expression, produc3on and polymorphisms in Q fever Anne F.M. Jansen1,2, Teske Schoffelen1,2, Julien Textoris3, Jean Louis Mege3, Chantal P. Bleeker-Rovers1,2, Esther van de Vosse4, Hendrik Jan Roest5, Marcel van Deuren1,2 1. Department of Internal Medicine, Division of Experimental Medicine, Radboud university medical center, Nijmegen, The Netherlands 2. Radboud Expert Centre for Q fever, Radboud university medical center, Nijmegen, the Netherlands, 3. URMITE, CNRS UMR 7278, IRD 198, INSERM 1095 Aix-Marseille University, Marseille, France 4. Department of Infec3ous Diseases, Leiden University Medical Center, Leiden, The Netherlands 5. Department of Bacteriology and TSEs, Central Veterinary Instute, part of Wageningen UR, Lelystad, the Netherlands Background C. burnei induces MMP-1 and MMP-9 produc3on in PBMCs Chronic Q fever is a life threatening condi3on caused by the Gram-negave bacterium Coxiella burnei, manifes3ng as an infec3on of aneurysms, aor3c prosthesis or heart valves. Matrix metalloproteinases (MMPs) are proteoly3c enzymes that cleave extracellular matrix and are implicated in the pathology of aneurysms and endocardi3s. Currently, the contribu3on of MMPs to the pathogenesis of chronic Q fever is unknown. Methods We inves3gated the C. burnei specific gene expression of MMPs in PBMCs and protein produc3on by ELISA in chronic Q fever paents (n=6, n=10, respec3vely), cardiovascular paents with a history of Q fever (n=10) and healthy controls (n=4, n=10, respec3vely), in some experiments, the controls had vascular disease (n=10). Circulang MMP levels were assessed with Luminex technology and these groups were also genotyped for 20 SNPs in MMP and Tissue Inhibitor of MMP (TIMP) genes.
    [Show full text]
  • Prognostic Significance of MMP-1 and MMP-3 Functional Promoter Polymorphisms in Colorectal Cancer
    594 Vol. 11, 594–599, January 15, 2005 Clinical Cancer Research Prognostic Significance of MMP-1 and MMP-3 Functional Promoter Polymorphisms in Colorectal Cancer Franck Zinzindohoue´,1 Thierry Lecomte,2 INTRODUCTION Jean-Marc Ferraz,2 Anne-Marie Houllier,1 Matrix metalloproteinase (MMP) belongs to a large group Paul-Henri Cugnenc,2 Anne Berger,2 of proteases, which includes over 22 known human zinc- He´le`ne Blons,1 and Pierre Laurent-Puig1 dependent proteolytic enzymes. These are capable of breaking essentially all components of the extracellular matrix (1, 2). 1INSERM U490 Laboratoire de toxicologie Mole´culaire and 2 MMPs take part in high tissue turnover and remodeling, both Poˆle de cance´rologie Hoˆpital europe´en Georges Pompidou, Paris, France physiologic and pathologic conditions such as cancer. MMPs are also implicated in all steps of tumorogenesis, cancer invasion, and metastasis (3). Tumor invasion and metastasis formation ABSTRACT always begin with blood and lymphatic vessel infiltration. As Purpose: Matrix metalloproteinase (MMP) belongs to a these processes involve proteolysis of the extracellular matrix, large group of proteases capable of breaking essentially all MMPs are suggested to play a major role in tumor progression. components of the extracellular matrix. They are implicated Colorectal cancer is the most common malignancy of the in all steps of tumorogenesis, cancer invasion, and gastrointestinal tract. It is the third cause of cancer overall and metastasis. Among them, metalloproteinase type 1 (MMP- the second leading cause of cancer-related death in the Europe 1) is implicated in tumor invasion and metastasis in and the United States with an incidence of 300,000 new cases different types of cancers including colorectal cancer in (4).
    [Show full text]
  • Role of MMP-1, MMP-8 and MMP-9 Gene Polymorphisms in Preterm Birth
    Journal of Genetics (2020)99:2 Ó Indian Academy of Sciences https://doi.org/10.1007/s12041-019-1161-7 (0123456789().,-volV)(0123456789().,-volV) RESEARCH ARTICLE Role of MMP-1, MMP-8 and MMP-9 gene polymorphisms in preterm birth MONIKA PANDEY* and SHALLY AWASTHI Department of Pediatrics, King George’s Medical University, Lucknow 226 003, India *For correspondence. E-mail: [email protected]. Received 6 January 2019; revised 17 August 2019; accepted 18 September 2019 Abstract. Novel approaches to preterm births are underway building upon our prior discoveries and probing into unknown discovery pathways. The recent findings showed a high affinity of MMP-9 in serum and its polymorphisms for preterm birth. This study, which is a hospital-based case– control study, aims to investigate the association of MMP-1, MMP-8 and MMP-9 polymorphisms, and levels of MMP-9 in preterm birth. Increased level of MMP-9 was reported in cases as compared to control. The significant association of MMP-9 (-1562) CT (P=0.001; OR = 1.44 (CI = 0.97–2.14)) and TT genotype (P=0.05; OR = 2.6 (CI = 1.46–4.69)) were reported in preterm birth. Our findings suggest that the MMP-9 plays an important role in contributing preterm labour and this can be used as a diagnostic tool during pregnancy. Keywords. infection; inflammation; matrix metalloproteinases; polymorphism; preterm birth. Introduction altered gene expression may be an attributing factor of causing preterm birth (Sheikh et al. 2016). The functional polymorphisms Preterm birth (PTB) is the most prevailing and persistent situated in MMP-1, MMP-8 and MMP-9 promoter region may be problem causing enormous morbidity and mortality among a contributing element (Fanjul-Ferna´ndez et al.
    [Show full text]
  • A Therapeutic Role for MMP Inhibitors in Lung Diseases?
    ERJ Express. Published on June 9, 2011 as doi: 10.1183/09031936.00027411 A therapeutic role for MMP inhibitors in lung diseases? Roosmarijn E. Vandenbroucke1,2, Eline Dejonckheere1,2 and Claude Libert1,2,* 1Department for Molecular Biomedical Research, VIB, Ghent, Belgium 2Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium *Corresponding author. Mailing address: DBMR, VIB & Ghent University Technologiepark 927 B-9052 Ghent (Zwijnaarde) Belgium Phone: +32-9-3313700 Fax: +32-9-3313609 E-mail: [email protected] 1 Copyright 2011 by the European Respiratory Society. A therapeutic role for MMP inhibitors in lung diseases? Abstract Disruption of the balance between matrix metalloproteinases and their endogenous inhibitors is considered a key event in the development of pulmonary diseases such as chronic obstructive pulmonary disease, asthma, interstitial lung diseases and lung cancer. This imbalance often results in elevated net MMP activity, making MMP inhibition an attractive therapeutic strategy. Although promising results have been obtained, the lack of selective MMP inhibitors together with the limited knowledge about the exact functions of a particular MMP hampers the clinical application. This review discusses the involvement of different MMPs in lung disorders and future opportunities and limitations of therapeutic MMP inhibition. 1. Introduction The family of matrix metalloproteinases (MMPs) is a protein family of zinc dependent endopeptidases. They can be classified into subgroups based on structure (Figure 1), subcellular location and/or function [1, 2]. Although it was originally believed that they are mainly involved in extracellular matrix (ECM) cleavage, MMPs have a much wider substrate repertoire, and their specific processing of bioactive molecules is their most important in vivo role [3, 4].
    [Show full text]
  • MMP-1/8 (A-7): Sc-137044
    SANTA CRUZ BIOTECHNOLOGY, INC. MMP-1/8 (A-7): sc-137044 BACKGROUND APPLICATIONS The matrix metalloproteinases (MMP) are a family of peptidase enzymes MMP-1/8 (A-7) is recommended for detection of MMP-1 of human origin and responsible for the degradation of extracellular matrix components, including MMP-8 of mouse, rat and human origin by Western Blotting (starting dilu- collagen, gelatin, fibronectin, laminin and proteoglycan. Transcription of tion 1:100, dilution range 1:100-1:1000), immunoprecipitation [1-2 µg per MMP genes is differentially activated by phorbol ester, lipopolysaccharide 100-500 µg of total protein (1 ml of cell lysate)], immunofluorescence (LPS) or staphylococcal enterotoxin B (SEB). MMP catalysis requires both (starting dilution 1:50, dilution range 1:50-1:500) and solid phase ELISA calcium and zinc. MMP-9 (also designated 92 kDa type IV collagenase or (starting dilution 1:30, dilution range 1:30-1:3000). gelatinase B) has been shown to degrade bone collagens in concert with Suitable for use as control antibody for MMP-8 siRNA (m): sc-35950, MMP-8 MMP-1 (also designated interstitial collagenase, fibroblast collagenase or shRNA Plasmid (m): sc-35950-SH and MMP-8 shRNA (m) Lentiviral Particles: collagenase-1) and cysteine proteases, and may play a role in bone osteo- sc-35950-V. clastic resorption. MMP-1 is downregulated by p53, and abnormality of p53 expression may contribute to joint degradation in rheumatoid arthritis by Molecular Weight of MMP-1: 52 kDa. regulating MMP-1 expression. MMP-8 (also designated neutrophil collage- Molecular Weight of latent MMP-8: 65 kDa.
    [Show full text]
  • Biochemical Characterization and Zinc Binding Group (Zbgs) Inhibition Studies on the Catalytic Domains of Mmp7 (Cdmmp7) and Mmp16 (Cdmmp16)
    MIAMI UNIVERSITY The Graduate School Certificate for Approving the Dissertation We hereby approve the Dissertation of Fan Meng Candidate for the Degree DOCTOR OF PHILOSOPHY ______________________________________ Director Dr. Michael W. Crowder ______________________________________ Dr. David L. Tierney ______________________________________ Dr. Carole Dabney-Smith ______________________________________ Dr. Christopher A. Makaroff ______________________________________ Graduate School Representative Dr. Hai-Fei Shi ABSTRACT BIOCHEMICAL CHARACTERIZATION AND ZINC BINDING GROUP (ZBGS) INHIBITION STUDIES ON THE CATALYTIC DOMAINS OF MMP7 (CDMMP7) AND MMP16 (CDMMP16) by Fan Meng Matrix metalloproteinase 7 (MMP7/matrilysin-1) and membrane type matrix metalloproteinase 16 (MMP16/MT3-MMP) have been implicated in the progression of pathological events, such as cancer and inflammatory diseases; therefore, these two MMPs are considered as viable drug targets. In this work, we (a) provide a review of the role(s) of MMPs in biology and of the previous efforts to target MMPs as therapeutics (Chapter 1), (b) describe our efforts at over-expression, purification, and characterization of the catalytic domains of MMP7 (cdMMP7) and MMP16 (cdMMP16) (Chapters 2 and 3), (c) present our efforts at the preparation and initial spectroscopic characterization of Co(II)-substituted analogs of cdMMP7 and cdMMP16 (Chapters 2 and 3), (d) present inhibition data on cdMMP7 and cdMMP16 using zinc binding groups (ZBG) as potential scaffolds for future inhibitors (Chapter 3), and (e) summarize our data in the context of previous results and suggest future directions (Chapter 4). The work described in this dissertation integrates biochemical (kinetic assays, inhibition studies, limited computational methods), spectroscopic (CD, UV-Vis, 1H-NMR, fluorescence, and EXAFS), and analytical (MALDI-TOF mass spectrometry, isothermal calorimetry) methods to provide a detailed structural and mechanistic view of these MMPs.
    [Show full text]
  • Circulating Proteomic Signature of Early Death in Heart Failure Patients with Reduced Ejection Fraction
    www.nature.com/scientificreports OPEN Circulating proteomic signature of early death in heart failure patients with reduced ejection fraction Marie Cuvelliez1,2,5, Vincent Vandewalle3,4,5, Maxime Brunin4, Olivia Beseme1,2, Audrey Hulot 4, Pascal de Groote1,2, Philippe Amouyel1, Christophe Bauters1,2,6, Guillemette Marot3,4,6 & Florence Pinet 1,2,6* Heart failure (HF) remains a main cause of mortality worldwide. Risk stratifcation of patients with systolic chronic HF is critical to identify those who may beneft from advanced HF therapies. The aim of this study is to identify plasmatic proteins that could predict the early death (within 3 years) of HF patients with reduced ejection fraction hospitalized in CHRU de Lille. The subproteome targeted by an aptamer-based technology, the Slow Of-rate Modifed Aptamer (SOMA) scan assay of 1310 proteins, was profled in blood samples from 168 HF patients, and 203 proteins were signifcantly modulated between patients who died of cardiovascular death and patients who were alive after 3 years of HF evaluation (Wilcoxon test, FDR 5%). A molecular network was built using these 203 proteins, and the resulting network contained 2281 molecules assigned to 34 clusters annotated to biological pathways by Gene Ontology. This network model highlighted extracellular matrix organization as the main mechanism involved in early death in HF patients. In parallel, an adaptive Least Absolute Shrinkage and Selection Operator (LASSO) was performed on these 203 proteins, and six proteins were selected as candidates to predict early death in HF patients: complement C3, cathepsin S and F107B were decreased and MAPK5, MMP1 and MMP7 increased in patients who died of cardiovascular causes compared with patients living 3 years after HF evaluation.
    [Show full text]
  • Polymorphisms of Genes Involved in Extracellular Matrix Remodeling And
    CORE Metadata, citation and similar papers at core.ac.uk Provided by Florence Research BASIC RESEARCH STUDIES Polymorphisms of genes involved in extracellular matrix remodeling and abdominal aortic aneurysm Claudia Saracini, PhD,a Paola Bolli, PhD,a Elena Sticchi, PhD,a,b Giovanni Pratesi, MD,c Raffaele Pulli, MD,d Francesco Sofi, PhD,a Carlo Pratesi, MD,d Gian Franco Gensini, MD,a,b Rosanna Abbate, MD,a and Betti Giusti, PhD,a Florence and Rome, Italy Background: Abdominal aortic aneurysm (AAA) has a multifactorial etiology and the relevance of genetic factors is getting increasing interest, in particular those related to the destructive remodeling of extracellular matrix. Methods: We performed a candidate gene association study of polymorphisms in genes coding matrix metalloproteinases (MMPs), tissue inhibitors of MMPs (TIMPs), and elastin (ELN) in AAA. DNA samples from 423 AAA patients and 423 ;controls were genotyped for 12 polymorphisms in 10 genes: MMP1 (؊1607G/GG), MMP2 (؊735C/T; ؊1306C/T ,(؊1575 G/A), MMP3 (5A/6A), MMP9 (؊1562C/T), MMP10 (A180G), MMP-12 (؊82A/G), MMP-13 (؊77A/G .(TIMP1 (C434T), TIMP3 (؊1296T/C), and ELN (G1355A Results: Genotype distribution was significantly different between patients and controls for the following polymorphisms: ؊1306C/T MMP2; 5A/6A MMP3; ؊77A/G MMP-13; G1355A ELN; and C434T TIMP1. In a multivariable logistic regression analysis adjusted for traditional cardiovascular risk factors and chronic obstructive pulmonary disease, confidence interval, CI .34-.85], P < .007) and G1355A ELN %95] 0.55 ؍ [؊1306C/T MMP2 (odds ratios [OR polymorphisms resulted in independent protective factors for abdominal aortic (046.
    [Show full text]
  • Serum MMP7, MMP10 and MMP12 Level As Negative Prognostic
    Klupp et al. BMC Cancer (2016) 16:494 DOI 10.1186/s12885-016-2515-7 RESEARCH ARTICLE Open Access Serum MMP7, MMP10 and MMP12 level as negative prognostic markers in colon cancer patients Fee Klupp1*, Lena Neumann1, Christoph Kahlert3, Johannes Diers1, Niels Halama2, Clemens Franz1, Thomas Schmidt1, Moritz Koch3ˆ, Juergen Weitz3, Martin Schneider1 and Alexis Ulrich1 Abstract Background: Matrixmetalloproteinases (MMPs) comprise a family of zinc-dependent endopeptidases which are involved in angiogenesis, tumor invasion and metastatic formation. Up to date, the prognostic relevance of MMPs in serum of patients with colon cancer remains unknown. Thus, we wanted to assess an expression pattern of MMPs in a homogenous cohort of colon cancer patients to assess their potential as prognostic biomarkers. Methods: Differences in the expression pattern of MMP7, MMP10 and MMP12 in 78 serum specimens of patients with an adenocarcinoma of the colon and serum specimens of a healthy control group were assessed using Luminex-100 technologies. Subsequently, we correlated these results with histopathological and clinical data of the patients. Results: Luminex based expression analysis revealed a significant overexpression of MMP7 and an overexpression of MMP10 and MMP12 in the sera of colon cancer patients compared to the healthy control group. Patients with vascular invasion showed a significantly higher MMP12 expression than V0-staged patients. Moreover overexpression of MMP7, MMP10 and MMP12 in colon cancer patients´ sera displayed a significantly impaired overall survival. Multivariate analysis revealed high MMP10 serum levels to be an independent adverse prognostic marker in colon cancer patients. Conclusions: Expression patterns of MMP7, MMP10 and MMP12 in colon cancer patients´ sera are different compared to serum specimens of healthy individuals.
    [Show full text]
  • BTXA Regulates the Epithelial–Mesenchymal Transition and Autophagy of Keloid Fibroblasts Via Modulating Mir-1587/Mir-2392 Targ
    Bioscience Reports (2019) 39 BSR20190679 https://doi.org/10.1042/BSR20190679 Research Article BTXA regulates the epithelial–mesenchymal transition and autophagy of keloid fibroblasts via modulating miR-1587/miR-2392 targeted ZEB2 1 1 2 Zhanying Hou , Feixiang Fan and Po Liu Downloaded from http://portlandpress.com/bioscirep/article-pdf/39/10/BSR20190679/858361/bsr-2019-0679.pdf by guest on 29 September 2021 1Department of Dermatology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, Guangdong, China; 2Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen 518110, Guangdong, China Correspondence: Zhanying Hou ([email protected], [email protected]) Keloids are very resistant to treatment in dermatology and plastic surgical practice. The present study aimed to explore the underlying mechanism of botulinum toxin A (BTXA) treated human skin keloid fibroblasts (HSFBs) proving some new insights into keloids treat- ment. Expression of miR-1587 and miR-2392 were significantly down-regulated in keloid tissues and HSFBs, while the ZEB2 was a target of both and up-regulated in keloid tis- sues and HSFBs compared with the normal controls. BTXA could significantly increase the expression of miR-1587 and miR-2392 but decrease the expression of ZEB2. BTXA could significantly inhibit the proliferation, cell cycle, and migration and promote apopto- sis and autophagy of HSFBs; however, miR-1587 and miR-2392 inhibitors could reverse these effects of BTXA on HSFBs. Silencing ZEB2 could significantly attenuate the effects of miR-1587 and miR-2392 inhibitors in promoting cell proliferation and migration and sup- pressing apoptosis and autophagy of HSFBs after treating with BTXA.
    [Show full text]
  • MMP-1 and Pro-MMP-10 As Potential Urinary Pharmacodynamic Biomarkers of FGFR3-Targeted Therapy in Patients with Bladder Cancer
    Published OnlineFirst October 17, 2014; DOI: 10.1158/1078-0432.CCR-13-3336 Clinical Cancer Personalized Medicine and Imaging Research MMP-1 and Pro-MMP-10 as Potential Urinary Pharmacodynamic Biomarkers of FGFR3-Targeted Therapy in Patients with Bladder Cancer Xiangnan Du1, Benjamin C. Lin2, Qian-Rena Wang1, Hao Li1, Ellen Ingalla1, Janet Tien1, Isabelle Rooney3, Avi Ashkenazi4, Elicia Penuel2, and Jing Qing1 Abstract Purpose: The aim of this study was to identify noninvasive pharmacodynamic biomarkers of FGFR3- targeted therapies in bladder cancer to facilitate the clinical development of experimental agent targeting FGFR3. Experimental Design: Potential soluble pharmacodynamic biomarkers of FGFR3 were identified using a combination of transcriptional profiling and biochemical analyses in preclinical models. Two matrix metalloproteinases (MMP), MMP-1 and MMP-10, were selected for further studies in human bladder cancer xenograft models treated with a specific anti-FGFR3 monoclonal antibody, R3Mab. Serum and urinary levels of MMP-1 and MMP-10 were determined in healthy donors and patients with bladder cancer. The modulation of MMP-1 and MMP-10 by R3Mab in patients with bladder cancer was further evaluated in a phase I dose-escalation study. Results: MMP-1 and MMP-10 mRNA and protein were downmodulated by FGFR3 shRNA and R3Mab in bladder cancer cell lines. FGFR3 signaling promoted the expression and secretion of MMP-1 and pro-MMP- 10 in a MEK-dependent fashion. In bladder cancer xenograft models, R3Mab substantially blocked tumor progression and reduced the protein levels of human MMP-1 and pro-MMP-10 in tumor tissues as well as in mouse serum.
    [Show full text]