1
|
Mongiat M, Pascal G, Poletto E, Williams DM, Iozzo RV. Proteoglycans of basement membranes: Crucial controllers of angiogenesis, neurogenesis, and autophagy. PROTEOGLYCAN RESEARCH 2024; 2:e22. [PMID: 39184370 PMCID: PMC11340296 DOI: 10.1002/pgr2.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/02/2024] [Indexed: 08/27/2024]
Abstract
Anti-angiogenic therapy is an established method for the treatment of several cancers and vascular-related diseases. Most of the agents employed target the vascular endothelial growth factor A, the major cytokine stimulating angiogenesis. However, the efficacy of these treatments is limited by the onset of drug resistance. Therefore, it is of fundamental importance to better understand the mechanisms that regulate angiogenesis and the microenvironmental cues that play significant role and influence patient treatment and outcome. In this context, here we review the importance of the three basement membrane heparan sulfate proteoglycans (HSPGs), namely perlecan, agrin and collagen XVIII. These HSPGs are abundantly expressed in the vasculature and, due to their complex molecular architecture, they interact with multiple endothelial cell receptors, deeply affecting their function. Under normal conditions, these proteoglycans exert pro-angiogenic functions. However, in pathological conditions such as cancer and inflammation, extracellular matrix remodeling leads to the degradation of these large precursor molecules and the liberation of bioactive processed fragments displaying potent angiostatic activity. These unexpected functions have been demonstrated for the C-terminal fragments of perlecan and collagen XVIII, endorepellin and endostatin. These bioactive fragments can also induce autophagy in vascular endothelial cells which contributes to angiostasis. Overall, basement membrane proteoglycans deeply affect angiogenesis counterbalancing pro-angiogenic signals during tumor progression, and represent possible means to develop new prognostic biomarkers and novel therapeutic approaches for the treatment of solid tumors.
Collapse
Affiliation(s)
- Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Gabriel Pascal
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Davion M. Williams
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Renato V. Iozzo
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
2
|
Nemoz-Billet L, Balland M, Gilquin L, Gillet B, Stévant I, Guillon E, Hughes S, Carpentier G, Vaganay E, Sohm F, Misiak V, Gonzalez-Melo MJ, Koch M, Ghavi-Helm Y, Bretaud S, Ruggiero F. Dual topologies of myotomal collagen XV and Tenascin C act in concert to guide and shape developing motor axons. Proc Natl Acad Sci U S A 2024; 121:e2314588121. [PMID: 38502691 PMCID: PMC10990108 DOI: 10.1073/pnas.2314588121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/26/2024] [Indexed: 03/21/2024] Open
Abstract
During development, motor axons are guided toward muscle target by various extrinsic cues including extracellular matrix (ECM) proteins whose identities and cellular source remain poorly characterized. Here, using single-cell RNAseq of sorted GFP+ cells from smyhc1:gfp-injected zebrafish embryos, we unravel the slow muscle progenitors (SMP) pseudotemporal trajectory at the single-cell level and show that differentiating SMPs are a major source of ECM proteins. The SMP core-matrisome was characterized and computationally predicted to form a basement membrane-like structure tailored for motor axon guidance, including basement membrane-associated ECM proteins, as collagen XV-B, one of the earliest core-matrisome gene transcribed in differentiating SMPs and the glycoprotein Tenascin C. To investigate how contact-mediated guidance cues are organized along the motor path to exert their function in vivo, we used microscopy-based methods to analyze and quantify motor axon navigation in tnc and col15a1b knock-out fish. We show that motor axon shape and growth rely on the timely expression of the attractive cue Collagen XV-B that locally provides axons with a permissive soft microenvironment and separately organizes the repulsive cue Tenascin C into a unique functional dual topology. Importantly, bioprinted micropatterns that mimic this in vivo ECM topology were sufficient to drive directional motor axon growth. Our study offers evidence that not only the composition of ECM cues but their topology critically influences motor axon navigation in vertebrates with potential applications in regenerative medicine for peripheral nerve injury as regenerating nerves follow their original path.
Collapse
Affiliation(s)
- Laurie Nemoz-Billet
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Martial Balland
- LIPphy: Interdisciplinary Laboratory of Physics, Université Grenoble Alpes, CNRS, GrenobleF-38000, France
| | - Laurent Gilquin
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Benjamin Gillet
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Isabelle Stévant
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Emilie Guillon
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Sandrine Hughes
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Gilles Carpentier
- Gly-CRRET: Glycobiology, Cell Growth and Tissue Repair Research Unit, Laboratoire Gly-CRRET Faculté des Sciences et Technologie, Université Paris Est-Créteil-Val de Marne, Créteil Cedex94010, France
| | - Elisabeth Vaganay
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Frédéric Sohm
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Vladimir Misiak
- LIPphy: Interdisciplinary Laboratory of Physics, Université Grenoble Alpes, CNRS, GrenobleF-38000, France
| | - Mary-Julieth Gonzalez-Melo
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Manuel Koch
- Institute for Experimental Dental Research and Oral Musculoskeletal Biology, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne50931, Germany
| | - Yad Ghavi-Helm
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Sandrine Bretaud
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Florence Ruggiero
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| |
Collapse
|
3
|
Xing T, Zhao Y, Wang L, Geng W, Liu W, Zhou J, Huang C, Wang W, Chu X, Liu B, Chen K, Zheng H, Li L. Fine-scale mapping of chromosome 9q22.33 identifies candidate causal variant in ovarian cancer. PeerJ 2024; 12:e16918. [PMID: 38371376 PMCID: PMC10874173 DOI: 10.7717/peerj.16918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/18/2024] [Indexed: 02/20/2024] Open
Abstract
Ovarian cancer is a complex polygenic disease in which genetic factors play a significant role in disease etiology. A genome-wide association study (GWAS) identified a novel variant on chromosome 9q22.33 as a susceptibility locus for epithelial ovarian cancer (EOC) in the Han Chinese population. However, the underlying mechanism of this genomic region remained unknown. In this study, we conducted a fine-mapping analysis of 130 kb regions, including 1,039 variants in 200 healthy women. Ten variants were selected to evaluate the association with EOC risk in 1,099 EOC cases and 1,591 controls. We identified two variants that were significantly associated with ovarian cancer risk (rs7027650, P = 1.91 × 10-7; rs1889268, P = 3.71 × 10-2). Expression quantitative trait locus (eQTL) analysis found that rs7027650 was significantly correlated with COL15A1 gene expression (P = 0.009). The Luciferase reporter gene assay confirmed that rs7027650 could interact with the promoter region of COL15A1, reducing its activity. An electrophoretic mobility shift assay (EMSA) showed the allele-specific binding capacity of rs7027650. These findings revealed that rs7027650 could be a potential causal variant at 9q22.33 region and may regulate the expression level of COL15A1. This study offered insight into the molecular mechanism behind a potential causal variant that affects the risk of ovarian cancer.
Collapse
Affiliation(s)
- Tongyu Xing
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yanrui Zhao
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Lili Wang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Wei Geng
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Wei Liu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Jingjing Zhou
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Caiyun Huang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Wei Wang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Xinlei Chu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Ben Liu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Hong Zheng
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Lian Li
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
4
|
Huang X, Huang J, Huang Q, Zhou S. A ten long noncoding RNA-based prognostic risk model construction and mechanism study in the basal-like immune-suppressed subtype of triple-negative breast cancer. Transl Cancer Res 2023; 12:3653-3671. [PMID: 38193005 PMCID: PMC10774046 DOI: 10.21037/tcr-23-147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 09/21/2023] [Indexed: 01/10/2024]
Abstract
Background According to the Fudan University Shanghai Cancer Center (FUSCC) system, triple-negative breast cancer (TNBC) is divided into four stable subtypes: (I) luminal androgen receptor, (II) immunomodulatory, (III) basal-like immune-suppressed (BLIS), and (IV) mesenchymal-like. However, the treatment outcomes of the corresponding targeted therapies are unsatisfactory, especially for the BLIS subtype. Therefore, we aimed to identify the key long noncoding RNAs (lncRNAs) to construct a prognostic model for BLIS subtype and discover potential targets to explore potential therapeutic strategies in this study. Methods The FUSCC cohort was used to establish a prognostic risk model via least absolute shrinkage and selection operator (LASSO) and Cox regression analysis. The Cancer Genome Atlas (TCGA) cohort was then used to evaluate and verify the model. To understand the functional aspects of the model, functional, immune landscape, mutation, and drug sensitivity analyses were performed between high- and low-risk groups. Results Ten prognostic-related lncRNAs identified, including C5ORF66-AS2, DIO3OS, FZD10-DT, LINC00393, LNC-ERI1-32, LNC-FOXO1-2, LNC-SPARCL1-1, HCG23, LNC-MMD-4 and LNC-TMEM106C-6, were selected for risk score system construction. The results showed that the model constructed could divide the patients with BLIS subtype into two groups of high and low risk, and patients with higher risk scores had shorter recurrence-free survival. In addition, drug sensitivity analysis identified 3 compounds, including BMS-754807, cytochalasin b, and linifanib, that could have a potential therapeutic effect on patients with the BLIS subtype. Conclusions The risk prognosis model showed good prognostic value for the BLIS subtype patients, and the ten lncRNAs may be potential therapeutic targets.
Collapse
Affiliation(s)
- Xiaoying Huang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jinlong Huang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qiuyan Huang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Shihao Zhou
- College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
5
|
Nelke C, Schroeter CB, Theissen L, Preusse C, Pawlitzki M, Räuber S, Dobelmann V, Cengiz D, Kleefeld F, Roos A, Schoser B, Brunn A, Neuen-Jacob E, Zschüntzsch J, Meuth SG, Stenzel W, Ruck T. Senescent fibro-adipogenic progenitors are potential drivers of pathology in inclusion body myositis. Acta Neuropathol 2023; 146:725-745. [PMID: 37773216 PMCID: PMC10564677 DOI: 10.1007/s00401-023-02637-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023]
Abstract
Inclusion body myositis (IBM) is unique across the spectrum of idiopathic inflammatory myopathies (IIM) due to its distinct clinical presentation and refractoriness to current treatment approaches. One explanation for this resistance may be the engagement of cell-autonomous mechanisms that sustain or promote disease progression of IBM independent of inflammatory activity. In this study, we focused on senescence of tissue-resident cells as potential driver of disease. For this purpose, we compared IBM patients to non-diseased controls and immune-mediated necrotizing myopathy patients. Histopathological analysis suggested that cellular senescence is a prominent feature of IBM, primarily affecting non-myogenic cells. In-depth analysis by single nuclei RNA sequencing allowed for the deconvolution and study of muscle-resident cell populations. Among these, we identified a specific cluster of fibro-adipogenic progenitors (FAPs) that demonstrated key hallmarks of senescence, including a pro-inflammatory secretome, expression of p21, increased β-galactosidase activity, and engagement of senescence pathways. FAP function is required for muscle cell health with changes to their phenotype potentially proving detrimental. In this respect, the transcriptomic landscape of IBM was also characterized by changes to the myogenic compartment demonstrating a pronounced loss of type 2A myofibers and a rarefication of acetylcholine receptor expressing myofibers. IBM muscle cells also engaged a specific pro-inflammatory phenotype defined by intracellular complement activity and the expression of immunogenic surface molecules. Skeletal muscle cell dysfunction may be linked to FAP senescence by a change in the collagen composition of the latter. Senescent FAPs lose collagen type XV expression, which is required to support myofibers' structural integrity and neuromuscular junction formation in vitro. Taken together, this study demonstrates an altered phenotypical landscape of muscle-resident cells and that FAPs, and not myofibers, are the primary senescent cell type in IBM.
Collapse
Affiliation(s)
- Christopher Nelke
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Christina B Schroeter
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Lukas Theissen
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Corinna Preusse
- Department of Neuropathology, Charité-University Medicine Berlin, Bonhoefferweg 3, 10117, Berlin, Germany
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Saskia Räuber
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Vera Dobelmann
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Derya Cengiz
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Felix Kleefeld
- Department of Neurology, Charité-University Medicine Berlin, Bonhoefferweg 3, 10117, Berlin, Germany
| | - Andreas Roos
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, Centre for Neuromuscular Disorders in Children, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Benedikt Schoser
- Friedrich Baur Institute at the Department of Neurology, LMU University Hospital, LMU Munich, 80336, Munich, Germany
| | - Anna Brunn
- Institute of Neuropathology, Heinrich Heine University, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Eva Neuen-Jacob
- Institute of Neuropathology, Heinrich Heine University, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Jana Zschüntzsch
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité-University Medicine Berlin, Bonhoefferweg 3, 10117, Berlin, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.
| |
Collapse
|
6
|
Rhee S, Paik DT, Yang JY, Nagelberg D, Williams I, Tian L, Roth R, Chandy M, Ban J, Belbachir N, Kim S, Zhang H, Phansalkar R, Wong KM, King DA, Valdez C, Winn VD, Morrison AJ, Wu JC, Red-Horse K. Endocardial/endothelial angiocrines regulate cardiomyocyte development and maturation and induce features of ventricular non-compaction. Eur Heart J 2021; 42:4264-4276. [PMID: 34279605 PMCID: PMC8560211 DOI: 10.1093/eurheartj/ehab298] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 04/21/2021] [Accepted: 05/15/2021] [Indexed: 12/15/2022] Open
Abstract
AIMS Non-compaction cardiomyopathy is a devastating genetic disease caused by insufficient consolidation of ventricular wall muscle that can result in inadequate cardiac performance. Despite being the third most common cardiomyopathy, the mechanisms underlying the disease, including the cell types involved, are poorly understood. We have previously shown that endothelial cell-specific deletion of the chromatin remodeller gene Ino80 results in defective coronary vessel development that leads to ventricular non-compaction in embryonic mouse hearts. We aimed to identify candidate angiocrines expressed by endocardial and endothelial cells (ECs) in wildtype and LVNC conditions in Tie2Cre;Ino80fl/fltransgenic embryonic mouse hearts, and test the effect of these candidates on cardiomyocyte proliferation and maturation. METHODS AND RESULTS We used single-cell RNA-sequencing to characterize endothelial and endocardial defects in Ino80-deficient hearts. We observed a pathological endocardial cell population in the non-compacted hearts and identified multiple dysregulated angiocrine factors that dramatically affected cardiomyocyte behaviour. We identified Col15a1 as a coronary vessel-secreted angiocrine factor, downregulated by Ino80-deficiency, that functioned to promote cardiomyocyte proliferation. Furthermore, mutant endocardial and endothelial cells up-regulated expression of secreted factors, such as Tgfbi, Igfbp3, Isg15, and Adm, which decreased cardiomyocyte proliferation and increased maturation. CONCLUSIONS These findings support a model where coronary endothelial cells normally promote myocardial compaction through secreted factors, but that endocardial and endothelial cells can secrete factors that contribute to non-compaction under pathological conditions.
Collapse
Affiliation(s)
- Siyeon Rhee
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - David T Paik
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Johnson Y Yang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Ian Williams
- Department of Biology, Stanford University, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lei Tian
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robert Roth
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Mark Chandy
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jiyeon Ban
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Nadjet Belbachir
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Seokho Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Hao Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ragini Phansalkar
- Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ka Man Wong
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Devin A King
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Caroline Valdez
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ashby J Morrison
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
7
|
Martínez-Nieto G, Heljasvaara R, Heikkinen A, Kaski HK, Devarajan R, Rinne O, Henriksson C, Thomson E, von Hertzen C, Miinalainen I, Ruotsalainen H, Pihlajaniemi T, Karppinen SM. Deletion of Col15a1 Modulates the Tumour Extracellular Matrix and Leads to Increased Tumour Growth in the MMTV-PyMT Mouse Mammary Carcinoma Model. Int J Mol Sci 2021; 22:9978. [PMID: 34576139 PMCID: PMC8467152 DOI: 10.3390/ijms22189978] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/05/2021] [Accepted: 09/10/2021] [Indexed: 12/18/2022] Open
Abstract
Basement membrane (BM) zone-associated collagen XV (ColXV) has been shown to suppress the malignancy of tumour cells, and its restin domain can inhibit angiogenesis. In human breast cancer, as well as in many other human carcinomas, ColXV is lost from the epithelial BM zone prior to tumour invasion. Here, we addressed the roles of ColXV in breast carcinogenesis using the transgenic MMTV-PyMT mouse mammary carcinoma model. We show here for the first time that the inactivation of Col15a1 in mice leads to changes in the fibrillar tumour matrix and to increased mammary tumour growth. ColXV is expressed by myoepithelial and endothelial cells in mammary tumours and is lost from the ductal BM along with the loss of the myoepithelial layer during cancer progression while persisting in blood vessels and capillaries, even in invasive tumours. However, despite the absence of anti-angiogenic restin domain, neovascularisation was reduced rather than increased in the ColXV-deficient mammary tumours compared to controls. We also show that, in robust tumour cell transplantation models or in a chemical-induced fibrosarcoma model, the inactivation of Col15a1 does not affect tumour growth or angiogenesis. In conclusion, our results support the proposed tumour suppressor function of ColXV in mammary carcinogenesis and reveal diverse roles of this collagen in different cancer types.
Collapse
MESH Headings
- Animals
- Antigens, Polyomavirus Transforming/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Carcinogenesis/pathology
- Cell Proliferation
- Collagen/deficiency
- Collagen/genetics
- Collagen/metabolism
- Disease Models, Animal
- Extracellular Matrix/metabolism
- Female
- Fibrosarcoma/pathology
- Fibrosis
- Gene Deletion
- Gene Expression Regulation, Neoplastic
- Humans
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/pathology
- Mammary Neoplasms, Animal/ultrastructure
- Mammary Tumor Virus, Mouse/physiology
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Pathologic/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Stromal Cells/pathology
- Stromal Cells/ultrastructure
- Survival Analysis
- Mice
Collapse
Affiliation(s)
- Guillermo Martínez-Nieto
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
| | - Ritva Heljasvaara
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
| | - Anne Heikkinen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
- Biocenter Oulu, University of Oulu, 90220 Oulu, Finland;
| | - Hanne-Kaisa Kaski
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
| | - Raman Devarajan
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
| | - Otto Rinne
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
| | - Charlotta Henriksson
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
| | - Emmi Thomson
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
| | - Camilla von Hertzen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
| | | | - Heli Ruotsalainen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
| | - Taina Pihlajaniemi
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
| | - Sanna-Maria Karppinen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland; (G.M.-N.); (R.H.); (A.H.); (H.-K.K.); (R.D.); (O.R.); (C.H.); (E.T.); (C.v.H.); (H.R.); (T.P.)
| |
Collapse
|
8
|
Karamanos NK, Theocharis AD, Piperigkou Z, Manou D, Passi A, Skandalis SS, Vynios DH, Orian-Rousseau V, Ricard-Blum S, Schmelzer CEH, Duca L, Durbeej M, Afratis NA, Troeberg L, Franchi M, Masola V, Onisto M. A guide to the composition and functions of the extracellular matrix. FEBS J 2021; 288:6850-6912. [PMID: 33605520 DOI: 10.1111/febs.15776] [Citation(s) in RCA: 362] [Impact Index Per Article: 120.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Extracellular matrix (ECM) is a dynamic 3-dimensional network of macromolecules that provides structural support for the cells and tissues. Accumulated knowledge clearly demonstrated over the last decade that ECM plays key regulatory roles since it orchestrates cell signaling, functions, properties and morphology. Extracellularly secreted as well as cell-bound factors are among the major members of the ECM family. Proteins/glycoproteins, such as collagens, elastin, laminins and tenascins, proteoglycans and glycosaminoglycans, hyaluronan, and their cell receptors such as CD44 and integrins, responsible for cell adhesion, comprise a well-organized functional network with significant roles in health and disease. On the other hand, enzymes such as matrix metalloproteinases and specific glycosidases including heparanase and hyaluronidases contribute to matrix remodeling and affect human health. Several cell processes and functions, among them cell proliferation and survival, migration, differentiation, autophagy, angiogenesis, and immunity regulation are affected by certain matrix components. Structural alterations have been also well associated with disease progression. This guide on the composition and functions of the ECM gives a broad overview of the matrisome, the major ECM macromolecules, and their interaction networks within the ECM and with the cell surface, summarizes their main structural features and their roles in tissue organization and cell functions, and emphasizes the importance of specific ECM constituents in disease development and progression as well as the advances in molecular targeting of ECM to design new therapeutic strategies.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems- Functional Molecular Systems, Eggenstein-Leopoldshafen, Germany
| | - Sylvie Ricard-Blum
- University of Lyon, UMR 5246, ICBMS, Université Lyon 1, CNRS, Villeurbanne Cedex, France
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany.,Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2: Matrix Aging and Vascular Remodelling, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, Sweden
| | - Nikolaos A Afratis
- Department Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich, UK
| | - Marco Franchi
- Department for Life Quality Study, University of Bologna, Rimini, Italy
| | | | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
9
|
Lang AE, Lundquist EA. The Collagens DPY-17 and SQT-3 Direct Anterior-Posterior Migration of the Q Neuroblasts in C. elegans. J Dev Biol 2021; 9:jdb9010007. [PMID: 33669899 PMCID: PMC8006237 DOI: 10.3390/jdb9010007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/31/2022] Open
Abstract
Cell adhesion molecules and their extracellular ligands control morphogenetic events such as directed cell migration. The migration of neuroblasts and neural crest cells establishes the structure of the central and peripheral nervous systems. In C. elegans, the bilateral Q neuroblasts and their descendants undergo long-range migrations with left/right asymmetry. QR and its descendants on the right migrate anteriorly, and QL and its descendants on the left migrate posteriorly, despite identical patterns of cell division, cell death, and neuronal generation. The initial direction of protrusion of the Q cells relies on the left/right asymmetric functions of the transmembrane receptors UNC-40/DCC and PTP-3/LAR in the Q cells. Here, we show that Q cell left/right asymmetry of migration is independent of the GPA-16/Gα pathway which regulates other left/right asymmetries, including nervous system L/R asymmetry. No extracellular cue has been identified that guides initial Q anterior versus posterior migrations. We show that collagens DPY-17 and SQT-3 control initial Q direction of protrusion. Genetic interactions with UNC-40/DCC and PTP-3/LAR suggest that DPY-17 and SQT-3 drive posterior migration and might act with both receptors or in a parallel pathway. Analysis of mutants in other collagens and extracellular matrix components indicated that general perturbation of collagens and the extracellular matrix (ECM) did not result in directional defects, and that the effect of DPY-17 and SQT-3 on Q direction is specific. DPY-17 and SQT-3 are components of the cuticle, but a role in the basement membrane cannot be excluded. Possibly, DPY-17 and SQT-3 are part of a pattern in the cuticle and/or basement membrane that is oriented to the anterior–posterior axis of the animal and that is deciphered by the Q cells in a left–right asymmetric fashion. Alternatively, DPY-17 and SQT-3 might be involved in the production or stabilization of a guidance cue that directs Q migrations. In any case, these results describe a novel role for the DPY-17 and SQT-3 collagens in directing posterior Q neuroblast migration.
Collapse
|
10
|
Martins Cavaco AC, Dâmaso S, Casimiro S, Costa L. Collagen biology making inroads into prognosis and treatment of cancer progression and metastasis. Cancer Metastasis Rev 2021; 39:603-623. [PMID: 32447477 DOI: 10.1007/s10555-020-09888-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Progression through dissemination to tumor-surrounding tissues and metastasis development is a hallmark of cancer that requires continuous cell-to-cell interactions and tissue remodeling. In fact, metastization can be regarded as a tissue disease orchestrated by cancer cells, leading to neoplastic colonization of new organs. Collagen is a major component of the extracellular matrix (ECM), and increasing evidence suggests that it has an important role in cancer progression and metastasis. Desmoplasia and collagen biomarkers have been associated with relapse and death in cancer patients. Despite the increasing interest in ECM and in the desmoplastic process in tumor microenvironment as prognostic factors and therapeutic targets in cancer, further research is required for a better understanding of these aspects of cancer biology. In this review, published evidence correlating collagen with cancer prognosis is retrieved and analyzed, and the role of collagen and its fragments in cancer pathophysiology is discussed.
Collapse
Affiliation(s)
- Ana C Martins Cavaco
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Sara Dâmaso
- Serviço de Oncologia, Hospital de Santa Maria-CHULN, 1649-028, Lisboa, Portugal
| | - Sandra Casimiro
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Luís Costa
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal.
- Serviço de Oncologia, Hospital de Santa Maria-CHULN, 1649-028, Lisboa, Portugal.
| |
Collapse
|
11
|
Villesen IF, Daniels SJ, Leeming DJ, Karsdal MA, Nielsen MJ. Review article: the signalling and functional role of the extracellular matrix in the development of liver fibrosis. Aliment Pharmacol Ther 2020; 52:85-97. [PMID: 32419162 DOI: 10.1111/apt.15773] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/17/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Patients with liver fibrosis show a large heterogeneity, and for that reason effective treatments are still lacking. Emerging data suggest that there is more to fibrosis than previously understood. Opposed to earlier belief of being a passive scaffold for cells to reside in, the extracellular matrix (ECM) is now known to hold both signalling and functional properties important for the development of fibrosis. The interaction between the ECM and the collagen-producing cells determines the course of the disease but is still poorly understood. Exploring the dynamics of this interplay will aid in the development of effective treatments. AIM To summarise and discuss the latest advances in the pathogenesis of liver fibrosis as well as key mediators of early disease progression. METHODS Through literature search using databases including PubMed and Google Scholar, manuscripts published between 1961 and 2019 were included to assess both well-established and recent theories of fibrosis development. Both pre-clinical and clinical studies were included. RESULTS Fibrosis alters the structure of the ECM releasing signalling fragments with the potential to escalate disease severity. In a diseased liver, hepatic stellate cells and other fibroblasts, together with hepatocytes and sinusoidal cells, produce an excessive amount of collagens. The cell-to-collagen interactions are unique in the different liver aetiologies, generating ECM profiles with considerable patient-monitoring potential. CONCLUSIONS The local milieu in the injured area affects the course of fibrosis development in a site-specific manner. Future research should focus on the dissimilarities in the ECM profile between different aetiologies of liver fibrosis.
Collapse
Affiliation(s)
- Ida Falk Villesen
- Nordic Bioscience A/S, Herlev, Denmark.,University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
12
|
Bretaud S, Guillon E, Karppinen SM, Pihlajaniemi T, Ruggiero F. Collagen XV, a multifaceted multiplexin present across tissues and species. Matrix Biol Plus 2020; 6-7:100023. [PMID: 33543021 PMCID: PMC7852327 DOI: 10.1016/j.mbplus.2020.100023] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/06/2020] [Accepted: 01/06/2020] [Indexed: 01/09/2023] Open
Abstract
Type XV collagen is a non-fibrillar collagen that is associated with basement membranes and belongs to the multiplexin subset of the collagen superfamily. Collagen XV was initially studied because of its sequence homology with collagen XVIII/endostatin whose anti-angiogenic and anti-tumorigenic properties were subjects of wide interest in the past years. But during the last fifteen years, collagen XV has gained growing attention with increasing number of studies that have attributed new functions to this widely distributed collagen/proteoglycan hybrid molecule. Despite the cumulative evidence of its functional pleiotropy and its evolutionary conserved function, no review compiling the current state of the art about collagen XV is currently available. Here, we thus provide the first comprehensive view of the knowledge gathered so far on the molecular structure, tissue distribution and functions of collagen XV in development, tissue homeostasis and disease with an evolutionary perspective. We hope that our review will open new roads for promising research on collagen XV in the coming years. Type XV collagen belongs to the multiplexin subset of the collagen superfamily. It is evolutionarily conserved collagen and associated with basement membranes. This collagen/proteoglycan hybrid molecule contains an anti-angiogenic restin domain. It has important functions in the cardiovascular and the neuromuscular systems. Its expression is dysregulated in various diseases including cancers.
Collapse
Key Words
- Animal models
- BM, basement membrane
- BMZ, basement membrane zone
- COL, collagenous domain
- CS, chondroitin sulfate
- CSPG, chondroitin sulfate proteoglycan
- Collagen-related disease
- Collagens
- Development
- ECM, extracellular matrix
- Evolution
- Extracellular matrix
- GAG, glycosaminoglycan
- HFD, High fat diet
- HS, heparan sulfate
- HSPG, heparan sulfate proteoglycan
- Multiplexin
- NC, non-collagenous domain
- TD, trimerization domain
- TSPN, Thrombospondin-1 N-terminal like domain
- dpf, day post-fertilization
Collapse
Affiliation(s)
- Sandrine Bretaud
- Institut de Génomique Fonctionnelle de Lyon, ENS de Lyon, UMR CNRS 5242, University of Lyon, Lyon 69364, France
| | - Emilie Guillon
- Institut de Génomique Fonctionnelle de Lyon, ENS de Lyon, UMR CNRS 5242, University of Lyon, Lyon 69364, France
| | - Sanna-Maria Karppinen
- Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7C, FI-90230 Oulu, Finland
| | - Taina Pihlajaniemi
- Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7C, FI-90230 Oulu, Finland
| | - Florence Ruggiero
- Institut de Génomique Fonctionnelle de Lyon, ENS de Lyon, UMR CNRS 5242, University of Lyon, Lyon 69364, France
| |
Collapse
|
13
|
Xu S, Xu H, Wang W, Li S, Li H, Li T, Zhang W, Yu X, Liu L. The role of collagen in cancer: from bench to bedside. J Transl Med 2019; 17:309. [PMID: 31521169 PMCID: PMC6744664 DOI: 10.1186/s12967-019-2058-1] [Citation(s) in RCA: 431] [Impact Index Per Article: 86.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/06/2019] [Indexed: 02/06/2023] Open
Abstract
Collagen is the major component of the tumor microenvironment and participates in cancer fibrosis. Collagen biosynthesis can be regulated by cancer cells through mutated genes, transcription factors, signaling pathways and receptors; furthermore, collagen can influence tumor cell behavior through integrins, discoidin domain receptors, tyrosine kinase receptors, and some signaling pathways. Exosomes and microRNAs are closely associated with collagen in cancer. Hypoxia, which is common in collagen-rich conditions, intensifies cancer progression, and other substances in the extracellular matrix, such as fibronectin, hyaluronic acid, laminin, and matrix metalloproteinases, interact with collagen to influence cancer cell activity. Macrophages, lymphocytes, and fibroblasts play a role with collagen in cancer immunity and progression. Microscopic changes in collagen content within cancer cells and matrix cells and in other molecules ultimately contribute to the mutual feedback loop that influences prognosis, recurrence, and resistance in cancer. Nanoparticles, nanoplatforms, and nanoenzymes exhibit the expected gratifying properties. The pathophysiological functions of collagen in diverse cancers illustrate the dual roles of collagen and provide promising therapeutic options that can be readily translated from bench to bedside. The emerging understanding of the structural properties and functions of collagen in cancer will guide the development of new strategies for anticancer therapy.
Collapse
Affiliation(s)
- Shuaishuai Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Huaxiang Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Shuo Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Hao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Tianjiao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wuhu Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, People's Republic of China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China. .,Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China. .,Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Liang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, People's Republic of China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China. .,Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China. .,Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
14
|
Sun F, Wang Z, Yang Z, Li Y, Cui H, Liu C, Gao D, Wang F, Tan H. Characterization, bioactivity and pharmacokinetic study of a novel carbohydrate-peptide polymer: Glycol-split heparin-endostatin2 (GSHP-ES2). Carbohydr Polym 2019; 207:79-90. [DOI: 10.1016/j.carbpol.2018.11.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/03/2018] [Accepted: 11/14/2018] [Indexed: 01/28/2023]
|
15
|
Emerging roles of proteoglycans in cardiac remodeling. Int J Cardiol 2018; 278:192-198. [PMID: 30528626 DOI: 10.1016/j.ijcard.2018.11.125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 11/12/2018] [Accepted: 11/27/2018] [Indexed: 02/07/2023]
Abstract
Cardiac remodeling is the response of the heart to a range of pathological stimuli. Cardiac remodeling is initially adaptive; however, if sustained, it ultimately causes adverse clinical outcomes. Cardiomyocyte loss or hypertrophy, inflammation and fibrosis are hallmarks of cardiac remodeling. Proteoglycans, which are composed of glycosaminoglycans and a core protein, are a non-structural component of the extracellular matrix. The lack of proteoglycans results in cardiovascular defects during development. Moreover, emerging evidence has indicated that proteoglycans act as significant modifiers in ischemia and pressure overload-related cardiac remodeling. Proteoglycans may also provide novel therapeutic strategies for further improvement in the prognosis of cardiovascular diseases.
Collapse
|
16
|
Karppinen SM, Honkanen HK, Heljasvaara R, Riihilä P, Autio-Harmainen H, Sormunen R, Harjunen V, Väisänen MR, Väisänen T, Hurskainen T, Tasanen K, Kähäri VM, Pihlajaniemi T. Collagens XV and XVIII show different expression and localisation in cutaneous squamous cell carcinoma: type XV appears in tumor stroma, while XVIII becomes upregulated in tumor cells and lost from microvessels. Exp Dermatol 2018; 25:348-54. [PMID: 26660139 DOI: 10.1111/exd.12913] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2015] [Indexed: 12/17/2022]
Abstract
As the second most common skin malignancy, cutaneous squamous cell carcinoma (cSCC) is an increasing health concern, while its pathogenesis at molecular level remains largely unknown. We studied the expression and localisation of two homologous basement membrane (BM) collagens, types XV and XVIII, at different stages of cSCC. These collagens are involved in angiogenesis and tumorigenesis, but their role in cancer development is incompletely understood. Quantitative RT-PCR analysis revealed upregulation of collagen XVIII, but not collagen XV, in primary cSCC cells in comparison with normal human epidermal keratinocytes. In addition, the Ha-ras-transformed invasive cell line II-4 expressed high levels of collagen XVIII mRNA, indicating upregulation in the course of malignant transformation. Immunohistochemical analyses of a large human tissue microarray material showed that collagen XVIII is expressed by tumor cells from grade 1 onwards, while keratinocytes in normal skin and in premalignant lesions showed negative staining for it. Collagen XV appeared instead as deposits in the tumor stroma. Our findings in human cSCCs and in mouse cSCCs from the DMBA-TPA skin carcinogenesis model showed that collagen XVIII, but not collagen XV or the BM markers collagen IV or laminin, was selectively reduced in the tumor vasculature, and this decrease associated significantly with cancer progression. Our results demonstrate that collagens XV and XVIII are expressed in different sites of cSCC and may contribute in a distinct manner to processes related to cSCC tumorigenesis, identifying these collagens as potential biomarkers in the disease.
Collapse
Affiliation(s)
- Sanna-Maria Karppinen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Hanne-Kaisa Honkanen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Ritva Heljasvaara
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Pilvi Riihilä
- MediCity Research Laboratory and Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland
| | | | - Raija Sormunen
- Department of Pathology, University of Oulu/Oulu University Hospital, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Vanessa Harjunen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | | | - Timo Väisänen
- Department of Pathology, University of Oulu/Oulu University Hospital, Oulu, Finland
| | - Tiina Hurskainen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Oulu, Finland.,Department of Dermatology, University of Oulu/Oulu University Hospital, Oulu, Finland
| | - Kaisa Tasanen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Oulu, Finland.,Department of Dermatology, University of Oulu/Oulu University Hospital, Oulu, Finland
| | - Veli-Matti Kähäri
- MediCity Research Laboratory and Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland
| | - Taina Pihlajaniemi
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
17
|
Theocharis AD, Karamanos NK. Proteoglycans remodeling in cancer: Underlying molecular mechanisms. Matrix Biol 2017; 75-76:220-259. [PMID: 29128506 DOI: 10.1016/j.matbio.2017.10.008] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 02/07/2023]
Abstract
Extracellular matrix is a highly dynamic macromolecular network. Proteoglycans are major components of extracellular matrix playing key roles in its structural organization and cell signaling contributing to the control of numerous normal and pathological processes. As multifunctional molecules, proteoglycans participate in various cell functions during morphogenesis, wound healing, inflammation and tumorigenesis. Their interactions with matrix effectors, cell surface receptors and enzymes enable them with unique properties. In malignancy, extensive remodeling of tumor stroma is associated with marked alterations in proteoglycans' expression and structural variability. Proteoglycans exert diverse functions in tumor stroma in a cell-specific and context-specific manner and they mainly contribute to the formation of a permissive provisional matrix for tumor growth affecting tissue organization, cell-cell and cell-matrix interactions and tumor cell signaling. Proteoglycans also modulate cancer cell phenotype and properties, the development of drug resistance and tumor stroma angiogenesis. This review summarizes the proteoglycans remodeling and their novel biological roles in malignancies with particular emphasis to the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
18
|
Yu Y, Sun F, Zhang C, Wang Z, Liu J, Tan H. Study on glyco-modification of endostatin-derived synthetic peptide endostatin2 (ES2) by soluble chitooligosaccharide. Carbohydr Polym 2016; 154:204-13. [DOI: 10.1016/j.carbpol.2016.08.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/26/2016] [Accepted: 08/14/2016] [Indexed: 10/21/2022]
|
19
|
Slow Muscle Precursors Lay Down a Collagen XV Matrix Fingerprint to Guide Motor Axon Navigation. J Neurosci 2016; 36:2663-76. [PMID: 26937007 DOI: 10.1523/jneurosci.2847-15.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
UNLABELLED The extracellular matrix (ECM) provides local positional information to guide motoneuron axons toward their muscle target. Collagen XV is a basement membrane component mainly expressed in skeletal muscle. We have identified two zebrafish paralogs of the human COL15A1 gene, col15a1a and col15a1b, which display distinct expression patterns. Here we show that col15a1b is expressed and deposited in the motor path ECM by slow muscle precursors also called adaxial cells. We further demonstrate that collagen XV-B deposition is both temporally and spatially regulated before motor axon extension from the spinal cord in such a way that it remains in this region after the adaxial cells have migrated toward the periphery of the myotome. Loss- and gain-of-function experiments in zebrafish embryos demonstrate that col15a1b expression and subsequent collagen XV-B deposition and organization in the motor path ECM depend on a previously undescribed two-step mechanism involving Hedgehog/Gli and unplugged/MuSK signaling pathways. In silico analysis predicts a putative Gli binding site in the col15a1b proximal promoter. Using col15a1b promoter-reporter constructs, we demonstrate that col15a1b participates in the slow muscle genetic program as a direct target of Hedgehog/Gli signaling. Loss and gain of col15a1b function provoke pathfinding errors in primary and secondary motoneuron axons both at and beyond the choice point where axon pathway selection takes place. These defects result in muscle atrophy and compromised swimming behavior, a phenotype partially rescued by injection of a smyhc1:col15a1b construct. These reveal an unexpected and novel role for collagen XV in motor axon pathfinding and neuromuscular development. SIGNIFICANCE STATEMENT In addition to the archetypal axon guidance cues, the extracellular matrix provides local information that guides motor axons from the spinal cord to their muscle targets. Many of the proteins involved are unknown. Using the zebrafish model, we identified an unexpected role of the extracellular matrix collagen XV in motor axon pathfinding. We show that the synthesis of collagen XV-B by slow muscle precursors and its deposition in the common motor path are dependent on a novel two-step mechanism that determines axon decisions at a choice point during motor axonogenesis. Zebrafish and humans use common molecular cues and regulatory mechanisms for the neuromuscular system development. And as such, our study reveals COL15A1 as a candidate gene for orphan neuromuscular disorders.
Collapse
|
20
|
Zaferani A, Talsma DT, Yazdani S, Celie JWAM, Aikio M, Heljasvaara R, Navis GJ, Pihlajaniemi T, van den Born J. Basement membrane zone collagens XV and XVIII/proteoglycans mediate leukocyte influx in renal ischemia/reperfusion. PLoS One 2014; 9:e106732. [PMID: 25188209 PMCID: PMC4154753 DOI: 10.1371/journal.pone.0106732] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 08/09/2014] [Indexed: 01/23/2023] Open
Abstract
Collagen type XV and XVIII are proteoglycans found in the basement membrane zones of endothelial and epithelial cells, and known for their cryptic anti-angiogenic domains named restin and endostatin, respectively. Mutations or deletions of these collagens are associated with eye, muscle and microvessel phenotypes. We now describe a novel role for these collagens, namely a supportive role in leukocyte recruitment. We subjected mice deficient in collagen XV or collagen XVIII, and their compound mutant, as well as the wild-type control mice to bilateral renal ischemia/reperfusion, and evaluated renal function, tubular injury, and neutrophil and macrophage influx at different time points after ischemia/reperfusion. Five days after ischemia/reperfusion, the collagen XV, collagen XVIII and the compound mutant mice showed diminished serum urea levels compared to wild-type mice (all p<0.05). Histology showed reduced tubular damage, and decreased inflammatory cell influx in all mutant mice, which were more pronounced in the compound mutant despite increased expression of MCP-1 and TNF-α in double mutant mice compared to wildtype mice. Both type XV and type XVIII collagen bear glycosaminoglycan side chains and an in vitro approach with recombinant collagen XVIII fragments with variable glycanation indicated a role for these side chains in leukocyte migration. Thus, basement membrane zone collagen/proteoglycan hybrids facilitate leukocyte influx and tubular damage after renal ischemia/reperfusion and might be potential intervention targets for the reduction of inflammation in this condition.
Collapse
Affiliation(s)
- Azadeh Zaferani
- Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- * E-mail:
| | - Ditmer T. Talsma
- Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Saleh Yazdani
- Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Johanna W. A. M. Celie
- Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Mari Aikio
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Ritva Heljasvaara
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Gerjan J. Navis
- Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Taina Pihlajaniemi
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Jacob van den Born
- Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
21
|
Monboisse JC, Oudart JB, Ramont L, Brassart-Pasco S, Maquart FX. Matrikines from basement membrane collagens: a new anti-cancer strategy. Biochim Biophys Acta Gen Subj 2014; 1840:2589-98. [PMID: 24406397 DOI: 10.1016/j.bbagen.2013.12.029] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/19/2013] [Accepted: 12/31/2013] [Indexed: 01/04/2023]
Abstract
BACKGROUND Tumor microenvironment is a complex system composed of a largely altered extracellular matrix with different cell types that determine angiogenic responses and tumor progression. Upon the influence of hypoxia, tumor cells secrete cytokines that activate stromal cells to produce proteases and angiogenic factors. In addition to stromal ECM breakdown, proteases exert various pro- or anti-tumorigenic functions and participate in the release of various ECM fragments, named matrikines or matricryptins, capable to act as endogenous angiogenesis inhibitors and to limit tumor progression. SCOPE OF REVIEW We will focus on the matrikines derived from the NC1 domains of the different constitutive chains of basement membrane-associated collagens and mainly collagen IV. MAJOR CONCLUSIONS The putative targets of the matrikine control are the proliferation and invasive properties of tumor or inflammatory cells, and the angiogenic and lymphangiogenic responses. Collagen-derived matrikines such as canstatin, tumstatin or tetrastatin for example, decrease tumor growth in various cancer models. Their anti-cancer activities comprise anti-proliferative effects on tumor or endothelial cells by induction of apoptosis or cell cycle blockade and the induction of a loss of their migratory phenotype. They were used in various preclinical therapeutic strategies: i) induction of their overexpression by cancer cells or by the host cells, ii) use of recombinant proteins or synthetic peptides or structural analogues designed from the structure of the active sequences, iii) used in combined therapies with conventional chemotherapy or radiotherapy. GENERAL SIGNIFICANCE Collagen-derived matrikines strongly inhibited tumor growth in many preclinical cancer models in mouse. They constitute a new family of anti-cancer agents able to limit cancer progression. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Jean Claude Monboisse
- FRE CNRS/URCA 7369, Université de Reims Champagne Ardenne, UFR Médecine, 51 Rue Cognacq Jay, 51095 Reims Cedex, France; Laboratoire Central de Biochimie, CHU de Reims, France
| | - Jean Baptiste Oudart
- FRE CNRS/URCA 7369, Université de Reims Champagne Ardenne, UFR Médecine, 51 Rue Cognacq Jay, 51095 Reims Cedex, France; Laboratoire Central de Biochimie, CHU de Reims, France
| | - Laurent Ramont
- FRE CNRS/URCA 7369, Université de Reims Champagne Ardenne, UFR Médecine, 51 Rue Cognacq Jay, 51095 Reims Cedex, France; Laboratoire Central de Biochimie, CHU de Reims, France
| | - Sylvie Brassart-Pasco
- FRE CNRS/URCA 7369, Université de Reims Champagne Ardenne, UFR Médecine, 51 Rue Cognacq Jay, 51095 Reims Cedex, France
| | - François Xavier Maquart
- FRE CNRS/URCA 7369, Université de Reims Champagne Ardenne, UFR Médecine, 51 Rue Cognacq Jay, 51095 Reims Cedex, France; Laboratoire Central de Biochimie, CHU de Reims, France.
| |
Collapse
|
22
|
Integrin Recognition Motifs in the Human Collagens. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 819:127-42. [DOI: 10.1007/978-94-017-9153-3_9] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
23
|
Connelly JJ, Cherepanova OA, Doss JF, Karaoli T, Lillard TS, Markunas CA, Nelson S, Wang T, Ellis PD, Langford CF, Haynes C, Seo DM, Goldschmidt-Clermont PJ, Shah SH, Kraus WE, Hauser ER, Gregory SG. Epigenetic regulation of COL15A1 in smooth muscle cell replicative aging and atherosclerosis. Hum Mol Genet 2013; 22:5107-20. [PMID: 23912340 DOI: 10.1093/hmg/ddt365] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Smooth muscle cell (SMC) proliferation is a hallmark of vascular injury and disease. Global hypomethylation occurs during SMC proliferation in culture and in vivo during neointimal formation. Regardless of the programmed or stochastic nature of hypomethylation, identifying these changes is important in understanding vascular disease, as maintenance of a cells' epigenetic profile is essential for maintaining cellular phenotype. Global hypomethylation of proliferating aortic SMCs and concomitant decrease of DNMT1 expression were identified in culture during passage. An epigenome screen identified regions of the genome that were hypomethylated during proliferation and a region containing Collagen, type XV, alpha 1 (COL15A1) was selected by 'genomic convergence' for characterization. COL15A1 transcript and protein levels increased with passage-dependent decreases in DNA methylation and the transcript was sensitive to treatment with 5-Aza-2'-deoxycytidine, suggesting DNA methylation-mediated gene expression. Phenotypically, knockdown of COL15A1 increased SMC migration and decreased proliferation and Col15a1 expression was induced in an atherosclerotic lesion and localized to the atherosclerotic cap. A sequence variant in COL15A1 that is significantly associated with atherosclerosis (rs4142986, P = 0.017, OR = 1.434) was methylated and methylation of the risk allele correlated with decreased gene expression and increased atherosclerosis in human aorta. In summary, hypomethylation of COL15A1 occurs during SMC proliferation and the consequent increased gene expression may impact SMC phenotype and atherosclerosis formation. Hypomethylated genes, such as COL15A1, provide evidence for concomitant epigenetic regulation and genetic susceptibility, and define a class of causal targets that sit at the intersection of genetic and epigenetic predisposition in the etiology of complex disease.
Collapse
|
24
|
Lord MS, Whitelock JM. Recombinant production of proteoglycans and their bioactive domains. FEBS J 2013; 280:2490-510. [DOI: 10.1111/febs.12197] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/04/2013] [Accepted: 02/15/2013] [Indexed: 12/11/2022]
Affiliation(s)
- Megan S. Lord
- Graduate School of Biomedical Engineering; The University of New South Wales; Sydney; NSW; Australia
| | - John M. Whitelock
- Graduate School of Biomedical Engineering; The University of New South Wales; Sydney; NSW; Australia
| |
Collapse
|
25
|
Chagnot C, Listrat A, Astruc T, Desvaux M. Bacterial adhesion to animal tissues: protein determinants for recognition of extracellular matrix components. Cell Microbiol 2012; 14:1687-96. [PMID: 22882798 DOI: 10.1111/cmi.12002] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 07/25/2012] [Accepted: 07/26/2012] [Indexed: 12/13/2022]
Abstract
The extracellular matrix (ECM) is present within all animal tissues and organs. Actually, it surrounds the eukaryotic cells composing the four basic tissue types, i.e. epithelial, muscle, nerve and connective. ECM does not solely refer to connective tissue but composes all tissues where its composition, structure and organization vary from one tissue to another. Constituted of the four main fibrous proteins, i.e. collagen, fibronectin, laminin and elastin, ECM components form a highly structured and functional network via specific interactions. From the basement membrane to interstitial matrix, further heterogeneity exists in the organization of the ECM in various tissues and organs also depending on their physiological state. Back to a molecular level, bacterial proteins represent the most significant part of the microbial surface components recognizing adhesive matrix molecules (MSCRAMM). These cell surface proteins are secreted and localized differently in monoderm and diderm-LPS bacteria. While one collagen-binding domain (CBD) and different fibronectin-binding domains (FBD1 to 8) have been registered in databases, much remains to be learned on specific binding to other ECM proteins via single or supramolecular protein structures. Besides theinteraction of bacterial proteins with individual ECM components, this review aims at stressing the importance of fully considering the ECM at supramolecular, cellular, tissue and organ levels. This conceptual view should not be overlooked to rigorously comprehend the physiology of bacterial interaction from commensal to pathogenic species.
Collapse
|
26
|
Boidin-Wichlacz C, Vergote D, Slomianny C, Jouy N, Salzet M, Tasiemski A. Morphological and functional characterization of leech circulating blood cells: role in immunity and neural repair. Cell Mol Life Sci 2012; 69:1717-31. [PMID: 22159559 PMCID: PMC11115165 DOI: 10.1007/s00018-011-0897-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 11/22/2011] [Accepted: 11/24/2011] [Indexed: 11/26/2022]
Abstract
Unlike most invertebrates, annelids possess a closed vascular system distinct from the coelomic liquid. The morphology and the function of leech blood cells are reported here. We have demonstrated the presence of a unique cell type which participates in various immune processes. In contrast to the mammalian spinal cord, the leech CNS is able to regenerate and restore function after injury. The close contact of the blood with the nerve cord also led us to explore the participation of blood in neural repair. Our data evidenced that, in addition to exerting peripheral immune functions, leech blood optimizes CNS neural repair through the release of neurotrophic substances. Circulating blood cells also appeared able to infiltrate the injured CNS where, in conjunction with microglia, they limit the formation of a scar. In mammals, CNS injury leads to the generation of a glial scar that blocks the mechanism of regeneration by preventing axonal regrowth. The results presented here constitute the first description of neuroimmune functions of invertebrate blood cells. Understanding the basic function of the peripheral circulating cells and their interactions with lesioned CNS in the leech would allow us to acquire insights into the complexity of the neuroimmune response of the injured mammalian brain.
Collapse
Affiliation(s)
- Céline Boidin-Wichlacz
- Laboratoire de Neuroimmunologie et Neurochimie Evolutive, CNRS, FRE3249, Université de Lille 1, Villeneuve d'Ascq, France
| | | | | | | | | | | |
Collapse
|
27
|
Mutolo MJ, Morris KJ, Leir SH, Caffrey TC, Lewandowska MA, Hollingsworth MA, Harris A. Tumor suppression by collagen XV is independent of the restin domain. Matrix Biol 2012; 31:285-9. [PMID: 22531369 DOI: 10.1016/j.matbio.2012.03.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 03/18/2012] [Accepted: 03/23/2012] [Indexed: 10/28/2022]
Abstract
Non-fibrillar collagen XV is a chondroitin sulfate modified glycoprotein that is associated with the basement membrane zone in many tissues. Its precise functions remain to be fully elucidated though it clearly plays a critical role in the structural integrity of the extracellular matrix. Loss of collagen XV from the basement membrane zone precedes invasion of a number of tumor types and we previously showed that collagen XV functions as a dose-dependent suppressor of tumorigenicity in cervical carcinoma cells. The carboxyl terminus of another non-fibrillar collagen (XVIII) is cleaved to produce endostatin, which has anti-angiogenic effects and thus may act as a tumor suppressor in vivo. Since collagen XV has structural similarity with collagen XVIII, its C-terminal restin domain could confer tumor suppressive functions on the molecule, though our previous data did not support this. We now show that expression of collagen XV enhances the adhesion of cervical carcinoma cells to collagen I in vitro as does the N-terminus and collagenous regions of collagen XV, but not the restin domain. Destruction of a cysteine residue in the collagenous region that is critical for intermolecular interactions of collagen XV abolished the enhanced adhesion to collagen I. Finally, we demonstrate that unlike full length collagen XV, expression of the restin domain alone does not suppress tumorigenicity of cervical carcinoma cells in vivo; hence, this process is dependent on functions and interactions of other parts of the protein.
Collapse
Affiliation(s)
- Michael J Mutolo
- Human Molecular Genetics Program, Children's Memorial Research Center, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, 2300 Children's Plaza #211, Chicago, IL 60614, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Abstract
Collagens are the most abundant proteins in mammals. The collagen family comprises 28 members that contain at least one triple-helical domain. Collagens are deposited in the extracellular matrix where most of them form supramolecular assemblies. Four collagens are type II membrane proteins that also exist in a soluble form released from the cell surface by shedding. Collagens play structural roles and contribute to mechanical properties, organization, and shape of tissues. They interact with cells via several receptor families and regulate their proliferation, migration, and differentiation. Some collagens have a restricted tissue distribution and hence specific biological functions.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- Institut de Biologie et Chimie des Protéines, UMR 5086 CNRS, Université Lyon 1, Lyon, 69367, France.
| |
Collapse
|
30
|
Lack of collagen XV impairs peripheral nerve maturation and, when combined with laminin-411 deficiency, leads to basement membrane abnormalities and sensorimotor dysfunction. J Neurosci 2010; 30:14490-501. [PMID: 20980607 DOI: 10.1523/jneurosci.2644-10.2010] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although the Schwann cell basement membrane (BM) is required for normal Schwann cell terminal differentiation, the role of BM-associated collagens in peripheral nerve maturation is poorly understood. Collagen XV is a BM zone component strongly expressed in peripheral nerves, and we show that its absence in mice leads to loosely packed axons in C-fibers and polyaxonal myelination. The simultaneous lack of collagen XV and another peripheral nerve component affecting myelination, laminin α4, leads to severely impaired radial sorting and myelination, and the maturation of the nerve is permanently compromised, contrasting with the slow repair observed in Lama4-/- single knock-out mice. Moreover, the Col15a1-/-;Lama4-/- double knock-out (DKO) mice initially lack C-fibers and, even over 1 year of age have only a few, abnormal C-fibers. The Lama4-/- knock-out results in motor and tactile sensory impairment, which is exacerbated by a simultaneous Col15a1-/- knock-out, whereas sensitivity to heat-induced pain is increased in the DKO mice. Lack of collagen XV results in slower sensory nerve conduction, whereas the Lama4-/- and DKO mice exhibit increased sensory nerve action potentials and decreased compound muscle action potentials; x-ray diffraction revealed less mature myelin in the sciatic nerves of the latter than in controls. Ultrastructural analyses revealed changes in the Schwann cell BM in all three mutants, ranging from severe (DKO) to nearly normal (Col15a1-/-). Collagen XV thus contributes to peripheral nerve maturation and C-fiber formation, and its simultaneous deletion from neural BM zones with laminin α4 leads to a DKO phenotype distinct from those of both single knock-outs.
Collapse
|
31
|
Rasi K, Piuhola J, Czabanka M, Sormunen R, Ilves M, Leskinen H, Rysä J, Kerkelä R, Janmey P, Heljasvaara R, Peuhkurinen K, Vuolteenaho O, Ruskoaho H, Vajkoczy P, Pihlajaniemi T, Eklund L. Collagen XV Is Necessary for Modeling of the Extracellular Matrix and Its Deficiency Predisposes to Cardiomyopathy. Circ Res 2010; 107:1241-52. [DOI: 10.1161/circresaha.110.222133] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Rationale:
The extracellular matrix (ECM) is a major determinant of the structural integrity and functional properties of the myocardium in common pathological conditions, and changes in vasculature contribute to cardiac dysfunction. Collagen (Col) XV is preferentially expressed in the ECM of cardiac muscle and microvessels.
Objective:
We aimed to characterize the ECM, cardiovascular function and responses to elevated cardiovascular load in mice lacking Col XV (
Col15a1
−/−
) to define its functional role in the vasculature and in age- and hypertension-associated myocardial remodeling.
Methods and Results:
Cardiac structure and vasculature were analyzed by light and electron microscopy. Cardiac function, intraarterial blood pressure, microhemodynamics, and gene expression profiles were studied using echocardiography, telemetry, intravital microscopy, and PCR, respectively. Experimental hypertension was induced with angiotensin II or with a nitric oxide synthesis inhibitor. Under basal conditions, lack of Col XV resulted in increased permeability and impaired microvascular hemodynamics, distinct early-onset and age-dependent defects in heart structure and function, a poorly organized fibrillar collagen matrix with marked interstitial deposition of nonfibrillar protein aggregates, increased tissue stiffness, and irregularly organized cardiomyocytes. In response to experimental hypertension,
Col15a1
gene expression was increased in the left ventricle of wild-type mice, and mRNA expression of natriuretic peptides (ANP and BNP) and ECM modeling were abnormal in
Col15a1
−/−
mice.
Conclusions:
Col XV is necessary for ECM organization in the heart, and for the structure and functions of microvessels. Col XV deficiency leads to a complex cardiac phenotype and predisposes the subject to pathological responses under cardiac stress.
Collapse
Affiliation(s)
- Karolina Rasi
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Jarkko Piuhola
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Marcus Czabanka
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Raija Sormunen
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Mika Ilves
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Hanna Leskinen
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Jaana Rysä
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Risto Kerkelä
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Paul Janmey
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Ritva Heljasvaara
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Keijo Peuhkurinen
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Olli Vuolteenaho
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Heikki Ruskoaho
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Peter Vajkoczy
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Taina Pihlajaniemi
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| | - Lauri Eklund
- From the Oulu Center for Cell-Matrix Research, Biocenter Oulu, and Department of Medical Biochemistry and Molecular Biology (K.R., R.H., T.P., L.E.); Biocenter Oulu and Department of Pharmacology and Toxicology (J.P., H.L., J.R., R.K., H.R.); Department of Internal Medicine, Division of Cardiology (J.P.); Biocenter Oulu and Department of Pathology (R.S.); and Department of Physiology (M.I., O.V.), University of Oulu, Finland; Department of Neurosurgery (M.C., P.V.), Charité-Universitätsmedizin
| |
Collapse
|