1
|
Yamamoto K, Scilabra SD, Bonelli S, Jensen A, Scavenius C, Enghild JJ, Strickland DK. Novel insights into the multifaceted and tissue-specific roles of the endocytic receptor LRP1. J Biol Chem 2024; 300:107521. [PMID: 38950861 PMCID: PMC11325810 DOI: 10.1016/j.jbc.2024.107521] [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: 03/13/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Receptor-mediated endocytosis provides a mechanism for the selective uptake of specific molecules thereby controlling the composition of the extracellular environment and biological processes. The low-density lipoprotein receptor-related protein 1 (LRP1) is a widely expressed endocytic receptor that regulates cellular events by modulating the levels of numerous extracellular molecules via rapid endocytic removal. LRP1 also participates in signalling pathways through this modulation as well as in the interaction with membrane receptors and cytoplasmic adaptor proteins. LRP1 SNPs are associated with several diseases and conditions such as migraines, aortic aneurysms, cardiopulmonary dysfunction, corneal clouding, and bone dysmorphology and mineral density. Studies using Lrp1 KO mice revealed a critical, nonredundant and tissue-specific role of LRP1 in regulating various physiological events. However, exactly how LRP1 functions to regulate so many distinct and specific processes is still not fully clear. Our recent proteomics studies have identified more than 300 secreted proteins that either directly interact with LRP1 or are modulated by LRP1 in various tissues. This review will highlight the remarkable ability of this receptor to regulate secreted molecules in a tissue-specific manner and discuss potential mechanisms underpinning such specificity. Uncovering the depth of these "hidden" specific interactions modulated by LRP1 will provide novel insights into a dynamic and complex extracellular environment that is involved in diverse biological and pathological processes.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.
| | - Simone D Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy
| | - Simone Bonelli
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Anders Jensen
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Goel K, Chhetri A, Ludhiadch A, Munshi A. Current Update on Categorization of Migraine Subtypes on the Basis of Genetic Variation: a Systematic Review. Mol Neurobiol 2024; 61:4804-4833. [PMID: 38135854 DOI: 10.1007/s12035-023-03837-3] [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: 09/26/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
Migraine is a complex neurovascular disorder that is characterized by severe behavioral, sensory, visual, and/or auditory symptoms. It has been labeled as one of the ten most disabling medical illnesses in the world by the World Health Organization (Aagaard et al Sci Transl Med 6(237):237ra65, 2014). According to a recent report by the American Migraine Foundation (Shoulson et al Ann Neurol 25(3):252-9, 1989), around 148 million people in the world currently suffer from migraine. On the basis of presence of aura, migraine is classified into two major subtypes: migraine with aura (Aagaard et al Sci Transl Med 6(237):237ra65, 2014) and migraine without aura. (Aagaard K et al Sci Transl Med 6(237):237ra65, 2014) Many complex genetic mechanisms have been proposed in the pathophysiology of migraine but specific pathways associated with the different subtypes of migraine have not yet been explored. Various approaches including candidate gene association studies (CGAS) and genome-wide association studies (Fan et al Headache: J Head Face Pain 54(4):709-715, 2014). have identified the genetic markers associated with migraine and its subtypes. Several single nucleotide polymorphisms (Kaur et al Egyp J Neurol, Psychiatry Neurosurg 55(1):1-7, 2019) within genes involved in ion homeostasis, solute transport, synaptic transmission, cortical excitability, and vascular function have been associated with the disorder. Currently, the diagnosis of migraine is majorly behavioral with no focus on the genetic markers and thereby the therapeutic intervention specific to subtypes. Therefore, there is a need to explore genetic variants significantly associated with MA and MO as susceptibility markers in the diagnosis and targets for therapeutic interventions in the specific subtypes of migraine. Although the proper characterization of pathways based on different subtypes is yet to be studied, this review aims to make a first attempt to compile the information available on various genetic variants and the molecular mechanisms involved with the development of MA and MO. An attempt has also been made to suggest novel candidate genes based on their function to be explored by future research.
Collapse
Affiliation(s)
- Kashish Goel
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Aakash Chhetri
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Abhilash Ludhiadch
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Anjana Munshi
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401.
| |
Collapse
|
3
|
Zelaya H, Grunz K, Nguyen TS, Habibi A, Witzler C, Reyda S, Gonzalez-Menendez I, Quintanilla-Martinez L, Bosmann M, Weiler H, Ruf W. Nucleic acid sensing promotes inflammatory monocyte migration through biased coagulation factor VIIa signaling. Blood 2024; 143:845-857. [PMID: 38096370 PMCID: PMC10940062 DOI: 10.1182/blood.2023021149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 11/30/2023] [Indexed: 03/08/2024] Open
Abstract
ABSTRACT Protease activated receptors (PARs) are cleaved by coagulation proteases and thereby connect hemostasis with innate immune responses. Signaling of the tissue factor (TF) complex with factor VIIa (FVIIa) via PAR2 stimulates extracellular signal-regulated kinase (ERK) activation and cancer cell migration, but functions of cell autonomous TF-FVIIa signaling in immune cells are unknown. Here, we show that myeloid cell expression of FVII but not of FX is crucial for inflammatory cell recruitment to the alveolar space after challenge with the double-stranded viral RNA mimic polyinosinic:polycytidylic acid [Poly(I:C)]. In line with these data, genetically modified mice completely resistant to PAR2 cleavage but not FXa-resistant PAR2-mutant mice are protected from lung inflammation. Poly(I:C)-stimulated migration of monocytes/macrophages is dependent on ERK activation and mitochondrial antiviral signaling (MAVS) but independent of toll-like receptor 3 (TLR3). Monocyte/macrophage-synthesized FVIIa cleaving PAR2 is required for integrin αMβ2-dependent migration on fibrinogen but not for integrin β1-dependent migration on fibronectin. To further dissect the downstream signaling pathway, we generated PAR2S365/T368A-mutant mice deficient in β-arrestin recruitment and ERK scaffolding. This mutation reduces cytosolic, but not nuclear ERK phosphorylation by Poly(I:C) stimulation, and prevents macrophage migration on fibrinogen but not fibronectin after stimulation with Poly(I:C) or CpG-B, a single-stranded DNA TLR9 agonist. In addition, PAR2S365/T368A-mutant mice display markedly reduced immune cell recruitment to the alveolar space after Poly(I:C) challenge. These results identify TF-FVIIa-PAR2-β-arrestin-biased signaling as a driver for lung infiltration in response to viral nucleic acids and suggest potential therapeutic interventions specifically targeting TF-VIIa signaling in thrombo-inflammation.
Collapse
Affiliation(s)
- Hortensia Zelaya
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- National Scientific and Technical Research Council (CONICET), Tucuman, Argentina
| | - Kristin Grunz
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - T. Son Nguyen
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Anxhela Habibi
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | - Claudius Witzler
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Sabine Reyda
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University, Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University, Tübingen, Germany
| | - Markus Bosmann
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Pulmonary Center, Department of Medicine and Department of Pathology & Laboratory Medicine, Boston University, Boston, MA
| | | | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
| |
Collapse
|
4
|
Pan S, Hale AT, Lemieux ME, Raval DK, Garton TP, Sadler B, Mahaney KB, Strahle JM. Iron homeostasis and post-hemorrhagic hydrocephalus: a review. Front Neurol 2024; 14:1287559. [PMID: 38283681 PMCID: PMC10811254 DOI: 10.3389/fneur.2023.1287559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
Iron physiology is regulated by a complex interplay of extracellular transport systems, coordinated transcriptional responses, and iron efflux mechanisms. Dysregulation of iron metabolism can result in defects in myelination, neurotransmitter synthesis, and neuronal maturation. In neonates, germinal matrix-intraventricular hemorrhage (GMH-IVH) causes iron overload as a result of blood breakdown in the ventricles and brain parenchyma which can lead to post-hemorrhagic hydrocephalus (PHH). However, the precise mechanisms by which GMH-IVH results in PHH remain elusive. Understanding the molecular determinants of iron homeostasis in the developing brain may lead to improved therapies. This manuscript reviews the various roles iron has in brain development, characterizes our understanding of iron transport in the developing brain, and describes potential mechanisms by which iron overload may cause PHH and brain injury. We also review novel preclinical treatments for IVH that specifically target iron. Understanding iron handling within the brain and central nervous system may provide a basis for preventative, targeted treatments for iron-mediated pathogenesis of GMH-IVH and PHH.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Andrew T. Hale
- Department of Neurosurgery, University of Alabama at Birmingham School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mackenzie E. Lemieux
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Dhvanii K. Raval
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Thomas P. Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Brooke Sadler
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Hematology and Oncology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Jennifer M. Strahle
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Orthopedic Surgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
5
|
Dutta SD, Ganguly K, Patil TV, Randhawa A, Lim KT. Unraveling the potential of 3D bioprinted immunomodulatory materials for regulating macrophage polarization: State-of-the-art in bone and associated tissue regeneration. Bioact Mater 2023; 28:284-310. [PMID: 37303852 PMCID: PMC10248805 DOI: 10.1016/j.bioactmat.2023.05.014] [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: 11/17/2022] [Revised: 04/29/2023] [Accepted: 05/20/2023] [Indexed: 06/13/2023] Open
Abstract
Macrophage-assisted immunomodulation is an alternative strategy in tissue engineering, wherein the interplay between pro-inflammatory and anti-inflammatory macrophage cells and body cells determines the fate of healing or inflammation. Although several reports have demonstrated that tissue regeneration depends on spatial and temporal regulation of the biophysical or biochemical microenvironment of the biomaterial, the underlying molecular mechanism behind immunomodulation is still under consideration for developing immunomodulatory scaffolds. Currently, most fabricated immunomodulatory platforms reported in the literature show regenerative capabilities of a particular tissue, for example, endogenous tissue (e.g., bone, muscle, heart, kidney, and lungs) or exogenous tissue (e.g., skin and eye). In this review, we briefly introduced the necessity of the 3D immunomodulatory scaffolds and nanomaterials, focusing on material properties and their interaction with macrophages for general readers. This review also provides a comprehensive summary of macrophage origin and taxonomy, their diverse functions, and various signal transduction pathways during biomaterial-macrophage interaction, which is particularly helpful for material scientists and clinicians for developing next-generation immunomodulatory scaffolds. From a clinical standpoint, we briefly discussed the role of 3D biomaterial scaffolds and/or nanomaterial composites for macrophage-assisted tissue engineering with a special focus on bone and associated tissues. Finally, a summary with expert opinion is presented to address the challenges and future necessity of 3D bioprinted immunomodulatory materials for tissue engineering.
Collapse
Affiliation(s)
- Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V. Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
6
|
Lin L, Hu K. Macrophage Function Modulated by tPA Signaling in Mouse Experimental Kidney Disease Models. Int J Mol Sci 2023; 24:11067. [PMID: 37446244 DOI: 10.3390/ijms241311067] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Macrophage infiltration and accumulation is a hallmark of chronic kidney disease. Tissue plasminogen activator (tPA) is a serine protease regulating the homeostasis of blood coagulation, fibrinolysis, and matrix degradation, and has been shown to act as a cytokine to trigger various receptor-mediated intracellular signal pathways, modulating macrophage function in response to kidney injury. In this review, we discuss the current understanding of tPA-modulated macrophage function and underlying signaling mechanisms during kidney fibrosis and inflammation.
Collapse
Affiliation(s)
- Ling Lin
- Division of Nephrology, Department of Medicine, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Kebin Hu
- Division of Nephrology, Department of Medicine, Penn State University College of Medicine, Hershey, PA 17033, USA
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
7
|
Langlois B, Martin J, Schneider C, Hachet C, Terryn C, Rioult D, Martiny L, Théret L, Salesse S, Dedieu S. LRP-1-dependent control of calpain expression and activity: A new mechanism regulating thyroid carcinoma cell adhesion. Front Oncol 2022; 12:981927. [PMID: 36052226 PMCID: PMC9424861 DOI: 10.3389/fonc.2022.981927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
The low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional endocytic receptor mediating the clearance of various molecules from the extracellular matrix. LRP1 also regulates cell surface expression of matrix receptors by modulating both extracellular and intracellular signals, though current knowledge of the underlying mechanisms remains partial in the frame of cancer cells interaction with matricellular substrates. In this study we identified that LRP1 downregulates calpain activity and calpain 2 transcriptional expression in an invasive thyroid carcinoma cell model. LRP1-dependent alleviation of calpain activity limits cell-matrix attachment strength and contributes to FTC133 cells invasive abilities in a modified Boyden chamber assays. In addition, using enzymatic assays and co-immunoprecipitation experiments, we demonstrated that LRP1 exerts post-translational inhibition of calpain activity through PKA-dependent phosphorylation of calpain-2. This LRP-1 dual mode of control of calpain activity fine-tunes carcinoma cell spreading. We showed that LRP1-mediated calpain inhibition participates in talin-positive focal adhesions dissolution and limits β1-integrin expression at carcinoma cell surface. In conclusion, we identified an additional and innovative intracellular mechanism which demonstrates LRP-1 pro-motile action in thyroid cancer cells. LRP-1 ability to specifically control calpain-2 expression and activity highlights a novel facet of its de-adhesion receptor status.
Collapse
Affiliation(s)
- Benoit Langlois
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
- *Correspondence: Benoit Langlois,
| | - Julie Martin
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Christophe Schneider
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Cathy Hachet
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Christine Terryn
- Plate-Forme Imagerie Cellulaire et Tissulaire (PICT), Université de Reims Champagne-Ardenne, UFR Médecine, Reims, France
| | - Damien Rioult
- Plateau Technique Mobile de Cytométrie Environnementale MOBICYTE, Université de Reims Champagne-Ardenne/INERIS, Reims, France
| | - Laurent Martiny
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Louis Théret
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Stéphanie Salesse
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Stéphane Dedieu
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| |
Collapse
|
8
|
Luo Z, Liu Y, Li H, Zhou Y, Peng Y, Lin X, Fang Y, Wan J, Wei B. Systematic Review and Meta-Analysis of SERPINE1 4G/5G Insertion/Deletion Variant With Circulating Lipid Levels. Front Cardiovasc Med 2022; 9:859979. [PMID: 35811710 PMCID: PMC9260103 DOI: 10.3389/fcvm.2022.859979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background Recent studies have shown that the 4G/5G insertion/deletion variant of SERPINE1 (rs1799889) is closely linked to coronary artery disease (CAD). This study aims to clarify the effects of the rs1799889 variant on lipid levels and to insight into the mechanisms underlying the rs1799889 variant and CAD. Methods and Results By searching PubMed and the Cochrane databases for studies published before 31 October 2021, 40 studies conducted on a total of 13,117 subjects were included for the analysis. The consistent findings for the effects of the 5G allele of rs1799889 variant on lipid metabolism were the significantly decreased triglycerides (TG) [standardized mean difference (SMD) = –0.12, 95% CI = –0.21 to 0.03, P = 0.01], total cholesterol (TC) (SMD = –0.12, 95% CI = –0.17 to 0.06, P < 0.001), and low-density lipoprotein cholesterol (LDL-C) (SMD = –0.13, 95% CI = –0.23 to 0.03, P = 0.01) levels. Intriguingly, the significant effects of the rs1799889 variant on LDL-C (SMD = –0.15, 95% CI = –0.26 to 0.05, P < 0.01) and TC (SMD = –0.17, 95% CI = –0.27 to 0.07, P < 0.01) levels were primarily observed in the Asian population. However, the significant effect of the rs1799889 variant on high-density lipoprotein cholesterol (HDL-C) (SMD = 0.26, 95% CI = 0.03–0.48, P = 0.03) levels was detected only in female subjects. Conclusion The rs1799889 variant of SERPINE1 is a protective genetic factor against CAD, the Asian population with the 5G allele of the rs1799889 variant may have a reduced CAD risk.
Collapse
Affiliation(s)
- Zhi Luo
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yang Liu
- Department of Endocrinology, China Resources and WISCO General Hospital, Wuhan, China
| | - Hang Li
- Department of Geratology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yawen Zhou
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yuanyuan Peng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Xuan Lin
- Department of Endocrinology, China Resources and WISCO General Hospital, Wuhan, China
| | - Ying Fang
- Department of Endocrinology, China Resources and WISCO General Hospital, Wuhan, China
| | - Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
- *Correspondence: Jing Wan,
| | - Baozhu Wei
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
- Baozhu Wei,
| |
Collapse
|
9
|
Tang MY, Gorin FA, Lein PJ. Review of evidence implicating the plasminogen activator system in blood-brain barrier dysfunction associated with Alzheimer's disease. AGEING AND NEURODEGENERATIVE DISEASES 2022; 2. [PMID: 35156107 PMCID: PMC8830591 DOI: 10.20517/and.2022.05] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Elucidating the pathogenic mechanisms of Alzheimer’s disease (AD) to identify therapeutic targets has been the focus of many decades of research. While deposition of extracellular amyloid-beta plaques and intraneuronal neurofibrillary tangles of hyperphosphorylated tau have historically been the two characteristic hallmarks of AD pathology, therapeutic strategies targeting these proteinopathies have not been successful in the clinics. Neuroinflammation has been gaining more attention as a therapeutic target because increasing evidence implicates neuroinflammation as a key factor in the early onset of AD disease progression. The peripheral immune response has emerged as an important contributor to the chronic neuroinflammation associated with AD pathophysiology. In this context, the plasminogen activator system (PAS), also referred to as the vasculature’s fibrinolytic system, is emerging as a potential factor in AD pathogenesis. Evolving evidence suggests that the PAS plays a role in linking chronic peripheral inflammatory conditions to neuroinflammation in the brain. While the PAS is better known for its peripheral functions, components of the PAS are expressed in the brain and have been demonstrated to alter neuroinflammation and blood-brain barrier (BBB) permeation. Here, we review plasmin-dependent and -independent mechanisms by which the PAS modulates the BBB in AD pathogenesis and discuss therapeutic implications of these observations.
Collapse
Affiliation(s)
- Mei-Yun Tang
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Fredric A Gorin
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA.,Department of Neurology, School of Medicine, University of California, Davis, CA 95616, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
10
|
Zhang L. Meet the Editorial Board Member. Curr Drug Targets 2022. [DOI: 10.2174/138945012302220118152057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- L. Zhang
- University of Maryland School of Medicine
Baltimore, MD
USA
| |
Collapse
|
11
|
Seillier C, Hélie P, Petit G, Vivien D, Clemente D, Le Mauff B, Docagne F, Toutirais O. Roles of the tissue-type plasminogen activator in immune response. Cell Immunol 2021; 371:104451. [PMID: 34781155 PMCID: PMC8577548 DOI: 10.1016/j.cellimm.2021.104451] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/06/2021] [Accepted: 10/29/2021] [Indexed: 11/30/2022]
Abstract
The COVID-19 pandemic has once again
brought to the forefront the existence of a tight link between the
coagulation/fibrinolytic system and the immunologic processes.
Tissue-type plasminogen activator (tPA) is a serine protease with a key
role in fibrinolysis by converting plasminogen into plasmin that can
finally degrade fibrin clots. tPA is released in the blood by endothelial
cells and hepatocytes but is also produced by various types of immune
cells including T cells and monocytes. Beyond its role on hemostasis, tPA
is also a potent modulator of inflammation and is involved in the
regulation of several inflammatory diseases. Here, after a brief
description of tPA structure, we review its new functions in adaptive
immunity focusing on T cells and antigen presenting cells. We intend to
synthesize the recent knowledge on proteolysis- and receptor-mediated
effects of tPA on immune response in physiological and pathological
context.
Collapse
Affiliation(s)
- Célia Seillier
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Pauline Hélie
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Gautier Petit
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France; Department of Immunology and Histocompatibility (HLA), Caen University Hospital, CHU Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France; Department of Clinical Research, Caen University Hospital, CHU Caen, France
| | - Diego Clemente
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Brigitte Le Mauff
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France; Department of Immunology and Histocompatibility (HLA), Caen University Hospital, CHU Caen, France
| | - Fabian Docagne
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Olivier Toutirais
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France; Department of Immunology and Histocompatibility (HLA), Caen University Hospital, CHU Caen, France.
| |
Collapse
|
12
|
Insulin resistance is linked to a specific profile of immune activation in human subjects. Sci Rep 2021; 11:12314. [PMID: 34112902 PMCID: PMC8192510 DOI: 10.1038/s41598-021-91758-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/19/2021] [Indexed: 11/08/2022] Open
Abstract
We tested the hypothesis that a particular immune activation profile might be correlated with insulin resistance in a general population. By measuring 43 markers of immune, endothelial, and coagulation activation, we have previously shown that five different immune activation profiles may be distinguished in 150 volunteers. One of these profiles, Profile 2, characterized by CD4+ T cell senescence, inflammation, monocyte, B cell, and endothelial activation, presented elevated insulinemia, glycemia, triglyceridemia, and γ-glutamyl transferase, a marker of liver injury, in comparison with other profiles. Our data are compatible with a model in which a particular immune activation profile might favor the development of insulin resistance and metabolic syndrome. In this hypothesis, identification of this profile, that is feasible with only 3 markers with an error rate of 5%, might allow to personalize the screening and prevention of metabolic syndrome-driven morbidities as liver steatosis.
Collapse
|
13
|
Chen J, Su Y, Pi S, Hu B, Mao L. The Dual Role of Low-Density Lipoprotein Receptor-Related Protein 1 in Atherosclerosis. Front Cardiovasc Med 2021; 8:682389. [PMID: 34124208 PMCID: PMC8192809 DOI: 10.3389/fcvm.2021.682389] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
Low-density lipoprotein receptor–related protein-1 (LRP1) is a large endocytic and signaling receptor belonging to the LDL receptor (LDLR) gene family and that is widely expressed in several tissues. LRP1 comprises a large extracellular domain (ECD; 515 kDa, α chain) and a small intracellular domain (ICD; 85 kDa, β chain). The deletion of LRP1 leads to embryonic lethality in mice, revealing a crucial but yet undefined role in embryogenesis and development. LRP1 has been postulated to participate in numerous diverse physiological and pathological processes ranging from plasma lipoprotein homeostasis, atherosclerosis, tumor evolution, and fibrinolysis to neuronal regeneration and survival. Many studies using cultured cells and in vivo animal models have revealed the important roles of LRP1 in vascular remodeling, foam cell biology, inflammation and atherosclerosis. However, its role in atherosclerosis remains controversial. LRP1 not only participates in the removal of atherogenic lipoproteins and proatherogenic ligands in the liver but also mediates the uptake of aggregated LDL to promote the formation of macrophage- and vascular smooth muscle cell (VSMC)-derived foam cells, which causes a prothrombotic transformation of the vascular wall. The dual and opposing roles of LRP1 may also represent an interesting target for atherosclerosis therapeutics. This review highlights the influence of LRP1 during atherosclerosis development, focusing on its dual role in vascular cells and immune cells.
Collapse
Affiliation(s)
- Jiefang Chen
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Su
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shulan Pi
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Mao
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Tseng YJ, Lee CH, Chen WY, Yang JL, Tzeng HT. Inhibition of PAI-1 Blocks PD-L1 Endocytosis and Improves the Response of Melanoma Cells to Immune Checkpoint Blockade. J Invest Dermatol 2021; 141:2690-2698.e6. [PMID: 34000287 DOI: 10.1016/j.jid.2021.03.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/17/2021] [Accepted: 03/28/2021] [Indexed: 12/28/2022]
Abstract
Immune checkpoint molecules, especially PD-1 and its ligand PD-L1, act as a major mechanism of cancer immune evasion. Although anti-PD-1/PD-L1 monotherapy increases therapeutic efficacy in melanoma treatment, only a subset of patients exhibits long-term tumor remission, and the underlying mechanism of resistance to PD-1/PD-L1 inhibitors remains unclear. In this study, we demonstrated that cell surface retention of PD-L1 is inversely correlated with PAI-1 expression in vitro, in vivo, and in clinical specimens. Moreover, extracellular PAI-1 induced the internalization of surface-expressed PD-L1 by triggering clathrin-mediated endocytosis. The endocytosed PD-L1 was transported to lysosomes for degradation by endolysosomal systems, resulting in the reduction of surface PD-L1. Notably, inhibition of PAI-1 by pharmacological inhibitor with tiplaxtinin led to elevated PD-L1 expression on the plasma membrane, both in vitro and in vivo. Strikingly, targeting PAI-1 by tiplaxtinin treatment synergizes with anti-PD-L1 immune checkpoint blockade therapy in a syngeneic murine model of melanoma. Our findings demonstrate a role for PAI-1 activity in immune checkpoint modulation by promoting surface PD-L1 for lysosomal degradation and provides an insight into the combination of PAI-1 inhibition and anti-PD-L1 immunotherapy as a promising therapeutic regimen for melanoma treatment.
Collapse
Affiliation(s)
- Yu-Ju Tseng
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City, Taiwan
| | - Chih-Hung Lee
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City, Taiwan
| | - Wei-Yu Chen
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City, Taiwan
| | - Jenq-Lin Yang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City, Taiwan
| | - Hong-Tai Tzeng
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City, Taiwan.
| |
Collapse
|
15
|
Lamers C, Plüss CJ, Ricklin D. The Promiscuous Profile of Complement Receptor 3 in Ligand Binding, Immune Modulation, and Pathophysiology. Front Immunol 2021; 12:662164. [PMID: 33995387 PMCID: PMC8118671 DOI: 10.3389/fimmu.2021.662164] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/12/2021] [Indexed: 12/19/2022] Open
Abstract
The β2-integrin receptor family has a broad spectrum of physiological functions ranging from leukocyte adhesion, cell migration, activation, and communication to the phagocytic uptake of cells and particles. Among the members of this family, complement receptor 3 (CR3; CD11b/CD18, Mac-1, αMβ2) is particularly promiscuous in its functional profile and ligand selectivity. There are close to 100 reported structurally unrelated ligands for CR3, and while many ligands appear to cluster at the αMI domain, molecular details about binding modes remain largely elusive. The versatility of CR3 is reflected in its functional portfolio, which includes prominent roles in the removal of invaders and cell debris, induction of tolerance and synaptic pruning, and involvement in the pathogenesis of numerous autoimmune and chronic inflammatory pathologies. While CR3 is an interesting therapeutic target for immune modulation due to these known pathophysiological associations, drug development efforts are limited by concerns of potential interference with host defense functions and, most importantly, an insufficient molecular understanding of the interplay between ligand binding and functional impact. Here, we provide a systematic summary of the various interaction partners of CR3 with a focus on binding mechanisms and functional implications. We also discuss the roles of CR3 as an immune receptor in health and disease, as an activation marker in research and diagnostics, and as a therapeutic target.
Collapse
Affiliation(s)
- Christina Lamers
- Molecular Pharmacy Unit, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | | | | |
Collapse
|
16
|
Das L, Banki MA, Azmoon P, Pizzo D, Gonias SL. Enzymatically Inactive Tissue-Type Plasminogen Activator Reverses Disease Progression in the Dextran Sulfate Sodium Mouse Model of Inflammatory Bowel Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:590-601. [PMID: 33465348 PMCID: PMC8101053 DOI: 10.1016/j.ajpath.2021.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
Enzymatically inactive tissue-type plasminogen activator (EI-tPA) does not activate fibrinolysis, but interacts with the N-methyl-d-aspartate receptor (NMDA-R) and low-density lipoprotein receptor-related protein-1 (LRP1) in macrophages to block innate immune system responses mediated by toll-like receptors. Herein, we examined the ability of EI-tPA to treat colitis in mice, induced by dextran sulfate sodium. In two separate studies, designed to generate colitis of differing severity, a single dose of EI-tPA administered after inflammation established significantly improved disease parameters. EI-tPA-treated mice demonstrated improved weight gain. Stools improved in character and became hemoccult negative. Abdominal tenderness decreased. Colon shortening significantly decreased in EI-tPA-treated mice, suggesting attenuation of irreversible tissue damage and remodeling. Furthermore, histopathologic evidence of disease decreased in the distal 25% of the colon in EI-tPA-treated mice. EI-tPA did not decrease the number of CD45-positive leukocytes or F4/80-positive macrophage-like cells detected in extracts of colons from dextran sulfate sodium-treated mice as assessed by flow cytometry. However, multiple colon cell types expressed the NMDA-R, suggesting the ability of diverse cells, including CD3-positive cells, CD103-positive cells, Ly6G-positive cells, and epithelial cell adhesion molecule-positive epithelial cells to respond to EI-tPA. Mesenchymal cells that line intestinal crypts and provide barrier function expressed LRP1, thereby representing another potential target for EI-tPA. These results demonstrate that the NMDA-R/LRP1 receptor system may be a target for drug development in diseases characterized by tissue damage and chronic inflammation.
Collapse
Affiliation(s)
- Lipsa Das
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Michael A Banki
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Pardis Azmoon
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Donald Pizzo
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Steven L Gonias
- Department of Pathology, University of California San Diego, La Jolla, California.
| |
Collapse
|
17
|
Plasminogen Activator Inhibitor-1 Secretion by Autophagy Contributes to Melanoma Resistance to Chemotherapy through Tumor Microenvironment Modulation. Cancers (Basel) 2021; 13:cancers13061253. [PMID: 33809137 PMCID: PMC7999393 DOI: 10.3390/cancers13061253] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/11/2021] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Dysregulation of tumor autophagy is implicated in cancer progression and chemotherapeutic response. It is unclear how tumor autophagy modulates tumor microenvironment and thereby contributes to chemoresistance. In this study, we found that autophagy-dependent plasminogen activator inhibitor (PAI)-1 secretion contributed to melanoma resistance to mitoxantrone (MitoX), a chemotherapeutic agent clinically used for treating various types of cancers (but not melanoma), by shaping a pro-tumoral microenvironment. Disruption of autophagy activity or targeting PAI-1 pharmacologically reshaped a tumor-suppressive immune microenvironment and enhanced the susceptibility of melanoma to MitoX in vivo. Overall, the results show that targeting autophagy/PAI-1 axis can serve as a novel strategy to repurpose MitoX-based chemotherapy. Abstract Autophagy plays a crucial role in maintenance of cellular homeostasis via intracellular signaling pathways, lysosomal degradation of selective cargo and mediating protein secretion. Dysregulation of autophagy has been implicated in tumorigenesis, tumor progression, and resistance to therapy. However, the mechanism of autophagy-dependent secretion involved in the responsiveness to chemotherapy is poorly understood. In this study, we showed that mitoxantrone (MitoX), a chemotherapeutic agent used for treating various cancers but not melanoma, induced autophagy in melanoma cells in vitro and in vivo. We also found that plasminogen activator inhibitor (PAI)-1 secretion by MitoX-induced autophagy modulated the pro-tumoral microenvironment. Attenuation of PAI-1 activity using a specific inhibitor, tiplaxtinin (TPX), or by targeting the autophagy gene, Becn1, induced efficient antitumor immunity, thereby overcoming the resistance to MitoX in vivo. Of note, the therapeutic efficacy of TPX was abolished in MitoX-treated Becn1-defective tumors. Collectively, our results demonstrate that tumor autophagy-dependent PAI-1 secretion impairs the therapeutic efficacy of MitoX and highlight targeting of tumor autophagy or its secretory cargo, PAI-1, as a novel strategy to repurpose MitoX-based chemotherapy for melanoma treatment.
Collapse
|
18
|
Sakamoto H, Koma YI, Higashino N, Kodama T, Tanigawa K, Shimizu M, Fujikawa M, Nishio M, Shigeoka M, Kakeji Y, Yokozaki H. PAI-1 derived from cancer-associated fibroblasts in esophageal squamous cell carcinoma promotes the invasion of cancer cells and the migration of macrophages. J Transl Med 2021; 101:353-368. [PMID: 33311557 PMCID: PMC7892342 DOI: 10.1038/s41374-020-00512-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 01/06/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) contribute to the progression of various cancers. Previously, we reported the significance of CAFs in esophageal squamous cell carcinoma (ESCC); however, the functions of CAFs in the ESCC microenvironment remain unknown. To investigate CAFs' function, we established an indirect coculture assay between human bone marrow-derived mesenchymal stem cells (MSCs) and ESCC cells. Cocultured MSCs expressed more fibroblast activation protein, one of the markers of CAFs, compared with monocultured MSCs. Therefore, we defined cocultured MSCs as CAF-like cells. To identify molecules associated with the ESCC progression in CAFs, we conducted a cDNA microarray analysis on monocultured MSCs and CAF-like cells to compare their gene expression profiles. We found that SERPINE1, which encodes plasminogen activator inhibitor-1 (PAI-1), was more abundant in CAF-like cells than in monocultured MSCs, and the PAI-1 derived from CAF-like cells induced the abilities of migration and invasion in both ESCC cells and macrophages by the Akt and Erk1/2 signaling pathways via the low-density lipoprotein receptor-related protein 1 (LRP1), which is a PAI-1 receptor. Based on immunohistochemistry assays of ESCC tissues, higher expression levels of PAI-1 and LRP1 were correlated with poor prognosis in ESCC patients. These results suggest that the PAI-1/LRP1 axis contributes to the progression of ESCC, making it a potential target for ESCC therapy.
Collapse
Affiliation(s)
- Hiroki Sakamoto
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yu-Ichiro Koma
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Nobuhide Higashino
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takayuki Kodama
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kohei Tanigawa
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Masaki Shimizu
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Masataka Fujikawa
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Mari Nishio
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Manabu Shigeoka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yoshihiro Kakeji
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
19
|
Mineo C. Lipoprotein receptor signalling in atherosclerosis. Cardiovasc Res 2021; 116:1254-1274. [PMID: 31834409 DOI: 10.1093/cvr/cvz338] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/01/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
The founding member of the lipoprotein receptor family, low-density lipoprotein receptor (LDLR) plays a major role in the atherogenesis through the receptor-mediated endocytosis of LDL particles and regulation of cholesterol homeostasis. Since the discovery of the LDLR, many other structurally and functionally related receptors have been identified, which include low-density lipoprotein receptor-related protein (LRP)1, LRP5, LRP6, very low-density lipoprotein receptor, and apolipoprotein E receptor 2. The scavenger receptor family members, on the other hand, constitute a family of pattern recognition proteins that are structurally diverse and recognize a wide array of ligands, including oxidized LDL. Among these are cluster of differentiation 36, scavenger receptor class B type I and lectin-like oxidized low-density lipoprotein receptor-1. In addition to the initially assigned role as a mediator of the uptake of macromolecules into the cell, a large number of studies in cultured cells and in in vivo animal models have revealed that these lipoprotein receptors participate in signal transduction to modulate cellular functions. This review highlights the signalling pathways by which these receptors influence the process of atherosclerosis development, focusing on their roles in the vascular cells, such as macrophages, endothelial cells, smooth muscle cells, and platelets. Human genetics of the receptors is also discussed to further provide the relevance to cardiovascular disease risks in humans. Further knowledge of the vascular biology of the lipoprotein receptors and their ligands will potentially enhance our ability to harness the mechanism to develop novel prophylactic and therapeutic strategies against cardiovascular diseases.
Collapse
Affiliation(s)
- Chieko Mineo
- Department of Pediatrics and Cell Biology, Center for Pulmonary and Vascular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA
| |
Collapse
|
20
|
Plasminogen activator inhibitor-1 reduces cardiac fibrosis and promotes M2 macrophage polarization in inflammatory cardiomyopathy. Basic Res Cardiol 2021; 116:1. [PMID: 33432417 PMCID: PMC7801308 DOI: 10.1007/s00395-020-00840-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/15/2020] [Indexed: 11/01/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) has a cardioprotective function in mice by repressing cardiac fibrosis through TGF-β and plasminogen-mediated pathways. In addition it is known to be involved in the recruitment and polarization of monocytes/macrophages towards a M2 phenotype in cancer. Here, we investigated the expression of PAI-1 in human dilated cardiomyopathy (DCM) and inflammatory dilated cardiomyopathy (DCMi) and its effect on cardiac fibrosis and macrophage polarization. We retrospectively analyzed endomyocardial biopsies (EMBs) of patients with DCM or DCMi for PAI-1 expression by immunohistochemistry. Furthermore, EMBs were evaluated for the content of fibrotic tissue, number of activated myofibroblasts, TGF-β expression, as well as for M1 and M2 macrophages. Patients with high-grade DCMi (DCMi-high, CD3+ lymphocytes > 30 cells/mm2) had significantly increased PAI-1 levels compared to DCM and low-grade DCMi patients (DCMi-low, CD3+ lymphocytes = 14-30 cells/mm2) (15.5 ± 0.4% vs. 1.0 ± 0.1% and 4.0 ± 0.1%, p ≤ 0.001). Elevated PAI-1 expression in DCMi-high subjects was associated with a diminished degree of cardiac fibrosis, decreased levels of TGF-β and reduced number of myofibroblasts. In addition, DCMi-high patients revealed an increased proportion of non-classical M2 macrophages towards classical M1 macrophages, indicating M2 macrophage-favoring properties of PAI-1 in inflammatory cardiomyopathies. Our findings give evidence that elevated expression of cardiac PAI-1 in subjects with high-grade DCMi suppresses fibrosis by inhibiting TGF-β and myofibroblast activation. Moreover, our data indicate that PAI-1 is involved in the polarization of M2 macrophages in the heart. Thus, PAI-1 could serve as a potential prognostic biomarker and as a possible therapeutic target in inflammatory cardiomyopathies.
Collapse
|
21
|
Mizrachi T, Gur-Wahnon D, Al-Roof Higazi A, Brenner T. Role of tissue plasminogen activator in clinical aggravation of experimental autoimmune encephalomyelitis and its therapeutic potential. Cell Immunol 2020; 348:104040. [PMID: 31955841 DOI: 10.1016/j.cellimm.2020.104040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/05/2020] [Accepted: 01/09/2020] [Indexed: 10/25/2022]
Abstract
Tissue plasminogen activator (tPA), a component of the plasminogen activator (PA) system, is elevated in inflammatory neurological disorders. In the present study, we explored the immunomodulatory activity of tPA in experimental autoimmune encephalomyelitis (EAE). The EAE was treated with two catalytic inactive tPA variant proteins: S(481)A and S(481)A + KHRR(296-299)AAAA. EAE-induced tPA-/- mice presented with markedly more severe disease than wt EAE mice. Further, treatment with tPA variants, demonstrated a significant suppression of disease severity in tPA-/- and wt mice. Immunological evaluation showed that specific T-cell reactivity was markedly reduced in the tPA-/- animals, as indicated by decreased T-cell reactivity and reduction in T-regulatory cells. The current findings indicate that tPA plays a role in the pathogenesis of EAE. Moreover, successful amelioration of EAE was achieved by administration of tPA variant proteins. This might mean that these proteins have potential for the immunomodulation of neuroinflammation.
Collapse
Affiliation(s)
- Tehila Mizrachi
- Laboratory of Neuroimmunology, Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, PO Box 12000, Jerusalem, Israel
| | - Devorah Gur-Wahnon
- Laboratory of Neuroimmunology, Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, PO Box 12000, Jerusalem, Israel
| | - Abd Al-Roof Higazi
- Department of Biochemistry, Hadassah-Hebrew University Medical Center, PO Box 12000, Jerusalem, Israel
| | - Talma Brenner
- Laboratory of Neuroimmunology, Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, PO Box 12000, Jerusalem, Israel.
| |
Collapse
|
22
|
Moschny N, Jahn K, Bajbouj M, Maier HB, Ballmaier M, Khan AQ, Pollak C, Bleich S, Frieling H, Neyazi A. DNA Methylation of the t-PA Gene Differs Between Various Immune Cell Subtypes Isolated From Depressed Patients Receiving Electroconvulsive Therapy. Front Psychiatry 2020; 11:571. [PMID: 32636772 PMCID: PMC7319092 DOI: 10.3389/fpsyt.2020.00571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 06/03/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Major depressive disorder (MDD) represents a tremendous health threat to the world's population. Electroconvulsive therapy (ECT) is the most effective treatment option for refractory MDD patients. Ample evidence suggests brain-derived neurotrophic factor (BDNF) to play a crucial role in ECT's mode of action. Tissue-type plasminogen activator (t-PA) and plasminogen activator inhibitor-1 (PAI-1) are involved in BDNF production. HYPOTHESIS The DNA methylation of gene regions encoding for t-PA and PAI-1 might be a suitable biomarker for ECT response prediction. METHODS We withdrew blood from two cohorts of treatment-resistant MDD patients receiving ECT. In the first cohort (n = 59), blood was collected at baseline only. To evaluate DNA methylation changes throughout the treatment course, we acquired a second group (n = 28) and took blood samples at multiple time points. DNA isolated from whole blood and defined immune cell subtypes (B cells, monocytes, natural killer cells, and T cells) served for epigenetic analyses. RESULTS Mixed linear models (corrected for multiple testing by Sidak's post-hoc test) revealed (1) no detectable baseline blood DNA methylation differences between ECT remitters (n = 33) and non-remitters (n = 53) in the regions analyzed, but (2) a significant difference in t-PA's DNA methylation between the investigated immune cell subtypes instead (p < 0.00001). This difference remained stable throughout the treatment course, showed no acute changes after ECT, and was independent of clinical remission. CONCLUSION DNA methylation of both proteins seems to play a minor role in ECT's mechanisms. Generally, we recommend using defined immune cell subtypes (instead of whole blood only) for DNA methylation analyses.
Collapse
Affiliation(s)
- Nicole Moschny
- Laboratory for Molecular Neurosciences, Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience, Hannover Graduate School for Veterinary Pathobiology, Neuroinfectiology, and Translational Medicine (HGNI), Hannover, Germany
| | - Kirsten Jahn
- Laboratory for Molecular Neurosciences, Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Malek Bajbouj
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Hannah Benedictine Maier
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | | | - Abdul Qayyum Khan
- Laboratory for Molecular Neurosciences, Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Christoph Pollak
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Stefan Bleich
- Center for Systems Neuroscience, Hannover Graduate School for Veterinary Pathobiology, Neuroinfectiology, and Translational Medicine (HGNI), Hannover, Germany.,Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Helge Frieling
- Laboratory for Molecular Neurosciences, Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience, Hannover Graduate School for Veterinary Pathobiology, Neuroinfectiology, and Translational Medicine (HGNI), Hannover, Germany.,Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Alexandra Neyazi
- Center for Systems Neuroscience, Hannover Graduate School for Veterinary Pathobiology, Neuroinfectiology, and Translational Medicine (HGNI), Hannover, Germany.,Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| |
Collapse
|
23
|
Kubala MH, Punj V, Placencio-Hickok VR, Fang H, Fernandez GE, Sposto R, DeClerck YA. Plasminogen Activator Inhibitor-1 Promotes the Recruitment and Polarization of Macrophages in Cancer. Cell Rep 2019; 25:2177-2191.e7. [PMID: 30463014 DOI: 10.1016/j.celrep.2018.10.082] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 06/27/2018] [Accepted: 10/23/2018] [Indexed: 12/14/2022] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) has a pro-tumorigenic function via its pro-angiogenic and anti-apoptotic activities. Here, we demonstrate that PAI-1 promotes the recruitment and M2 polarization of monocytes/macrophages through different structural domains. Its LRP1 interacting domain regulated macrophage migration, while its C-terminal uPA interacting domain promoted M2 macrophage polarization through activation of p38MAPK and nuclear factor κB (NF-κB) and induction of an autocrine interleukin (IL)-6/STAT3 activation pathway. We then show in several experiments in mice that expression of PAI-1 is associated with increased tumorigenicity, increased presence of M2 macrophages, higher levels of IL-6, and increased STAT3 phosphorylation in macrophages. Strong positive correlations between PAI-1, IL-6, and CD163 (M2 marker) expression were also found by meta-analysis of transcriptome data in many human cancers. Altogether, these data provide evidence for a mechanism explaining the paradoxical pro-tumorigenic function of PAI-1 in cancer.
Collapse
Affiliation(s)
- Marta Helena Kubala
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, CA 90033, USA; The Saban Research Institute of Children's Hospital, Los Angeles, CA 90027, USA
| | - Vasu Punj
- Division of Hematology, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Veronica Rae Placencio-Hickok
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, CA 90033, USA; The Saban Research Institute of Children's Hospital, Los Angeles, CA 90027, USA
| | - Hua Fang
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, CA 90033, USA; The Saban Research Institute of Children's Hospital, Los Angeles, CA 90027, USA
| | - G Esteban Fernandez
- The Saban Research Institute of Children's Hospital, Los Angeles, CA 90027, USA
| | - Richard Sposto
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, CA 90033, USA; The Saban Research Institute of Children's Hospital, Los Angeles, CA 90027, USA; Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yves Albert DeClerck
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, CA 90033, USA; The Saban Research Institute of Children's Hospital, Los Angeles, CA 90027, USA; Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
24
|
Takahashi T, Minematsu T, Murayama R, Nakagami G, Mori T, Sanada H. Catheter tips are a possible resource for biological study on catheter failure. Drug Discov Ther 2019; 13:280-287. [PMID: 31723100 DOI: 10.5582/ddt.2019.01073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Few studies have investigated the molecular mechanisms of catheter failure (CF). Herein, we performed histological and molecular biological analyses of the catheter tip to demonstrate its potential as a resource for biological investigation. Additionally, we searched for risk factors for the development of inflammation and coagulation, which are pathological conditions clarified by biological analysis. The CF group included 30 failed catheters involving thrombus and subcutaneous edema identified by ultrasonography. The No-CF group included 26 catheters with no complications. The removed catheter tips were fixed for hematoxylin-eosin (HE) staining with the application of a real-time reverse transcriptase polymerase chain reaction for eukaryotic 18S ribosomal RNA (rRNA), interleukin 1β, tumor necrosis factor α, tissue plasminogen activator, and plasminogen activator inhibitor 1 (SERPINE1). HE staining identified attached nuclear cells on the inner surfaces of both CF and No-CF catheters. The 18S rRNA was amplified in all samples. The expression level of SERPINE1 was significantly higher in the CF group than in the No-CF group (p = 0.01), whereas the expression levels of other genes did not differ between the groups. Symptoms of CF associated with the expression of SERPINE1 were analyzed. The catheter being in contact with blood vessels during placement was a suggested factor related to the high expression of SERPINE1 (p = 0.04). Catheter tips are a potential resource for biological investigation, and expression analysis of the attached cells can reflect the pathological condition of the catheterized tissue. Further studies using catheter tips are required to elucidate the molecular mechanisms of CF.
Collapse
Affiliation(s)
- Toshiaki Takahashi
- Department of Life Support Technology (Molten), Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeo Minematsu
- Department of Skincare Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Care Innovation, Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryoko Murayama
- Division of Care Innovation, Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Advanced Nursing Technology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Gojiro Nakagami
- Division of Care Innovation, Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taketoshi Mori
- Department of Life Support Technology (Molten), Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Care Innovation, Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiromi Sanada
- Division of Care Innovation, Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
25
|
Li W, Lu Y, Lou Y, Zhao S, Cui W, Wang Y, Luo M, Sun J, Miao L. FFNT25 ameliorates unilateral ureteral obstruction-induced renal fibrosis. Ren Fail 2019; 41:419-426. [PMID: 31140898 PMCID: PMC6566665 DOI: 10.1080/0886022x.2019.1612430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 01/06/2023] Open
Abstract
Renal fibrosis is a common pathological feature of chronic kidney disease (CKD) patients who progress to end-stage renal disease (ESRD). With the increasing incidence of CKD, it is of importance to develop effective therapies that blunt development of renal fibrosis. FFNT25 is a newly developed molecular compound that could be used to prevent fibrosis. In this study, we administered FFNT25 to rats following unilateral ureteral obstruction (UUO) to investigate its anti-fibrosis mechanism. Thirty-two Sprague-Dawley rats were randomly divided into four groups: (1) control (normal rats), (2) sham-operated, (3) UUO-operated + vehicle, and (4) UUO-operated + FFNT25. Two weeks after UUO, the rats were gavaged with either FFNT25 (20.6 mg/kg/day) or vehicle for two weeks. Serum, urine, and kidney samples were collected at the end of the study. FFNT25 reduced levels of renal fibrosis and decreased mRNA and protein levels of extracellular matrix (ECM) markers α-smooth muscle actin (α-SMA) and plasminogen activator inhibitor-1 (PAI-1) following UUO compared to vehicle treatment (n = 8, p<.05). The current results indicate that FFNT25 can affect both the production and degradation of collagen fibers to reduce fibrosis.
Collapse
Affiliation(s)
- Wen Li
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Yue Lu
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Yan Lou
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Shiyue Zhao
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Wenpeng Cui
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Yangwei Wang
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Manyu Luo
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Jing Sun
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Lining Miao
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Au DT, Arai AL, Fondrie WE, Muratoglu SC, Strickland DK. Role of the LDL Receptor-Related Protein 1 in Regulating Protease Activity and Signaling Pathways in the Vasculature. Curr Drug Targets 2019; 19:1276-1288. [PMID: 29749311 DOI: 10.2174/1389450119666180511162048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 04/24/2018] [Accepted: 04/24/2018] [Indexed: 12/22/2022]
Abstract
Aortic aneurysms represent a significant clinical problem as they largely go undetected until a rupture occurs. Currently, an understanding of mechanisms leading to aneurysm formation is limited. Numerous studies clearly indicate that vascular smooth muscle cells play a major role in the development and response of the vasculature to hemodynamic changes and defects in these responses can lead to aneurysm formation. The LDL receptor-related protein 1 (LRP1) is major smooth muscle cell receptor that has the capacity to mediate the endocytosis of numerous ligands and to initiate and regulate signaling pathways. Genetic evidence in humans and mouse models reveal a critical role for LRP1 in maintaining the integrity of the vasculature. Understanding the mechanisms by which this is accomplished represents an important area of research, and likely involves LRP1's ability to regulate levels of proteases known to degrade the extracellular matrix as well as its ability to modulate signaling events.
Collapse
Affiliation(s)
- Dianaly T Au
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - Allison L Arai
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - William E Fondrie
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - Selen C Muratoglu
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States.,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States
| |
Collapse
|
27
|
Wang Y, Ding L, Li Z, Chen G, Sun M, Oupicky D. Treatment of acute lung injury and early- and late-stage pulmonary fibrosis with combination emulsion siRNA polyplexes. J Control Release 2019; 314:12-24. [PMID: 31644934 DOI: 10.1016/j.jconrel.2019.10.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/02/2019] [Accepted: 10/15/2019] [Indexed: 01/05/2023]
Abstract
Acute lung injury (ALI) and idiopathic pulmonary fibrosis (IPF) are severe lung diseases causing irreversible lung damage and premature death. Both diseases share multiple pathological features, including overexpression of C-X-C chemokine receptor type 4 (CXCR4) and upregulation of plasminogen activator inhibitor-1 (PAI-1). The goal of the present study was to evaluate therapeutic potential of pulmonary treatment with combined inhibition of CXCR4 and PAI-1 in ALI and various disease stages of IPF. We report preparation of perfluorocarbon emulsion polyplexes containing a fluorinated polymeric CXCR4 antagonist (F-PAMD) as an siRNA carrier suitable for pulmonary delivery. In vitro testing of the emulsion polyplexes in primary lung fibroblasts from IPF mice showed high cellular uptake and promising antifibrotic effect as indicated by the decreased expression of α smooth muscle actin, when compared with conventional siRNA polyplexes. Biodistribution analysis in mice with IPF showed prolonged lung retention and widespread lung distribution following intratracheal administration of the formulations. The emulsion polyplexes showed promising therapeutic efficacy in ALI and in early fibrinogenic stage of IPF. Increased survival was observed in the model of late-stage IPF. The use of perfluorocarbon emulsion polyplexes to achieve combined CXCR4 antagonism and PAI-1 inhibition is a promising strategy for treatment of ALI and IPF.
Collapse
Affiliation(s)
- Yixin Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Ling Ding
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China; Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha NE 68198, USA
| | - Zhaoting Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Gang Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China; Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Minjie Sun
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - David Oupicky
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha NE 68198, USA.
| |
Collapse
|
28
|
Bres EE, Faissner A. Low Density Receptor-Related Protein 1 Interactions With the Extracellular Matrix: More Than Meets the Eye. Front Cell Dev Biol 2019; 7:31. [PMID: 30931303 PMCID: PMC6428713 DOI: 10.3389/fcell.2019.00031] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a biological substrate composed of collagens, proteoglycans and glycoproteins that ensures proper cell migration and adhesion and keeps the cell architecture intact. The regulation of the ECM composition is a vital process strictly controlled by, among others, proteases, growth factors and adhesion receptors. As it appears, ECM remodeling is also essential for proper neuronal and glial development and the establishment of adequate synaptic signaling. Hence, disturbances in ECM functioning are often present in neurodegenerative diseases like Alzheimer’s disease. Moreover, mutations in ECM molecules are found in some forms of epilepsy and malfunctioning of ECM-related genes and pathways can be seen in, for example, cancer or ischemic injury. Low density lipoprotein receptor-related protein 1 (Lrp1) is a member of the low density lipoprotein receptor family. Lrp1 is involved not only in ligand uptake, receptor mediated endocytosis and lipoprotein transport—functions shared by low density lipoprotein receptor family members—but also regulates cell surface protease activity, controls cellular entry and binding of toxins and viruses, protects against atherosclerosis and acts on many cell signaling pathways. Given the plethora of functions, it is not surprising that Lrp1 also impacts the ECM and is involved in its remodeling. This review focuses on the role of Lrp1 and some of its major ligands on ECM function. Specifically, interactions with two Lrp1 ligands, integrins and tissue plasminogen activator are described in more detail.
Collapse
Affiliation(s)
- Ewa E Bres
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
29
|
Liu Y, Wang L, Luo M, Chen N, Deng X, He J, Zhang L, Luo P, Wu J. Inhibition of PAI-1 attenuates perirenal fat inflammation and the associated nephropathy in high-fat diet-induced obese mice. Am J Physiol Endocrinol Metab 2019; 316:E260-E267. [PMID: 30532990 DOI: 10.1152/ajpendo.00387.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is increasingly recognized as a mediator in extracellular matrix (ECM) accumulation in diabetic nephropathy. Previous studies have implicated PAI-1 in adipose tissue (AT) expansion, while also contributing to insulin resistance. As inflammation is also known to occur in perirenal AT during obesity, we hypothesized that in a high-fat diet (HFD)-induced obese mouse model, PAI-1 contributes to macrophage-mediated inflammation and diabetic nephropathy. The HFD mice showed increased expression of PAI-1 in perirenal fat and also displayed increased fat weight and macrophage numbers. We found that the macrophage polarization, proinflammatory macrophage-M1-phenotype, including CD11c, IL-6, and monocyte chemoattractant protein-1, were increased by an HFD and decreased by either the genetic depletion of PAI-1 or treatment with the PAI-1 inhibitor, PAI-039. Similarly, an enhanced anti-inflammatory M2-phenotype, including CD206 and IL-10, was accompanied by either the genetic deletion of PAI-1 or PAI-039 treatment. Furthermore, the inhibition of PAI-1 reduced HFD-induced renal histological lesions and abated profibrotic/extracellular-matrix protein. Collectively, our findings provide support that PAI-1 contributes to the development of inflammation in perirenal fat and correlates with the development of diabetic nephropathy in HFD-induced obesity.
Collapse
Affiliation(s)
- Yong Liu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People's Republic of China
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macau, People's Republic of China
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Lin Wang
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Mao Luo
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Ni Chen
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Xin Deng
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Jing He
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Liping Zhang
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Pei Luo
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People's Republic of China
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macau, People's Republic of China
| | - Jianbo Wu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People's Republic of China
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macau, People's Republic of China
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Department of Medicine and Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine and Research Service , Columbia, Missouri
- Harry S. Truman Memorial Veterans Hospital , Columbia, Missouri
| |
Collapse
|
30
|
Signaling pathways involved in the expression of SZNF and the target genes binding with SZNF related to cyadox. Biomed Pharmacother 2018; 108:1879-1893. [DOI: 10.1016/j.biopha.2018.09.141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 09/11/2018] [Accepted: 09/26/2018] [Indexed: 11/22/2022] Open
|
31
|
Meyers JL, Winans B, Kelsaw E, Murthy A, Gerber S, Lawrence BP. Environmental cues received during development shape dendritic cell responses later in life. PLoS One 2018; 13:e0207007. [PMID: 30412605 PMCID: PMC6226176 DOI: 10.1371/journal.pone.0207007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022] Open
Abstract
Environmental signals mediated via the aryl hydrocarbon receptor (AHR) shape the developing immune system and influence immune function. Developmental exposure to AHR binding chemicals causes persistent changes in CD4+ and CD8+ T cell responses later in life, including dampened clonal expansion and differentiation during influenza A virus (IAV) infection. Naïve T cells require activation by dendritic cells (DCs), and AHR ligands modulate the function of DCs from adult organisms. Yet, the consequences of developmental AHR activation by exogenous ligands on DCs later in life has not been examined. We report here that early life activation of AHR durably reduces the ability of DC to activate naïve IAV-specific CD8+ T cells; however, activation of naïve CD4+ T cells was not impaired. Also, DCs from developmentally exposed offspring migrated more poorly than DCs from control dams in both in vivo and ex vivo assessments of DC migration. Conditional knockout mice, which lack Ahr in CD11c lineage cells, suggest that dampened DC emigration is intrinsic to DCs. Yet, levels of chemokine receptor 7 (CCR7), a key regulator of DC trafficking, were generally unaffected. Gene expression analyses reveal changes in Lrp1, Itgam, and Fcgr1 expression, and point to alterations in genes that regulate DC migration and antigen processing and presentation as being among pathways disrupted by inappropriate AHR signaling during development. These studies establish that AHR activation during development causes long-lasting changes to DCs, and provide new information regarding how early life environmental cues shape immune function later in life.
Collapse
Affiliation(s)
- Jessica L. Meyers
- Department of Environmental Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York, United States of America
| | - Bethany Winans
- Department of Environmental Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York, United States of America
| | - Erin Kelsaw
- Department of Microbiology and Immunology, University of Rochester School of Medicine & Dentistry, Rochester, New York, United States of America
| | - Aditi Murthy
- Department of Microbiology and Immunology, University of Rochester School of Medicine & Dentistry, Rochester, New York, United States of America
| | - Scott Gerber
- Department of Microbiology and Immunology, University of Rochester School of Medicine & Dentistry, Rochester, New York, United States of America
- Department of Surgery, University of Rochester School of Medicine & Dentistry, Rochester, New York, United States of America
| | - B. Paige Lawrence
- Department of Environmental Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York, United States of America
- Department of Microbiology and Immunology, University of Rochester School of Medicine & Dentistry, Rochester, New York, United States of America
| |
Collapse
|
32
|
Au DT, Ying Z, Hernández-Ochoa EO, Fondrie WE, Hampton B, Migliorini M, Galisteo R, Schneider MF, Daugherty A, Rateri DL, Strickland DK, Muratoglu SC. LRP1 (Low-Density Lipoprotein Receptor-Related Protein 1) Regulates Smooth Muscle Contractility by Modulating Ca 2+ Signaling and Expression of Cytoskeleton-Related Proteins. Arterioscler Thromb Vasc Biol 2018; 38:2651-2664. [PMID: 30354243 PMCID: PMC6214382 DOI: 10.1161/atvbaha.118.311197] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 09/12/2018] [Indexed: 01/12/2023]
Abstract
Objective- Mutations affecting contractile-related proteins in the ECM (extracellular matrix), microfibrils, or vascular smooth muscle cells can predispose the aorta to aneurysms. We reported previously that the LRP1 (low-density lipoprotein receptor-related protein 1) maintains vessel wall integrity, and smLRP1-/- mice exhibited aortic dilatation. The current study focused on defining the mechanisms by which LRP1 regulates vessel wall function and integrity. Approach and Results- Isometric contraction assays demonstrated that vasoreactivity of LRP1-deficient aortic rings was significantly attenuated when stimulated with vasoconstrictors, including phenylephrine, thromboxane receptor agonist U-46619, increased potassium, and L-type Ca2+ channel ligand FPL-64176. Quantitative proteomics revealed proteins involved in actin polymerization and contraction were significantly downregulated in aortas of smLRP1-/- mice. However, studies with calyculin A indicated that although aortic muscle from smLRP1-/- mice can contract in response to calyculin A, a role for LRP1 in regulating the contractile machinery is not revealed. Furthermore, intracellular calcium imaging experiments identified defects in calcium release in response to a RyR (ryanodine receptor) agonist in smLRP1-/- aortic rings and cultured vascular smooth muscle cells. Conclusions- These results identify a critical role for LRP1 in modulating vascular smooth muscle cell contraction by regulating calcium signaling events that potentially protect against aneurysm development.
Collapse
MESH Headings
- Actin Cytoskeleton/drug effects
- Actin Cytoskeleton/genetics
- Actin Cytoskeleton/metabolism
- Actin Cytoskeleton/ultrastructure
- Animals
- Aorta/metabolism
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Calcium Signaling/drug effects
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Female
- Gene Expression Regulation
- Low Density Lipoprotein Receptor-Related Protein-1
- Male
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/ultrastructure
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Ryanodine Receptor Calcium Release Channel/genetics
- Ryanodine Receptor Calcium Release Channel/metabolism
- Tissue Culture Techniques
- Tumor Suppressor Proteins/deficiency
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Vasoconstriction/drug effects
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- Dianaly T. Au
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zhekang Ying
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Erick O. Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - William E. Fondrie
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Brian Hampton
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mary Migliorini
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Rebeca Galisteo
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Martin F. Schneider
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Debra L. Rateri
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Selen C. Muratoglu
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
33
|
Kikuchi K, Setoyama K, Tanaka E, Otsuka S, Terashi T, Nakanishi K, Takada S, Sakakima H, Ampawong S, Kawahara KI, Nagasato T, Hosokawa K, Harada Y, Yamamoto M, Kamikokuryo C, Kiyama R, Morioka M, Ito T, Maruyama I, Tancharoen S. Uric acid enhances alteplase-mediated thrombolysis as an antioxidant. Sci Rep 2018; 8:15844. [PMID: 30367108 PMCID: PMC6203847 DOI: 10.1038/s41598-018-34220-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/15/2018] [Indexed: 01/01/2023] Open
Abstract
Uric acid (UA) therapy may prevent early ischemic worsening after acute stroke in thrombolysis patients. The aim of this study was to examine the influence of UA on the thrombolytic efficacy of alteplase in human blood samples by measuring thrombolysis under flow conditions using a newly developed microchip-based flow-chamber assay. Human blood samples from healthy volunteers were exposed to UA, alteplase, or a combination of UA and alteplase. Whole blood and platelet-rich plasma were perfused over a collagen- and thromboplastin-coated microchip, and capillary occlusion was monitored with a video microscope and flow-pressure sensor. The area under the curve (extent of thrombogenesis or thrombolysis) at 30 minutes was 92% lower in the UA-alteplase-treated group compared with the alteplase-treated group. D-dimers were measured to evaluate these effects in human platelet-poor plasma samples. Although hydrogen peroxide significantly decreased the elevation of D-dimers by alteplase, UA significantly inhibited the effect of hydrogen peroxide. Meanwhile, rat models of thromboembolic cerebral ischemia were treated with either alteplase or UA-alteplase combination therapy. Compared with alteplase alone, the combination therapy reduced the infarct volume and inhibited haemorrhagic transformation. UA enhances alteplase-mediated thrombolysis, potentially by preventing oxidative stress, which inhibits fibrinolysis by alteplase in thrombi.
Collapse
Affiliation(s)
- Kiyoshi Kikuchi
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, Kurume, Japan.,Department of Neurosurgery, Kurume University School of Medicine, Kurume, Japan.,Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan.,Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Kentaro Setoyama
- Natural Science Center for Research and Education, Division of Laboratory Animal Science, Kagoshima University, Kagoshima, Japan
| | - Eiichiro Tanaka
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, Kurume, Japan
| | - Shotaro Otsuka
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Takuto Terashi
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Kazuki Nakanishi
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Seiya Takada
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Harutoshi Sakakima
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, (S.A.), Mahidol University, Bangkok, Thailand
| | - Ko-Ichi Kawahara
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan.,Laboratory of Functional Foods, Department of Biomedical Engineering, Osaka Institute of Technology, Osaka, Japan
| | - Tomoka Nagasato
- Research Institute, Fujimori Kogyo Co., Yokohama, Kanagawa, Japan
| | - Kazuya Hosokawa
- Research Institute, Fujimori Kogyo Co., Yokohama, Kanagawa, Japan
| | - Yoichiro Harada
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Mika Yamamoto
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Chinatsu Kamikokuryo
- Department of Emergency and Critical Care Medicine, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Ryoji Kiyama
- School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Motohiro Morioka
- Department of Neurosurgery, Kurume University School of Medicine, Kurume, Japan
| | - Takashi Ito
- Department of Emergency and Critical Care Medicine, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Salunya Tancharoen
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
34
|
CTRP5 promotes transcytosis and oxidative modification of low-density lipoprotein and the development of atherosclerosis. Atherosclerosis 2018; 278:197-209. [PMID: 30300788 DOI: 10.1016/j.atherosclerosis.2018.09.037] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/05/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Increased transcytosis of low-density lipoprotein (LDL) across the endothelium and oxidation of LDL deposited within the subendothelial space are crucial early events in atherogenesis. C1q/TNF-related protein (CTRP) 5 is a novel secreted glycoprotein and its biological functions are largely undefined. METHODS Expression of CTRP5 was analyzed in sera and atherosclerotic plaques of patients with coronary artery disease (CAD). The role of CTRP5 in atherogenesis was investigated in vitro and in vivo. RESULTS We found CTRP5 serum levels were higher in patients with than without CAD (247.26 ± 61.71 vs. 167.81 ± 68.08 ng/mL, p < 0.001), and were positively correlated with the number of diseased vessels (Spearman's r = 0.611, p < 0.001). Increased expression of CTRP5 was detected in human coronary endarterectomy specimens as compared to non-atherosclerotic arteries. Immunofluorescence further showed that CTRP5 was predominantly localized in the endothelium, infiltrated macrophages and smooth muscle cells in the neointima. In vivo and in vitro experiments demonstrated that CTRP5 promoted transcytosis of LDL across endothelial monolayers, as well as the oxidative modification of LDL in endothelial cells. Mechanistically, we found that CTRP5 up-regulated 12/15-lipoxygenase (LOX), a key enzyme in mediating LDL trafficking and oxidation, through STAT6 signaling. Genetic or pharmacological inhibition of 12/15-LOX dramatically attenuated the deposition of oxidized LDL in the subendothelial space and the development of atherosclerosis. CONCLUSIONS These data indicate that CTRP5 is a novel pro-atherogenic cytokine and promotes transcytosis and oxidation of LDL in endothelial cells via up-regulation of 12/15-LOX.
Collapse
|
35
|
Wang L, Chen L, Liu Z, Liu Y, Luo M, Chen N, Deng X, Luo Y, He J, Zhang L, Hill MA, Li R, Wu J. PAI-1 Exacerbates White Adipose Tissue Dysfunction and Metabolic Dysregulation in High Fat Diet-Induced Obesity. Front Pharmacol 2018; 9:1087. [PMID: 30319420 PMCID: PMC6169321 DOI: 10.3389/fphar.2018.01087] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 09/06/2018] [Indexed: 01/29/2023] Open
Abstract
Background: Plasminogen activator inhibitor (PAI)-1 levels and activity are known to increase during metabolic syndrome and obesity. In addition, previous studies have implicated PAI-1 in adipose tissue (AT) expansion while also contributing to insulin resistance. As inflammation is also known to occur in AT during obesity, we hypothesized that in a high-fat diet (HFD)-induced obese mouse model PAI-1 contributes to macrophage-mediated inflammation and metabolic dysfunction. Methods: Four- to five-weeks-old male C57B6/6J mice were fed a HFD (45%) for 14 weeks, while age-matched control mice were fed a standard laboratory chow diet (10% fat). Additional studies were performed in PAI-1 knockout mice and wild type mice treated with an inhibitor (PAI-039) of PAI-1. Macrophage polarization were measured by real time PCR. Results: HFD mice showed increased expression of PAI-1 in visceral white AT (WAT) that also displayed increased macrophage numbers. PAI-1 deficient mice exhibited increased numbers of anti-inflammatory macrophages in WAT and were resistant to HFD-induced obesity. Similarly, pharmacological inhibition of PAI-1 using PAI-039 significantly decreased macrophage infiltration in WAT and improved metabolic status in HFD-induced wild-type mice. Importantly, the numbers of M1 macrophages appeared to be increased by the HFD and decreased by either genetic PAI-1 depletion or PAI-039 treatment. Conclusions: Collectively, our findings provide support for PAI-1 contributing to the development of inflammation in adipose tissue and explain the mechanism of inflammation modulated by PAI-1 in the disordered metabolism in HFD-induced obesity.
Collapse
Affiliation(s)
- Lin Wang
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Liyuan Chen
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zheran Liu
- Queen Mary School, Medical College of Nanchang University, Nanchang, China
| | - Yaofang Liu
- Department of Gynecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Mao Luo
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Ni Chen
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xin Deng
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yulin Luo
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing He
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Liping Zhang
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Michael A Hill
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Rong Li
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianbo Wu
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
36
|
Cisplatin-activated PAI-1 secretion in the cancer-associated fibroblasts with paracrine effects promoting esophageal squamous cell carcinoma progression and causing chemoresistance. Cell Death Dis 2018; 9:759. [PMID: 29988148 PMCID: PMC6037765 DOI: 10.1038/s41419-018-0808-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/17/2018] [Accepted: 06/15/2018] [Indexed: 01/12/2023]
Abstract
Preoperative chemotherapy is a promising strategy for the treatment of esophageal squamous cell carcinoma (ESCC). Acquired resistance to chemotherapy is a major obstacle in improving patient prognosis. Cancer-associated fibroblasts (CAFs) are the primary components of the tumor microenvironment and play a crucial role in tumor development; these cells are also potential therapeutic targets for cancer. Using protein arrays, we identified a key secreted cytokine, PAI-1, from CAFs pretreated with cisplatin that was induced after DNA damage of CAFs. The PAI-1 in the tumor microenvironment promoted tumor growth and attenuated the effects of cisplatin treatment. Extracellular PAI-1 activated the AKT and ERK1/2 signaling pathways and inhibited caspase-3 activity and reactive oxygen species accumulation. Tiplaxtinin as a PAI-1 inhibitor could play synergistic effects with cisplatin in vitro and in vivo. In clinical samples, ESCC patients with high expression of PAI-1 in CAFs presented a significantly worse progression-free survival. Taken together, our results showed that PAI-1 secreted from cisplatin-activated CAFs promoted tumor growth and reduced the effects of cisplatin in a paracrine manner, establishing a preclinical rationale to target this cytokine to further improve the clinical response of esophageal squamous cell carcinoma.
Collapse
|
37
|
Wujak L, Böttcher RT, Pak O, Frey H, El Agha E, Chen Y, Schmitt S, Bellusci S, Schaefer L, Weissmann N, Fässler R, Wygrecka M. Low density lipoprotein receptor-related protein 1 couples β1 integrin activation to degradation. Cell Mol Life Sci 2018; 75:1671-1685. [PMID: 29116364 PMCID: PMC11105666 DOI: 10.1007/s00018-017-2707-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/19/2017] [Accepted: 11/01/2017] [Indexed: 01/09/2023]
Abstract
Low density lipoprotein receptor-related protein (LRP) 1 modulates cell adhesion and motility under normal and pathological conditions. Previous studies documented that LRP1 binds several integrin receptors and mediates their trafficking to the cell surface and endocytosis. However, the mechanism by which LRP1 may regulate integrin activation remains unknown. Here we report that LRP1 promotes the activation and subsequent degradation of β1 integrin and thus supports cell adhesion, spreading, migration and integrin signaling on fibronectin. LRP1 interacts with surface β1 integrin, binds the integrin activator kindlin2 and stimulates β1 integrin-kindlin2 complex formation. Specifically, serine 76 in the LRP1 cytoplasmic tail is crucial for the interaction with kindlin2, β1 integrin activation and cell adhesion. Interestingly, a loss of LRP1 induces the accumulation of several integrin receptors on the cell surface. Following internalization, intracellular trafficking of integrins is driven by LRP1 in a protein kinase C- and class II myosin-dependent manner. Ultimately, LRP1 dictates the fate of endocytosed β1 integrin by directing it down the pathway of lysosomal and proteasomal degradation. We propose that LRP1 mediates cell adhesion by orchestrating a multi-protein pathway to activate, traffic and degrade integrins. Thus, LRP1 may serve as a focal point in the integrin quality control system to ensure a firm connection to the extracellular matrix.
Collapse
Affiliation(s)
- Lukasz Wujak
- Department of Biochemistry, Justus Liebig University, 35392, Giessen, Germany
| | - Ralph T Böttcher
- Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Oleg Pak
- Excellence Cluster Cardio-Pulmonary System, Justus Liebig University, 35392, Giessen, Germany
| | - Helena Frey
- Institute of Pharmacology and Toxicology, Goethe University School of Medicine, University Hospital, 60590, Frankfurt am Main, Germany
| | - Elie El Agha
- Excellence Cluster Cardio-Pulmonary System, Justus Liebig University, 35392, Giessen, Germany
| | - Ying Chen
- Department of Biochemistry, Justus Liebig University, 35392, Giessen, Germany
| | - Sigrid Schmitt
- Department of Biochemistry, Justus Liebig University, 35392, Giessen, Germany
| | - Saverio Bellusci
- Excellence Cluster Cardio-Pulmonary System, Justus Liebig University, 35392, Giessen, Germany
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University School of Medicine, University Hospital, 60590, Frankfurt am Main, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System, Justus Liebig University, 35392, Giessen, Germany
| | - Reinhard Fässler
- Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Justus Liebig University, 35392, Giessen, Germany.
| |
Collapse
|
38
|
Kang J, Kim W, Kwon T, Youn H, Kim JS, Youn B. Plasminogen activator inhibitor-1 enhances radioresistance and aggressiveness of non-small cell lung cancer cells. Oncotarget 2018; 7:23961-74. [PMID: 27004408 PMCID: PMC5029677 DOI: 10.18632/oncotarget.8208] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/04/2016] [Indexed: 12/20/2022] Open
Abstract
Acquired resistance of tumor cells during treatment limits the clinical efficacy of radiotherapy. Recent studies to investigate acquired resistance under treatment have focused on intercellular communication because it promotes survival and aggressiveness of tumor cells, causing therapy failure and tumor relapse. Accordingly, a better understanding of the functional communication between subpopulations of cells within a tumor is essential to development of effective cancer treatment strategies. Here, we found that conditioned media (CM) from radioresistant non-small cell lung cancer (NSCLC) cells increased survival of radiosensitive cells. Comparative proteomics analysis revealed plasminogen activator inhibitor-1 (PAI-1) as a key molecule in the secretome that acts as an extracellular signaling trigger to strengthen resistance to radiation. Our results revealed that expression and secretion of PAI-1 in radioresistant cells was increased by radiation-induced transcription factors, including p53, HIF-1α, and Smad3. When CM from radioresistant cells was applied to radiosensitive cells, extracellular PAI-1 activated the AKT and ERK1/2 signaling pathway and inhibited caspase-3 activity. Our study also proposed that PAI-1 activates the signaling pathway in radiosensitive cells via extracellular interaction with its binding partners, not clathrin-mediated endocytosis. Furthermore, secreted PAI-1 increased cell migration capacity and expression of EMT markers in vitro and in vivo. Taken together, our findings demonstrate that PAI-1 secreted from radioresistant NSCLC cells reduced radiosensitivity of nearby cells in a paracrine manner, indicating that functional inhibition of PAI-1 signaling has therapeutic potential because it prevents sensitive cells from acquiring radioresistance.
Collapse
Affiliation(s)
- JiHoon Kang
- Department of Integrated Biological Science, Pusan National University, Busan, 46241, Republic of Korea
| | - Wanyeon Kim
- Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea.,Nuclear Science Research Institute, Pusan National University, Busan 46241, Republic of Korea
| | - TaeWoo Kwon
- Department of Integrated Biological Science, Pusan National University, Busan, 46241, Republic of Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Joong Sun Kim
- Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan, 46241, Republic of Korea.,Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea.,Nuclear Science Research Institute, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
39
|
Wujak L, Schnieder J, Schaefer L, Wygrecka M. LRP1: A chameleon receptor of lung inflammation and repair. Matrix Biol 2017; 68-69:366-381. [PMID: 29262309 DOI: 10.1016/j.matbio.2017.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 12/17/2022]
Abstract
The lung displays a remarkable capability to regenerate following injury. Considerable effort has been made thus far to understand the cardinal processes underpinning inflammation and reconstruction of lung tissue. However, the factors determining the resolution or persistence of inflammation and efficient wound healing or aberrant remodeling remain largely unknown. Low density lipoprotein receptor-related protein 1 (LRP1) is an endocytic/signaling cell surface receptor which controls cellular and molecular mechanisms driving the physiological and pathological inflammatory reactions and tissue remodeling in several organs. In this review, we will discuss the impact of LRP1 on the consecutive steps of the inflammatory response and its role in the balanced tissue repair and aberrant remodeling in the lung.
Collapse
Affiliation(s)
- Lukasz Wujak
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Jennifer Schnieder
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Liliana Schaefer
- Goethe University School of Medicine, University Hospital, Theodor-Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany; Member of the German Center for Lung Research (DZL), Germany.
| |
Collapse
|
40
|
Luo M, Ji Y, Luo Y, Li R, Fay WP, Wu J. Plasminogen activator inhibitor-1 regulates the vascular expression of vitronectin. J Thromb Haemost 2017; 15:2451-2460. [PMID: 29028290 PMCID: PMC5716874 DOI: 10.1111/jth.13869] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Indexed: 11/30/2022]
Abstract
Essentials Vitronectin (VN) is produced by smooth muscle cells (SMCs) and promotes neointima formation. We studied the regulation of vascular VN expression by plasminogen activator inhibitor-1 (PAI-1). PAI-1 stimulates VN gene expression in SMCs by binding LDL receptor-related protein 1. Stimulation of VN gene expression may be a mechanism by which PAI-1 controls vascular remodeling. SUMMARY Background Increased expression of vitronectin (VN) by smooth muscle cells (SMCs) promotes neointima formation after vascular injury, and may contribute to chronic vascular diseases, such as atherosclerosis. However, the molecular regulation of vascular VN expression is poorly defined. Given the overlapping expression profiles and functions of VN and plasminogen activator inhibitor (PAI)-1, we hypothesized that PAI-1 regulates vascular VN expression. Objectives To determine whether PAI-1 regulates VN expression in SMCs and in vivo. Methods The effects of genetic alterations in PAI-1 expression, pharmacologic PAI-1 inhibition and recombinant PAI-1 on SMC VN expression were studied, and vascular VN expression in wild-type (WT) and PAI-1-deficient mice was assessed. Results VN expression was significantly lower in PAI-1-deficient SMCs and significantly increased in PAI-1-overexpressing SMCs. PAI-1 small interfering RNA and pharmacologic PAI-1 inhibition significantly decreased SMC VN expression. Recombinant PAI-1 stimulated VN expression by binding LDL receptor-related protein-1 (LRP1), but another LRP1 ligand, α2 -macroglobulin, did not. As compared with WT controls, carotid artery VN expression was significantly lower in PAI-1-deficient mice and significantly higher in PAI-1-transgenic mice. In a vein graft (VG) model of intimal hyperplasia, VN expression was significantly attenuated in PAI-1-deficient VGs as compared with WT controls. The plasma VN concentration was significantly decreased in PAI-1-deficient mice versus WT controls at 4 weeks, but not at 5 days or 8 weeks, after surgery. Conclusions PAI-1 stimulates SMC VN expression by binding LRP1, and controls vascular VN expression in vivo. Autocrine regulation of vascular VN expression by PAI-1 may play important roles in vascular homeostasis and pathologic vascular remodeling.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Gene Expression Regulation
- Humans
- Low Density Lipoprotein Receptor-Related Protein-1
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Neointima/etiology
- Neointima/genetics
- Neointima/metabolism
- RNA, Small Interfering/genetics
- Receptors, LDL/metabolism
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Serpin E2/deficiency
- Serpin E2/genetics
- Serpin E2/metabolism
- Tumor Suppressor Proteins/metabolism
- Vascular Remodeling
- Vitronectin/deficiency
- Vitronectin/genetics
- Vitronectin/metabolism
Collapse
Affiliation(s)
- M Luo
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Y Ji
- Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Y Luo
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China
| | - R Li
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China
| | - W P Fay
- Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
- Department of Medical Pharmacology & Physiology, University of Missouri School of Medicine, Columbia, MO, USA
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - J Wu
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
41
|
Kubala MH, DeClerck YA. Conditional Knockdown of Gene Expression in Cancer Cell Lines to Study the Recruitment of Monocytes/Macrophages to the Tumor Microenvironment. J Vis Exp 2017. [PMID: 29286360 DOI: 10.3791/56333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
siRNA and shRNA-mediated knock down (KD) methods of regulating gene expression are invaluable tools for understanding gene and protein function. However, in the case that the KD of the protein of interest has a lethal effect on cells or the anticipated effect of the KD is time-dependent, unconditional KD methods are not appropriate. Conditional systems are more suitable in these cases and have been the subject of much interest. These include Ecdysone-inducible overexpression systems, Cytochrome P-450 induction system1, and the tetracycline regulated gene expression systems. The tetracycline regulated gene expression system enables reversible control over protein expression by induction of shRNA expression in the presence of tetracycline. In this protocol, we present an experimental design using functional Tet-ON system in human cancer cell lines for conditional regulation of gene expression. We then demonstrate the use of this system in the study of tumor cell-monocyte interaction.
Collapse
Affiliation(s)
- Marta H Kubala
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Department of Pediatrics, University of Southern California; The Saban Research Institute of Children's Hospital Los Angeles;
| | - Yves A DeClerck
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Department of Pediatrics, University of Southern California; The Saban Research Institute of Children's Hospital Los Angeles; Department of Biochemistry and Molecular Medicine, University of Southern California
| |
Collapse
|
42
|
Edaravone, a Synthetic Free Radical Scavenger, Enhances Alteplase-Mediated Thrombolysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6873281. [PMID: 29259732 PMCID: PMC5702421 DOI: 10.1155/2017/6873281] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/07/2017] [Indexed: 12/12/2022]
Abstract
The combination of alteplase, a recombinant tissue plasminogen activator, and edaravone, an antioxidant, reportedly enhances recanalization after acute ischemic stroke. We examined the influence of edaravone on the thrombolytic efficacy of alteplase by measuring thrombolysis using a newly developed microchip-based flow-chamber assay. Rat models of embolic cerebral ischemia were treated with either alteplase or alteplase-edaravone combination therapy. The combination therapy significantly reduced the infarct volume and improved neurological deficits. Human blood samples from healthy volunteers were exposed to edaravone, alteplase, or a combination of alteplase and edaravone or hydrogen peroxide. Whole blood was perfused over a collagen- and thromboplastin-coated microchip; capillary occlusion was monitored with a video microscope and flow-pressure sensor. The area under the curve (extent of thrombogenesis or thrombolysis) at 30 minutes was 69.9% lower in the edaravone-alteplase- than alteplase-treated group. The thrombolytic effect of alteplase was significantly attenuated in the presence of hydrogen peroxide, suggesting that oxidative stress might hinder thrombolysis. D-dimers were measured to evaluate these effects in human platelet-poor plasma samples. Although hydrogen peroxide significantly decreased the elevation of D-dimers by alteplase, edaravone significantly inhibited the decrease. Edaravone enhances alteplase-mediated thrombolysis, likely by preventing oxidative stress, which inhibits fibrinolysis by alteplase in thrombi.
Collapse
|
43
|
Lin L, Hu K. Tissue-type plasminogen activator modulates macrophage M2 to M1 phenotypic change through annexin A2-mediated NF-κB pathway. Oncotarget 2017; 8:88094-88103. [PMID: 29152144 PMCID: PMC5675696 DOI: 10.18632/oncotarget.21510] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/31/2017] [Indexed: 11/25/2022] Open
Abstract
Macrophage accumulation is one of the hallmarks of progressive kidney disease. In response to injury, macrophages undergo a phenotypic polarization to become two functionally distinct subsets: M1 and M2 macrophages. Macrophage polarization is a dynamic process, and recent work indicates that macrophages, in response to kidney injury, can shift their polarity. However, the underlying mechanisms remain largely unknown. Tissue-type plasminogen activator (tPA), a protease up-regulated in the chronically injured kidneys, has been shown to preferably promote M1 macrophage accumulation and renal inflammation. We hypothesized that tPA may be an endogenous factor that modulates macrophage M2 to M1 phenotypic change contributing to the accumulation of M1 macrophages in the injured kidneys. It was found that obstruction-induced renal M1 chemokine expression was alleviated in tPA knockout mice, and these knockout mice displayed increased M2 markers. In vitro, resting J774 macrophages were treated with IL-4 to induce M2 phenotype as indicated by de novo expression of arginase 1, Ym1, and IL-10, as well as suppression of iNOS, TNF-α, and IL-1β. Intriguingly, these IL-4-induced M2 macrophages, after tPA treatment, not only lost their M2 markers such as arginase 1, Ym1, and IL-10, but also displayed increased M1 chemokines including iNOS, TNF-α, and IL-1β. Possible endotoxin contamination was also excluded as heat-inactivated tPA lost its effect. Additionally, tPA-mediated macrophage M2 to M1 phenotypic change required its receptor annexin A2, and SN50, a specific NF-κB inhibitor, abolished tPA's effect. Thus, it's clear that tPA promotes macrophage M2 to M1 phenotypic change through annexin A2-mediated NF-κB pathway.
Collapse
Affiliation(s)
- Ling Lin
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Kebin Hu
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, Pennsylvania, USA.,Division of Nephrology, Department of Medicine, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
44
|
Microglial-mediated PDGF-CC activation increases cerebrovascular permeability during ischemic stroke. Acta Neuropathol 2017; 134:585-604. [PMID: 28725968 PMCID: PMC5587628 DOI: 10.1007/s00401-017-1749-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 12/25/2022]
Abstract
Treatment of acute ischemic stroke with the thrombolytic tissue plasminogen activator (tPA) can significantly improve neurological outcomes; however, thrombolytic therapy is associated with an increased risk of intra-cerebral hemorrhage (ICH). Previously, we demonstrated that during stroke tPA acting on the parenchymal side of the neurovascular unit (NVU) can increase blood–brain barrier (BBB) permeability and ICH through activation of latent platelet-derived growth factor-CC (PDGF-CC) and signaling by the PDGF receptor-α (PDGFRα). However, in vitro, activation of PDGF-CC by tPA is very inefficient and the mechanism of PDGF-CC activation in the NVU is not known. Here, we show that the integrin Mac-1, expressed on brain microglia/macrophages (denoted microglia throughout), acts together with the endocytic receptor LRP1 in the NVU to promote tPA-mediated activation of PDGF-CC. Mac-1-deficient mice (Mac-1−/−) are protected from tPA-induced BBB permeability but not from permeability induced by intracerebroventricular injection of active PDGF-CC. Immunofluorescence analysis demonstrates that Mac-1, LRP1, and the PDGFRα all localize to the NVU of arterioles, and following middle cerebral artery occlusion (MCAO) Mac-1−/− mice show significantly less PDGFRα phosphorylation, BBB permeability, and infarct volume compared to wild-type mice. Bone-marrow transplantation studies indicate that resident CD11b+ cells, but not bone-marrow-derived leukocytes, mediate the early activation of PDGF-CC by tPA after MCAO. Finally, using a model of thrombotic stroke with late thrombolysis, we show that wild-type mice have an increased incidence of spontaneous ICH following thrombolysis with tPA 5 h after MCAO, whereas Mac-1−/− mice are resistant to the development of ICH even with late tPA treatment. Together, these results indicate that Mac-1 and LRP1 act as co-factors for the activation of PDGF-CC by tPA in the NVU, and suggest a novel mechanism for tightly regulating PDGFRα signaling in the NVU and controlling BBB permeability.
Collapse
|
45
|
TGF-β-induced intracellular PAI-1 is responsible for retaining hematopoietic stem cells in the niche. Blood 2017; 130:2283-2294. [PMID: 28821477 DOI: 10.1182/blood-2017-02-767384] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 08/10/2017] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) reside in the supportive stromal niche in bone marrow (BM); when needed, however, they are rapidly mobilized into the circulation, suggesting that HSPCs are intrinsically highly motile but usually stay in the niche. We questioned what determines the motility of HSPCs. Here, we show that transforming growth factor (TGF)-β-induced intracellular plasminogen activator inhibitor (PAI)-1 activation is responsible for keeping HSPCs in the BM niche. We found that the expression of PAI-1, a downstream target of TGF-β signaling, was selectively augmented in niche-residing HSPCs. Functional inhibition of the TGF-β-PAI-1 signal increased MT1-MMP-dependent cellular motility, causing a detachment of HSPCs from the TGF-β-expressing niche cells, such as megakaryocytes. Furthermore, consistently high motility in PAI-1-deficient HSPCs was demonstrated by both a transwell migration assay and reciprocal transplantation experiments, indicating that intracellular, not extracellular, PAI-1 suppresses the motility of HSPCs, thereby causing them to stay in the niche. Mechanistically, intracellular PAI-1 inhibited the proteolytic activity of proprotein convertase Furin, diminishing MT1-MMP activity. This reduced expression of MT1-MMP in turn affected the expression levels of several adhesion/deadhesion molecules for determination of HSPC localization, such as CD44, VLA-4, and CXCR4, which then promoted the retention of HSPCs in the niche. Our findings open up a new field for the study of intracellular proteolysis as a regulatory mechanism of stem cell fate, which has the potential to improve clinical HSPC mobilization and transplantation protocols.
Collapse
|
46
|
Theret L, Jeanne A, Langlois B, Hachet C, David M, Khrestchatisky M, Devy J, Hervé E, Almagro S, Dedieu S. Identification of LRP-1 as an endocytosis and recycling receptor for β1-integrin in thyroid cancer cells. Oncotarget 2017; 8:78614-78632. [PMID: 29108253 PMCID: PMC5667986 DOI: 10.18632/oncotarget.20201] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/25/2017] [Indexed: 12/14/2022] Open
Abstract
LRP-1 is a large endocytic receptor mediating the clearance of various molecules from the extracellular matrix. LRP-1 was reported to control focal adhesion turnover to optimize the adhesion-deadhesion balance to support invasion. To better understand how LRP-1 coordinates cell-extracellular matrix interface, we explored its ability to regulate cell surface integrins in thyroid carcinomas. Using an antibody approach, we demonstrated that β1-integrin levels were increased at the plasma membrane under LRP1 silencing or upon RAP treatment, used as LRP-1 antagonist. Our data revealed that LRP-1 binds with both inactive and active β1-integrin conformations and identified the extracellular ligand-binding domains II or IV of LRP-1 as sufficient to bind β1-integrin. Using a recombinant β1-integrin, we demonstrated that LRP-1 acts as a regulator of β1-integrin intracellular traffic. Moreover, RAP or LRP-1 blocking antibodies decreased up to 36% the number of β1-integrin-containing endosomes. LRP-1 blockade did not significantly affect the levels of β1-integrin-containing lysosomes while decreasing localization of β1-integrin within Rab-11 positive vesicles. Overall, we identified an original molecular process in which LRP-1 acts as a main regulator of β1-integrin internalization and recycling in thyroid cancer cells.
Collapse
Affiliation(s)
- Louis Theret
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Albin Jeanne
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France.,SATT Nord, Lille, France
| | - Benoit Langlois
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Cathy Hachet
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Marion David
- VECT-HORUS SAS, Faculté de Médecine Secteur Nord, Marseille, France
| | | | - Jérôme Devy
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Emonard Hervé
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Sébastien Almagro
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Stéphane Dedieu
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| |
Collapse
|
47
|
Rabieian R, Boshtam M, Zareei M, Kouhpayeh S, Masoudifar A, Mirzaei H. Plasminogen Activator Inhibitor Type-1 as a Regulator of Fibrosis. J Cell Biochem 2017; 119:17-27. [PMID: 28520219 DOI: 10.1002/jcb.26146] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/17/2017] [Indexed: 12/17/2022]
Abstract
Fibrosis is known as a frequent and irreversible pathological condition which is associated with organ failure. Tissue fibrosis is a central process in a variety of chronic progressive diseases such as diabetes, hypertension, and persistent inflammation. This state could contribute to chronic injury and the initiation of tissue repair. Fibrotic disorders represent abnormal wound healing with defective matrix turnover and clearance that lead to excessive accumulation of extracellular matrix components. A variety of identified growth factors, cytokines, and persistently activated myofibroblasts have critical roles in the pathogenesis of fibrosis. Irrespective of etiology, the transforming growth factor-β pathway is the major driver of fibrotic response. Plasminogen activator inhibitor-1 (PAI-1) is a crucial downstream target of this pathway. Transforming growth factor-β positively regulates PAI-1 gene expression via two main pathways including Smad-mediated canonical and non-canonical pathways. Overexpression of PAI-1 reduces extracellular matrix degradation via perturbing the plasminogen activation system. Indeed, elevated PAI-1 levels inhibit proteolytic activity of tissue plasminogen activator and urokinase plasminogen activator which could contribute to a variety of inflammatory elements in the injury site and to excessive matrix deposition. This review summarizes the current knowledge of critical pathways that regulate PAI-1 gene expression and suggests effective approaches for the treatment of fibrotic disease. J. Cell. Biochem. 119: 17-27, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Reyhaneh Rabieian
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahshid Zareei
- Department of Biology, School of Sciences, University of Isfahan, Isfahan, Iran
| | - Shirin Kouhpayeh
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aria Masoudifar
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Hamed Mirzaei
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
48
|
Five-year follow-up of a case of lipoprotein glomerulopathy with APOE Kyoto mutation. CEN Case Rep 2017; 5:148-153. [PMID: 28508969 DOI: 10.1007/s13730-016-0214-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/09/2016] [Indexed: 10/22/2022] Open
Abstract
We report the case of a 34-year-old Japanese male with lipoprotein glomerulopathy (LPG). Renal biopsy showed LPG, and followed by a genetic analysis revealed a mutation in apolipoprotein E gene (APOE Kyoto; Arg25Cys). We started treatment with probucol, bezafibrate, losartan, and allopurinol. Urinary protein decreased in response to treatment but has remained at about 1.27 ± 0.71 g/gCr, and a repeat biopsy which was performed 1 year after the first biopsy showed no clear evidence of pathological remission and complication of other glomerular disease. After 5 years of follow-up after the start of treatment, renal function has almost maintained without apparent deterioration. Interestingly, the course of the urinary protein level closely paralleled his triglyceride and cholesterol levels in a long-term. This observation suggests the importance of tight control of lipid profiles as a means of renoprotection in LPG patient.
Collapse
|
49
|
Honjo K, Munakata S, Tashiro Y, Salama Y, Shimazu H, Eiamboonsert S, Dhahri D, Ichimura A, Dan T, Miyata T, Takeda K, Sakamoto K, Hattori K, Heissig B. Plasminogen activator inhibitor‐1 regulates macrophage‐dependent postoperative adhesion by enhancing EGF‐HER1 signaling in mice. FASEB J 2017; 31:2625-2637. [DOI: 10.1096/fj.201600871rr] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 02/21/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Kumpei Honjo
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Coloproctological Surgery Tokyo Japan
| | - Shinya Munakata
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Coloproctological Surgery Tokyo Japan
| | - Yoshihiko Tashiro
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Coloproctological Surgery Tokyo Japan
| | - Yousef Salama
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Hiroshi Shimazu
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Salita Eiamboonsert
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Douaa Dhahri
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Atsuhiko Ichimura
- United Centers for Advanced Research and Translational MedicineGraduate School of Medicine, Tohoku University Sendai Japan
| | - Takashi Dan
- United Centers for Advanced Research and Translational MedicineGraduate School of Medicine, Tohoku University Sendai Japan
| | - Toshio Miyata
- United Centers for Advanced Research and Translational MedicineGraduate School of Medicine, Tohoku University Sendai Japan
| | - Kazuyoshi Takeda
- Department of Immunology and Atopy CenterGraduate School of Medicine, Juntendo University Tokyo Japan
| | | | - Koichi Hattori
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Center for Genomic and Regenerative MedicineFaculty of Medicine Tokyo Japan
| | - Beate Heissig
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Immunology and Atopy CenterGraduate School of Medicine, Juntendo University Tokyo Japan
| |
Collapse
|
50
|
Leonetti C, Macrez R, Pruvost M, Hommet Y, Bronsard J, Fournier A, Perrigault M, Machin I, Vivien D, Clemente D, De Castro F, Maubert E, Docagne F. Tissue-type plasminogen activator exerts EGF-like chemokinetic effects on oligodendrocytes in white matter (re)myelination. Mol Neurodegener 2017; 12:20. [PMID: 28231842 PMCID: PMC5322587 DOI: 10.1186/s13024-017-0160-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/10/2017] [Indexed: 01/12/2023] Open
Abstract
Background The ability of oligodendrocyte progenitor cells (OPCs) to give raise to myelin forming cells during developmental myelination, normal adult physiology and post-lesion remyelination in white matter depends on factors which govern their proliferation, migration and differentiation. Tissue plasminogen activator (tPA) is a serine protease expressed in the central nervous system (CNS), where it regulates cell fate. In particular, tPA has been reported to protect oligodendrocytes from apoptosis and to facilitate the migration of neurons. Here, we investigated whether tPA can also participate in the migration of OPCs during CNS development and during remyelination after focal white matter lesion. Methods OPC migration was estimated by immunohistological analysis in spinal cord and corpus callosum during development in mice embryos (E13 to P0) and after white matter lesion induced by the stereotactic injection of lysolecithin in adult mice (1 to 21 days post injection). Migration was compared in these conditions between wild type and tPA knock-out animals. The action of tPA was further investigated in an in vitro chemokinesis assay. Results OPC migration along vessels is delayed in tPA knock-out mice during development and during remyelination. tPA enhances OPC migration via an effect dependent on the activation of epidermal growth factor receptor. Conclusion Endogenous tPA facilitates the migration of OPCs during development and during remyelination after white matter lesion by the virtue of its epidermal growth factor-like domain. Electronic supplementary material The online version of this article (doi:10.1186/s13024-017-0160-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Camille Leonetti
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Richard Macrez
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Mathilde Pruvost
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Yannick Hommet
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Jérémie Bronsard
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Antoine Fournier
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Maxime Perrigault
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Isabel Machin
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos-SESCAM, Toledo, Spain.,Grupo de Neuroinmuno-reparación, Hospital Nacional de Parapléjicos, Toledo, Spain
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Diego Clemente
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos-SESCAM, Toledo, Spain.,Grupo de Neuroinmuno-reparación, Hospital Nacional de Parapléjicos, Toledo, Spain
| | - Fernando De Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos-SESCAM, Toledo, Spain.,Grupo de Neurobiología del Desarrollo (GNDe), Instituto Cajal, CSIC, Madrid, Spain
| | - Eric Maubert
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Fabian Docagne
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France. .,Inserm, Centre Cyceron, Bvd Becquerel, BP5229, Caen Cedex, 14074, France.
| |
Collapse
|