1
|
Zou Y, Zhuo M, Chen W, Song W, Jiang Y, Xu J, Wang J. Multiomics analysis of O-GlcNAcylation in podocytes of diabetic kidney disease. Diabetes Obes Metab 2025; 27:2708-2719. [PMID: 39950214 DOI: 10.1111/dom.16274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 04/04/2025]
Abstract
AIM To investigate the role of O-GlcNAc transferase (OGT)-mediated protein O-GlcNAcylation in podocyte injury during the progression of diabetic kidney disease (DKD). MATERIALS AND METHODS Proteomic and O-glycoproteomic analyses were conducted on high glucose (HG)-stimulated podocytes with OGT knockdown. Differentially expressed proteins and O-GlcNAcylated peptides/proteins were identified, and functional enrichment (GO, KEGG, COG/KOG) and motif analysis (motif-x) were performed using bioinformatics analysis. Co-immunoprecipitation (Co-IP) was used to validate O-GlcNAcylation of candidate proteins. RESULTS OGT knockdown in HG-treated podocytes resulted in 128 upregulated and 45 downregulated proteins. Glycoproteomics revealed 32 glycopeptides/21 glycoproteins upregulated and 37 glycopeptides/22 glycoproteins downregulated. The focus was on down-regulated glycosylated proteins without changes in their protein levels. These proteins are predominantly enriched in translation factor activity, RNA binding, and ECM-receptor interactions pathways. Among these proteins, Caprin1, Lrp1, and Sil1 were modified by O-GlcNAcylation. CONCLUSION OGT-driven O-GlcNAcylation exacerbates podocyte injury in DKD by post-translationally modifying key regulators of translational machinery and ECM signalling. Precision targeting of O-GlcNAc dynamics represents a promising therapeutic strategy to attenuate DKD.
Collapse
Affiliation(s)
- Yun Zou
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, People's Republic of China
- Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, People's Republic of China
| | - Mengyao Zhuo
- Department of Medical Genetics and Cell Biology, Medical College of Nanchang University, Nanchang, People's Republic of China
| | - Wen Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Wenze Song
- Department of Medical Genetics and Cell Biology, Medical College of Nanchang University, Nanchang, People's Republic of China
| | - Yanxia Jiang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Jixiong Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, People's Republic of China
- Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, People's Republic of China
| | - Jiao Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, People's Republic of China
- Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, People's Republic of China
| |
Collapse
|
2
|
Timilsina S, Amara AR, Abu R, Spring BQ. Identification of potential cell surface targets in patient-derived cultures toward photoimmunotherapy of high-grade serous ovarian cancer. Photochem Photobiol 2025. [PMID: 40205302 DOI: 10.1111/php.14091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 04/11/2025]
Abstract
Tumor-targeted, activatable photoimmunotherapy (taPIT) has shown promise in preclinical models to selectively eliminate drug-resistant micrometastases that evade standard treatments. Moreover, taPIT has the potential to resensitize chemo-resistant tumor cells to chemotherapy, making it a complementary modality for treating recurrent high-grade serous ovarian cancer (HGSOC). However, the established implementation of taPIT relies on the overexpression of EGFR in tumor cells, which is not universally observed in HGSOCs. Motivated by the need to expand taPIT applications beyond EGFR, we conducted mRNA-sequencing and proteomics to identify alternative cell surface targets for taPIT in patient-derived HGSOC cell cultures with weak EGFR expression and lacking expression of other cell surface proteins commonly reported in the literature as overexpressed in ovarian cancers, such as FOLR1 and EpCAM. Our findings highlight TFRC and LRP1 as promising alternative targets. Notably, TFRC was overexpressed in 100% (N = 5) of the patient-derived HGSOC models tested, whereas only 60% of models had high EpCAM expression, suggesting that future larger cohort studies should include TFRC. While this study focuses on target identification, future work will expand the approaches developed here to larger HGSOC biopsy repositories and will also develop and evaluate antibody-photosensitizer conjugates targeting these proteins for taPIT applications.
Collapse
Affiliation(s)
- Sudip Timilsina
- Department of Physics, Northeastern University, Boston, Massachusetts, USA
- Translational Biophotonics Cluster, Northeastern University, Boston, Massachusetts, USA
| | - Anish Raju Amara
- Translational Biophotonics Cluster, Northeastern University, Boston, Massachusetts, USA
- Department of Bioinformatics, Northeastern University, Boston, Massachusetts, USA
| | - Rafay Abu
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, USA
- Mass Spectrometry Core, Barnett Institute for Chemical & Biological Analysis, Northeastern University, Boston, Massachusetts, USA
| | - Bryan Q Spring
- Department of Physics, Northeastern University, Boston, Massachusetts, USA
- Translational Biophotonics Cluster, Northeastern University, Boston, Massachusetts, USA
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Kakogiannos N, Scalise AA, Martini E, Maderna C, Benvenuto AF, D’Antonio M, Carmignani L, Magni S, Gullotta GS, Lampugnani MG, Iannelli F, Beznoussenko GV, Mironov AA, Cerutti C, Bentley K, Philippides A, Zanardi F, Bacigaluppi M, Sigismund S, Bassani C, Farina C, Martino G, De Giovanni M, Dejana E, Iannacone M, Inverso D, Giannotta M. GPR126 is a specifier of blood-brain barrier formation in the mouse central nervous system. J Clin Invest 2024; 134:e165368. [PMID: 39087467 PMCID: PMC11290973 DOI: 10.1172/jci165368] [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: 09/13/2022] [Accepted: 06/04/2024] [Indexed: 08/02/2024] Open
Abstract
The blood-brain barrier (BBB) acquires unique properties to regulate neuronal function during development. The formation of the BBB, which occurs in tandem with angiogenesis, is directed by the Wnt/β-catenin signaling pathway. Yet the exact molecular interplay remains elusive. Our study reveals the G protein-coupled receptor GPR126 as a critical target of canonical Wnt signaling, essential for the development of the BBB's distinctive vascular characteristics and its functional integrity. Endothelial cell-specific deletion of the Gpr126 gene in mice induced aberrant vascular morphogenesis, resulting in disrupted BBB organization. Simultaneously, heightened transcytosis in vitro compromised barrier integrity, resulting in enhanced vascular permeability. Mechanistically, GPR126 enhanced endothelial cell migration, pivotal for angiogenesis, acting through an interaction between LRP1 and β1 integrin, thereby balancing the levels of β1 integrin activation and recycling. Overall, we identified GPR126 as a specifier of an organotypic vascular structure, which sustained angiogenesis and guaranteed the acquisition of the BBB properties during development.
Collapse
Affiliation(s)
| | | | - Emanuele Martini
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Claudio Maderna
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Michele D’Antonio
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Carmignani
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Serena Magni
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Giorgia Serena Gullotta
- Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS, San Raffaele Hospital, Milan, Italy
| | | | - Fabio Iannelli
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | | | | | - Camilla Cerutti
- Department of Experimental Oncology, European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Katie Bentley
- The Francis Crick Institute, London, United Kingdom
- Department of Informatics, King’s College London, London, United Kingdom
| | - Andrew Philippides
- Department of Informatics, University of Sussex, Brighton, United Kingdom
| | - Federica Zanardi
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Marco Bacigaluppi
- Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS, San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Sara Sigismund
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Claudia Bassani
- Immunobiology of Neurological Disorders Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cinthia Farina
- Immunobiology of Neurological Disorders Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianvito Martino
- Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS, San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Marco De Giovanni
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Matteo Iannacone
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Donato Inverso
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Monica Giannotta
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
4
|
Ramírez‐Sánchez DA, Canizalez‐Román A, León‐Sicairos N, Pérez Martínez G. The anticancer activity of bovine lactoferrin is reduced by deglycosylation and it follows a different pathway in cervix and colon cancer cells. Food Sci Nutr 2024; 12:3516-3528. [PMID: 38726451 PMCID: PMC11077203 DOI: 10.1002/fsn3.4020] [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: 09/13/2023] [Revised: 01/15/2024] [Accepted: 01/27/2024] [Indexed: 05/12/2024] Open
Abstract
Bovine lactoferrin (bLF) is a glycosylated protein with purported beneficial properties. The aim of this work was to determine the role of bLF glycosylation in the adhesion, internalization, and growth inhibition of cancer cells. The viability of cervix (HeLa) and colon (Caco-2) cancer cells (MTT assay and epifluorescence microscopy) was inhibited by bLF, while deglycosylated bLF (bLFdeg) had no effect. Adhesion to cell surfaces was quantified by immunofluorescence assay and showed that bLF was able to bind more efficiently to both cell lines than bLFdeg. Microscopic observations indicated that bLF glycosylation favored bLF binding to epithelial cells and that it was endocytosed through caveolin-1-mediated internalization. In addition, the mechanism of action of bLF on cancer cell proliferation was investigated by determining the amount of phosphorylated intermediates of signaling pathways such as epidermal growth factor receptor (EGFR), extracellular signal-regulated kinase (ERK) and protein kinase B (known as Akt). Chemoluminescence immunoassay of phosphorylated intermediates showed that bLF inhibited Akt phosphorylation, consistent with its growth inhibiting activity. This assay also indicated that the bLF receptor/signaling pathways may be different in the two cell lines, Caco-2 and HeLa. This work confirmed the effect of glycosylated bLF in inhibiting cancer cell growth and that glycosylation is required for optimal surface adhesion, internalization, and inhibition of the ERK/Akt pathway of cell proliferation through glycosylated cell surface receptors.
Collapse
Affiliation(s)
- Diana A. Ramírez‐Sánchez
- Programa Regional de Noroeste para el Doctorado en BiotecnologíaUniversidad Autónoma de Sinaloa Facultad de Ciencias Químico BiológicasCuliacanMexico
| | - Adrián Canizalez‐Román
- Unidad de Investigación, Facultad de MedicinaUniversidad Autónoma de SinaloaCuliacanMexico
- Servicios de Salud de SinaloaHospital de la MujerCuliacanMexico
| | - Nidia León‐Sicairos
- Unidad de Investigación, Facultad de MedicinaUniversidad Autónoma de SinaloaCuliacanMexico
- Servicios de Salud de Sinaloa, Departamento de Investigación del Hospital Pediátrico de SinaloaCuliacanMexico
| | - Gaspar Pérez Martínez
- Consejo Superior de Investigaciones CientificasInstituto de Agroquímica y Tecnología de AlimentosPaternaSpain
| |
Collapse
|
5
|
Xu W, Liu F, Li Q, Li L, Liu X. Integrated Analysis of miRNA and mRNA Regulation Network in Hypertension. Biochem Genet 2023; 61:2566-2579. [PMID: 37165183 DOI: 10.1007/s10528-023-10389-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/18/2023] [Indexed: 05/12/2023]
Abstract
Hypertension is the most common chronic disease. Early diagnosis is helpful for early medical intervention. The miRNAs and the messenger RNAs (mRNAs) network may be valuable disease diagnosis markers. We aimed to explore the diagnostic value of the miRNA-mRNA network for hypertension patients. Data of miRNAs and mRNAs expression were obtained from the Gene Expression Omnibus database. The weighted gene co-expression network analysis was performed to screen hypertension-related gene modules, and these genes undergone functional enrichment analysis using "clusterProfiler" package. Differential expression analysis was applied on miRNAs expression profiles using "limma" package. TargetScanHuman and miRDB databases were used to select target mRNAs. Cytoscape software was used to visualize the miRNA-mRNA regulation network. P value < 0.05 was considered statistically significant after t test. There were 123 screened mRNAs which were enriched in 161 Gene ontology (GO) terms and 14 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Thirty-five differentially expressed miRNAs (DEMs) are found in the GSE75670. Totally 36 miRNA-mRNA pairs were obtained after the integrated analysis, and three mRNAs and the hsa-miRNA-5589-5p were identified as key joints. Hub genes, KIAA0513, ARID3A and LRPAP1, and key hsa-miRNA-5589-5p are potential diagnostic biomarkers for hypertension. Our findings are promising in the clinical application, conducive to early detection and prompt intervention of hypertension.
Collapse
Affiliation(s)
- Weijuan Xu
- Department of Geriatrics, Zibo Central Hospital, No 54 Gongqingtuan West Road, 255036, Zibo, People's Republic of China
| | - Feng Liu
- Department of Emergency, Zibo Central Hospital, No 54 Gongqingtuan West Road, 255036, Zibo, People's Republic of China
| | - Qinghua Li
- Department of Emergency, Zibo Central Hospital, No 54 Gongqingtuan West Road, 255036, Zibo, People's Republic of China
| | - Li Li
- Department of Geriatrics, Zibo Central Hospital, No 54 Gongqingtuan West Road, 255036, Zibo, People's Republic of China.
| | - Xiqiang Liu
- Department of Geriatrics, Zibo Central Hospital, No 54 Gongqingtuan West Road, 255036, Zibo, People's Republic of China
| |
Collapse
|
6
|
Soler Y, Rodriguez M, Austin D, Gineste C, Gelber C, El-Hage N. SERPIN-Derived Small Peptide (SP16) as a Potential Therapeutic Agent against HIV-Induced Inflammatory Molecules and Viral Replication in Cells of the Central Nervous System. Cells 2023; 12:cells12040632. [PMID: 36831299 PMCID: PMC9954444 DOI: 10.3390/cells12040632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/18/2023] Open
Abstract
Despite the success of combined antiretroviral therapy (cART) increasing the survival rate in human immunodeficiency virus (HIV) patients, low levels of viremia persist in the brain of patients leading to glia (microglia and astrocytes)-induced neuroinflammation and consequently, the reactivation of HIV and neuronal injury. Here, we tested the therapeutic efficacy of a Low-Density Lipoprotein Receptor-Related Protein 1 (LRP-1) agonistic small peptide drug (SP16) in attenuating HIV replication and the secretion of inflammatory molecules in brain reservoirs. SP16 was developed by Serpin Pharma and is derived from the pentapeptide sequence of the serine protease inhibitor alpha-1-antitrypsin (A1AT). The SP16 peptide sequence was subsequently modified to improve the stability, bioavailability, efficacy, and binding to LRP-1; a scavenger regulatory receptor that internalizes ligands to induce anti-viral, anti-inflammatory, and pro-survival signals. Using glial cells infected with HIV, we showed that: (i) SP16 attenuated viral-induced secretion of pro-inflammatory molecules; and (ii) SP16 attenuated viral replication. Using an artificial 3D blood-brain barrier (BBB) system, we showed that: (i) SP16 was transported across the BBB; and (ii) restored the permeability of the BBB compromised by HIV. Mechanistically, we showed that SP16 interaction with LRP-1 and binding lead to: (i) down-regulation in the expression levels of nuclear factor-kappa beta (NF-κB); and (ii) up-regulation in the expression levels of Akt. Using an in vivo mouse model, we showed that SP16 was transported across the BBB after intranasal delivery, while animals infected with EcoHIV undergo a reduction in (i) viral replication and (ii) viral secreted inflammatory molecules, after exposure to SP16 and antiretrovirals. Overall, these studies confirm a therapeutic response of SP16 against HIV-associated inflammatory effects in the brain.
Collapse
Affiliation(s)
- Yemmy Soler
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Miami, FL 33199, USA
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, USA
| | - Myosotys Rodriguez
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Miami, FL 33199, USA
| | - Dana Austin
- Serpin Pharma, 9501 Discovery Blvd Suite 120, Manassas, VA 20109, USA
| | - Cyrille Gineste
- Serpin Pharma, 9501 Discovery Blvd Suite 120, Manassas, VA 20109, USA
| | - Cohava Gelber
- Serpin Pharma, 9501 Discovery Blvd Suite 120, Manassas, VA 20109, USA
| | - Nazira El-Hage
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Miami, FL 33199, USA
- Correspondence: ; Tel.: +1-(305)-348-4346; Fax: +1-(305)-348-1109
| |
Collapse
|
7
|
Baraniecki Ł, Tokarz-Deptuła B, Syrenicz A, Deptuła W. Macrophage efferocytosis in atherosclerosis. Scand J Immunol 2022; 97:e13251. [PMID: 36583598 DOI: 10.1111/sji.13251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/17/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022]
Abstract
This paper presents the role of macrophage efferocytosis, the process of elimination of apoptotic bodies-elements formed during vascular atherosclerosis. The mechanisms of macrophage efferocytosis are presented, introducing the specific signals of this process, that is, 'find me', 'eat me' and 'don't eat me'. The role of the process of efferocytosis in the formation of vascular atherosclerosis is also presented, including the factors and mechanisms that determine it, as well as the factors that determine the maintenance of homeostasis in the vessels, including the formation of vascular atherosclerosis.
Collapse
Affiliation(s)
| | | | - Anhelli Syrenicz
- Department of Endocrinology, Metabolic Diseases and Internal Diseases, Pomeranian Medical University, Szczecin, Poland
| | - Wiesław Deptuła
- Faculty of Biological and Veterinary Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, Toruń, Poland
| |
Collapse
|
8
|
Friedmacher F, Rolle U, Puri P. Genetically Modified Mouse Models of Congenital Diaphragmatic Hernia: Opportunities and Limitations for Studying Altered Lung Development. Front Pediatr 2022; 10:867307. [PMID: 35633948 PMCID: PMC9136148 DOI: 10.3389/fped.2022.867307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/18/2022] [Indexed: 11/21/2022] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a relatively common and life-threatening birth defect, characterized by an abnormal opening in the primordial diaphragm that interferes with normal lung development. As a result, CDH is accompanied by immature and hypoplastic lungs, being the leading cause of morbidity and mortality in patients with this condition. In recent decades, various animal models have contributed novel insights into the pathogenic mechanisms underlying CDH and associated pulmonary hypoplasia. In particular, the generation of genetically modified mouse models, which show both diaphragm and lung abnormalities, has resulted in the discovery of multiple genes and signaling pathways involved in the pathogenesis of CDH. This article aims to offer an up-to-date overview on CDH-implicated transcription factors, molecules regulating cell migration and signal transduction as well as components contributing to the formation of extracellular matrix, whilst also discussing the significance of these genetic models for studying altered lung development with regard to the human situation.
Collapse
Affiliation(s)
- Florian Friedmacher
- Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Udo Rolle
- Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Prem Puri
- Beacon Hospital, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
9
|
Calvier L, Herz J, Hansmann G. Interplay of Low-Density Lipoprotein Receptors, LRPs, and Lipoproteins in Pulmonary Hypertension. JACC Basic Transl Sci 2022; 7:164-180. [PMID: 35257044 PMCID: PMC8897182 DOI: 10.1016/j.jacbts.2021.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/21/2022]
Abstract
The low-density lipoprotein receptor (LDLR) gene family includes LDLR, very LDLR, and LDL receptor-related proteins (LRPs) such as LRP1, LRP1b (aka LRP-DIT), LRP2 (aka megalin), LRP4, and LRP5/6, and LRP8 (aka ApoER2). LDLR family members constitute a class of closely related multifunctional, transmembrane receptors, with diverse functions, from embryonic development to cancer, lipid metabolism, and cardiovascular homeostasis. While LDLR family members have been studied extensively in the systemic circulation in the context of atherosclerosis, their roles in pulmonary arterial hypertension (PAH) are understudied and largely unknown. Endothelial dysfunction, tissue infiltration of monocytes, and proliferation of pulmonary artery smooth muscle cells are hallmarks of PAH, leading to vascular remodeling, obliteration, increased pulmonary vascular resistance, heart failure, and death. LDLR family members are entangled with the aforementioned detrimental processes by controlling many pathways that are dysregulated in PAH; these include lipid metabolism and oxidation, but also platelet-derived growth factor, transforming growth factor β1, Wnt, apolipoprotein E, bone morpohogenetic proteins, and peroxisome proliferator-activated receptor gamma. In this paper, we discuss the current knowledge on LDLR family members in PAH. We also review mechanisms and drugs discovered in biological contexts and diseases other than PAH that are likely very relevant in the hypertensive pulmonary vasculature and the future care of patients with PAH or other chronic, progressive, debilitating cardiovascular diseases.
Collapse
Key Words
- ApoE, apolipoprotein E
- Apoer2
- BMP
- BMPR, bone morphogenetic protein receptor
- BMPR2
- COPD, chronic obstructive pulmonary disease
- CTGF, connective tissue growth factor
- HDL, high-density lipoprotein
- KO, knockout
- LDL receptor related protein
- LDL, low-density lipoprotein
- LDLR
- LDLR, low-density lipoprotein receptor
- LRP
- LRP, low-density lipoprotein receptor–related protein
- LRP1
- LRP1B
- LRP2
- LRP4
- LRP5
- LRP6
- LRP8
- MEgf7
- Mesd, mesoderm development
- PAH
- PAH, pulmonary arterial hypertension
- PASMC, pulmonary artery smooth muscle cell
- PDGF
- PDGFR-β, platelet-derived growth factor receptor-β
- PH, pulmonary hypertension
- PPARγ
- PPARγ, peroxisome proliferator-activated receptor gamma
- PVD
- RV, right ventricle/ventricular
- RVHF
- RVSP, right ventricular systolic pressure
- TGF-β1
- TGF-β1, transforming growth factor β1
- TGFBR, transforming growth factor β1 receptor
- TNF, tumor necrosis factor receptor
- VLDLR
- VLDLR, very low density lipoprotein receptor
- VSMC, vascular smooth muscle cell
- Wnt
- apolipoprotein E receptor 2
- endothelial cell
- gp330
- low-density lipoprotein receptor
- mRNA, messenger RNA
- megalin
- monocyte
- multiple epidermal growth factor-like domains 7
- pulmonary arterial hypertension
- pulmonary vascular disease
- right ventricle heart failure
- smooth muscle cell
- very low density lipoprotein receptor
- β-catenin
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
- Pulmonary Vascular Research Center, Hannover Medical School, Hannover, Germany
| |
Collapse
|
10
|
The essential anti-angiogenic strategies in cartilage engineering and osteoarthritic cartilage repair. Cell Mol Life Sci 2022; 79:71. [PMID: 35029764 PMCID: PMC9805356 DOI: 10.1007/s00018-021-04105-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/01/2021] [Accepted: 12/18/2021] [Indexed: 01/16/2023]
Abstract
In the cartilage matrix, complex interactions occur between angiogenic and anti-angiogenic components, growth factors, and environmental stressors to maintain a proper cartilage phenotype that allows for effective load bearing and force distribution. However, as seen in both degenerative disease and tissue engineering, cartilage can lose its vascular resistance. This vascularization then leads to matrix breakdown, chondrocyte apoptosis, and ossification. Research has shown that articular cartilage inflammation leads to compromised joint function and decreased clinical potential for regeneration. Unfortunately, few articles comprehensively summarize what we have learned from previous investigations. In this review, we summarize our current understanding of the factors that stabilize chondrocytes to prevent terminal differentiation and applications of these factors to rescue the cartilage phenotype during cartilage engineering and osteoarthritis treatment. Inhibiting vascularization will allow for enhanced phenotypic stability so that we are able to develop more stable implants for cartilage repair and regeneration.
Collapse
|
11
|
Campion O, Thevenard Devy J, Billottet C, Schneider C, Etique N, Dupuy JW, Raymond AA, Boulagnon Rombi C, Meunier M, Djermoune EH, Lelièvre E, Wahart A, Bour C, Hachet C, Cairo S, Bikfalvi A, Dedieu S, Devy J. LRP-1 Matricellular Receptor Involvement in Triple Negative Breast Cancer Tumor Angiogenesis. Biomedicines 2021; 9:biomedicines9101430. [PMID: 34680548 PMCID: PMC8533426 DOI: 10.3390/biomedicines9101430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 01/15/2023] Open
Abstract
Background: LRP-1 is a multifunctional scavenger receptor belonging to the LDLR family. Due to its capacity to control pericellular levels of various growth factors and proteases, LRP-1 plays a crucial role in membrane proteome dynamics, which appears decisive for tumor progression. Methods: LRP-1 involvement in a TNBC model was assessed using an RNA interference strategy in MDA-MB-231 cells. In vivo, tumorigenic and angiogenic effects of LRP-1-repressed cells were evaluated using an orthotopic xenograft model and two angiogenic assays (Matrigel® plugs, CAM). DCE-MRI, FMT, and IHC were used to complete a tumor longitudinal follow-up and obtain morphological and functional vascular information. In vitro, HUVECs’ angiogenic potential was evaluated using a tumor secretome, subjected to a proteomic analysis to highlight LRP-1-dependant signaling pathways. Results: LRP-1 repression in MDA-MB-231 tumors led to a 60% growth delay because of, inter alia, morphological and functional vascular differences, confirmed by angiogenic models. In vitro, the LRP-1-repressed cells secretome restrained HUVECs’ angiogenic capabilities. A proteomics analysis revealed that LRP-1 supports tumor growth and angiogenesis by regulating TGF-β signaling and plasminogen/plasmin system. Conclusions: LRP-1, by its wide spectrum of interactions, emerges as an important matricellular player in the control of cancer-signaling events such as angiogenesis, by supporting tumor vascular morphology and functionality.
Collapse
Affiliation(s)
- Océane Campion
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Jessica Thevenard Devy
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Clotilde Billottet
- INSERM, LAMC, U1029, Université de Bordeaux, 33600 Pessac, France; (C.B.); (A.B.)
| | - Christophe Schneider
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Nicolas Etique
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | | | | | - Camille Boulagnon Rombi
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
- Laboratoire d’Anatomie Pathologie, CHU Reims, 51100 Reims, France
| | - Marie Meunier
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | | | - Elodie Lelièvre
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Amandine Wahart
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Camille Bour
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Cathy Hachet
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | | | - Andréas Bikfalvi
- INSERM, LAMC, U1029, Université de Bordeaux, 33600 Pessac, France; (C.B.); (A.B.)
| | - Stéphane Dedieu
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Jérôme Devy
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
- Correspondence:
| |
Collapse
|
12
|
Storck SE, Kurtyka M, Pietrzik CU. Brain endothelial LRP1 maintains blood-brain barrier integrity. Fluids Barriers CNS 2021; 18:27. [PMID: 34147102 PMCID: PMC8214794 DOI: 10.1186/s12987-021-00260-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 06/07/2021] [Indexed: 11/28/2022] Open
Abstract
The entry of blood-borne molecules into the brain is restricted by the blood–brain barrier (BBB). Various physical, transport and immune properties tightly regulate molecule movement between the blood and the brain to maintain brain homeostasis. A recent study utilizing a pan-endothelial, constitutive Tie2-Cre showed that paracellular passage of blood proteins into the brain is governed by endocytic and cell signaling protein low-density lipoprotein receptor–related protein 1 (LRP1). Taking advantage of conditional Slco1c1-CreERT2 specific to CNS endothelial cells and choroid plexus epithelial cells we now supplement previous results and show that brain endothelial Lrp1 ablation results in protease-mediated tight junction degradation, P-glycoprotein (P-gp) reduction and a loss of BBB integrity.
Collapse
Affiliation(s)
- Steffen E Storck
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Duesbergweg 6, 55099, Mainz, Germany.
| | - Magdalena Kurtyka
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Duesbergweg 6, 55099, Mainz, Germany
| | - Claus U Pietrzik
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Duesbergweg 6, 55099, Mainz, Germany.
| |
Collapse
|
13
|
Munshaw S, Bruche S, Redpath AN, Jones A, Patel J, Dubé KN, Lee R, Hester SS, Davies R, Neal G, Handa A, Sattler M, Fischer R, Channon KM, Smart N. Thymosin β4 protects against aortic aneurysm via endocytic regulation of growth factor signaling. J Clin Invest 2021; 131:127884. [PMID: 33784254 PMCID: PMC8121525 DOI: 10.1172/jci127884] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/23/2021] [Indexed: 01/06/2023] Open
Abstract
Vascular stability and tone are maintained by contractile smooth muscle cells (VSMCs). However, injury-induced growth factors stimulate a contractile-synthetic phenotypic modulation which increases susceptibility to abdominal aortic aneurysm (AAA). As a regulator of embryonic VSMC differentiation, we hypothesized that Thymosin β4 (Tβ4) may function to maintain healthy vasculature throughout postnatal life. This was supported by the identification of an interaction with low density lipoprotein receptor related protein 1 (LRP1), an endocytic regulator of platelet-derived growth factor BB (PDGF-BB) signaling and VSMC proliferation. LRP1 variants have been implicated by genome-wide association studies with risk of AAA and other arterial diseases. Tβ4-null mice displayed aortic VSMC and elastin defects that phenocopy those of LRP1 mutants, and their compromised vascular integrity predisposed them to Angiotensin II-induced aneurysm formation. Aneurysmal vessels were characterized by enhanced VSMC phenotypic modulation and augmented PDGFR-β signaling. In vitro, enhanced sensitivity to PDGF-BB upon loss of Tβ4 was associated with dysregulated endocytosis, with increased recycling and reduced lysosomal targeting of LRP1-PDGFR-β. Accordingly, the exacerbated aneurysmal phenotype in Tβ4-null mice was rescued upon treatment with the PDGFR-β antagonist Imatinib. Our study identifies Tβ4 as a key regulator of LRP1 for maintaining vascular health, and provides insights into the mechanisms of growth factor-controlled VSMC phenotypic modulation underlying aortic disease progression.
Collapse
MESH Headings
- Angiotensin II/adverse effects
- Angiotensin II/pharmacology
- Animals
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/prevention & control
- Becaplermin/genetics
- Becaplermin/metabolism
- Low Density Lipoprotein Receptor-Related Protein-1/genetics
- Low Density Lipoprotein Receptor-Related Protein-1/metabolism
- Male
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Thymosin/genetics
- Thymosin/metabolism
- Thymosin/pharmacology
Collapse
Affiliation(s)
- Sonali Munshaw
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Susann Bruche
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Andia N. Redpath
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Alisha Jones
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Munich, Germany
- Biomolecular NMR and Center for Integrated Protein Science Munich at Chemistry Department, Technical University of Munich, Garching, Munich, Germany
| | - Jyoti Patel
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | | | - Regent Lee
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Svenja S. Hester
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Rachel Davies
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Giles Neal
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Ashok Handa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Munich, Germany
- Biomolecular NMR and Center for Integrated Protein Science Munich at Chemistry Department, Technical University of Munich, Garching, Munich, Germany
| | - Roman Fischer
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Keith M. Channon
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Nicola Smart
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| |
Collapse
|
14
|
He Z, Wang G, Wu J, Tang Z, Luo M. The molecular mechanism of LRP1 in physiological vascular homeostasis and signal transduction pathways. Biomed Pharmacother 2021; 139:111667. [PMID: 34243608 DOI: 10.1016/j.biopha.2021.111667] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/07/2021] [Accepted: 04/23/2021] [Indexed: 01/10/2023] Open
Abstract
Interactions between vascular smooth muscle cells (VSMCs), endothelial cells (ECs), pericytes (PCs) and macrophages (MФ), the major components of blood vessels, play a crucial role in maintaining vascular structural and functional homeostasis. Low-density lipoprotein (LDL) receptor-related protein-1 (LRP1), a transmembrane receptor protein belonging to the LDL receptor family, plays multifunctional roles in maintaining endocytosis, homeostasis, and signal transduction. Accumulating evidence suggests that LRP1 modulates vascular homeostasis mainly by regulating vasoactive substances and specific intracellular signaling pathways, including the plasminogen activator inhibitor 1 (PAI-1) signaling pathway, platelet-derived growth factor (PDGF) signaling pathway, transforming growth factor-β (TGF-β) signaling pathway and vascular endothelial growth factor (VEGF) signaling pathway. The aim of the present review is to focus on recent advances in the discovery and mechanism of vascular homeostasis regulated by LRP1-dependent signaling pathways. These recent discoveries expand our understanding of the mechanisms controlling LRP1 as a target for studies on vascular complications.
Collapse
Affiliation(s)
- Zhaohui He
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Department of Clinical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Wang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianbo Wu
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Zonghao Tang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Mao Luo
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
15
|
Heissig B, Salama Y, Takahashi S, Osada T, Hattori K. The multifaceted role of plasminogen in inflammation. Cell Signal 2020; 75:109761. [PMID: 32861744 PMCID: PMC7452830 DOI: 10.1016/j.cellsig.2020.109761] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 01/01/2023]
Abstract
A fine-tuned activation and deactivation of proteases and their inhibitors are involved in the execution of the inflammatory response. The zymogen/proenzyme plasminogen is converted to the serine protease plasmin, a key fibrinolytic factor by plasminogen activators including tissue-type plasminogen activator (tPA). Plasmin is part of an intricate protease network controlling proteins of initial hemostasis/coagulation, fibrinolytic and complement system. Activation of these protease cascades is required to mount a proper inflammatory response. Although best known for its ability to dissolve clots and cleave fibrin, recent studies point to the importance of fibrin-independent functions of plasmin during acute inflammation and inflammation resolution. In this review, we provide an up-to-date overview of the current knowledge of the enzymatic and cytokine-like effects of tPA and describe the role of tPA and plasminogen receptors in the regulation of the inflammatory response with emphasis on the cytokine storm syndrome such as observed during coronavirus disease 2019 or macrophage activation syndrome. We discuss tPA as a modulator of Toll like receptor signaling, plasmin as an activator of NFkB signaling, and summarize recent studies on the role of plasminogen receptors as controllers of the macrophage conversion into the M2 type and as mediators of efferocytosis during inflammation resolution.
Collapse
Affiliation(s)
- Beate Heissig
- Department of Immunological Diagnosis, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan.
| | - Yousef Salama
- An-Najah Center for Cancer and Stem Cell Research, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine.
| | - Satoshi Takahashi
- Department of Hematology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| | - Taro Osada
- Department of Gastroenterology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi, 279-0021 Chiba, Japan.
| | - Koichi Hattori
- Center for Genomic & Regenerative Medicine, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan.
| |
Collapse
|
16
|
Chieosilapatham P, Yue H, Ikeda S, Ogawa H, Niyonsaba F. Involvement of the lipoprotein receptor LRP1 in AMP-IBP5-mediated migration and proliferation of human keratinocytes and fibroblasts. J Dermatol Sci 2020; 99:158-167. [DOI: 10.1016/j.jdermsci.2020.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 05/13/2020] [Accepted: 07/13/2020] [Indexed: 12/25/2022]
|
17
|
De La-Rocque S, Moretto E, Butnaru I, Schiavo G. Knockin' on heaven's door: Molecular mechanisms of neuronal tau uptake. J Neurochem 2020; 156:563-588. [PMID: 32770783 PMCID: PMC8432157 DOI: 10.1111/jnc.15144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022]
Abstract
Since aggregates of the microtubule‐binding protein tau were found to be the main component of neurofibrillary tangles more than 30 years ago, their contribution to neurodegeneration in Alzheimer's disease (AD) and tauopathies has become well established. Recent work shows that both tau load and its distribution in the brain of AD patients correlate with cognitive decline more closely compared to amyloid plaque deposition. In addition, the amyloid cascade hypothesis has been recently challenged because of disappointing results of clinical trials designed to treat AD by reducing beta‐amyloid levels, thus fuelling a renewed interest in tau. There is now robust evidence to indicate that tau pathology can spread within the central nervous system via a prion‐like mechanism following a stereotypical pattern, which can be explained by the trans‐synaptic inter‐neuronal transfer of pathological tau. In the receiving neuron, tau has been shown to take multiple routes of internalisation, which are partially dependent on its conformation and aggregation status. Here, we review the emerging mechanisms proposed for the uptake of extracellular tau in neurons and the requirements for the propagation of its pathological conformers, addressing how they gain access to physiological tau monomers in the cytosol. Furthermore, we highlight some of the key mechanistic gaps of the field, which urgently need to be addressed to expand our understanding of tau propagation and lead to the identification of new therapeutic strategies for tauopathies.
Collapse
Affiliation(s)
- Samantha De La-Rocque
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Edoardo Moretto
- UK Dementia Research Institute, University College London, London, UK
| | - Ioana Butnaru
- UK Dementia Research Institute, University College London, London, UK
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
18
|
Campion O, Al Khalifa T, Langlois B, Thevenard-Devy J, Salesse S, Savary K, Schneider C, Etique N, Dedieu S, Devy J. Contribution of the Low-Density Lipoprotein Receptor Family to Breast Cancer Progression. Front Oncol 2020; 10:882. [PMID: 32850302 PMCID: PMC7406569 DOI: 10.3389/fonc.2020.00882] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/05/2020] [Indexed: 12/18/2022] Open
Abstract
The low-density lipoprotein receptor (LDLR) family comprises 14 single-transmembrane receptors sharing structural homology and common repeats. These receptors specifically recognize and internalize various extracellular ligands either alone or complexed with membrane-spanning co-receptors that are then sorted for lysosomal degradation or cell-surface recovery. As multifunctional endocytic receptors, some LDLR members from the core family were first considered as potential tumor suppressors due to their clearance activity against extracellular matrix-degrading enzymes. LDLRs are also involved in pleiotropic functions including growth factor signaling, matricellular proteins, and cell matrix adhesion turnover and chemoattraction, thereby affecting both tumor cells and their surrounding microenvironment. Therefore, their roles could appear controversial and dependent on the malignancy state. In this review, recent advances highlighting the contribution of LDLR members to breast cancer progression are discussed with focus on (1) specific expression patterns of these receptors in primary cancers or distant metastasis and (2) emerging mechanisms and signaling pathways. In addition, potential diagnosis and therapeutic options are proposed.
Collapse
Affiliation(s)
- Océane Campion
- Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Tesnim Al Khalifa
- Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Benoit Langlois
- Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Jessica Thevenard-Devy
- Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Stéphanie Salesse
- Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Katia Savary
- Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Christophe Schneider
- Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Nicolas Etique
- Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Stéphane Dedieu
- Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Jérôme Devy
- Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| |
Collapse
|
19
|
Rothe R, Schulze S, Neuber C, Hauser S, Rammelt S, Pietzsch J. Adjuvant drug-assisted bone healing: Part II - Modulation of angiogenesis. Clin Hemorheol Microcirc 2020; 73:409-438. [PMID: 31177206 DOI: 10.3233/ch-199103] [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] [Indexed: 12/12/2022]
Abstract
The treatment of critical-size bone defects following complicated fractures, infections or tumor resections is a major challenge. The same applies to fractures in patients with impaired bone healing due to systemic inflammatory and metabolic diseases. Despite considerable progress in development and establishment of new surgical techniques, design of bone graft substitutes and imaging techniques, these scenarios still represent unresolved clinical problems. However, the development of new active substances offers novel potential solutions for these issues. This work discusses therapeutic approaches that influence angiogenesis or hypoxic situations in healing bone and surrounding tissue. In particular, literature on sphingosine-1-phosphate receptor modulators and nitric oxide (NO•) donors, including bi-functional (hybrid) compounds like NO•-releasing cyclooxygenase-2 inhibitors, was critically reviewed with regard to their local and systemic mode of action.
Collapse
Affiliation(s)
- Rebecca Rothe
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sabine Schulze
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Tatzberg 4, Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
20
|
Saeui CT, Cho KC, Dharmarha V, Nairn AV, Galizzi M, Shah SR, Gowda P, Park M, Austin M, Clarke A, Cai E, Buettner MJ, Ariss R, Moremen KW, Zhang H, Yarema KJ. Cell Line-, Protein-, and Sialoglycosite-Specific Control of Flux-Based Sialylation in Human Breast Cells: Implications for Cancer Progression. Front Chem 2020; 8:13. [PMID: 32117864 PMCID: PMC7013041 DOI: 10.3389/fchem.2020.00013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022] Open
Abstract
Sialylation, a post-translational modification that impacts the structure, activity, and longevity of glycoproteins has been thought to be controlled primarily by the expression of sialyltransferases (STs). In this report we explore the complementary impact of metabolic flux on sialylation using a glycoengineering approach. Specifically, we treated three human breast cell lines (MCF10A, T-47D, and MDA-MB-231) with 1,3,4-O-Bu3ManNAc, a "high flux" metabolic precursor for the sialic acid biosynthetic pathway. We then analyzed N-glycan sialylation using solid phase extraction of glycopeptides (SPEG) mass spectrometry-based proteomics under conditions that selectively captured sialic acid-containing glycopeptides, referred to as "sialoglycosites." Gene ontology (GO) analysis showed that flux-based changes to sialylation were broadly distributed across classes of proteins in 1,3,4-O-Bu3ManNAc-treated cells. Only three categories of proteins, however, were "highly responsive" to flux (defined as two or more sialylation changes of 10-fold or greater). Two of these categories were cell signaling and cell adhesion, which reflect well-known roles of sialic acid in oncogenesis. A third category-protein folding chaperones-was unexpected because little precedent exists for the role of glycosylation in the activity of these proteins. The highly flux-responsive proteins were all linked to cancer but sometimes as tumor suppressors, other times as proto-oncogenes, or sometimes both depending on sialylation status. A notable aspect of our analysis of metabolically glycoengineered breast cells was decreased sialylation of a subset of glycosites, which was unexpected because of the increased intracellular levels of sialometabolite "building blocks" in the 1,3,4-O-Bu3ManNAc-treated cells. Sites of decreased sialylation were minor in the MCF10A (<25% of all glycosites) and T-47D (<15%) cells but dominated in the MDA-MB-231 line (~60%) suggesting that excess sialic acid could be detrimental in advanced cancer and cancer cells can evolve mechanisms to guard against hypersialylation. In summary, flux-driven changes to sialylation offer an intriguing and novel mechanism to switch between context-dependent pro- or anti-cancer activities of the several oncoproteins identified in this study. These findings illustrate how metabolic glycoengineering can uncover novel roles of sialic acid in oncogenesis.
Collapse
Affiliation(s)
- Christopher T Saeui
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Kyung-Cho Cho
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Vrinda Dharmarha
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Alison V Nairn
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Melina Galizzi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Sagar R Shah
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Prateek Gowda
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Marian Park
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Melissa Austin
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Amelia Clarke
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Edward Cai
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Matthew J Buettner
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Ryan Ariss
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Hui Zhang
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Kevin J Yarema
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States.,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD, United States.,Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
21
|
Du C, Liu X, Hu H, Li H, Yu L, Geng D, Chen Y, Zhang J. Dual-targeting and excretable ultrasmall SPIONs for T1-weighted positive MR imaging of intracranial glioblastoma cells by targeting the lipoprotein receptor-related protein. J Mater Chem B 2020; 8:2296-2306. [PMID: 32100784 DOI: 10.1039/c9tb02391g] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A multifunctional targeted nanoprobe composed of ultrasmall superparamagnetic iron oxide nanoparticles with surface-conjugated Angiopep-2 was successfully constructed for targeted MR imaging of intracranial glioblastoma.
Collapse
Affiliation(s)
- Chengjuan Du
- Department of Radiology
- Huashan Hospital
- Shanghai
- P. R. China
| | - Xianping Liu
- Department of Radiology
- Huashan Hospital
- Shanghai
- P. R. China
| | - Hui Hu
- Department of Radiology
- The Affiliated Renmin Hospital of Jiangsu University
- Zhenjiang
- China
| | - Huiming Li
- Department of Radiology
- Huashan Hospital
- Shanghai
- P. R. China
| | - Luodan Yu
- State Laboratory of High Performance Ceramics and Superfine Microstructure
- Shanghai Institute of Ceramics
- Chinese Academy of Sciences
- Shanghai
- P. R. China
| | - Daoying Geng
- Department of Radiology
- Huashan Hospital
- Shanghai
- P. R. China
| | - Yu Chen
- State Laboratory of High Performance Ceramics and Superfine Microstructure
- Shanghai Institute of Ceramics
- Chinese Academy of Sciences
- Shanghai
- P. R. China
| | - Jun Zhang
- Department of Radiology
- Huashan Hospital
- Shanghai
- P. R. China
| |
Collapse
|
22
|
Potere N, Del Buono MG, Mauro AG, Abbate A, Toldo S. Low Density Lipoprotein Receptor-Related Protein-1 in Cardiac Inflammation and Infarct Healing. Front Cardiovasc Med 2019; 6:51. [PMID: 31080804 PMCID: PMC6497734 DOI: 10.3389/fcvm.2019.00051] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/09/2019] [Indexed: 01/07/2023] Open
Abstract
Acute myocardial infarction (AMI) leads to myocardial cell death and ensuing sterile inflammatory response, which represents an attempt to clear cellular debris and promote cardiac repair. However, an overwhelming, unopposed or unresolved inflammatory response following AMI leads to further injury, worse remodeling and heart failure (HF). Additional therapies are therefore warranted to blunt the inflammatory response associated with ischemia and reperfusion and prevent long-term adverse events. Low-density lipoprotein receptor-related protein 1 (LRP1) is a ubiquitous endocytic cell surface receptor with the ability to recognize a wide range of structurally and functionally diverse ligands. LRP1 transduces multiple intracellular signal pathways regulating the inflammatory reaction, tissue remodeling and cell survival after organ injury. In preclinical studies, activation of LRP1-mediated signaling in the heart with non-selective and selective LRP1 agonists is linked with a powerful cardioprotective effect, reducing infarct size and cardiac dysfunction after AMI. The data from early phase clinical studies with plasma-derived α1-antitrypsin (AAT), an endogenous LRP1 agonist, and SP16 peptide, a synthetic LRP1 agonist, support the translational value of LRP1 as a novel therapeutic target in AMI. In this review, we will summarize the cellular and molecular bases of LRP1 functions in modulating the inflammatory reaction and the reparative process after injury in various peripheral tissues, and discuss recent evidences implicating LRP1 in myocardial inflammation and infarct healing.
Collapse
Affiliation(s)
- Nicola Potere
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Marco Giuseppe Del Buono
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Adolfo Gabriele Mauro
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
23
|
Over-expression of low-density lipoprotein receptor-related Protein-1 is associated with poor prognosis and invasion in pancreatic ductal adenocarcinoma. Pancreatology 2019; 19:429-435. [PMID: 30902418 DOI: 10.1016/j.pan.2019.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 02/16/2019] [Accepted: 02/23/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Low-density lipoprotein receptor-Related Protein-1 (LRP-1) has been reported to involve in tumor development. However, its role in pancreatic cancer has not been elucidated. The present study was designed to evaluate the expression of LRP-1 in Pancreatic Ductal Adenocarcinoma Cancer (PDAC) as well as its association with prognosis. METHODS Here, 478 pancreatic cancers were screened for suitable primary PDAC tumors. The samples were analyzed using qRT-PCR, western blotting, and Immunohistochemistry (IHC) staining as well as LRP-1 expression in association with clinicopathological features. RESULTS The relative LRP-1 mRNA expression was up-regulated in 82.3% (42/51) of the PDAC tumors and its expression (3.72 ± 1.25) was significantly higher than that in pancreatic normal margins (1.0 ± 0.23, P < 0.05). This up-regulation was stage dependent (P < 0.05). A similar pattern of LRP-1 protein expression was discovered (P < 0.05). The high expression of LRP-1 in the PDAC tissues was strongly correlated with the low survival time (P = 0.001), TNM classification (P = 0.001), low differentiations status (P = 0.001), lymphatic invasion (P = 0.01) and Perineural Invasion (PNI) status (P = 0.001). CONCLUSIONS Our finding for the first time revealed that LRP-1 expression inversely associated with poor prognosis and PNI in PDAC tumor.
Collapse
|
24
|
Potere N, Del Buono MG, Niccoli G, Crea F, Toldo S, Abbate A. Developing LRP1 Agonists into a Therapeutic Strategy in Acute Myocardial Infarction. Int J Mol Sci 2019; 20:E544. [PMID: 30696029 PMCID: PMC6387161 DOI: 10.3390/ijms20030544] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/14/2019] [Accepted: 01/25/2019] [Indexed: 12/16/2022] Open
Abstract
Cardioprotection refers to a strategy aimed at enhancing survival pathways in the injured yet salvageable myocardium following ischemia-reperfusion. Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional receptor that can be targeted following reperfusion, to induce a cardioprotective signaling through the activation of the reperfusion injury salvage kinase (RISK) pathway. The data from preclinical studies with non-selective and selective LRP1 agonists are promising, showing a large therapeutic window for intervention to reduce infarct size after ischemia-reperfusion. A pilot clinical trial with plasma derived α1-antitrypsin (AAT), a naturally occurring LRP1 agonist, supports the translational value of LRP1 as a novel therapeutic target for cardioprotection. A phase I study with a selective LRP1 agonist has been completed showing no toxicity. These findings may open the way to early phase clinical studies with pharmacologic LRP1 activation in patients with acute myocardial infarction (AMI).
Collapse
Affiliation(s)
- Nicola Potere
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
- Unit of Cardiovascular Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy.
| | - Marco Giuseppe Del Buono
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Giampaolo Niccoli
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
25
|
Overexpression of low density lipoprotein receptor-related protein 1 (LRP1) is associated with worsened prognosis and decreased cancer immunity in clear-cell renal cell carcinoma. Biochem Biophys Res Commun 2018; 503:1537-1543. [PMID: 30033103 DOI: 10.1016/j.bbrc.2018.07.076] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022]
Abstract
AIM Clear-cell renal cell carcinoma (ccRCC) is characterized with underlying genetic disorders and the role of low density lipoprotein receptor-related protein 1 (LRP1) in ccRCC is unknown. METHOD An in silico exploratory analysis using multiple public genetic datasets was used to establish association between LRP1 expression and clinicopathological parameters. Associations of interest were validated using 155 ccRCC samples using immunohistochemistry. RESULTS LRP1 was overexpressed in tumor compared with normal kidney tissue. Increased LRP1 expression in ccRCC was associated with advanced stage, grade and worsened overall survival and progression-free survival. Functional annotation indicated an immune-modulatory role of LRP1 in ccRCC. LRP1 expression was significantly correlated with expressions of PBRM1, SETD2, and KDM5C. Positive correlations between LRP1 and pro-angiogenic factors ERAP1, SCG2, STAB1, and RUNX1 were observed. LRP1 expression was positively correlated with PD-L2 level. Negative correlations between LRP1 and anti-angiogenic factors EMCN and IL18 were observed. LRP1 expression was not associated with microvessel density (MVD) yet was negatively correlated with tumor-infiltrating lymphocytes (TIL). CONCLUSION LRP1 is associated with worsened prognosis in ccRCC and is related to cancer immune modulation. LRP1-targeted therapy can be of therapeutic potential.
Collapse
|
26
|
Ruan H, Chai Z, Shen Q, Chen X, Su B, Xie C, Zhan C, Yao S, Wang H, Zhang M, Ying M, Lu W. A novel peptide ligand RAP12 of LRP1 for glioma targeted drug delivery. J Control Release 2018; 279:306-315. [PMID: 29679668 DOI: 10.1016/j.jconrel.2018.04.035] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 03/22/2018] [Accepted: 04/17/2018] [Indexed: 12/18/2022]
Abstract
The receptor associated protein (RAP) is a 39 kDa chaperone protein, binding tightly to low-density lipoprotein receptor-related protein-1 (LRP1) that is overexpressed in glioma, tumor neovasculature, vasculogenic mimicry (VM), the blood-brain barrier (BBB) and the blood-brain tumor barrier (BBTB). Herein, we miniaturized the RAP protein into a short peptide RAP12 (EAKIEKHNHYQK) aiding by computer-aided peptide design technique. RAP12 contained the essential lysines at the positions 253 and 256. The binding affinity of RAP12 to LRP1 was theoretically and experimentally evaluated. In cellular level, RAP12 could effectively internalize into U87, HUVEC and bEnd.3 cells. When modified on the surface of PEG-PLA micelles (RAP12-PEG-PLA), RAP12 could effectively facilitate the penetration of micelles through the BBB/BBTB in vitro/vivo. Paclitaxel-loaded RAP12-PEG-PLA could remarkably inhibit the growth of glioma cells and the formation of tumor neovasculature and VM, significantly prolong the median survival time of nude mice bearing intracranial glioma in comparison to model mice treated with plain micelles or Taxol. These results suggested that the RAP12 held the potential for multifunctional glioma-targeted drug delivery.
Collapse
Affiliation(s)
- Huitong Ruan
- Key Laboratory of Smart Drug Delivery of the Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Zhilan Chai
- Key Laboratory of Smart Drug Delivery of the Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Qing Shen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xishan Chen
- Key Laboratory of Smart Drug Delivery of the Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Bingxia Su
- Key Laboratory of Smart Drug Delivery of the Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Cao Xie
- Key Laboratory of Smart Drug Delivery of the Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Changyou Zhan
- Key Laboratory of Smart Drug Delivery of the Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Shengyu Yao
- Key Laboratory of Smart Drug Delivery of the Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Huan Wang
- Key Laboratory of Smart Drug Delivery of the Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Mingfei Zhang
- Key Laboratory of Smart Drug Delivery of the Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Man Ying
- Key Laboratory of Smart Drug Delivery of the Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Weiyue Lu
- Key Laboratory of Smart Drug Delivery of the Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; Minhang Hospital, Fudan University, Shanghai 201199, China; State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; The Institutes of Integrative Medicine of Fudan University, Shanghai 200040, China.
| |
Collapse
|
27
|
Bahreyni A, Khazaei M, Rajabian M, Ryzhikov M, Avan A, Hassanian SM. Therapeutic potency of pharmacological adenosine receptor agonist/antagonist in angiogenesis, current status and perspectives. ACTA ACUST UNITED AC 2017; 70:191-196. [PMID: 29057476 DOI: 10.1111/jphp.12844] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/30/2017] [Indexed: 01/19/2023]
Abstract
OBJECTIVES Adenosine concentration significantly increases in tumour microenvironment contributing to tumorigenic processes including cell proliferation, survival, invasion and of special interest in this review angiogenesis. KEY FINDINGS This review summarizes the role of pharmacological adenosine receptor agonist and antagonist in regulating angiogenesis for a better understanding and hence a better management of angiogenesis-associated disorders. SUMMARY Depending upon the pharmacological characteristics of adenosine receptor subtypes, adenosine elicits anti- or pro-angiogenic responses in stimulated cells. Inhibition of the stimulatory effect of adenosine signalling on angiogenesis using specific pharmacological adenosine receptor agonist, and antagonist is a potentially novel strategy to suppress angiogenesis in tumours.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Faculty of Medicine, Department of Clinical Biochemistry and Immunogenetic Research Center, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran
| | - Majid Khazaei
- Faculty of Medicine, Department of Medical Physiology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Rajabian
- Department of Biochemistry, Payam-e-Noor University, Mashhad, Iran
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, Washington University, School of Medicine, Saint Louis, MO, USA
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed M Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Faculty of Medicine, Department of Medical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran.,Microanatomy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
28
|
Storck SE, Pietrzik CU. Endothelial LRP1 - A Potential Target for the Treatment of Alzheimer's Disease : Theme: Drug Discovery, Development and Delivery in Alzheimer's Disease Guest Editor: Davide Brambilla. Pharm Res 2017; 34:2637-2651. [PMID: 28948494 DOI: 10.1007/s11095-017-2267-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/15/2017] [Indexed: 12/19/2022]
Abstract
The accumulation of the neurotoxin beta-amyloid (Aβ) is a major hallmark in Alzheimer's disease (AD). Aβ homeostasis in the brain is governed by its production and various clearance mechanisms. Both pathways are influenced by the ubiquitously expressed low-density lipoprotein receptor-related protein 1 (LRP1). In cerebral blood vessels, LRP1 is an important mediator for the rapid removal of Aβ from brain via transport across the blood-brain barrier (BBB). Here, we summarize recent findings on LRP1 function and discuss the targeting of LRP1 as a modulator for AD pathology and drug delivery into the brain.
Collapse
Affiliation(s)
- Steffen E Storck
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Duesbergweg 6, 55099, Mainz, Germany
| | - Claus U Pietrzik
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Duesbergweg 6, 55099, Mainz, Germany.
| |
Collapse
|