1
|
Zhu Y, Zhao L, Yan W, Ma H, Zhao W, Qu J, Zheng W, Zhang C, Du H, Yu M, Wan N, Ye H, Xie Y, Ke B, Xu Q, Sun H, Sun Y, Ouyang Z. Celastrol directly targets LRP1 to inhibit fibroblast-macrophage crosstalk and ameliorates psoriasis progression. Acta Pharm Sin B 2025; 15:876-891. [PMID: 40177548 PMCID: PMC11959968 DOI: 10.1016/j.apsb.2024.12.041] [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: 07/29/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
Psoriasis is an incurable chronic inflammatory disease that requires new interventions. Here, we found that fibroblasts exacerbate psoriasis progression by promoting macrophage recruitment via CCL2 secretion by single-cell multi-omics analysis. The natural small molecule celastrol was screened to interfere with the secretion of CCL2 by fibroblasts and improve the psoriasis-like symptoms in both murine and cynomolgus monkey models. Mechanistically, celastrol directly bound to the low-density lipoprotein receptor-related protein 1 (LRP1) β-chain and abolished its binding to the transcription factor c-Jun in the nucleus, which in turn inhibited CCL2 production by skin fibroblasts, blocked fibroblast-macrophage crosstalk, and ameliorated psoriasis progression. Notably, fibroblast-specific LRP1 knockout mice exhibited a significant reduction in psoriasis like inflammation. Taken together, from clinical samples and combined with various mouse models, we revealed the pathogenesis of psoriasis from the perspective of fibroblast-macrophage crosstalk, and provided a foundation for LRP1 as a novel potential target for psoriasis treatment.
Collapse
Affiliation(s)
- Yuyu Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Lixin Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Wei Yan
- Department of Dermatology and Venereology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongyue Ma
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wanjun Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Wei Zheng
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Chenyang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Haojie Du
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Meng Yu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ning Wan
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Ye
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yicheng Xie
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Bowen Ke
- Department of Anesthesiology, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Haiyan Sun
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Zijun Ouyang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China
| |
Collapse
|
2
|
Akula S, Alvarado-Vazquez A, Haide Mendez Enriquez E, Bal G, Franke K, Wernersson S, Hallgren J, Pejler G, Babina M, Hellman L. Characterization of Freshly Isolated Human Peripheral Blood B Cells, Monocytes, CD4+ and CD8+ T Cells, and Skin Mast Cells by Quantitative Transcriptomics. Int J Mol Sci 2024; 25:13050. [PMID: 39684762 DOI: 10.3390/ijms252313050] [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: 11/12/2024] [Revised: 11/25/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Quantitative transcriptomics offers a new way to obtain a detailed picture of freshly isolated cells. By direct isolation, the cells are unaffected by in vitro culture, and the isolation at cold temperatures maintains the cells relatively unaltered in phenotype by avoiding activation through receptor cross-linking or plastic adherence. Simultaneous analysis of several cell types provides the opportunity to obtain detailed pictures of transcriptomic differences between them. Here, we present such an analysis focusing on four human blood cell populations and compare those to isolated human skin mast cells. Pure CD19+ peripheral blood B cells, CD14+ monocytes, and CD4+ and CD8+ T cells were obtained by fluorescence-activated cell sorting, and KIT+ human connective tissue mast cells (MCs) were purified by MACS sorting from healthy skin. Detailed information concerning expression levels of the different granule proteases, protease inhibitors, Fc receptors, other receptors, transcription factors, cell signaling components, cytoskeletal proteins, and many other protein families relevant to the functions of these cells were obtained and comprehensively discussed. The MC granule proteases were found exclusively in the MC samples, and the T-cell granzymes in the T cells, of which several were present in both CD4+ and CD8+ T cells. High levels of CD4 were also observed in MCs and monocytes. We found a large variation between the different cell populations in the expression of Fc receptors, as well as for lipid mediators, proteoglycan synthesis enzymes, cytokines, cytokine receptors, and transcription factors. This detailed quantitative comparative analysis of more than 780 proteins of importance for the function of these populations can now serve as a good reference material for research into how these entities shape the role of these cells in immunity and tissue homeostasis.
Collapse
Affiliation(s)
- Srinivas Akula
- Department of Cell and Molecular Biology, Uppsala University, The Biomedical Center, Box 596, SE-751 24 Uppsala, Sweden
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Box 7023, SE-75007 Uppsala, Sweden
| | - Abigail Alvarado-Vazquez
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Erika Haide Mendez Enriquez
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Gürkan Bal
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Kristin Franke
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Sara Wernersson
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Box 7023, SE-75007 Uppsala, Sweden
| | - Jenny Hallgren
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Magda Babina
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Lars Hellman
- Department of Cell and Molecular Biology, Uppsala University, The Biomedical Center, Box 596, SE-751 24 Uppsala, Sweden
| |
Collapse
|
3
|
Dutka M, Zimmer K, Ćwiertnia M, Ilczak T, Bobiński R. The role of PCSK9 in heart failure and other cardiovascular diseases-mechanisms of action beyond its effect on LDL cholesterol. Heart Fail Rev 2024; 29:917-937. [PMID: 38886277 PMCID: PMC11306431 DOI: 10.1007/s10741-024-10409-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
Proprotein convertase subtilisin/kexin type-9 (PCSK9) is a protein that regulates low-density lipoprotein (LDL) cholesterol metabolism by binding to the hepatic LDL receptor (LDLR), ultimately leading to its lysosomal degradation and an increase in LDL cholesterol (LDLc) levels. Treatment strategies have been developed based on blocking PCSK9 with specific antibodies (alirocumab, evolocumab) and on blocking its production with small regulatory RNA (siRNA) (inclisiran). Clinical trials evaluating these drugs have confirmed their high efficacy in reducing serum LDLc levels and improving the prognosis in patients with atherosclerotic cardiovascular diseases. Most studies have focused on the action of PCSK9 on LDLRs and the subsequent increase in LDLc concentrations. Increasing evidence suggests that the adverse cardiovascular effects of PCSK9, particularly its atherosclerotic effects on the vascular wall, may also result from mechanisms independent of its effects on lipid metabolism. PCSK9 induces the expression of pro-inflammatory cytokines contributing to inflammation within the vascular wall and promotes apoptosis, pyroptosis, and ferroptosis of cardiomyocytes and is thus involved in the development and progression of heart failure. The elimination of PCSK9 may, therefore, not only be a treatment for hypercholesterolaemia but also for atherosclerosis and other cardiovascular diseases. The mechanisms of action of PCSK9 in the cardiovascular system are not yet fully understood. This article reviews the current understanding of the mechanisms of PCSK9 action in the cardiovascular system and its contribution to cardiovascular diseases. Knowledge of these mechanisms may contribute to the wider use of PCSK9 inhibitors in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland.
| | - Karolina Zimmer
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Michał Ćwiertnia
- Department of Emergency Medicine, Faculty of Health Sciences, University of Bielsko-Biala, 43-309, Bielsko-Biała, Poland
| | - Tomasz Ilczak
- Department of Emergency Medicine, Faculty of Health Sciences, University of Bielsko-Biala, 43-309, Bielsko-Biała, Poland
| | - Rafał Bobiński
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| |
Collapse
|
4
|
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
|
5
|
Azargoonjahromi A. The duality of amyloid-β: its role in normal and Alzheimer's disease states. Mol Brain 2024; 17:44. [PMID: 39020435 PMCID: PMC11256416 DOI: 10.1186/s13041-024-01118-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024] Open
Abstract
Alzheimer's disease (AD) is a degenerative neurological condition that gradually impairs cognitive abilities, disrupts memory retention, and impedes daily functioning by impacting the cells of the brain. A key characteristic of AD is the accumulation of amyloid-beta (Aβ) plaques, which play pivotal roles in disease progression. These plaques initiate a cascade of events including neuroinflammation, synaptic dysfunction, tau pathology, oxidative stress, impaired protein clearance, mitochondrial dysfunction, and disrupted calcium homeostasis. Aβ accumulation is also closely associated with other hallmark features of AD, underscoring its significance. Aβ is generated through cleavage of the amyloid precursor protein (APP) and plays a dual role depending on its processing pathway. The non-amyloidogenic pathway reduces Aβ production and has neuroprotective and anti-inflammatory effects, whereas the amyloidogenic pathway leads to the production of Aβ peptides, including Aβ40 and Aβ42, which contribute to neurodegeneration and toxic effects in AD. Understanding the multifaceted role of Aβ, particularly in AD, is crucial for developing effective therapeutic strategies that target Aβ metabolism, aggregation, and clearance with the aim of mitigating the detrimental consequences of the disease. This review aims to explore the mechanisms and functions of Aβ under normal and abnormal conditions, particularly in AD, by examining both its beneficial and detrimental effects.
Collapse
|
6
|
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
|
7
|
Boregowda SV, Haga CL, Supper VM, Booker CN, Phinney DG. Novel role for alpha-2-macroglobulin (A2M) as a disease modifying protein in senile osteoporosis. Front Cell Dev Biol 2023; 11:1294438. [PMID: 37965574 PMCID: PMC10642388 DOI: 10.3389/fcell.2023.1294438] [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: 09/14/2023] [Accepted: 10/17/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction: In the rapidly aging U.S. population, age-induced bone loss (senile osteoporosis) represents a major public health concern that is associated with a significant increased risk for low trauma fragility fractures, which are debilitating to patients, cause significant morbidity and mortality, and are costly to treat and manage. While various treatments exist to slow bone loss in osteoporosis patients, these suffer from poor tolerability and label restrictions that limit their overall effectiveness. Over the past decade, skeletal stem/progenitor cells (SSPCs), which are the main precursor of osteoblasts and adipocytes in adult bone marrow (BM), have emerged as important players in osteoporosis. Methods: Age-induced skeletal pathology was quantified in elderly (24-month-old) vs. mature (3-month-old) mice by micro-CT and changes in SSPC abundance in the BM of these mice was quantified by fluorescence-activated cell sorting (FACS). SSPCs from elderly vs. mature mice were also analyzed by RNA-Seq to identify differentially expressed genes (DEGs), and gain and loss-of-function studies were performed in human BM-derived mesenchymal stromal cells (BM-MSCs) to assess A2M function. Results: Elderly mice were shown to exhibit significant age-induced skeletal pathology, which correlated with a significant increase in SSPC abundance in BM. RNA-seq analysis identified alpha-2-macroglobulin (A2M), a pan-protease inhibitor that also binds inflammatory cytokines, as one of the most downregulated transcripts in SSPCs isolated from the BM of elderly vs. mature mice, and silencing of A2M expression in human BM-MSCs induced their proliferation and skewed their lineage bifurcation toward adipogenesis at the expense of osteogenesis thereby recapitulating critical aspects of age-induced stem cell dysfunction. Conclusion: These findings identify A2M as a novel disease modifying protein in osteoporosis, downregulation of which in bone marrow promotes SSPC dysfunction and imbalances in skeletal homeostasis.
Collapse
Affiliation(s)
| | | | | | | | - Donald G. Phinney
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, United States
| |
Collapse
|
8
|
Babenko VA, Fedulova KS, Silachev DN, Rahimi-Moghaddam P, Kalyuzhnaya YN, Demyanenko SV, Plotnikov EY. The Role of Matrix Metalloproteinases in Hemorrhagic Transformation in the Treatment of Stroke with Tissue Plasminogen Activator. J Pers Med 2023; 13:1175. [PMID: 37511788 PMCID: PMC10381732 DOI: 10.3390/jpm13071175] [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: 06/17/2023] [Revised: 07/15/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Ischemic stroke is a leading cause of disability and mortality worldwide. The only approved treatment for ischemic stroke is thrombolytic therapy with tissue plasminogen activator (tPA), though this approach often leads to a severe complication: hemorrhagic transformation (HT). The pathophysiology of HT in response to tPA is complex and not fully understood. However, numerous scientific findings suggest that the enzymatic activity and expression of matrix metalloproteinases (MMPs) in brain tissue play a crucial role. In this review article, we summarize the current knowledge of the functioning of various MMPs at different stages of ischemic stroke development and their association with HT. We also discuss the mechanisms that underlie the effect of tPA on MMPs as the main cause of the adverse effects of thrombolytic therapy. Finally, we describe recent research that aimed to develop new strategies to modulate MMP activity to improve the efficacy of thrombolytic therapy. The ultimate goal is to provide more targeted and personalized treatment options for patients with ischemic stroke to minimize complications and improve clinical outcomes.
Collapse
Affiliation(s)
- Valentina A Babenko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Ksenia S Fedulova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Denis N Silachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Parvaneh Rahimi-Moghaddam
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Yulia N Kalyuzhnaya
- Academy of Biology and Biotechnology, Southern Federal University, 344090 Rostov-on-Don, Russia
| | - Svetlana V Demyanenko
- Academy of Biology and Biotechnology, Southern Federal University, 344090 Rostov-on-Don, Russia
| | - Egor Y Plotnikov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
9
|
Zhang T, Wang X, Li X, Li YN, Li Y, Wu S, Xu L, Zhou R, Yang J, Li G, Liu X, Zheng X, Zhang Z, Zhang H. MoLrp1-mediated signaling induces nuclear accumulation of MoMsn2 to facilitate fatty acid oxidation for infectious growth of the rice blast fungus. PLANT COMMUNICATIONS 2023:100561. [PMID: 36774535 PMCID: PMC10363509 DOI: 10.1016/j.xplc.2023.100561] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 01/05/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Fatty acid β-oxidation is critical for fatty acid degradation and cellular development. In the rice blast fungus Magnaporthe oryzae, fatty acid β-oxidation is reported to be important mainly for turgor generation in the appressorium. However, the role of fatty acid β-oxidation during invasive hyphal growth is rarely documented. We demonstrated that blocking peroxisomal fatty acid β-oxidation impaired lipid droplet (LD) degradation and infectious growth of M. oryzae. We found that the key regulator of pathogenesis, MoMsn2, which we identified previously, is involved in fatty acid β-oxidation by targeting MoDCI1 (encoding dienoyl-coenzyme A [CoA] isomerase), which is also important for LD degradation and infectious growth. Cytological observations revealed that MoMsn2 accumulated from the cytosol to the nucleus during early infection or upon treatment with oleate. We determined that the low-density lipoprotein receptor-related protein MoLrp1, which is also involved in fatty acid β-oxidation and infectious growth, plays a critical role in the accumulation of MoMsn2 from the cytosol to the nucleus by activating the cyclic AMP signaling pathway. Our results provide new insights into the importance of fatty acid oxidation during invasive hyphal growth, which is modulated by MoMsn2 and its related signaling pathways in M. oryzae.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Xingyu Wang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Xue Li
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Ya-Nan Li
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Yuhe Li
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Shuang Wu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Lele Xu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Ruiwen Zhou
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Jing Yang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Guotian Li
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, the Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science & Technology, Huazhong Agricultural University, Wuhan, China
| | - Xinyu Liu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Xiaobo Zheng
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Zhengguang Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Haifeng Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
10
|
Zheng Y, Meng L, Qu L, Zhao C, Wang L, Liu C, Shou C. Anti-PAI-1 Monoclonal Antibody Inhibits the Metastasis and Growth of Esophageal Squamous Cell Carcinoma. J Cancer 2023; 14:114-128. [PMID: 36605486 PMCID: PMC9809335 DOI: 10.7150/jca.77888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/20/2022] [Indexed: 01/04/2023] Open
Abstract
Plasminogen activator inhibitor (PAI-1) is highly expressed in esophageal squamous cell carcinoma (ESCC) and strongly contributes to metastasis, making it a potential target for ESCC therapy. However, the antibodies and inhibitors targeting PAI-1 have not shown good therapeutic effect in the in vivo experiments yet. Here, we generated a panel of novel monoclonal antibodies (mAbs) against PAI-1. Analysis of PAI-1 expression in 90 tissue specimens and 128 serum specimens from ESCC patients with these mAbs confirmed that PAI-1 levels was significantly correlated with metastasis and poor survival. In addition, we found that high PAI-1 expression contributed to the enhanced motility and invasiveness of two ESCC cell lines. Next, mAb-1E2 and mAb-2E3, which have highest affinity with PAI-1, were shown to possess strong inhibitory effects on ESCC migration and invasion. Anti-tumor and anti-metastatic effects of mAb-2E3 were further demonstrated in the experimental animal models. Finally, LRP1 was identified as key factor mediating the pro-invasive function of PAI-1 and the anti-invasive capacity of mAb-2E3 in ESCC cells. The mAb-2E3 markedly decreased STAT1 phosphorylation levels and blocked the binding between PAI-1 and LRP1-ClusterII domain. Collectively, mAb-2E3 developed by our lab may be an effective antibody drug which can be used for anti-metastatic therapy in ESCC.
Collapse
Affiliation(s)
| | | | | | | | | | - Caiyun Liu
- ✉ Corresponding authors: Chengchao Shou (E-mail: ), Caiyun Liu (E-mail: )
| | - Chengchao Shou
- ✉ Corresponding authors: Chengchao Shou (E-mail: ), Caiyun Liu (E-mail: )
| |
Collapse
|
11
|
Liu HJ, Xu P. Strategies to overcome/penetrate the BBB for systemic nanoparticle delivery to the brain/brain tumor. Adv Drug Deliv Rev 2022; 191:114619. [PMID: 36372301 PMCID: PMC9724744 DOI: 10.1016/j.addr.2022.114619] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/23/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
Despite its prevalence in the management of peripheral tumors, compared to surgery and radiation therapy, chemotherapy is still a suboptimal intervention in fighting against brain cancer and cancer brain metastases. This discrepancy is mainly derived from the complicatedly physiological characteristic of intracranial tumors, including the presence of blood-brain barrier (BBB) and limited enhanced permeability and retention (EPR) effect attributed to blood-brain tumor barrier (BBTB), which largely lead to insufficient therapeutics penetrating to tumor lesions to produce pharmacological effects. Therefore, dependable methodologies that can boost the efficacy of chemotherapy for brain tumors are urgently needed. Recently, nanomedicines have shown great therapeutic potential in brain tumors by employing various transcellular strategies, paracellular strategies, and their hybrids, such as adsorptive-mediated transcytosis, receptor-mediated transcytosis, BBB disruption technology, and so on. It is compulsory to comprehensively summarize these practices to shed light on future directions in developing therapeutic regimens for brain tumors. In this review, the biological and pathological characteristics of brain tumors, including BBB and BBTB, are illustrated. After that, the emerging delivery strategies for brain tumor management are summarized into different classifications and supported with detailed examples. Finally, the potential challenges and prospects for developing and clinical application of brain tumor-oriented nanomedicine are discussed.
Collapse
Affiliation(s)
- Hai-Jun Liu
- Department of Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter, Columbia, SC 29208, USA
| | - Peisheng Xu
- Department of Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter, Columbia, SC 29208, USA.
| |
Collapse
|
12
|
N. R. S, Behera MM, Naik SK, Das SK, Gopan S, Ghosh A, Sahu RN, Patra S, Purkait S. Elevated expression of cholesterol transporter LRP-1 is crucially implicated in the pathobiology of glioblastoma. Front Neurol 2022; 13:1003730. [PMID: 36267880 PMCID: PMC9576951 DOI: 10.3389/fneur.2022.1003730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor with a grave prognosis. The present study evaluated the expression of Cholesterol transporter [importer -Lipoprotein Receptor-related Protein-1 (LRP-1) and exporter -ATP-binding cassette transporters-1 (ABCA-1)] in GBM and their implications in tumor-biology, clinical outcome and therapeutic potentials. The mRNA and protein expression was assessed by qRT-PCR and immunohistochemistry, respectively, in 85 GBMs. For comparison, 25 lower-grade astrocytomas (IDH-mutant, grade-2/3) [LGA] 16 cases of high-grade astrocytomas (IDH-mutant, grade-4) [HGA] were also evaluated. In-vitro analysis was performed on U87MG and LN229 glioma cell line. The expression of LRP-1 (mRNA and protein) was significantly higher in GBM than LGA, HGA and normal brain (NB) [p-values 0.007, 0.003 and <0.001 for mRNA; 0.024, <0.001 and <0.001 for immunohistochemistry]. Majority of the GBMs (82.4%) showed strong immunoreactivity for LRP-1, and all tumor cases were positive while the normal brain was negative. LRP-1 immunoreactivity positively correlated with the MIB-1 labeling index (p-value-0.013). LRP-1 knockdown in-vitro was associated with decreased cell survival, proliferation, migration, invasion, and increased apoptosis. Similar effect was also demonstrated by Receptor Associated Protein (RAP), a LRP-1 inhibitory drug. The silencing of LRP-1 was also associated with decreased cholesterol level. The ABCA-1 expression was higher in GBM than LGA and NB (p-value 0.011 and <0.001), however there was no significant association with other parameters. LRP-1 showed a positive correlation with ABCA-1 and associated with decreased expression with LRP-1 knock-down in-vitro. The expression of LRP-1 and ABCA-1 didn't correlate with overall survival in GBMs. Hence, LRP-1 is crucial for the tumor cells' survival and aggressive biological behavior which is maintain through the regulation of high intracellular cholesterol import. Its expression is significantly higher in GBMs and also implicated in the regulation of ABCA-1 expression. Considering its immune-positivity only in the neoplastic cell and strong positivity in GBM it may be a useful adjunct to the diagnosis. For the first time, the present study emphasized its role as a potential therapeutic target in the form of RAP which is presently being used in other neurological diseases under clinical trials.
Collapse
Affiliation(s)
- Shruthi N. R.
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Minakshi M. Behera
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Sanoj Kumar Naik
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Sunil Kumar Das
- Department of Neurosurgery, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Sooraj Gopan
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Amit Ghosh
- Department of Physiology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Rabi Narayan Sahu
- Department of Neurosurgery, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Susama Patra
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Suvendu Purkait
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
- *Correspondence: Suvendu Purkait
| |
Collapse
|
13
|
Identification of a De Novo LRP1 mutation in a Saudi Family with Tetralogy of Fallot. Gene X 2022; 851:146909. [PMID: 36162527 DOI: 10.1016/j.gene.2022.146909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/19/2022] [Accepted: 09/19/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Tetralogy of Fallot (TOF) is a rare, complex congenital heart defect caused by genetic and environmental interactions that results in abnormal heart development during the early stages of pregnancy. Genetic basis of TOF in Saudi populations is not yet studied. Therefore, the objective of this study is to screen for the molecular defects causing TOF in Saudi patients. METHODS A family with non-syndromic TOF was recruited from the Western region of Saudi Arabia. Whole exome sequencing (WES) was performed on the proband and her parents. The identified candidate variant was verified by sanger sequencing. Also, different computational biology tools were used to figure out how candidate variants affect the structure and function of candidate protein involved in TOF. RESULTS A novel heterozygous de novo mutation in LRP1 (p. G3311D) gene was identified in the index case. Also, this variant was absent in the in-house exome sequencing data of 80 healthy Saudi individuals. This variant was predicted to be likely pathogenic, as it negatively affects the biophysical chemical properties and stability of the protein. Furthermore, functional biology data from knock out mouse models confirms that molecular defects in LRP1 gene leads to cardiac defects and lethality. This variant was not previously reported in both Arab and global population genetic databases. CONCLUSION The findings in this study postulate that the LRP1 variant has a role in TOF pathogenesis and facilitate accurate diagnosis as well as the understanding of underlying molecular mechanisms and pathophysiology of the disease.
Collapse
|
14
|
Xiong H, Liu X, Xie Z, Zhu L, Lu H, Wang C, Yao J. Metabolic Symbiosis-Blocking Nano-Combination for Tumor Vascular Normalization Treatment. Adv Healthc Mater 2022; 11:e2102724. [PMID: 35708141 DOI: 10.1002/adhm.202102724] [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: 12/14/2021] [Revised: 06/02/2022] [Indexed: 01/27/2023]
Abstract
The clinical anti-vascular endothelial growth factor (anti-VEGF) drugs and metronomic chemotherapy (MET) induced tumor vascular normalization treatment (TVNT) are easily antagonized by tumor microenvironment metabolic cross-talk between tumor cells and endothelial cells (ECs). To overcome this dilemma, nanodrug with the ability of ECs targeted glycolysis inhibition and nanodrug with the ability of tumor cell glycolysis inhibition, anti-VEGF, and MET are combined to prepare Nano-combination the pathways related to angiogenesis, tumor cell proliferation, and immunosuppression and breaking the negative sugar-lipid-protein metabolism balance in tumor microenvironment. Thus, stronger and more lasting normalized tumor vascular network and remarkable antitumor efficacy are obtained after treatment, constructing a positive feedback loop between TVNT and anti-tumor therapy. Above all, this study provides a new insight for solving the bottleneck of clinical TVNT.
Collapse
Affiliation(s)
- Hui Xiong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Xiaoyan Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Zuohan Xie
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Linyuan Zhu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Haipeng Lu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Cheng Wang
- School of Pharmacy, Changzhou University, No. 21 Middle Gehu Road, Changzhou, 213164, P. R. China
| | - Jing Yao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| |
Collapse
|
15
|
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: 1] [Impact Index Per Article: 0.3] [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
|
16
|
Ameka M, Hasty AH. Paying the Iron Price: Liver Iron Homeostasis and Metabolic Disease. Compr Physiol 2022; 12:3641-3663. [PMID: 35766833 PMCID: PMC10155403 DOI: 10.1002/cphy.c210039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Iron is an essential metal element whose bioavailability is tightly regulated. Under normal conditions, systemic and cellular iron homeostases are synchronized for optimal function, based on the needs of each system. During metabolic dysfunction, this synchrony is lost, and markers of systemic iron homeostasis are no longer coupled to the iron status of key metabolic organs such as the liver and adipose tissue. The effects of dysmetabolic iron overload syndrome in the liver have been tied to hepatic insulin resistance, nonalcoholic fatty liver disease, and nonalcoholic steatohepatitis. While the existence of a relationship between iron dysregulation and metabolic dysfunction has long been acknowledged, identifying correlative relationships is complicated by the prognostic reliance on systemic measures of iron homeostasis. What is lacking and perhaps more informative is an understanding of how cellular iron homeostasis changes with metabolic dysfunction. This article explores bidirectional relationships between different proteins involved in iron homeostasis and metabolic dysfunction in the liver. © 2022 American Physiological Society. Compr Physiol 12:3641-3663, 2022.
Collapse
Affiliation(s)
- Magdalene Ameka
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA.,VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
17
|
Role of the LRP1-pPyk2-MMP9 pathway in hyperoxia-induced lung injury in neonatal rats. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:1289-1294. [PMID: 34911615 PMCID: PMC8690715 DOI: 10.7499/j.issn.1008-8830.2108125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVES To study the role of the low-density lipoprotein receptor-related protein 1 (LRP1)-proline-rich tyrosine kinase 2 phosphorylation (pPyk2)-matrix metalloproteinases 9 (MMP9) pathway in hyperoxia-induced lung injury in neonatal rats. METHODS A total of 16 neonatal rats were randomly placed in chambers containing room air (air group) or 95% medical oxygen (hyperoxia group) immediately after birth, with 8 rats in each group. All of the rats were sacrificed on day 8 of life. Hematoxylin and eosin staining was used to observe the pathological changes of lung tissue. ELISA was used to measure the levels of soluble LRP1 (sLRP1) and MMP9 in serum and bronchoalveolar lavage fluid (BALF). Western blot was used to measure the protein expression levels of LRP1, MMP9, Pyk2, and pPyk2 in lung tissue. RT-PCR was used to measure the mRNA expression levels of LRP1 and MMP9 in lung tissue. RESULTS The hyperoxia group had significantly higher levels of sLRP1 and MMP9 in serum and BALF than the air group (P<0.05). Compared with the air group, the hyperoxia group had significant increases in the protein expression levels of LRP1, MMP9, and pPyk2 in lung tissue (P<0.05). The hyperoxia group had significantly higher relative mRNA expression levels of LRP1 and MMP9 in lung tissue than the air group (P<0.05). CONCLUSIONS The activation of the LRP1-pPyk2-MMP9 pathway is enhanced in hyperoxia-induced lung injury in neonatal rats, which may be involved in the pathogenesis of bronchopulmonary dysplasia.
Collapse
|
18
|
Gindina S, Barron AO, Hu Y, Dimopoulos A, Danias J. Tissue plasminogen activator rescues steroid-induced outflow facility reduction via non-enzymatic action. Mol Vis 2021; 27:691-705. [PMID: 35002214 PMCID: PMC8684809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/05/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Tissue plasminogen activator (tPA) prevents steroid-induced reduction in aqueous humor outflow facility; however, its mechanism of action at the trabecular meshwork (TM) remains unclear. Enzymatic and non-enzymatic domains allow tPA to function as both an enzyme and a cytokine. This study sought to determine whether cytokine activity is sufficient to rescue steroid-induced outflow facility reduction. METHODS Outflow facility was measured in C57BL/6J mice following triamcinolone acetonide exposure and either transfection of the TM using adenoviral vectors, encoding for enzymatically active and inactive tPA, or administration of the respective proteins. Protein injections were also administered to tPA deficient (PlatKO) and Mmp-9 deficient (Mmp-9KO) mice to determine the potential to rescue reductions in outflow facility and determine downstream mechanisms. Gene expression of matrix metalloproteinases (Mmp-2, -9, and -13) was measured in angle ring tissues containing the TM. RESULTS Enzymatically active and inactive tPA (either produced after TM transfection or after direct administration) were equally effective in attenuating steroid-induced outflow facility reduction in C57BL/6J mice. They were also equally effective in rescuing outflow reduction in PlatKO mice and causing enhanced expression of matrix metalloproteinases. However, both enzymatically active and enzymatically inactive tPA did not improve outflow reduction in Mmp-9KO mice or increase the baseline outflow facility in naïve C57BL/6J mice. CONCLUSIONS tPA enzymatic activity is not necessary in the regulation of aqueous humor outflow. tPA can increase the expression of matrix metalloproteinases in a cytokine-mediated fashion. This cascade of events may eventually lead to extracellular matrix remodeling at the TM, which reverses outflow facility reduction caused by steroids.
Collapse
Affiliation(s)
- Sofya Gindina
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY,Department of Ophthalmology, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Arturo O. Barron
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY,Department of Ophthalmology, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Yan Hu
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY,Department of Ophthalmology, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Antonios Dimopoulos
- Department of Ophthalmology, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - John Danias
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY,Department of Ophthalmology, SUNY Downstate Health Sciences University, Brooklyn, NY
| |
Collapse
|
19
|
Role of Endocytosis Proteins in Gefitinib-Mediated EGFR Internalisation in Glioma Cells. Cells 2021; 10:cells10113258. [PMID: 34831480 PMCID: PMC8618144 DOI: 10.3390/cells10113258] [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: 10/14/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/23/2022] Open
Abstract
EGFR (epidermal growth factor receptor), a member of the ErbB tyrosine kinase receptor family, is a clinical therapeutic target in numerous solid tumours. EGFR overexpression in glioblastoma (GBM) drives cell invasion and tumour progression. However, clinical trials were disappointing, and a molecular basis to explain these poor results is still missing. EGFR endocytosis and membrane trafficking, which tightly regulate EGFR oncosignaling, are often dysregulated in glioma. In a previous work, we showed that EGFR tyrosine kinase inhibitors, such as gefitinib, lead to enhanced EGFR endocytosis into fused early endosomes. Here, using pharmacological inhibitors, siRNA-mediated silencing, or expression of mutant proteins, we showed that dynamin 2 (DNM2), the small GTPase Rab5 and the endocytosis receptor LDL receptor-related protein 1 (LRP-1), contribute significantly to gefitinib-mediated EGFR endocytosis in glioma cells. Importantly, we showed that inhibition of DNM2 or LRP-1 also decreased glioma cell responsiveness to gefitinib during cell evasion from tumour spheroids. By highlighting the contribution of endocytosis proteins in the activity of gefitinib on glioma cells, this study suggests that endocytosis and membrane trafficking might be an attractive therapeutic target to improve GBM treatment.
Collapse
|
20
|
A Possible Role for PAI-1 Blockade in Melanoma Immunotherapy. J Invest Dermatol 2021; 141:2566-2568. [PMID: 34688409 DOI: 10.1016/j.jid.2021.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/04/2021] [Accepted: 05/04/2021] [Indexed: 11/23/2022]
Abstract
In their new article in the Journal of Investigative Dermatology, Tseng et al. (2021) confirm that the sensitivity of melanoma cells to anti‒PD-L1 checkpoint inhibitor therapy is correlated with high PD-L1 surface expression. By blocking PD-L1 membrane clearing, controlled by LRP1 and PAI-1, the expression of high-cell-surface levels of PD-L1 was maintained.
Collapse
|
21
|
Yamamoto K, Wilkinson D, Bou-Gharios G. Targeting Dysregulation of Metalloproteinase Activity in Osteoarthritis. Calcif Tissue Int 2021; 109:277-290. [PMID: 32772139 PMCID: PMC8403128 DOI: 10.1007/s00223-020-00739-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 07/24/2020] [Indexed: 02/06/2023]
Abstract
Metalloproteinases were first identified as collagen cleaving enzymes and are now appreciated to play important roles in a wide variety of biological processes. The aberrant activity and dysregulation of the metalloproteinase family are linked to numerous diseases including cardiovascular and pulmonary diseases, chronic wounds, cancer, fibrosis and arthritis. Osteoarthritis (OA) is the most prevalent age-related joint disorder that causes pain and disability, but there are no disease-modifying drugs available. The hallmark of OA is loss of articular cartilage and elevated activities of matrix-degrading metalloproteinases are responsible. These enzymes do not exist in isolation and their activity is tightly regulated by a number of processes, such as transcription, proteolytic activation, interaction with their inhibitors, cell surface and extracellular matrix molecules, and endocytic clearance from the extracellular milieu. Here, we describe the functions and roles of metalloproteinase family in OA pathogenesis. We highlight recent studies that have illustrated novel mechanisms regulating their extracellular activity and impairment of such regulations that lead to the development of OA. We also discuss how to stop or slow down the degenerative processes by targeting aberrant metalloproteinase activity, which may in future become therapeutic interventions for the disease.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| | - David Wilkinson
- Institute of Life Course and Medical Sciences, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - George Bou-Gharios
- Institute of Life Course and Medical Sciences, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| |
Collapse
|
22
|
Extracellular Metalloproteinases in the Plasticity of Excitatory and Inhibitory Synapses. Cells 2021; 10:cells10082055. [PMID: 34440823 PMCID: PMC8391609 DOI: 10.3390/cells10082055] [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: 07/01/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
Long-term synaptic plasticity is shaped by the controlled reorganization of the synaptic proteome. A key component of this process is local proteolysis performed by the family of extracellular matrix metalloproteinases (MMPs). In recent years, considerable progress was achieved in identifying extracellular proteases involved in neuroplasticity phenomena and their protein substrates. Perisynaptic metalloproteinases regulate plastic changes at synapses through the processing of extracellular and membrane proteins. MMP9 was found to play a crucial role in excitatory synapses by controlling the NMDA-dependent LTP component. In addition, MMP3 regulates the L-type calcium channel-dependent form of LTP as well as the plasticity of neuronal excitability. Both MMP9 and MMP3 were implicated in memory and learning. Moreover, altered expression or mutations of different MMPs are associated with learning deficits and psychiatric disorders, including schizophrenia, addiction, or stress response. Contrary to excitatory drive, the investigation into the role of extracellular proteolysis in inhibitory synapses is only just beginning. Herein, we review the principal mechanisms of MMP involvement in the plasticity of excitatory transmission and the recently discovered role of proteolysis in inhibitory synapses. We discuss how different matrix metalloproteinases shape dynamics and turnover of synaptic adhesome and signal transduction pathways in neurons. Finally, we discuss future challenges in exploring synapse- and plasticity-specific functions of different metalloproteinases.
Collapse
|
23
|
Rong J, Tao X, Lin Y, Zheng H, Ning L, Lu HS, Daugherty A, Shi P, Mullick AE, Chen S, Zhang Z, Xu Y, Wang J. Loss of Hepatic Angiotensinogen Attenuates Sepsis-Induced Myocardial Dysfunction. Circ Res 2021; 129:547-564. [PMID: 34238019 DOI: 10.1161/circresaha.120.318075] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale: The renin-angiotensin system (RAS) is a complex regulatory network that maintains normal physiological functions. The role of the RAS in sepsis-induced myocardial dysfunction (SIMD) is poorly defined. Angiotensinogen (AGT) is the unique precursor of the RAS and gives rise to all angiotensin peptides. The effects and mechanisms of AGT in development of SIMD have not been defined. Objective: To determine a role of AGT in SIMD and investigate the underlying mechanisms. Methods and Results: Either intraperitoneal injection of lipopolysaccharide (LPS) or cecal ligation and puncture (CLP) significantly enhanced AGT abundances in liver, heart, and plasma. Deficiency of hepatocyte-derived AGT (hepAGT), rather than cardiomyocyte-derived AGT (carAGT), alleviated septic cardiac dysfunction in mice and prolonged survival time. Further investigations revealed that the effects of hepAGT on SIMD were partially associated with augmented angiotensin II (AngII) production in circulation. In addition, hepAGT was internalized by LDL receptor-related protein 1 (LRP1) in cardiac fibroblasts (CF), and subsequently activated NLRP3 inflammasome via an AngII-independent pathway, ultimately promoting SIMD by suppressing Sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a (SERCA2a) abundances in cardiomyocytes (CM). Conclusions: HepAGT promoted SIMD via both AngII-dependent and AngII-independent pathways. We identified a liver-heart axis by which AGT regulated development of SIMD. Our study may provide a potential novel therapeutic target for SIMD.
Collapse
Affiliation(s)
- Jiabing Rong
- Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, CHINA
| | - Xinran Tao
- Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine
| | - Yao Lin
- Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine
| | - Haiqiong Zheng
- Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, College of Medicine, Zhejiang university, CHINA
| | - Le Ning
- Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine
| | - Hong S Lu
- Physiology, University of Kentucky, UNITED STATES
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, UNITED STATES
| | - Peng Shi
- Institute of Translational Medicine, Zhejiang University, CHINA
| | - Adam E Mullick
- Antisense Drug Discovery, Ionis Pharmaceuticals, UNITED STATES
| | - Sicong Chen
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, CHINA
| | - Zhaocai Zhang
- Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, CHINA
| | - Yinchuan Xu
- Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang university, CHINA
| | - Jian'an Wang
- Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, CHINA
| |
Collapse
|
24
|
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: 4.3] [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
|
25
|
Jankowska U, Skupien-Rabian B, Swiderska B, Prus G, Dziedzicka-Wasylewska M, Kedracka-Krok S. Proteome Analysis of PC12 Cells Reveals Alterations in Translation Regulation and Actin Signaling Induced by Clozapine. Neurochem Res 2021; 46:2097-2111. [PMID: 34024016 PMCID: PMC8254727 DOI: 10.1007/s11064-021-03348-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/19/2021] [Accepted: 05/12/2021] [Indexed: 12/18/2022]
Abstract
Although antipsychotics are routinely used in the treatment of schizophrenia for the last decades, their precise mechanism of action is still unclear. In this study, we investigated changes in the PC12 cells’ proteome under the influence of clozapine, risperidone, and haloperidol to identify protein pathways regulated by antipsychotics. Analysis of the protein profiles in two time points: after 12 and 24 h of incubation with drugs revealed significant alterations in 510 proteins. Further canonical pathway analysis revealed an inhibition of ciliary trophic factor signaling after treatment with haloperidol and showed a decrease in acute phase response signaling in the risperidone group. Interestingly, all tested drugs have caused changes in PC12 proteome which correspond to inhibition of cytokines: tumor necrosis factor (TNF) and transforming growth factor beta 1 (TGF-β1). We also found that the 12-h incubation with clozapine caused up-regulation of protein kinase A signaling and translation machinery. After 24 h of treatment with clozapine, the inhibition of the actin cytoskeleton signaling and Rho proteins signaling was revealed. The obtained results suggest that the mammalian target of rapamycin complex 1 (mTORC1) and 2 (mTORC2) play a central role in the signal transduction of clozapine.
Collapse
Affiliation(s)
- Urszula Jankowska
- Proteomics and Mass Spectrometry Core Facility, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a str, 30-387, Krakow, Poland.
| | - Bozena Skupien-Rabian
- Proteomics and Mass Spectrometry Core Facility, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a str, 30-387, Krakow, Poland
| | - Bianka Swiderska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics Polish Academy of Sciences, Pawinskiego 5a, Warsaw, Poland
| | - Gabriela Prus
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, Poland
| | - Marta Dziedzicka-Wasylewska
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, Poland
| | - Sylwia Kedracka-Krok
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, Poland
| |
Collapse
|
26
|
Yap ZJ, Sharif M, Bashir M. Is there an immunogenomic difference between thoracic and abdominal aortic aneurysms? J Card Surg 2021; 36:1520-1530. [PMID: 33604952 DOI: 10.1111/jocs.15440] [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: 09/30/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND AIM Aortic aneurysms most commonly occur in the infra-renal and proximal thoracic regions. While generally asymptomatic, progressive aneurysmal dilation can become rapidly lethal when dissection or ruptures occurs, highlighting the need for more robust screening. Abdominal aortic aneurysm (AAA) is more prevalent compared to thoracic aortic aneurysm (TAA). The true incidence of TAA is underreported due to the absence of population screening and the silent nature of TAA. To achieve the optimum survival rate in aortic aneurysms, knowledge of natural course, genetic association, and surgical results are needed to be applied with adequate medical treatment and careful selection of patients for operation. The purpose of this paper is to provide a comprehensive review of the literature on natural history, immunology, and genetic differences between thoracic and AAAs. METHOD The literature was collected from OVID, SCOPUS, and PubMed. RESULTS (1) AAA expands faster than TAA. AAA expands at approximately 0.3-0.45 cm annually, depending on various factors (advancing age, diameter of aorta, smoking etc.). TAA expands up to 0.3 cm annually in a non-bicuspid aortic valve patient. (2) An increase in Matrix metallopeptidase 1, 2, 9, 12, 14 led to degrading extracellular matrix of the aortic vessel wall. This significantly contributed to the pathogenesis in AAA, whereas overactive Transforming growth factor-beta played a major role in the pathogenesis of TAA. CONCLUSION In the future, genetic testing may be the gold standard for tackling the geneticheterogeneity of aneurysms, therefore, identifying at-risk individuals developing TAA andAAA earlier.
Collapse
Affiliation(s)
- Zhi Jiun Yap
- Department of Anaesthetic, Dorset County Hospital, Dorset, England
| | - Monira Sharif
- Department of Molecular & Clinical Medicine, Ninewells Hospital and Medical School, Dundee, Scotland
| | - Mohamad Bashir
- Department of Emergency Medicine and Surgery, Royal Blackburn Teaching Hospital, Blackburn, England
| |
Collapse
|
27
|
Husain MA, Laurent B, Plourde M. APOE and Alzheimer's Disease: From Lipid Transport to Physiopathology and Therapeutics. Front Neurosci 2021; 15:630502. [PMID: 33679311 PMCID: PMC7925634 DOI: 10.3389/fnins.2021.630502] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/20/2021] [Indexed: 12/23/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder characterized by extracellular amyloid β (Aβ) and intraneuronal tau protein aggregations. One risk factor for developing AD is the APOE gene coding for the apolipoprotein E protein (apoE). Humans have three versions of APOE gene: ε2, ε3, and ε4 allele. Carrying the ε4 allele is an AD risk factor while carrying the ε2 allele is protective. ApoE is a component of lipoprotein particles in the plasma at the periphery, as well as in the cerebrospinal fluid (CSF) and in the interstitial fluid (ISF) of brain parenchyma in the central nervous system (CNS). ApoE is a major lipid transporter that plays a pivotal role in the development, maintenance, and repair of the CNS, and that regulates multiple important signaling pathways. This review will focus on the critical role of apoE in AD pathogenesis and some of the currently apoE-based therapeutics developed in the treatment of AD.
Collapse
Affiliation(s)
- Mohammed Amir Husain
- Centre de Recherche Sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada.,Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Benoit Laurent
- Centre de Recherche Sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada.,Département de Biochimie et Génomique Fonctionnelle, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mélanie Plourde
- Centre de Recherche Sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada.,Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
28
|
Hsu HW, Rodriguez-Ortiz CJ, Zumkehr J, Kitazawa M. Inflammatory Cytokine IL-1β Downregulates Endothelial LRP1 via MicroRNA-mediated Gene Silencing. Neuroscience 2021; 453:69-80. [PMID: 33246059 PMCID: PMC7796931 DOI: 10.1016/j.neuroscience.2020.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/26/2022]
Abstract
Effective clearance of neurotoxic amyloid-beta (Aβ) from the brain is a critical process to prevent Alzheimer's disease (AD). One major clearance mechanism is Aβ transcytosis mediated by low-density lipoprotein receptor-related protein 1 (LRP1) in capillary endothelial cells. A marked loss of endothelial LRP1 is found in AD brains and is believed to significantly impair Aβ clearance. Recently, we demonstrated that pro-inflammatory cytokines IL-1β, IL-6 and TNF-α, significantly down-regulated LRP1 in human primary microvascular endothelial cells (MVECs). In this study, we sought to determine the underlying molecular mechanism by which IL-1β led to LRP1 loss in MVECs. Reduced LRP1 protein and transcript were detected up to 24 h post-exposure and returned to the baseline levels after 48 h post-exposure with 1 ng/ml IL-1β. This reduction was in part mediated by microRNA-205-5p, -200b-3p, and -200c-3p, as these microRNAs were concomitantly upregulated in MVECs exposed to IL-1β. Synthetic microRNA-205-5p, -200b-3p, and -200c-3p mimics recapitulated LRP1 loss in MVECs without IL-1β, and their synthetic antagomirs effectively reversed IL-1β-mediated LRP1 loss. Importantly, we found that the expression of these three microRNAs was controlled by NF-κB as pharmacological NF-κB inhibitor, BMS-345541, inhibited the IL-1β-mediated upregulation of these microRNAs and rescued LRP1 expression. siRNA-mediated silencing of IκB in MVECs elevated microRNA-200b-3p and decreased LRP1 transcript, partially confirming our overall findings. In conclusion, our study provides a mechanism by which pro-inflammatory IL-1β instigates the suppression of LRP1 expression in MVECs. Our findings could implicate spatiotemporal loss of LRP1 and impairment of the LRP1-mediated clearance mechanism by endothelial cells.
Collapse
Affiliation(s)
- Heng-Wei Hsu
- Center for Occupational and Environmental Health, Department of Environmental and Occupational Health and Department of Medicine, University of California, Irvine, CA, United States
| | - Carlos J Rodriguez-Ortiz
- Center for Occupational and Environmental Health, Department of Environmental and Occupational Health and Department of Medicine, University of California, Irvine, CA, United States
| | - Joannee Zumkehr
- Center for Occupational and Environmental Health, Department of Environmental and Occupational Health and Department of Medicine, University of California, Irvine, CA, United States
| | - Masashi Kitazawa
- Center for Occupational and Environmental Health, Department of Environmental and Occupational Health and Department of Medicine, University of California, Irvine, CA, United States.
| |
Collapse
|
29
|
Li K, Liu CJ, Zhang XZ. Multifunctional peptides for tumor therapy. Adv Drug Deliv Rev 2020; 160:36-51. [PMID: 33080257 DOI: 10.1016/j.addr.2020.10.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022]
Abstract
Controlled nano-systems for drug delivery are designed to deliver therapeutical drugs to desirable sites on demand. Due to the diverse physiological functions of peptides, it is reasonable to introduce peptides into anti-tumor nano-system. The integration of peptides into nanomaterials has complementary advantages, which not only avoids the rapid degradation of peptides in vivo, but also improves the intelligence and functionality of the nano-system. We summarized the functional peptides with targeting and stimulus-responsive properties, and the present review outlined the most relevant and recent developed peptide-based multifunctional nanomaterials for tumor therapy.
Collapse
|
30
|
Red Blood Cells and Hemoglobin in Human Atherosclerosis and Related Arterial Diseases. Int J Mol Sci 2020; 21:ijms21186756. [PMID: 32942605 PMCID: PMC7554753 DOI: 10.3390/ijms21186756] [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: 06/17/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
As the main particulate component of the circulating blood, RBCs play major roles in physiological hemodynamics and impact all arterial wall pathologies. RBCs are the main determinant of blood viscosity, defining the frictional forces exerted by the blood on the arterial wall. This function is used in phylogeny and ontogeny of the cardiovascular (CV) system, allowing the acquisition of vasomotricity adapted to local metabolic demands, and systemic arterial pressure after birth. In pathology, RBCs collide with the arterial wall, inducing both local retention of their membranous lipids and local hemolysis, releasing heme-Fe++ with a high toxicity for arterial cells: endothelial and smooth muscle cells (SMCs) cardiomyocytes, neurons, etc. Specifically, overloading of cells by Fe++ promotes cell death. This local hemolysis is an event associated with early and advanced stages of human atherosclerosis. Similarly, the permanent renewal of mural RBC clotting is the major support of oxidation in abdominal aortic aneurysm. In parallel, calcifications promote intramural hemorrhages, and hemorrhages promote an osteoblastic phenotypic shift of arterial wall cells. Different plasma or tissue systems are able, at least in part, to limit this injury by acting at the different levels of this system.
Collapse
|
31
|
Guo Y, Yan B, Gui Y, Tang Z, Tai S, Zhou S, Zheng XL. Physiology and role of PCSK9 in vascular disease: Potential impact of localized PCSK9 in vascular wall. J Cell Physiol 2020; 236:2333-2351. [PMID: 32875580 DOI: 10.1002/jcp.30025] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/12/2020] [Accepted: 08/16/2020] [Indexed: 12/26/2022]
Abstract
Proprotein convertase subtilisin/kexin type-9 (PCSK9), a member of the proprotein convertase family, is an important drug target because of its crucial role in lipid metabolism. Emerging evidence suggests a direct role of localized PCSK9 in the pathogenesis of vascular diseases. With this in our consideration, we reviewed PCSK9 physiology with respect to recent development and major studies (clinical and experimental) on PCSK9 functionality in vascular disease. PCSK9 upregulates low-density lipoprotein (LDL)-cholesterol levels by binding to the LDL-receptor (LDLR) and facilitating its lysosomal degradation. PCSK9 gain-of-function mutations have been confirmed as a novel genetic mechanism for familial hypercholesterolemia. Elevated serum PCSK9 levels in patients with vascular diseases may contribute to coronary artery disease, atherosclerosis, cerebrovascular diseases, vasculitis, aortic diseases, and arterial aging pathogenesis. Experimental models of atherosclerosis, arterial aneurysm, and coronary or carotid artery ligation also support PCSK9 contribution to inflammatory response and disease progression, through LDLR-dependent or -independent mechanisms. More recently, several clinical trials have confirmed that anti-PCSK9 monoclonal antibodies can reduce systemic LDL levels, total nonfatal cardiovascular events, and all-cause mortality. Interaction of PCSK9 with other receptor proteins (LDLR-related proteins, cluster of differentiation family members, epithelial Na+ channels, and sortilin) may underlie its roles in vascular disease. Improved understanding of PCSK9 roles and molecular mechanisms in various vascular diseases will facilitate advances in lipid-lowering therapy and disease prevention.
Collapse
Affiliation(s)
- Yanan Guo
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China.,Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada.,Department of Physiology & Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada
| | - Binjie Yan
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada.,Department of Physiology & Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada.,Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Yu Gui
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada.,Department of Physiology & Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada
| | - Zhihan Tang
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada.,Department of Physiology & Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada.,Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Shi Tai
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China.,Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada.,Department of Physiology & Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada
| | - Shenghua Zhou
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xi-Long Zheng
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada.,Department of Physiology & Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
32
|
Paraoan L, Sharif U, Carlsson E, Supharattanasitthi W, Mahmud NM, Kamalden TA, Hiscott P, Jackson M, Grierson I. Secretory proteostasis of the retinal pigmented epithelium: Impairment links to age-related macular degeneration. Prog Retin Eye Res 2020; 79:100859. [PMID: 32278708 DOI: 10.1016/j.preteyeres.2020.100859] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022]
Abstract
Secretory proteostasis integrates protein synthesis, processing, folding and trafficking pathways that are essential for efficient cellular secretion. For the retinal pigment epithelium (RPE), secretory proteostasis is of vital importance for the maintenance of the structural and functional integrity of apical (photoreceptors) and basal (Bruch's membrane/choroidal blood supply) sides of the environment it resides in. This integrity is achieved through functions governed by RPE secreted proteins, which include extracellular matrix modelling/remodelling, angiogenesis and immune response modulation. Impaired RPE secretory proteostasis affects not only the extracellular environment, but leads to intracellular protein aggregation and ER-stress with subsequent cell death. Ample recent evidence implicates dysregulated proteostasis as a key factor in the development of age-related macular degeneration (AMD), the leading cause of blindness in the developed world, and research aiming to characterise the roles of various proteins implicated in AMD-associated dysregulated proteostasis unveiled unexpected facets of the mechanisms involved in degenerative pathogenesis. This review analyses cellular processes unveiled by the study of the top 200 transcripts most abundantly expressed by the RPE/choroid in the light of the specialised secretory nature of the RPE. Functional roles of these proteins and the mechanisms of their impaired secretion, due to age and genetic-related causes, are analysed in relation to AMD development. Understanding the importance of RPE secretory proteostasis in relation to maintaining retinal health and how it becomes impaired in disease is of paramount importance for the development and assessment of future therapeutic advancements involving gene and cell therapies.
Collapse
Affiliation(s)
- Luminita Paraoan
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom.
| | - Umar Sharif
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Emil Carlsson
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Wasu Supharattanasitthi
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom; Department of Physiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Nur Musfirah Mahmud
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Tengku Ain Kamalden
- Eye Research Centre, Department of Ophthalmology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Paul Hiscott
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Malcolm Jackson
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Ian Grierson
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
33
|
Mohd Yunos RI, Ab Mutalib NS, Tieng FYF, Abu N, Jamal R. Actionable Potentials of Less Frequently Mutated Genes in Colorectal Cancer and Their Roles in Precision Medicine. Biomolecules 2020; 10:biom10030476. [PMID: 32245111 PMCID: PMC7175115 DOI: 10.3390/biom10030476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 02/06/2023] Open
Abstract
Global statistics have placed colorectal cancer (CRC) as the third most frequently diagnosed cancer and the fourth principal cause of cancer-related deaths worldwide. Improving survival for CRC is as important as early detection. Personalized medicine is important in maximizing an individual's treatment success and minimizing the risk of adverse reactions. Approaches in achieving personalized therapy in CRC have included analyses of specific genes with its clinical implications. Tumour genotyping via next-generation sequencing has become a standard practice to guide clinicians into predicting tumor behaviour, disease prognosis, and treatment response. Nevertheless, better prognostic markers are necessary to further stratify patients for personalized treatment plans. The discovery of new markers remains indispensable in providing the most effective chemotherapy in order to improve the outcomes of treatment and survival in CRC patients. This review aims to compile and discuss newly discovered, less frequently mutated genes in CRC. We also discuss how these mutations are being used to assist therapeutic decisions and their potential prospective clinical utilities. In addition, we will summarize the importance of profiling the large genomic rearrangements, gene amplification, and large deletions and how these alterations may assist in determining the best treatment option for CRC patients.
Collapse
Affiliation(s)
| | | | | | | | - Rahman Jamal
- Correspondence: (N.S.A.M.); (R.J.); Tel.: +60-3-91459073 (N.S.A.M.); +60-3-91459000 (R.J.)
| |
Collapse
|
34
|
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: 0.8] [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
|
35
|
Henriet P, Emonard H. Matrix metalloproteinase-2: Not (just) a "hero" of the past. Biochimie 2019; 166:223-232. [PMID: 31362036 DOI: 10.1016/j.biochi.2019.07.019] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 07/22/2019] [Indexed: 01/13/2023]
Abstract
The 72-kDa type IV collagenase or gelatinase A is the second member of the matrix metalloproteinase family, MMP-2. Since the discovery of its first two substrates within components of the extracellular matrix, denatured interstitial type I collagen and native type IV collagen, the roles and various levels of regulation of MMP-2 have been intensively studied, mainly in vitro. Its (over)expression in most if not all tumors was considered a hallmark of cancer aggressiveness and boosted investigations aiming at its inhibition. Unfortunately, the enthusiasm subsided like a soufflé after clinical trial failures, mostly because of insufficient knowledge of in vivo MMP-2 activities and detrimental side effects of broad-spectrum MMP inhibition. Nowadays, MMP-2 remains a major topic of interest in research, the second in the MMP family after MMP-9. This review presents a broad overview of the major features of this protease. This knowledge is crucial to identify diagnostic or therapeutic strategies focusing on MMP-2. In this sense, recent publications and clinical trials underline the potential value of measuring circulating or tissular MMP-2 levels as diagnostic or prognostic tools, or as a useful secondary outcome for therapies against other primary targets. Direct MMP-2 inhibition has benefited from substantial progress in the design of more specific inhibitors but their in vivo application remains challenging but certainly worth the efforts it receives.
Collapse
Affiliation(s)
- Patrick Henriet
- de Duve Institute, Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Hervé Emonard
- CNRS and Université de Reims Champagne-Ardenne, UMR 7369, 51100, Reims, France.
| |
Collapse
|
36
|
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
|
37
|
Ruiz-Gómez G, Vogel S, Möller S, Pisabarro MT, Hempel U. Glycosaminoglycans influence enzyme activity of MMP2 and MMP2/TIMP3 complex formation - Insights at cellular and molecular level. Sci Rep 2019; 9:4905. [PMID: 30894640 PMCID: PMC6426840 DOI: 10.1038/s41598-019-41355-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/08/2019] [Indexed: 01/01/2023] Open
Abstract
The extracellular matrix (ECM) is a highly dynamic network constantly remodeled by a fine-tuned protein formation and degradation balance. Matrix metalloproteinases (MMPs) constitute key orchestrators of ECM degradation. Their activity is controlled by tissue inhibitors of metalloproteinases (TIMPs) and glycosaminoglycans (GAG). Here, we investigated the molecular interplay of MMP2 with different GAG (chondroitin sulfate, hyaluronan (HA), sulfated hyaluronan (SH) and heparin (HE)) and the impact of GAG on MMP2/TIMP3 complex formation using in vitro-experiments with human bone marrow stromal cells, in silico docking and molecular dynamics simulations. SH and HE influenced MMP2 and TIMP3 protein levels and MMP2 activity. Only SH supported the alignment of both proteins in fibrillar-like structures, which, based on our molecular models, would be due to a stabilization of the interactions between MMP2-hemopexin domain and TIMP3-C-terminal tail. Dependent on the temporal sequential order in which the final ternary complex was formed, our models indicated that SH and HA can affect TIMP3-induced MMP2 inhibition through precluding or supporting their interactions, respectively. Our combined experimental and theoretical approach provides valuable new insights on how GAG interfere with MMP2 activity and MMP2/TIMP3 complex formation. The results obtained evidence GAG as promising molecules for fine-balanced intervention of ECM remodeling.
Collapse
Affiliation(s)
- Gloria Ruiz-Gómez
- Structural Bioinformatics, BIOTEC TU Dresden, Tatzberg 47-51, 01307, Dresden, Germany
| | - Sarah Vogel
- Medical Department, Institute of Physiological Chemistry, TU Dresden, Fiedlerstraße 42, 01307, Dresden, Germany
| | - Stephanie Möller
- Biomaterials Department, INNOVENT e.V., Prüssingstraße 27 B, 07745, Jena, Germany
| | - M Teresa Pisabarro
- Structural Bioinformatics, BIOTEC TU Dresden, Tatzberg 47-51, 01307, Dresden, Germany
| | - Ute Hempel
- Medical Department, Institute of Physiological Chemistry, TU Dresden, Fiedlerstraße 42, 01307, Dresden, Germany.
| |
Collapse
|
38
|
|
39
|
Grünwald B, Schoeps B, Krüger A. Recognizing the Molecular Multifunctionality and Interactome of TIMP-1. Trends Cell Biol 2019; 29:6-19. [DOI: 10.1016/j.tcb.2018.08.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/24/2018] [Accepted: 08/28/2018] [Indexed: 01/31/2023]
|
40
|
Snigireva AV, Vrublevskaya VV, Zhmurina MA, Skarga YY, Morenkov OS. The Mechanisms of Stimulation of Migration and Invasion of Tumor Cells by Extracellular Heat Shock Protein 90 (eHsp90) in vitro. Biophysics (Nagoya-shi) 2018. [DOI: 10.1134/s0006350918060258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
41
|
Barry AM, Sondermann JR, Sondermann JH, Gomez-Varela D, Schmidt M. Region-Resolved Quantitative Proteome Profiling Reveals Molecular Dynamics Associated With Chronic Pain in the PNS and Spinal Cord. Front Mol Neurosci 2018; 11:259. [PMID: 30154697 PMCID: PMC6103001 DOI: 10.3389/fnmol.2018.00259] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/10/2018] [Indexed: 12/27/2022] Open
Abstract
To obtain a thorough understanding of chronic pain, large-scale molecular mapping of the pain axis at the protein level is necessary, but has not yet been achieved. We applied quantitative proteome profiling to build a comprehensive protein compendium of three regions of the pain neuraxis in mice: the sciatic nerve (SN), the dorsal root ganglia (DRG), and the spinal cord (SC). Furthermore, extensive bioinformatics analysis enabled us to reveal unique protein subsets which are specifically enriched in the peripheral nervous system (PNS) and SC. The immense value of these datasets for the scientific community is highlighted by validation experiments, where we monitored protein network dynamics during neuropathic pain. Here, we resolved profound region-specific differences and distinct changes of PNS-enriched proteins under pathological conditions. Overall, we provide a unique and validated systems biology proteome resource (summarized in our online database painproteome.em.mpg.de), which facilitates mechanistic insights into somatosensory biology and chronic pain—a prerequisite for the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Allison M Barry
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - Julia R Sondermann
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - Jan-Hendrik Sondermann
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - David Gomez-Varela
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - Manuela Schmidt
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| |
Collapse
|
42
|
Zhao R, Ghazzawi N, Wu J, Le K, Li C, Moghadasian MH, Siow YL, Apea-Bah FB, Beta T, Yin Z, Shen GX. Germinated Brown Rice Attenuates Atherosclerosis and Vascular Inflammation in Low-Density Lipoprotein Receptor-Knockout Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:4512-4520. [PMID: 29656648 DOI: 10.1021/acs.jafc.8b00005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The present study investigates the impact of germinated brown rice (GBR) on atherosclerosis and the underlying mechanism in low-density lipoprotein receptor-knockout (LDLr-KO) mice. The intensity of atherosclerosis in aortas of LDLr-KO mice receiving diet supplemented with 60% GBR (weight/weight) was significantly less than that in mice fed with 60% white rice (WR) or control diet ( p < 0.05); all diets contained 0.06% cholesterol. WR or GBR diet did not significantly alter plasma total or LDL-cholesterol, fecal sterols, or glucose, or the activities of antioxidant enzymes, compared to the control diet. The adhesion of monocytes to aortas from LDLr-KO mice fed with WR diet was significantly more than that from mice receiving the control diet ( p < 0.01). GBR diet decreased monocyte adhesion to aortas compared to WR diet ( p < 0.01). GBR diet also reduced the levels of plasminogen activator inhibitor-1 (PAI-1), monocyte chemotactic protein-1 (MCP-1), and tumor necrosis factor-α (TNF-α) in plasma, and the abundances of MCP-1, PAI-1, TNF-α, intracellular cell adhesion molecule-1, toll-like receptor-4, PAI-1, LDLr-like protein, and urokinase plasminogen activator and its receptor in aortas or hearts from LDLr-KO mice in comparison to the WR diet ( p < 0.05, 0.01, respectively). The findings suggest that GBR administration attenuated atherosclerosis and vascular inflammation in LDLr-KO mice compared to WR. The anti-atherosclerotic effect of GBR in LDLr-KO mice at least in part results from its anti-inflammatory activity.
Collapse
Affiliation(s)
| | | | - Jiansu Wu
- Jiangsu Ruiguanlong Ecological Agricultural Science and Technological Center , Jiangsu Academy of Agriculture and Sciences , Nanjing , 211500 China
| | | | - Chunyang Li
- Institute of Farm Product Processing, Jiangsu Academy of Agriculture and Sciences , Nanjing , 210014 China
| | | | | | | | | | - Zhengfeng Yin
- Jiangsu Ruiguanlong Ecological Agricultural Science and Technological Center , Jiangsu Academy of Agriculture and Sciences , Nanjing , 211500 China
| | | |
Collapse
|
43
|
Paik S, Somvanshi RK, Kumar U. Somatostatin Maintains Permeability and Integrity of Blood-Brain Barrier in β-Amyloid Induced Toxicity. Mol Neurobiol 2018; 56:292-306. [PMID: 29700775 DOI: 10.1007/s12035-018-1045-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 03/27/2018] [Indexed: 12/11/2022]
Abstract
In Alzheimer's disease (AD), the impaired clearance of β-amyloid peptide (Aβ) due to disrupted tight junction and transporter proteins is the prominent cause of disease progression. Somatostatin (SST) blocks the aggregation of Aβ and inflammation whereas reduction of SST levels in the CSF and brain tissue is associated with impaired cognitive function and memory loss. However, the role of SST in preservation of blood-brain barrier (BBB) integrity and functionality in Aβ-induced toxicity is not known. In the present study using human CMEC/D3 cells, we demonstrate that SST prevents Aβ-induced BBB permeability by regulating LRP1 and RAGE expression and improving the disrupted tight junction proteins. Furthermore, SST abrogates Aβ-induced JNK phosphorylation and expression of MMP2. Taken together, results presented here suggest that SST might serve as a therapeutic intervention in AD via targeting multiple pathways responsible for neurotoxicity, impaired BBB function, and disease progression.
Collapse
Affiliation(s)
- Seungil Paik
- Faculty of Pharmaceutical Sciences, The University of British Columbia, V6T1Z3, Vancouver, BC, Canada
| | - Rishi K Somvanshi
- Faculty of Pharmaceutical Sciences, The University of British Columbia, V6T1Z3, Vancouver, BC, Canada
| | - Ujendra Kumar
- Faculty of Pharmaceutical Sciences, The University of British Columbia, V6T1Z3, Vancouver, BC, Canada.
| |
Collapse
|
44
|
Remacle AG, Hullugundi SK, Dolkas J, Angert M, Chernov AV, Strongin AY, Shubayev VI. Acute- and late-phase matrix metalloproteinase (MMP)-9 activity is comparable in female and male rats after peripheral nerve injury. J Neuroinflammation 2018; 15:89. [PMID: 29558999 PMCID: PMC5859418 DOI: 10.1186/s12974-018-1123-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 03/08/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND In the peripheral nerve, pro-inflammatory matrix metalloproteinase (MMP)-9 performs essential functions in the acute response to injury. Whether MMP-9 activity contributes to late-phase injury or whether MMP-9 expression or activity after nerve injury is sexually dimorphic remains unknown. METHODS Patterns of MMP-9 expression, activity and excretion were assessed in a model of painful peripheral neuropathy, sciatic nerve chronic constriction injury (CCI), in female and male rats. Real-time Taqman RT-PCR for MMP-9 and its endogenous inhibitor, tissue inhibitor of metalloproteinase-1 (TIMP-1) of nerve samples over a 2-month time course of CCI was followed by gelatin zymography of crude nerve extracts and purified MMP-9 from the extracts using gelatin Sepharose-beads. MMP excretion was determined using protease activity assay of urine in female and male rats with CCI. RESULTS The initial upsurge in nerve MMP-9 expression at day 1 post-CCI was superseded more than 100-fold at day 28 post-CCI. The high level of MMP-9 expression in late-phase nerve injury was accompanied by the reduction in TIMP-1 level. The absence of MMP-9 in the normal nerve and the presence of multiple MMP-9 species (the proenzyme, mature enzyme, homodimers, and heterodimers) was observed at day 1 and day 28 post-CCI. The MMP-9 proenzyme and mature enzyme species dominated in the early- and late-phase nerve injury, consistent with the high and low level of TIMP-1 expression, respectively. The elevated nerve MMP-9 levels corresponded to the elevated urinary MMP excretion post-CCI. All of these findings were comparable in female and male rodents. CONCLUSION The present study offers the first evidence for the excessive, uninhibited proteolytic MMP-9 activity during late-phase painful peripheral neuropathy and suggests that the pattern of MMP-9 expression, activity, and excretion after peripheral nerve injury is universal in both sexes.
Collapse
Affiliation(s)
- Albert G Remacle
- Infectious and Inflammatory Disease Center/Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, San Diego, CA, 92037, USA
| | - Swathi K Hullugundi
- Department of Anesthesiology, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA, 92093-0629, USA.,VA San Diego Healthcare System, La Jolla, San Diego, CA, 92037, USA
| | - Jennifer Dolkas
- Department of Anesthesiology, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA, 92093-0629, USA.,VA San Diego Healthcare System, La Jolla, San Diego, CA, 92037, USA
| | - Mila Angert
- Department of Anesthesiology, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA, 92093-0629, USA.,VA San Diego Healthcare System, La Jolla, San Diego, CA, 92037, USA
| | - Andrei V Chernov
- Infectious and Inflammatory Disease Center/Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, San Diego, CA, 92037, USA
| | - Alex Y Strongin
- Infectious and Inflammatory Disease Center/Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, San Diego, CA, 92037, USA.
| | - Veronica I Shubayev
- Department of Anesthesiology, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA, 92093-0629, USA. .,VA San Diego Healthcare System, La Jolla, San Diego, CA, 92037, USA.
| |
Collapse
|
45
|
Meng W, Adams MJ, Hebert HL, Deary IJ, McIntosh AM, Smith BH. A Genome-Wide Association Study Finds Genetic Associations with Broadly-Defined Headache in UK Biobank (N=223,773). EBioMedicine 2018; 28:180-186. [PMID: 29397368 PMCID: PMC5898025 DOI: 10.1016/j.ebiom.2018.01.023] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/20/2018] [Accepted: 01/20/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Headache is the most common neurological symptom and a leading cause of years lived with disability. We sought to identify the genetic variants associated with a broadly-defined headache phenotype in 223,773 subjects from the UK Biobank cohort. METHODS We defined headache based on a specific question answered by the UK Biobank participants. We performed a genome-wide association study of headache as a single entity, using 74,461 cases and 149,312 controls. RESULTS We identified 3343 SNPs which reached the genome-wide significance level of P<5×10-8. The SNPs were located in 28 loci, with the top SNP of rs11172113 in the LRP1 gene having a P value of 4.92×10-47. Of the 28 loci, 14 have previously been associated with migraine. Among 14 new loci, rs77804065 with a P value of 5.87×10-15 in the LINC02210-CRHR1 gene was the top SNP. Significant relationships between multiple brain tissues and genetic associations were identified through tissue expression analysis. We also identified significant positive genetic correlations between headache and many psychological traits. CONCLUSIONS Our results suggest that brain function is closely related to broadly-defined headache. In addition, we found that many psychological traits have genetic correlations with headache.
Collapse
Affiliation(s)
- Weihua Meng
- Division of Population Health Sciences, School of Medicine, University of Dundee, Dundee DD2 4BF, UK.
| | - Mark J Adams
- Division of Psychiatry, Edinburgh Medical School, University of Edinburgh, Edinburgh EH10 5HF, UK
| | - Harry L Hebert
- Division of Population Health Sciences, School of Medicine, University of Dundee, Dundee DD2 4BF, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Andrew M McIntosh
- Division of Psychiatry, Edinburgh Medical School, University of Edinburgh, Edinburgh EH10 5HF, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Blair H Smith
- Division of Population Health Sciences, School of Medicine, University of Dundee, Dundee DD2 4BF, UK
| |
Collapse
|
46
|
Travis T, Ghassemi P, Prindeze N, Moffatt L, Carney B, Alkhalil A, Ramella-Roman J, Shupp J. Matrix Metalloproteinases Are Differentially Regulated and Responsive to Compression Therapy in a Red Duroc Model of Hypertrophic Scar. EPLASTY 2018; 18:e1. [PMID: 29375731 PMCID: PMC5765626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
Abstract
Objective: Proteins of the matrix metalloproteinases family play a vital role in extracellular matrix maintenance and basic physiological processes in tissue homeostasis. The function and activities of matrix metalloproteinases in response to compression therapies have yet to be defined. Here, a swine model of hypertrophic scar was used to profile the transcription of all known 26 matrix metalloproteinases in scars treated with a precise compression dose. Methods: Full-thickness excisional wounds were created. Wounds underwent healing and scar formation. A subset of scars underwent 2 weeks of compression therapy. Biopsy specimens were preserved, and microarrays, reverse transcription-polymerase chain reaction, Western blotting, and immunohistochemistry were performed to characterize the transcription and expression of various matrix metalloproteinase family members. Results: Microarray results showed that 13 of the known 26 matrix metalloproteinases were differentially transcribed in wounds relative to the preinjury skin. The predominant upregulation of these matrix metalloproteinases during early wound-healing stages declined gradually in later stages of wound healing. The use of compression therapy reduced this decline in 10 of the 13 differentially regulated matrix metalloproteinases. Further investigation of MMP7 using reverse transcription-polymerase chain reaction confirmed the effect of compression on transcript levels. Assessment of MMP7 at the protein level using Western blotting and immunohistochemistry was concordant. Conclusions: In a swine model of hypertrophic scar, the application of compression to hypertrophic scar attenuated a trend of decreasing levels of matrix metalloproteinases during the process of hypertrophic wound healing, including MMP7, whose enzyme regulation was confirmed at the protein level.
Collapse
Affiliation(s)
- Taryn E. Travis
- aThe Burn Center, Department of Surgery, MedStar Washington Hospital Center, Washington, DC,bFirefighters’ Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC
| | - Pejhman Ghassemi
- cDepartment of Biomedical Engineering, Catholic University of America, Washington, DC
| | - Nicholas J. Prindeze
- bFirefighters’ Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC
| | - Lauren T. Moffatt
- bFirefighters’ Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC
| | - Bonnie C. Carney
- bFirefighters’ Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC
| | - Abdulnaser Alkhalil
- bFirefighters’ Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC
| | | | - Jeffrey W. Shupp
- aThe Burn Center, Department of Surgery, MedStar Washington Hospital Center, Washington, DC,bFirefighters’ Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC,cDepartment of Biomedical Engineering, Catholic University of America, Washington, DC,Correspondence:
| |
Collapse
|
47
|
Ce O, Rs P, Ab W, S D, Cj W, Qm M, D L. Potential Link Between Proprotein Convertase Subtilisin/Kexin Type 9 and Alzheimer's Disease. ACTA ACUST UNITED AC 2018; 1. [PMID: 32352077 DOI: 10.31531/2581-4745.1000106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Alzheimer's disease [AD] is not only the most common neurodegenerative disease but is also currently incurable. Proprotein convertase subtilisin/kexin-9 [PCSK9] is an indirect regulator of plasma low density lipoprotein [LDL] levels controlling LDL receptor expression at the plasma membrane. PCSK9 also appears to regulate the development of glucose intolerance, insulin resistance, abdominal obesity, inflammation, and hypertension, conditions that have been identified as risk factors for AD. PCSK9 levels also depend on age, sex, and ethnic background, factors associated with AD. Herein, we will review indirect evidence that suggests a link between PCSK9 levels and AD.
Collapse
Affiliation(s)
- Oldham Ce
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Powell Rs
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Williams Ab
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Dixon S
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Wooten Cj
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Melendez Qm
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Lopez D
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| |
Collapse
|
48
|
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: 28] [Impact Index Per Article: 3.5] [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
|
49
|
Abstract
PURPOSE OF REVIEW Cardiometabolic diseases increasingly afflict our aging, dysmetabolic population. Complex signals regulating low-density lipoprotein receptor-related protein (LRP) and frizzled protein family members - the plasma membrane receptors for the cadre of Wnt polypeptide morphogens - contribute to the control of cardiovascular homeostasis. RECENT FINDINGS Both canonical (β-catenin-dependent) and noncanonical (β-catenin-independent) Wnt signaling programs control vascular smooth muscle (VSM) cell phenotypic modulation in cardiometabolic disease. LRP6 limits VSM proliferation, reduces arteriosclerotic transcriptional reprogramming, and preserves insulin sensitivity while LRP5 restrains foam cell formation. Adipose, skeletal muscle, macrophages, and VSM have emerged as important sources of circulating Wnt ligands that are dynamically regulated during the prediabetes-diabetes transition with cardiometabolic consequences. Platelets release Dkk1, a LRP5/LRP6 inhibitor that induces endothelial inflammation and the prosclerotic endothelial-mesenchymal transition. By contrast, inhibitory secreted frizzled-related proteins shape the Wnt signaling milieu to limit myocardial inflammation with ischemia-reperfusion injury. VSM sclerostin, an inhibitor of canonical Wnt signaling in bone, restrains remodeling that predisposes to aneurysm formation, and is downregulated in aneurysmal vessels by epigenetic methylation. SUMMARY Components of the Wnt signaling cascade represent novel targets for pharmacological intervention in cardiometabolic disease. Conversely, strategies targeting the Wnt signaling cascade for other therapeutic purposes will have cardiovascular consequences that must be delineated to establish clinically useful pharmacokinetic-pharmacodynamic relationships.
Collapse
Affiliation(s)
- Austin Gay
- Department of Internal Medicine-Endocrine Division, UT Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
50
|
Stepanova V, Dergilev KV, Holman KR, Parfyonova YV, Tsokolaeva ZI, Teter M, Atochina-Vasserman EN, Volgina A, Zaitsev SV, Lewis SP, Zabozlaev FG, Obraztsova K, Krymskaya VP, Cines DB. Urokinase-type plasminogen activator (uPA) is critical for progression of tuberous sclerosis complex 2 (TSC2)-deficient tumors. J Biol Chem 2017; 292:20528-20543. [PMID: 28972182 DOI: 10.1074/jbc.m117.799593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/20/2017] [Indexed: 12/20/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a fatal lung disease associated with germline or somatic inactivating mutations in tuberous sclerosis complex genes (TSC1 or TSC2). LAM is characterized by neoplastic growth of smooth muscle-α-actin-positive cells that destroy lung parenchyma and by the formation of benign renal neoplasms called angiolipomas. The mammalian target of rapamycin complex 1 (mTORC1) inhibitor rapamycin slows progression of these diseases but is not curative and associated with notable toxicity at clinically effective doses, highlighting the need for better understanding LAM's molecular etiology. We report here that LAM lesions and angiomyolipomas overexpress urokinase-type plasminogen activator (uPA). Tsc1-/- and Tsc2-/- mouse embryonic fibroblasts expressed higher uPA levels than their WT counterparts, resulting from the TSC inactivation. Inhibition of uPA expression in Tsc2-null cells reduced the growth and invasiveness and increased susceptibility to apoptosis. However, rapamycin further increased uPA expression in TSC2-null tumor cells and immortalized TSC2-null angiomyolipoma cells, but not in cells with intact TSC. Induction of glucocorticoid receptor signaling or forkhead box (FOXO) 1/3 inhibition abolished the rapamycin-induced uPA expression in TSC-compromised cells. Moreover, rapamycin-enhanced migration of TSC2-null cells was inhibited by the uPA inhibitor UK122, dexamethasone, and a FOXO inhibitor. uPA-knock-out mice developed fewer and smaller TSC2-null lung tumors, and introduction of uPA shRNA in tumor cells or amiloride-induced uPA inhibition reduced tumorigenesis in vivo These findings suggest that interference with the uPA-dependent pathway, when used along with rapamycin, might attenuate LAM progression and potentially other TSC-related disorders.
Collapse
Affiliation(s)
| | - Konstantin V Dergilev
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Kelci R Holman
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Yelena V Parfyonova
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Zoya I Tsokolaeva
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Mimi Teter
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Elena N Atochina-Vasserman
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Alla Volgina
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | | | - Shane P Lewis
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Fedor G Zabozlaev
- the Department of Pathology, Federal Research Clinical Center Federal Medical and Biological Agency of Russia, Moscow 115682, Russia
| | - Kseniya Obraztsova
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Vera P Krymskaya
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Douglas B Cines
- From the Department of Pathology and Laboratory Medicine and
| |
Collapse
|