1
|
Lins A, Keuter L, Mulac D, Humpf HU, Langer K. Are stabilizers, located on the surface of PLGA nanoparticles, able to modify the protein adsorption pattern? Int J Pharm 2025; 674:125488. [PMID: 40107467 DOI: 10.1016/j.ijpharm.2025.125488] [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: 12/06/2024] [Revised: 01/23/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Poly(lactic-co-glycolic acid) (PLGA) is an FDA-approved, biodegradable, and biocompatible polymer, which makes it a promising starting material for the development of nanoparticles. However, in vivo studies have revealed a short biological half-life due to recognition and consequently internalization of these nanoparticles by cells of the mononuclear phagocyte system, resulting in their accumulation in the liver and spleen. In this study, we analyzed the adsorption pattern of proteins on PLGA nanoparticles after incubation with human plasma and human serum. For this analysis, different nanoparticle stabilizer systems were manufactured, and the adsorbed protein amounts were determined after incubation. Additionally, the adsorbed proteins were identified and enrichment and depletion processes of specific proteins that take place during protein incubation were measured via LC-MS/MS. The results showed a high enrichment of several opsonins on the nanoparticle surface and a depletion of most dysopsonins. Therefore, we hypothesize that an explanation for the unfavorable in vivo behavior of PLGA nanoparticles could be the formation of a biomolecular corona with a preferential adsorption of opsonins. Furthermore, we aimed to analyze whether different stabilizers, located on the surface of PLGA nanoparticles, were able to modify the protein adsorption pattern. Our findings suggest that the use of different stabilizers can influence the amount of total bound proteins on the nanoparticle surface. However, the change of stabilizers has only a minor impact on the composition of the biomolecular corona.
Collapse
Affiliation(s)
- Anika Lins
- Institute of Pharmaceutical Technology and Biopharmaceutics, University of Münster, Corrensstr. 48, Muenster 48149, Germany
| | - Lucas Keuter
- Institute of Food Chemistry, University of Münster, Corrensstr. 45, Muenster 48149, Germany
| | - Dennis Mulac
- Institute of Pharmaceutical Technology and Biopharmaceutics, University of Münster, Corrensstr. 48, Muenster 48149, Germany
| | - Hans-Ulrich Humpf
- Institute of Food Chemistry, University of Münster, Corrensstr. 45, Muenster 48149, Germany
| | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmaceutics, University of Münster, Corrensstr. 48, Muenster 48149, Germany.
| |
Collapse
|
2
|
Chen Y, Li K, Huang R, Xiong L, Li R, Jiang L, Xun Y, Wan W, Hu K. Proteomics identifies hypothermia induced adiponectin protects corneal endothelial cells via AMPK mediated autophagy in phacoemulsification. Graefes Arch Clin Exp Ophthalmol 2024; 262:3207-3217. [PMID: 38850333 DOI: 10.1007/s00417-024-06542-6] [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: 10/26/2023] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
AIM To explore the molecular mechanism underlying the protective effect of hypothermic perfusion on the corneal endothelium during phacoemulsification. METHODS Phacoemulsification was performed on New Zealand white rabbits. Perfusate at different temperatures was used during the operation, and the aqueous humor was collected for proteomic sequencing after the operation. Corneal endothelial cell injury was simulated by a corneal endothelial cell oxygen-glucose deprivation/reoxygenation (OGD/R) model in vitro. Flow cytometry and evaluation of fluorescent LC3B puncta were used to detect apoptosis and autophagy, and western blotting was used to detect protein expression. RESULTS A total of 381 differentially expressed proteins were identified between the two groups. In vitro, 4 ℃ hypothermia significantly reduced apoptosis and promoted autophagy. Apoptosis increased after autophagy was inhibited by 3-Methyladenine (3-MA). Furthermore, adiponectin (ADIPOQ) knockdown inhibited phospho-AMPK and blocked the protective effect of hypothermia on corneal endothelial cells. CONCLUSIONS We investigated the differential expression of proteins between the hypothermia group and normothermia group by proteomics. Moreover, hypothermia-induced ADIPOQ can reduce apoptosis by promoting AMPK-mediated autophagy.
Collapse
Affiliation(s)
- Yanyi Chen
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Kewei Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Rongxi Huang
- Department of Endocrinology, Chongqing General Hospital, Chongqing, China
| | - Liang Xiong
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Ruonan Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Lu Jiang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Yan Xun
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Wenjuan Wan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China.
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China.
| |
Collapse
|
3
|
Bourdin A, Brusselle G, Couillard S, Fajt ML, Heaney LG, Israel E, McDowell PJ, Menzies-Gow A, Martin N, Mitchell PD, Petousi N, Quirce S, Schleich F, Pavord ID. Phenotyping of Severe Asthma in the Era of Broad-Acting Anti-Asthma Biologics. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:809-823. [PMID: 38280454 DOI: 10.1016/j.jaip.2024.01.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/20/2023] [Accepted: 01/01/2024] [Indexed: 01/29/2024]
Abstract
Severe asthma is associated with significant morbidity and mortality despite the maximal use of inhaled corticosteroids and additional controller medications, and has a high economic burden. Biologic therapies are recommended for the management of severe, uncontrolled asthma to help to prevent exacerbations and to improve symptoms and health-related quality of life. The effective management of severe asthma requires consideration of clinical heterogeneity that is driven by varying clinical and inflammatory phenotypes, which are reflective of distinct underlying disease mechanisms. Phenotyping patients using a combination of clinical characteristics such as the age of onset or comorbidities and biomarker profiles, including blood eosinophil counts and levels of fractional exhaled nitric oxide and serum total immunoglobulin E, is important for the differential diagnosis of asthma. In addition, phenotyping is beneficial for risk assessment, selection of treatment, and monitoring of the treatment response in patients with asthma. This review describes the clinical and inflammatory phenotypes of asthma, provides an overview of biomarkers routinely used in clinical practice and those that have recently been explored for phenotyping, and aims to assess the value of phenotyping in severe asthma management in the current era of biologics.
Collapse
Affiliation(s)
- Arnaud Bourdin
- PhyMedExp, University of Montpellier, CNRS, INSERM, CHU Montpellier, Montpellier, France
| | - Guy Brusselle
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Simon Couillard
- Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Merritt L Fajt
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Liam G Heaney
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Elliot Israel
- Pulmonary and Critical Care Medicine, Allergy & Immunology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - P Jane McDowell
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Andrew Menzies-Gow
- Respiratory and Immunology, BioPharmaceuticals Medical, AstraZeneca, Cambridge, United Kingdom; Royal Brompton and Harefield Hospitals, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Neil Martin
- Respiratory and Immunology, BioPharmaceuticals Medical, AstraZeneca, Cambridge, United Kingdom; University of Leicester, Leicester, United Kingdom
| | | | - Nayia Petousi
- Respiratory Medicine, NIHR Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Santiago Quirce
- Department of Allergy, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Florence Schleich
- Department of Respiratory Medicine, CHU Liege, GIGA I3 Lab, University of Liege, Liege, Belgium
| | - Ian D Pavord
- Respiratory Medicine, NIHR Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
4
|
Rezaeeyan H, Arabfard M, Rasouli HR, Shahriary A, Gh BFNM. Evaluation of common protein biomarkers involved in the pathogenesis of respiratory diseases with proteomic methods: A systematic review. Immun Inflamm Dis 2023; 11:e1090. [PMID: 38018577 PMCID: PMC10659759 DOI: 10.1002/iid3.1090] [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: 05/15/2023] [Revised: 09/22/2023] [Accepted: 11/04/2023] [Indexed: 11/30/2023] Open
Abstract
AIM Respiratory disease (RD) is one of the most common diseases characterized by lung dysfunction. Many diagnostic mechanisms have been used to identify the pathogenic agents of responsible for RD. Among these, proteomics emerges as a valuable diagnostic method for pinpointing the specific proteins involved in RD pathogenesis. Therefore, in this study, for the first time, we examined the protein markers involved in the pathogenesis of chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), asthma, bronchiolitis obliterans (BO), and chemical warfare victims exposed to mustard gas, using the proteomics method as a systematic study. MATERIALS AND METHODS A systematic search was performed up to September 2023 on several databases, including PubMed, Scopus, ISI Web of Science, and Cochrane. In total, selected 4246 articles were for evaluation according to the criteria. Finally, 119 studies were selected for this systematic review. RESULTS A total of 13,806 proteins were identified, 6471 in COPD, 1603 in Asthma, 5638 in IPF, three in BO, and 91 in mustard gas exposed victims. Alterations in the expression of these proteins were observed in the respective diseases. After evaluation, the results showed that 31 proteins were found to be shared among all five diseases. CONCLUSION Although these 31 proteins regulate different factors and molecular pathways in all five diseases, they ultimately lead to the regulation of inflammatory pathways. In other words, the expression of some proteins in COPD and mustard-exposed patients increases inflammatory reactions, while in IPF, they cause lung fibrosis. Asthma, causes allergic reactions due to T-cell differentiation toward Th2.
Collapse
Affiliation(s)
- Hadi Rezaeeyan
- Chemical Injuries Research Center, Systems Biology and Poisonings InstituteBaqiyatallah University of Medical SciencesTehranIran
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion MedicineIranian Blood Transfusion Organization (IBTO)TehranIran
| | - Masoud Arabfard
- Chemical Injuries Research Center, Systems Biology and Poisonings InstituteBaqiyatallah University of Medical SciencesTehranIran
| | - Hamid R. Rasouli
- Trauma Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | - Alireza Shahriary
- Chemical Injuries Research Center, Systems Biology and Poisonings InstituteBaqiyatallah University of Medical SciencesTehranIran
| | - B. Fatemeh Nobakht M. Gh
- Chemical Injuries Research Center, Systems Biology and Poisonings InstituteBaqiyatallah University of Medical SciencesTehranIran
| |
Collapse
|
5
|
Zhang Q, Song J, Cao L, Sun M, Xu T, Yang S, Li S, Wang H, Fu X. RNF113A targeted by miR-197 promotes proliferation and inhibits autophagy via CXCR4/CXCL12/AKT/ERK/Beclin1 axis in cervical cancer. Exp Cell Res 2023; 428:113632. [PMID: 37164050 DOI: 10.1016/j.yexcr.2023.113632] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 05/03/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
Ring Finger Protein 113 (RNF113A), an ubiquitin E3 ligase, is genetically associated with many biological processes, including proliferation, differentiation, cell death, and neurogenesis. Recently, RNF113A has been found to be an abnormal expression in many diseases, such as X-linked trichothiodystrophy syndrome and esophageal cancer. Here, we explore the potential mechanism of RNF113A in the progression of cervical cancer (CC). In this study, we evaluated the expression level and biological function of RNF113A in CC both in vitro and in vivo by bioinformatic prediction, DIA proteomic analysis, compensation experiment, Co-IP, dual-luciferase reporter assay and nude mouse xenograft to identify the RNF113A-associated autophagy pathways involved with tumorigenesis. Consistent with the prediction from biological information analysis, we found that RNF113A was highly expressed in human CC tissues and cells. In addition, this study illustrated that the high expression of RNF113A dramatically promoted proliferation and suppressed autophagy both in vitro and in vivo. In contrast, low expression of RNF113A enhanced autophagy activities and inhibited tumor growth in CC. We also found that miRNA-197, the level of which (negative correlation with RNF113A) declined in human CC, directly restrained the expression of RNF113A. Mechanistically, proteomic and mechanistic assays uncovered that RNF113A confirmed as the direct downstream target of miR-197, promoted proliferation and restrained autophagy in CC not through direct ubiquitination degradation of autophagy marker Beclin1 but via CXCR4/CXCL12/AKT/ERK/Beclin1 signal transduction axis. In summary, we found a new miR-197/RNF113 A/CXCR4/CXCL12/AKT/ERK/Beclin1 regulation pathway that plays an important part in the survival and progression of CC.
Collapse
Affiliation(s)
- Qingwei Zhang
- Department of Obstetrics and Gynaecology, Luohe Central Hospital, Luohe, 462000, Henan, China; Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe, 462000, China
| | - Jiayu Song
- Department of Pharmacology, Luohe Medical College, Luohe, 462000, Henan, China; School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, 030001, China.
| | - Liejia Cao
- Department of Obstetrics and Gynaecology, Luohe Central Hospital, Luohe, 462000, Henan, China; Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe, 462000, China
| | - Mingzheng Sun
- Department of Pharmacology, Luohe Medical College, Luohe, 462000, Henan, China
| | - Tenghan Xu
- Department of Obstetrics and Gynaecology, Luohe Central Hospital, Luohe, 462000, Henan, China; Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe, 462000, China
| | - Shaozhe Yang
- Department of Obstetrics and Gynaecology, Luohe Central Hospital, Luohe, 462000, Henan, China; Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe, 462000, China
| | - Suhong Li
- Department of Obstetrics and Gynaecology, Luohe Central Hospital, Luohe, 462000, Henan, China; Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe, 462000, China
| | - Huifen Wang
- Department of Obstetrics and Gynaecology, Luohe Central Hospital, Luohe, 462000, Henan, China; Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe, 462000, China
| | - Xiuhong Fu
- Department of Obstetrics and Gynaecology, Luohe Central Hospital, Luohe, 462000, Henan, China; Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe, 462000, China.
| |
Collapse
|
6
|
Velázquez-Enríquez JM, Ramírez-Hernández AA, Navarro LMS, Reyes-Avendaño I, González-García K, Jiménez-Martínez C, Castro-Sánchez L, Sánchez-Chino XM, Vásquez-Garzón VR, Baltiérrez-Hoyos R. Proteomic Analysis Reveals Differential Expression Profiles in Idiopathic Pulmonary Fibrosis Cell Lines. Int J Mol Sci 2022; 23:ijms23095032. [PMID: 35563422 PMCID: PMC9105114 DOI: 10.3390/ijms23095032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/18/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, irreversible lung disorder of unknown cause. This disease is characterized by profibrotic activation of resident pulmonary fibroblasts resulting in aberrant deposition of extracellular matrix (ECM) proteins. However, although much is known about the pathophysiology of IPF, the cellular and molecular processes that occur and allow aberrant fibroblast activation remain an unmet need. To explore the differentially expressed proteins (DEPs) associated with aberrant activation of these fibroblasts, we used the IPF lung fibroblast cell lines LL97A (IPF-1) and LL29 (IPF-2), compared to the normal lung fibroblast cell line CCD19Lu (NL-1). Protein samples were quantified and identified using a label-free quantitative proteomic analysis approach by liquid chromatography-tandem mass spectrometry (LC-MS/MS). DEPs were identified after pairwise comparison, including all experimental groups. Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG), and Protein–Protein Interaction (PPI) network construction were used to interpret the proteomic data. Eighty proteins expressed exclusively in the IPF-1 and IPF-2 clusters were identified. In addition, 19 proteins were identified up-regulated in IPF-1 and 10 in IPF-2; 10 proteins were down-regulated in IPF-1 and 2 in IPF-2 when compared to the NL-1 proteome. Using the search tool for retrieval of interacting genes/proteins (STRING) software, a PPI network was constructed between the DEPs and the 80 proteins expressed exclusively in the IPF-2 and IPF-1 clusters, containing 115 nodes and 136 edges. The 10 hub proteins present in the IPP network were identified using the CytoHubba plugin of the Cytoscape software. GO and KEGG pathway analyses showed that the hub proteins were mainly related to cell adhesion, integrin binding, and hematopoietic cell lineage. Our results provide relevant information on DEPs present in IPF lung fibroblast cell lines when compared to the normal lung fibroblast cell line that could play a key role during IPF pathogenesis.
Collapse
Affiliation(s)
- Juan Manuel Velázquez-Enríquez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico; (J.M.V.-E.); (A.A.R.-H.); (I.R.-A.); (K.G.-G.)
| | - Alma Aurora Ramírez-Hernández
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico; (J.M.V.-E.); (A.A.R.-H.); (I.R.-A.); (K.G.-G.)
| | | | - Itayetzi Reyes-Avendaño
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico; (J.M.V.-E.); (A.A.R.-H.); (I.R.-A.); (K.G.-G.)
| | - Karina González-García
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico; (J.M.V.-E.); (A.A.R.-H.); (I.R.-A.); (K.G.-G.)
| | - Cristian Jiménez-Martínez
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Zacatenco, Av. Wilfrido Massieu Esq. Cda. Miguel Stampa S/N, Alcaldía Gustavo A. Madero, Mexico City 07738, Mexico;
| | - Luis Castro-Sánchez
- Conacyt-Centro Universitario de Investigaciones Biomédicas “CUIB”, Universidad de Colima, Colima 28045, Mexico;
| | - Xariss Miryam Sánchez-Chino
- Catedra-Conacyt, Departamento de Salud El Colegio de La Frontera Sur, Unidad Villahermosa, Tabasco 86280, Mexico;
| | | | - Rafael Baltiérrez-Hoyos
- Conacyt-Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico;
- Correspondence:
| |
Collapse
|
7
|
Suzuki M, Cole JJ, Konno S, Makita H, Kimura H, Nishimura M, Maciewicz RA. Large-scale plasma proteomics can reveal distinct endotypes in chronic obstructive pulmonary disease and severe asthma. Clin Transl Allergy 2021; 11:e12091. [PMID: 34962717 PMCID: PMC8686766 DOI: 10.1002/clt2.12091] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/09/2021] [Accepted: 12/07/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Chronic airway diseases including chronic obstructive pulmonary disease (COPD) and asthma are heterogenous in nature and endotypes within are underpinned by complex biology. This study aimed to investigate the utility of proteomic profiling of plasma combined with bioinformatic mining, and to define molecular endotypes and expand our knowledge of the underlying biology in chronic respiratory diseases. METHODS The plasma proteome was evaluated using an aptamer-based affinity proteomics platform (SOMAscan®), representing 1238 proteins in 34 subjects with stable COPD and 51 subjects with stable but severe asthma. For each disease, we evaluated a range of clinical/demographic characteristics including bronchodilator reversibility, blood eosinophilia levels, and smoking history. We applied modified bioinformatic approaches used in the evaluation of RNA transcriptomics. RESULTS Subjects with COPD and severe asthma were distinguished from each other by 365 different protein abundancies, with differential pathway networks and upstream modulators. Furthermore, molecular endotypes within each disease could be defined. The protein groups that defined these endotypes had both known and novel biology including groups significantly enriched in exosomal markers derived from immune/inflammatory cells. Finally, we observed associations to clinical characteristics that previously have been under-explored. CONCLUSION This investigational study evaluating the plasma proteome in clinically-phenotyped subjects with chronic airway diseases provides support that such a method can be used to define molecular endotypes and pathobiological mechanisms that underpins these endotypes. It provided new concepts about the complexity of molecular pathways that define these diseases. In the longer term, such information will help to refine treatment options for defined groups.
Collapse
Affiliation(s)
- Masaru Suzuki
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| | - John J. Cole
- GLAZgo Discovery CentreUniversity of GlasgowGlasgowUK
| | - Satoshi Konno
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| | - Hironi Makita
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
- Hokkaido Medical Research Institute for Respiratory DiseasesSapporoJapan
| | - Hiroki Kimura
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| | - Masaharu Nishimura
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
- Hokkaido Medical Research Institute for Respiratory DiseasesSapporoJapan
| | - Rose A. Maciewicz
- GLAZgo Discovery CentreUniversity of GlasgowGlasgowUK
- Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech UnitAstraZenecaGothenburgSweden
| |
Collapse
|
8
|
Qin W, Wang T, Liu G, Sun L, Han W, Gao Y. Dynamic Urinary Proteome Changes in Ovalbumin-Induced Asthma Mouse Model Using Data-Independent Acquisition Proteomics. J Asthma Allergy 2021; 14:1355-1366. [PMID: 34785909 PMCID: PMC8590963 DOI: 10.2147/jaa.s330054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 10/30/2021] [Indexed: 01/09/2023] Open
Abstract
Background In this work, we aim to investigate dynamic urinary proteome changes during asthma development and to identify potential urinary protein biomarkers for the diagnosis of asthma. Methods An ovalbumin (OVA)-induced mouse model was used to mimic asthma. The urinary proteome from asthma and control mice was determined using data-independent acquisition combined with high-resolution tandem mass spectrometry. Results Overall, 331 proteins were identified, among which 53 were differentially expressed (26, 24, 14 and 20 on days 2, 8, 15 and 18, respectively; 1.5-fold change, adjust P<0.05). Gene Ontology annotation of the differential proteins showed that the acute-phase response, innate immune response, B cell receptor signaling pathway, and complement activation were significantly enriched. Protein–protein interaction network revealed that these differential proteins were partially biologically connected in OVA-induced asthma, as a group. On days 2 and 8, after two episodes of OVA sensitization, six differential proteins (CRAMP, ECP, HP, F2, AGP1, and CFB) were also reported to be closely associated with asthma. These proteins may hold the potential for the early screening of asthma. On days 15 and 18, after challenged with 1% OVA by inhalation, seven differential proteins (VDBP, HP, CTSE, PIGR, AAT, TRFE, and HPX) were also reported to be closely associated with asthma. Thus, these proteins hold the potential to be biomarkers for the diagnosis of asthma attack. Conclusion Our results indicate that the urinary proteome could reflect dynamic pathophysiological changes in asthma progression.
Collapse
Affiliation(s)
- Weiwei Qin
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, People's Republic of China.,Department of Biochemistry and Molecular Biology, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Ting Wang
- Department of Biochemistry and Molecular Biology, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Lixin Sun
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Wei Han
- Department of Respiratory Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Youhe Gao
- Department of Biochemistry and Molecular Biology, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, 100875, People's Republic of China
| |
Collapse
|
9
|
Fanidis D, Moulos P, Aidinis V. Fibromine is a multi-omics database and mining tool for target discovery in pulmonary fibrosis. Sci Rep 2021; 11:21712. [PMID: 34741074 PMCID: PMC8571330 DOI: 10.1038/s41598-021-01069-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/21/2021] [Indexed: 11/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a lethal lung fibroproliferative disease with limited therapeutic options. Differential expression profiling of affected sites has been instrumental for involved pathogenetic mechanisms dissection and therapeutic targets discovery. However, there have been limited efforts to comparatively analyse/mine the numerous related publicly available datasets, to fully exploit their potential on the validation/creation of novel research hypotheses. In this context and towards that goal, we present Fibromine, an integrated database and exploration environment comprising of consistently re-analysed, manually curated transcriptomic and proteomic pulmonary fibrosis datasets covering a wide range of experimental designs in both patients and animal models. Fibromine can be accessed via an R Shiny application (http://www.fibromine.com/Fibromine) which offers dynamic data exploration and real-time integration functionalities. Moreover, we introduce a novel benchmarking system based on transcriptomic datasets underlying characteristics, resulting to dataset accreditation aiming to aid the user on dataset selection. Cell specificity of gene expression can be visualised and/or explored in several scRNA-seq datasets, in an effort to link legacy data with this cutting-edge methodology and paving the way to their integration. Several use case examples are presented, that, importantly, can be reproduced on-the-fly by a non-specialist user, the primary target and potential user of this endeavour.
Collapse
Affiliation(s)
- Dionysios Fanidis
- Institute for Bioinnovation, Biomedical Sciences Research Center ″Alexander Fleming″, 16672, Athens, Greece
| | - Panagiotis Moulos
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center ″Alexander Fleming″, 16672, Athens, Greece.
| | - Vassilis Aidinis
- Institute for Bioinnovation, Biomedical Sciences Research Center ″Alexander Fleming″, 16672, Athens, Greece.
| |
Collapse
|
10
|
Horimasu Y, Yamaguchi K, Sakamoto S, Masuda T, Miyamoto S, Nakashima T, Iwamoto H, Fujitaka K, Hamada H, Hattori N. Quantitative parameters of lymphocyte nuclear morphology in bronchoalveolar lavage fluid as novel biomarkers for sarcoidosis. Orphanet J Rare Dis 2021; 16:298. [PMID: 34217348 PMCID: PMC8254956 DOI: 10.1186/s13023-021-01926-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/27/2021] [Indexed: 12/03/2022] Open
Abstract
Background Bronchoalveolar lavage (BAL) is one of the fundamental examinations for the differential diagnosis of interstitial lung diseases (ILDs), and lymphocytosis strongly indicates alternative diagnoses rather than idiopathic pulmonary fibrosis. However, the BALF lymphocytosis is observed in several ILDs. We considered that quantitative evaluation of the BALF lymphocyte nuclear morphology would be useful in the differential diagnosis of ILDs with increased BALF lymphocyte fraction. Results One hundred and twenty-one patients with ILDs having increased BALF lymphocyte fraction were recruited (68 in the development cohort and 53 in the validation cohort). In the development cohort, BALF lymphocyte nuclei in sarcoidosis patients showed significantly smaller areas, shorter perimeters, lower radius ratios, and increased roundness than those of other ILD patients (p < 0.001 for each). Next, the fractions of lymphocytes with small areas, short perimeters, low radius ratios, and increased roundness, which were determined based on receiver operating characteristic (ROC) analyses-based thresholds, were demonstrated to be higher in sarcoidosis patients than in the other ILD patients (p < 0.001 for each). Furthermore, when we combined size-representing parameters with shape-representing parameters, the fraction of lymphocytes with small and round nuclei showed approximately 0.90 of area under the ROC curve in discriminating sarcoidosis both in the development cohort and the validation cohort. Conclusion This study is the first to demonstrate the usefulness of quantitative parameters of BALF lymphocyte nuclear morphology as novel biomarkers for sarcoidosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01926-x.
Collapse
Affiliation(s)
- Yasushi Horimasu
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima, 734-8551, Japan.
| | - Kakuhiro Yamaguchi
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima, 734-8551, Japan
| | - Shinjiro Sakamoto
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima, 734-8551, Japan
| | - Takeshi Masuda
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima, 734-8551, Japan
| | - Shintaro Miyamoto
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima, 734-8551, Japan
| | - Taku Nakashima
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima, 734-8551, Japan
| | - Hiroshi Iwamoto
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima, 734-8551, Japan
| | - Kazunori Fujitaka
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima, 734-8551, Japan
| | - Hironobu Hamada
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima, 734-8551, Japan
| | - Noboru Hattori
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima, 734-8551, Japan
| |
Collapse
|
11
|
Wen XP, Zhang YZ, Wan QQ. Non-targeted proteomics of acute respiratory distress syndrome: clinical and research applications. Proteome Sci 2021; 19:5. [PMID: 33743690 PMCID: PMC7980750 DOI: 10.1186/s12953-021-00174-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/11/2021] [Indexed: 01/08/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by refractory hypoxemia caused by accumulation of pulmonary fluid with a high mortality rate, but the underlying mechanism is not yet fully understood, causing absent specific therapeutic drugs to treat with ARDS. In recent years, more and more studies have applied proteomics to ARDS. Non-targeted studies of proteomics in ARDS are just beginning and have the potential to identify novel drug targets and key pathways in this disease. This paper will provide a brief review of the recent advances in the application of non-targeted proteomics to ARDS.
Collapse
Affiliation(s)
- Xu-Peng Wen
- Transplantation Center, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Yue-Zhong Zhang
- Clinical Medicine, Xiangya School of Medicine, Central South University, Changsha, 410083, Hunan, China
| | - Qi-Quan Wan
- Transplantation Center, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
12
|
Abolfathi H, Sheikhpour M, Shahraeini SS, Khatami S, Nojoumi SA. Studies in lung cancer cytokine proteomics: a review. Expert Rev Proteomics 2021; 18:49-64. [PMID: 33612047 DOI: 10.1080/14789450.2021.1892491] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Proteins are molecules that have role in the progression of the diseases. Proteomics is a tool that can play an effective role in identifying diagnostic and therapeutic biomarkers for lung cancer. Cytokines are proteins that play a decisive role in activating body's immune system in lung cancer. They can increase the growth of the tumor (oncogenic cytokines) or limit tumor growth (anti-tumor cytokines) by regulating related signaling pathways such as proliferation, growth, metastasis, and apoptosis. AREAS COVERED In the present study, a total of 223 papers including 196 research papers and 27 review papers, extracted from PubMed and Scopus and published from 1997 to present, are reviewed. The most important involved-cytokines in lung cancer including TNF-α, IFN- γ, TGF-β, VEGF and interleukins such as IL-6, IL-17, IL-8, IL-10, IL-22, IL-1β and IL-18 are introduced. Also, the pathological and biological role of such cytokines in cancer signaling pathways is explained. EXPERT OPINION In lung cancer, the cytokine expression changes under the physiological conditions of the immune system, and inflammatory cytokines are associated with the progression of lung cancer. Therefore, the cytokine expression profile can be used in the diagnosis, prognosis, prediction of therapeutic responses, and survival of patients with lung cancer.
Collapse
Affiliation(s)
- Hanie Abolfathi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Mojgan Sheikhpour
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Sadegh Shahraeini
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Shohreh Khatami
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Ali Nojoumi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
13
|
Ebrahimi B, Nazarinia M, Molayem M. Calprotectin, an available prognostic biomarker in systemic sclerosis: a systematic review. Clin Rheumatol 2020; 40:1709-1715. [PMID: 33044726 DOI: 10.1007/s10067-020-05446-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Finding easier and less invasive biologic biomarker in the clinical specimen of systemic sclerosis (SSc) patients can be effective in diagnosing and treating SSc-associated multisystem diseases. The complex of S100A8 and S100A9 (Calprotectin) is an easily available prognostic biomarker that secretes from immune cells and is necessary for initiating the immune response to noninfectious inflammation processes. The present study aims to evaluate the effectiveness of Calprotectin in specimen of SSc patients. We reviewed the evidence for Calprotectin in diagnostic and prognostic of SSc patients. METHODS This systematic review was done to identify studies on "Calprotectin" within "SSc" patients. PubMed, Web of knowledge, and Scopus were searched for this purpose. A standardized form was used to extract diseases, sample size, biomarkers identified, source of biomarker, and its effects. RESULTS Overall, the 16 articles selected show that the main sources of Calprotectin were plasma, bronchoalveolar lavage fluid, and especially stool. CONCLUSION The best source of Calprotectin was fecal Calprotectin that could show the inflammation and small intestinal bacterial overgrowth (SIBO) on SSc patients. Also, the most arguable source is plasma because of its low sample size. Comparing the Calprotectin level in different rheumatic diseases showed the specificity of fecal Calprotectin for SSc disease. Nevertheless, it has to be noted that Calprotectin correlates with some other factors such as age, PIP drug, and nonsteroidal anti-inflammatory drugs.
Collapse
Affiliation(s)
- Bahareh Ebrahimi
- Shiraz Geriatric Research Center, Shiraz University of Medical Sciences, Shiraz, 71936-35899, Iran
| | - MohamadAli Nazarinia
- Shiraz Geriatric Research Center, Shiraz University of Medical Sciences, Shiraz, 71936-35899, Iran.
- Department of Internal Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mina Molayem
- Shiraz Geriatric Research Center, Shiraz University of Medical Sciences, Shiraz, 71936-35899, Iran
| |
Collapse
|
14
|
Moodley Y, Yang IA. 'Omics': The new language in medicine that we all must learn. Respirology 2019; 25:137-138. [PMID: 31840902 DOI: 10.1111/resp.13754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/10/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Yuben Moodley
- Department of Respiratory Medicine, Fiona Stanley Hospital, Perth, WA, Australia.,School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Ian A Yang
- Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
15
|
Peripheral blood proteomic profiling of idiopathic pulmonary fibrosis biomarkers in the multicentre IPF-PRO Registry. Respir Res 2019; 20:227. [PMID: 31640794 PMCID: PMC6805665 DOI: 10.1186/s12931-019-1190-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/20/2019] [Indexed: 01/28/2023] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease for which diagnosis and management remain challenging. Defining the circulating proteome in IPF may identify targets for biomarker development. We sought to quantify the circulating proteome in IPF, determine differential protein expression between subjects with IPF and controls, and examine relationships between protein expression and markers of disease severity. Methods This study involved 300 patients with IPF from the IPF-PRO Registry and 100 participants without known lung disease. Plasma collected at enrolment was analysed using aptamer-based proteomics (1305 proteins). Linear regression was used to determine differential protein expression between participants with IPF and controls and associations between protein expression and disease severity measures (percent predicted values for forced vital capacity [FVC] and diffusion capacity of the lung for carbon monoxide [DLco]; composite physiologic index [CPI]). Multivariable models were fit to select proteins that best distinguished IPF from controls. Results Five hundred fifty one proteins had significantly different levels between IPF and controls, of which 47 showed a |log2(fold-change)| > 0.585 (i.e. > 1.5-fold difference). Among the proteins with the greatest difference in levels in patients with IPF versus controls were the glycoproteins thrombospondin 1 and von Willebrand factor and immune-related proteins C-C motif chemokine ligand 17 and bactericidal permeability-increasing protein. Multivariable classification modelling identified nine proteins that, when considered together, distinguished IPF versus control status with high accuracy (area under receiver operating curve = 0.99). Among participants with IPF, 14 proteins were significantly associated with FVC % predicted, 23 with DLco % predicted, 14 with CPI. Four proteins (roundabout homolog-2, spondin-1, polymeric immunoglobulin receptor, intercellular adhesion molecule 5) demonstrated the expected relationship across all three disease severity measures. When considered in pathways analyses, proteins associated with the presence or severity of IPF were enriched in pathways involved in platelet and haemostatic responses, vascular or platelet derived growth factor signalling, immune activation, and extracellular matrix organisation. Conclusions Patients with IPF have a distinct circulating proteome and can be distinguished using a nine-protein profile. Several proteins strongly associate with disease severity. The proteins identified may represent biomarker candidates and implicate pathways for further investigation. Trial registration ClinicalTrials.gov (NCT01915511).
Collapse
|
16
|
Yue X, Guidry JJ. Differential Protein Expression Profiles of Bronchoalveolar Lavage Fluid Following Lipopolysaccharide-Induced Direct and Indirect Lung Injury in Mice. Int J Mol Sci 2019; 20:ijms20143401. [PMID: 31373289 PMCID: PMC6679226 DOI: 10.3390/ijms20143401] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/01/2019] [Accepted: 07/10/2019] [Indexed: 12/29/2022] Open
Abstract
The pathogenic mechanisms of acute lung injury due to direct and indirect pulmonary insults are incompletely understood. Using an unbiased, discovery and quantitative proteomic approach, we examined bronchoalveolar lavage fluid (BALF) proteome following lipopolysaccharide (LPS)-induced direct and indirect lung injury in mice. A total of 1017 proteins were both identified and quantitated in BALF from control, intratracheal (I.T., direct) and intraperitoneal (I.P., indirect) LPS-treated mice. The two LPS groups shared 13 up-regulated and 22 down-regulated proteins compared to the control group. Ingenuity pathway analysis revealed that acute-phase response signaling was activated by both I.T. and I.P. LPS; however, the magnitude of activation was much greater in the I.T. LPS group. Intriguingly, two canonical signaling pathways, liver X receptor/retinoid X receptor activation, and the production of nitric oxide and reactive oxygen species in macrophages, were activated by I.T. but suppressed by I.P. LPS. Cxcl15 (also known as lungkine) was also up-regulated by I.T. but down-regulated by I.P. LPS. In conclusion, our quantitative discovery-based proteomic approach identified commonalities, as well as significant differences in BALF protein expression profiles between LPS-induced direct and indirect lung injury, and importantly, LPS-induced indirect lung injury resulted in suppression of select components of lung innate immunity.
Collapse
Affiliation(s)
- Xinping Yue
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Jessie J Guidry
- Department of Biochemistry and The Proteomic Core Facility, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
17
|
Affiliation(s)
- Albert B. Arul
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Renã A. S. Robinson
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Vanderbilt Memory & Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, Tennessee 37212, United States
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37235, United States
| |
Collapse
|