1
|
Ma X, Zhang L, Gao F, Jia W, Li C. Salvia miltiorrhiza and Tanshinone IIA reduce endothelial inflammation and atherosclerotic plaque formation through inhibiting COX-2. Biomed Pharmacother 2023; 167:115501. [PMID: 37713995 DOI: 10.1016/j.biopha.2023.115501] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023] Open
Abstract
The mechanisms of Salvia miltiorrhiza (SM) and Tanshinone IIA (Tan IIA) in the treatment of atherosclerosis was examined by combining network pharmacology and molecular biology experiments. The TCMSP and BATMAN-TCM databases provided 104 SM candidate ingredients and 813 target genes, while GEO and GeneCards databases identified 35 overlapping targets between SM and coronary artery disease (CAD). From these data, we constructed a CAD-target-active ingredient network, and using Gene Ontology (GO) and KEGG pathway analysis, 211 GO terms and 43 pathways were identified, which facilitated the construction of a key active ingredient-target-pathway network. We then constructed a protein-protein interaction (PPI) network and performed molecular docking simulations between Tan IIA and 10 key target proteins to analyze the interactions between the molecule and the protein. SM was found to alleviate CAD by reducing the expression of key pro-inflammatory factors, such as COX-2 (PTGS2), MMP9, ICAM1, TNF-α, and NF-κB. Tan IIA was identified as the primary effective component of SM in treating CAD, with TNF and PTGS2 being its main targets. We further validated these findings using in vitro/in vivo experiments. The results showed that both SM and Tan IIA attenuated the buildup of plaque and the accumulation of lipids in ApoE-/- mice. In addition, SM and Tan IIA reduced vascular inflammatory factors expression in ApoE-/- mice and ox-LDL-cultured HUVECs. Furthermore, our findings showed that Tan IIA reduced vascular endothelial inflammation and prevented plaque formation via COX-2/TNF-a/NF-κB signaling pathway. We have demonstrated for the first time that Tan IIA plays a vital role in attenuating atherosclerosis by downregulating COX-2 expression.
Collapse
Affiliation(s)
- Xiangke Ma
- Beijing Shijingshan Hospital, Capital Medical University, Beijing 100040, China.
| | - Lei Zhang
- Shandong University of Traditional Chinese Medicine, Jinan 250000, China
| | - Fujun Gao
- Shandong University of Traditional Chinese Medicine, Jinan 250000, China
| | - Weihua Jia
- Beijing Shijingshan Hospital, Capital Medical University, Beijing 100040, China
| | - Chao Li
- Shandong University of Traditional Chinese Medicine, Jinan 250000, China.
| |
Collapse
|
2
|
Forbes T, Pauza AG, Adams JC. In the balance: how do thrombospondins contribute to the cellular pathophysiology of cardiovascular disease? Am J Physiol Cell Physiol 2021; 321:C826-C845. [PMID: 34495764 DOI: 10.1152/ajpcell.00251.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Thrombospondins (TSPs) are multidomain, secreted proteins that associate with cell surfaces and extracellular matrix. In mammals, there is a large body of data on functional roles of various TSP family members in cardiovascular disease (CVD), including stroke, cardiac remodeling and fibrosis, atherosclerosis, and aortic aneurysms. Coding single nucleotide polymorphisms (SNPs) of TSP1 or TSP4 are also associated with increased risk of several forms of CVD. Whereas interactions and functional effects of TSPs on a variety of cell types have been studied extensively, the molecular and cellular basis for the differential effects of the SNPs remains under investigation. Here, we provide an integrative review on TSPs, their roles in CVD and cardiovascular cell physiology, and known properties and mechanisms of TSP SNPs relevant to CVD. In considering recent expansions to knowledge of the fundamental cellular roles and mechanisms of TSPs, as well as the effects of wild-type and variant TSPs on cells of the cardiovascular system, we aim to highlight knowledge gaps and areas for future research or of translational potential.
Collapse
Affiliation(s)
- Tessa Forbes
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Audrys G Pauza
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
3
|
Lamarche J, Ronga L, Szpunar J, Lobinski R. Characterization and Quantification of Selenoprotein P: Challenges to Mass Spectrometry. Int J Mol Sci 2021; 22:ijms22126283. [PMID: 34208081 PMCID: PMC8230778 DOI: 10.3390/ijms22126283] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/13/2022] Open
Abstract
Selenoprotein P (SELENOP) is an emerging marker of the nutritional status of selenium and of various diseases, however, its chemical characteristics still need to be investigated and methods for its accurate quantitation improved. SELENOP is unique among selenoproteins, as it contains multiple genetically encoded SeCys residues, whereas all the other characterized selenoproteins contain just one. SELENOP occurs in the form of multiple isoforms, truncated species and post-translationally modified variants which are relatively poorly characterized. The accurate quantification of SELENOP is contingent on the availability of specific primary standards and reference methods. Before recombinant SELENOP becomes available to be used as a primary standard, careful investigation of the characteristics of the SELENOP measured by electrospray MS and strict control of the recoveries at the various steps of the analytical procedures are strongly recommended. This review critically discusses the state-of-the-art of analytical approaches to the characterization and quantification of SELENOP. While immunoassays remain the standard for the determination of human and animal health status, because of their speed and simplicity, mass spectrometry techniques offer many attractive and complementary features that are highlighted and critically evaluated.
Collapse
Affiliation(s)
- Jérémy Lamarche
- IPREM UMR5254, E2S UPPA, Institut des Sciences Analytiques et de Physico-Chimie Pour l’Environnement et les Matériaux, CNRS, Université de Pau et des Pays de l’Adour, Hélioparc, 64053 Pau, France; (L.R.); (J.S.); (R.L.)
- Correspondence:
| | - Luisa Ronga
- IPREM UMR5254, E2S UPPA, Institut des Sciences Analytiques et de Physico-Chimie Pour l’Environnement et les Matériaux, CNRS, Université de Pau et des Pays de l’Adour, Hélioparc, 64053 Pau, France; (L.R.); (J.S.); (R.L.)
| | - Joanna Szpunar
- IPREM UMR5254, E2S UPPA, Institut des Sciences Analytiques et de Physico-Chimie Pour l’Environnement et les Matériaux, CNRS, Université de Pau et des Pays de l’Adour, Hélioparc, 64053 Pau, France; (L.R.); (J.S.); (R.L.)
| | - Ryszard Lobinski
- IPREM UMR5254, E2S UPPA, Institut des Sciences Analytiques et de Physico-Chimie Pour l’Environnement et les Matériaux, CNRS, Université de Pau et des Pays de l’Adour, Hélioparc, 64053 Pau, France; (L.R.); (J.S.); (R.L.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
- Chair of Analytical Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| |
Collapse
|
4
|
Olkowicz M, Czyzynska-Cichon I, Szupryczynska N, Kostogrys RB, Kochan Z, Debski J, Dadlez M, Chlopicki S, Smolenski RT. Multi-omic signatures of atherogenic dyslipidaemia: pre-clinical target identification and validation in humans. J Transl Med 2021; 19:6. [PMID: 33407555 PMCID: PMC7789501 DOI: 10.1186/s12967-020-02663-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Dyslipidaemia is a major risk factor for atherosclerosis and cardiovascular diseases. The molecular mechanisms that translate dyslipidaemia into atherogenesis and reliable markers of its progression are yet to be fully elucidated. To address this issue, we conducted a comprehensive metabolomic and proteomic analysis in an experimental model of dyslipidaemia and in patients with familial hypercholesterolemia (FH). METHODS Liquid chromatography/mass spectrometry (LC/MS) and immunoassays were used to find out blood alterations at metabolite and protein levels in dyslipidaemic ApoE-/-/LDLR-/- mice and in FH patients to evaluate their human relevance. RESULTS We identified 15 metabolites (inhibitors and substrates of nitric oxide synthase (NOS), low-molecular-weight antioxidants (glutamine, taurine), homocysteine, methionine, 1-methylnicotinamide, alanine and hydroxyproline) and 9 proteins (C-reactive protein, proprotein convertase subtilisin/kexin type 9, apolipoprotein C-III, soluble intercellular adhesion molecule-1, angiotensinogen, paraoxonase-1, fetuin-B, vitamin K-dependent protein S and biglycan) that differentiated FH patients from healthy controls. Most of these changes were consistently found in dyslipidaemic mice and were further amplified if mice were fed an atherogenic (Western or low-carbohydrate, high-protein) diet. CONCLUSIONS The alterations highlighted the involvement of an immune-inflammatory response system, oxidative stress, hyper-coagulation and impairment in the vascular function/regenerative capacity in response to dyslipidaemia that may also be directly engaged in development of atherosclerosis. Our study further identified potential biomarkers for an increased risk of atherosclerosis that may aid in clinical diagnosis or in the personalized treatment.
Collapse
Affiliation(s)
- Mariola Olkowicz
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 1 Debinki St, 80-211, Gdansk, Poland. .,Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego St., 30-348, Krakow, Poland.
| | - Izabela Czyzynska-Cichon
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego St., 30-348, Krakow, Poland
| | - Natalia Szupryczynska
- Department of Nutritional Biochemistry, Faculty of Health Sciences, Medical University of Gdansk, 7 Debinki St., 80-211, Gdansk, Poland
| | - Renata B Kostogrys
- Department of Human Nutrition and Dietetics, Faculty of Food Technology, University of Agriculture in Krakow, 122 Balicka St., 30-149, Krakow, Poland
| | - Zdzislaw Kochan
- Department of Nutritional Biochemistry, Faculty of Health Sciences, Medical University of Gdansk, 7 Debinki St., 80-211, Gdansk, Poland
| | - Janusz Debski
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 5a Pawinskiego St., 02-106, Warsaw, Poland
| | - Michal Dadlez
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 5a Pawinskiego St., 02-106, Warsaw, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego St., 30-348, Krakow, Poland.,Chair of Pharmacology, Jagiellonian University Medical College, 16 Grzegorzecka St., 31-531, Krakow, Poland
| | - Ryszard T Smolenski
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 1 Debinki St, 80-211, Gdansk, Poland.
| |
Collapse
|
5
|
Zhang Y, Ou M, Lin H, Lai L, Chen H, Chen J, Sui W, Xue W, Zhang R, Gan Q, Tang D, Sun X, Dong J, Yan Q, Dai Y. Proteomic analysis of differentially expressed proteins in the serum of patients with acute renal allograft rejection using iTRAQ labelling technology. Mol Med Rep 2020; 22:2329-2341. [PMID: 32705285 PMCID: PMC7411402 DOI: 10.3892/mmr.2020.11299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 04/07/2020] [Indexed: 12/24/2022] Open
Abstract
Transplantation is currently the best treatment for patients with end‑stage renal disease. However, acute rejection (AR) is the major source of failure in renal transplantation. The current best practice for the diagnosis of AR involves renal biopsy, but it is invasive, time‑consuming, costly and inconvenient. Sensitive and less invasive detection of AR episodes in renal transplant patients is essential to preserve allograft function. The present study applied isobaric tags for relative and absolute quantitation (iTRAQ) mass spectrometry to analyze serum protein expression in patients with AR and healthy controls. Overall, 1,399 proteins were identified. Using a cut‑off of Q<0.05 and a fold change of >1.2 for the variation in expression, 109 proteins were identified to be differentially expressed between the AR and control groups, 72 of which were upregulated and 37 were downregulated. Several proteins, including properdin, keratin 1, lipoprotein(a) and vitamin D‑binding protein, may have roles in the pathogenesis of AR. The present study focused on iTRAQ‑based proteomic profiling of serum samples in AR. Insight from the present study may help advance the understanding of the molecular mechanisms of AR and identify potential novel biomarkers of AR for further characterization.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| | - Minglin Ou
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| | - Hua Lin
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| | - Liusheng Lai
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| | - Huaizhou Chen
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| | - Jiejing Chen
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| | - Weiguo Sui
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| | - Wen Xue
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| | - Ruohan Zhang
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| | - Qing Gan
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| | - Donge Tang
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, P.R. China
| | - Xuyong Sun
- Department of Organ Transplantation, Institute of Transplant Medicine, No. 923 Hospital of People's Liberation Army, Guangxi Key Laboratory for Transplantation Medicine, Guangxi Transplantation Medicine Research Center of Engineering Technology, Nanning, Guangxi 530021, P.R. China
| | - Jianhui Dong
- Department of Organ Transplantation, Institute of Transplant Medicine, No. 923 Hospital of People's Liberation Army, Guangxi Key Laboratory for Transplantation Medicine, Guangxi Transplantation Medicine Research Center of Engineering Technology, Nanning, Guangxi 530021, P.R. China
| | - Qiang Yan
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| | - Yong Dai
- Department of Nephrology, Guilin No. 924 Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Disease Research, Guilin, Guangxi 541002, P.R. China
| |
Collapse
|
6
|
Valdés A, Bergström Lind S. Mass Spectrometry-Based Analysis of Time-Resolved Proteome Quantification. Proteomics 2019; 20:e1800425. [PMID: 31652013 DOI: 10.1002/pmic.201800425] [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: 06/28/2019] [Revised: 09/20/2019] [Indexed: 11/09/2022]
Abstract
The aspect of time is essential in biological processes and thus it is important to be able to monitor signaling molecules through time. Proteins are key players in cellular signaling and they respond to many stimuli and change their expression in many time-dependent processes. Mass spectrometry (MS) is an important tool for studying proteins, including their posttranslational modifications and their interaction partners-both in qualitative and quantitative ways. In order to distinguish the different trends over time, proteins, modification sites, and interacting proteins must be compared between different time points, and therefore relative quantification is preferred. In this review, the progress and challenges for MS-based analysis of time-resolved proteome dynamics are discussed. Further, aspects on model systems, technologies, sampling frequencies, and presentation of the dynamic data are discussed.
Collapse
Affiliation(s)
- Alberto Valdés
- Department of Analytical Chemistry, Physical Chemistry and Chemical Engineering, University of Alcalá, Ctra. Madrid-Barcelona, Km. 33.600, 28871, Alcalá de Henares, Madrid, Spain
| | - Sara Bergström Lind
- Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Box 599, 75124, Uppsala, Sweden
| |
Collapse
|
7
|
Moulder R, Bhosale SD, Goodlett DR, Lahesmaa R. Analysis of the plasma proteome using iTRAQ and TMT-based Isobaric labeling. MASS SPECTROMETRY REVIEWS 2018; 37:583-606. [PMID: 29120501 DOI: 10.1002/mas.21550] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/26/2017] [Indexed: 05/23/2023]
Abstract
Over the past decade, chemical labeling with isobaric tandem mass tags, such as isobaric tags for relative and absolute quantification reagents (iTRAQ) and tandem mass tag (TMT) reagents, has been employed in a wide range of different clinically orientated serum and plasma proteomics studies. In this review the scope of these works is presented with attention to the areas of research, methods employed and performance limitations. These applications have covered a wide range of diseases, disorders and infections, and have implemented a variety of different preparative and mass spectrometric approaches. In contrast to earlier works, which struggled to quantify more than a few hundred proteins, increasingly these studies have provided deeper insight into the plasma proteome extending the numbers of quantified proteins to over a thousand.
Collapse
Affiliation(s)
- Robert Moulder
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Santosh D Bhosale
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | | | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| |
Collapse
|
8
|
Molecular phenotyping of laboratory mouse strains using 500 multiple reaction monitoring mass spectrometry plasma assays. Commun Biol 2018; 1:78. [PMID: 30271959 PMCID: PMC6123701 DOI: 10.1038/s42003-018-0087-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/28/2018] [Indexed: 12/18/2022] Open
Abstract
Mouse is the predominant experimental model for the study of human disease due, in part, to phylogenetic relationship, ease of breeding, and the availability of molecular tools for genetic manipulation. Advances in genome-editing methodologies, such as CRISPR-Cas9, enable the rapid production of new transgenic mouse strains, necessitating complementary high-throughput and systematic phenotyping technologies. In contrast to traditional protein phenotyping techniques, multiple reaction monitoring (MRM) mass spectrometry can be highly multiplexed without forgoing specificity or quantitative precision. Here we present MRM assays for the quantitation of 500 proteins and subsequently determine reference concentration values for plasma proteins across five laboratory mouse strains that are typically used in biomedical research, revealing inter-strain and intra-strain phenotypic differences. These 500 MRM assays will have a broad range of research applications including high-throughput phenotypic validation of novel transgenic mice, identification of candidate biomarkers, and general research applications requiring multiplexed and precise protein quantification.
Collapse
|
9
|
Herrington DM, Mao C, Parker SJ, Fu Z, Yu G, Chen L, Venkatraman V, Fu Y, Wang Y, Howard TD, Jun G, Zhao CF, Liu Y, Saylor G, Spivia WR, Athas GB, Troxclair D, Hixson JE, Vander Heide RS, Wang Y, Van Eyk JE. Proteomic Architecture of Human Coronary and Aortic Atherosclerosis. Circulation 2018; 137:2741-2756. [PMID: 29915101 PMCID: PMC6011234 DOI: 10.1161/circulationaha.118.034365] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/12/2018] [Indexed: 12/26/2022]
Abstract
BACKGOUND The inability to detect premature atherosclerosis significantly hinders implementation of personalized therapy to prevent coronary heart disease. A comprehensive understanding of arterial protein networks and how they change in early atherosclerosis could identify new biomarkers for disease detection and improved therapeutic targets. METHODS Here we describe the human arterial proteome and proteomic features strongly associated with early atherosclerosis based on mass spectrometry analysis of coronary artery and aortic specimens from 100 autopsied young adults (200 arterial specimens). Convex analysis of mixtures, differential dependent network modeling, and bioinformatic analyses defined the composition, network rewiring, and likely regulatory features of the protein networks associated with early atherosclerosis and how they vary across 2 anatomic distributions. RESULTS The data document significant differences in mitochondrial protein abundance between coronary and aortic samples (coronary>>aortic), and between atherosclerotic and normal tissues (atherosclerotic< CONCLUSIONS The human arterial proteome can be viewed as a complex network whose architectural features vary considerably as a function of anatomic location and the presence or absence of atherosclerosis. The data suggest important reductions in mitochondrial protein abundance in early atherosclerosis and also identify a subset of plasma proteins that are highly predictive of angiographically defined coronary disease.
Collapse
Affiliation(s)
- David M Herrington
- Section on Cardiovascular Medicine, Department of Internal Medicine (D.M.H., C.F.Z., G.S.)
| | - Chunhong Mao
- Biocomplexity Institute of Virginia Tech, Virginia Tech, Blacksburg (C.M.)
| | - Sarah J Parker
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Heart Institute, and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (S.T.P., V.V., W.R.S., J.E.V.E.)
| | - Zongming Fu
- Johns Hopkins Medical Institute, Baltimore, MD (Z.F.)
| | - Guoqiang Yu
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington (G.Y., L.C., Y.F., Yizhi Wang, Yue Wang)
| | - Lulu Chen
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington (G.Y., L.C., Y.F., Yizhi Wang, Yue Wang)
| | - Vidya Venkatraman
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Heart Institute, and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (S.T.P., V.V., W.R.S., J.E.V.E.)
| | - Yi Fu
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington (G.Y., L.C., Y.F., Yizhi Wang, Yue Wang)
| | - Yizhi Wang
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington (G.Y., L.C., Y.F., Yizhi Wang, Yue Wang)
| | | | - Goo Jun
- Department of Epidemiology, Human Genetics and Environmental Sciences, Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston (G.J., J.E.H.)
| | - Caroline F Zhao
- Section on Cardiovascular Medicine, Department of Internal Medicine (D.M.H., C.F.Z., G.S.)
| | - Yongmei Liu
- Department of Epidemiology, Division of Public Health Sciences (Y.L.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Georgia Saylor
- Section on Cardiovascular Medicine, Department of Internal Medicine (D.M.H., C.F.Z., G.S.)
| | - Weston R Spivia
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Heart Institute, and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (S.T.P., V.V., W.R.S., J.E.V.E.)
| | - Grace B Athas
- Department of Pathology, Louisiana State Health Science Center, New Orleans (G.B.A., D.T., R.C.V.H.)
| | - Dana Troxclair
- Department of Pathology, Louisiana State Health Science Center, New Orleans (G.B.A., D.T., R.C.V.H.)
| | - James E Hixson
- Department of Epidemiology, Human Genetics and Environmental Sciences, Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston (G.J., J.E.H.)
| | - Richard S Vander Heide
- Department of Pathology, Louisiana State Health Science Center, New Orleans (G.B.A., D.T., R.C.V.H.)
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington (G.Y., L.C., Y.F., Yizhi Wang, Yue Wang)
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Heart Institute, and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (S.T.P., V.V., W.R.S., J.E.V.E.)
| |
Collapse
|
10
|
Ibrahim NE, Januzzi JL, Magaret CA, Gaggin HK, Rhyne RF, Gandhi PU, Kelly N, Simon ML, Motiwala SR, Belcher AM, van Kimmenade RR. A Clinical and Biomarker Scoring System to Predict the Presence of Obstructive Coronary Artery Disease. J Am Coll Cardiol 2017; 69:1147-1156. [DOI: 10.1016/j.jacc.2016.12.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 01/09/2023]
|
11
|
Godzien J, Ciborowski M, Armitage EG, Jorge I, Camafeita E, Burillo E, Martín-Ventura JL, Rupérez FJ, Vázquez J, Barbas C. A Single In-Vial Dual Extraction Strategy for the Simultaneous Lipidomics and Proteomics Analysis of HDL and LDL Fractions. J Proteome Res 2016; 15:1762-75. [DOI: 10.1021/acs.jproteome.5b00898] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Joanna Godzien
- CEMBIO,
Centre for Metabolomics and Bioanalysis, San Pablo CEU University, Madrid 28003, Spain
| | - Michal Ciborowski
- Clinical
Research Centre, Medical University of Bialystok, Bialystok 12-089, Poland
| | - Emily Grace Armitage
- CEMBIO,
Centre for Metabolomics and Bioanalysis, San Pablo CEU University, Madrid 28003, Spain
| | - Inmaculada Jorge
- Cardiovascular
Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Emilio Camafeita
- Cardiovascular
Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Elena Burillo
- Vascular
Research Laboratory, IIS-Fundación Jiménez Díaz-Autonoma University, 28040 Madrid, Spain
| | - Jose Luis Martín-Ventura
- Vascular
Research Laboratory, IIS-Fundación Jiménez Díaz-Autonoma University, 28040 Madrid, Spain
| | - Francisco J. Rupérez
- CEMBIO,
Centre for Metabolomics and Bioanalysis, San Pablo CEU University, Madrid 28003, Spain
| | - Jesús Vázquez
- Cardiovascular
Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Coral Barbas
- CEMBIO,
Centre for Metabolomics and Bioanalysis, San Pablo CEU University, Madrid 28003, Spain
| |
Collapse
|
12
|
Thomas S, Hao L, Ricke WA, Li L. Biomarker discovery in mass spectrometry-based urinary proteomics. Proteomics Clin Appl 2016; 10:358-70. [PMID: 26703953 DOI: 10.1002/prca.201500102] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 12/05/2015] [Accepted: 12/21/2015] [Indexed: 01/03/2023]
Abstract
Urinary proteomics has become one of the most attractive topics in disease biomarker discovery. MS-based proteomic analysis has advanced continuously and emerged as a prominent tool in the field of clinical bioanalysis. However, only few protein biomarkers have made their way to validation and clinical practice. Biomarker discovery is challenged by many clinical and analytical factors including, but not limited to, the complexity of urine and the wide dynamic range of endogenous proteins in the sample. This article highlights promising technologies and strategies in the MS-based biomarker discovery process, including study design, sample preparation, protein quantification, instrumental platforms, and bioinformatics. Different proteomics approaches are discussed, and progresses in maximizing urinary proteome coverage and standardization are emphasized in this review. MS-based urinary proteomics has great potential in the development of noninvasive diagnostic assays in the future, which will require collaborative efforts between analytical scientists, systems biologists, and clinicians.
Collapse
Affiliation(s)
- Samuel Thomas
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Ling Hao
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - William A Ricke
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA.,Department of Urology, University of Wisconsin-Madison, Madison, WI, USA
| | - Lingjun Li
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA.,Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
13
|
Hanzawa H, Sakamoto T, Kaneko A, Manri N, Zhao Y, Zhao S, Tamaki N, Kuge Y. Combined Plasma and Tissue Proteomic Study of Atherogenic Model Mouse: Approach To Elucidate Molecular Determinants in Atherosclerosis Development. J Proteome Res 2015; 14:4257-69. [PMID: 26323832 DOI: 10.1021/acs.jproteome.5b00405] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Atherogenic cardiovascular diseases are the major cause of mortality. Prevention and prediction of incidents is important; however, biomarkers that directly reflect the disease progression remain poorly investigated. To elucidate molecular determinants of atherogenesis, proteomic approaches are advantageous by using model animals for comparing changes occurring systematically (bloodstream) and locally (lesion) in accordance with the disease progression stages. We conducted differential mass spectrometric analysis between apolipoprotein E deficient (apoED) and wild-type (wt) mice using the plasma and arterial tissue of both types of mice obtained at four pathognomonic time points of the disease. A total of 100 proteins in the plasma and 390 in the arterial tissues were continuously detected throughout the four time points; 29 were identified in common. Of those, 13 proteins in the plasma and 36 in the arterial tissues showed significant difference in abundance between the apoED and wt mice at certain time points. Importantly, we found that quantitative variation patterns regarding the pathognomonic time points did not always correspond between the plasma and arterial tissues, resulting in gaining insight into atherosclerotic plaque progression. These characteristic proteins were found to be components of inflammation, thrombus formation, and vascular remodeling, suggesting drastic and integrative alteration in accordance with atherosclerosis development.
Collapse
Affiliation(s)
- Hiroko Hanzawa
- Center for Exploratory Research, Research & Development Group, Hitachi, Ltd. , 350-0395 Hatoyama, Saitama Japan.,Central Institute of Isotope Science, Hokkaido University , 060-0814 Sapporo, Japan
| | - Takeshi Sakamoto
- Center for Technology Innovation - Healthcare, Research & Development Group, Hitachi, Ltd. , 185-8601 Kokubunji, Japan.,Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University , 060-8638 Sapporo, Japan
| | - Akihito Kaneko
- Center for Technology Innovation - Healthcare, Research & Development Group, Hitachi, Ltd. , 185-8601 Kokubunji, Japan
| | - Naomi Manri
- Center for Technology Innovation - Healthcare, Research & Development Group, Hitachi, Ltd. , 185-8601 Kokubunji, Japan.,Central Institute of Isotope Science, Hokkaido University , 060-0814 Sapporo, Japan
| | - Yan Zhao
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University , 060-8638 Sapporo, Japan
| | - Songji Zhao
- Department of Tracer Kinetics & Bio-analysis, Graduate School of Medicine, Hokkaido University , 060-8638 Sapporo, Japan
| | - Nagara Tamaki
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University , 060-8638 Sapporo, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University , 060-0814 Sapporo, Japan.,Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University , 060-8638 Sapporo, Japan
| |
Collapse
|
14
|
Vascular Proteomics Reveal Novel Proteins Involved in SMC Phenotypic Change: OLR1 as a SMC Receptor Regulating Proliferation and Inflammatory Response. PLoS One 2015; 10:e0133845. [PMID: 26305474 PMCID: PMC4548952 DOI: 10.1371/journal.pone.0133845] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 07/03/2015] [Indexed: 01/21/2023] Open
Abstract
Neointimal hyperplasia of vascular smooth muscle cells (VSMC) plays a critical role in atherosclerotic plaque formation and in-stent restenosis, but the underlying mechanisms are still incompletely understood. We performed a proteomics study to identify novel signaling molecules organizing the VSMC hyperplasia. The differential proteomics analysis in a balloon-induced injury model of rat carotid artery revealed that the expressions of 44 proteins are changed within 3 days post injury. The combination of cellular function assays and a protein network analysis further demonstrated that 27 out of 44 proteins constitute key signaling networks orchestrating the phenotypic change of VSMC from contractile to epithelial-like synthetic. Among the list of proteins, the in vivo validation specifically revealed that six proteins (Rab15, ITR, OLR1, PDHβ, PTPε) are positive regulators for VSMC hyperplasia. In particular, the OLR1 played dual roles in the VSMC hyperplasia by directly mediating oxidized LDL-induced monocyte adhesion via NF-κB activation and by assisting the PDGF-induced proliferation/migration. Importantly, OLR1 and PDGFRβ were associated in close proximity in the plasma membrane. Thus, this study elicits the protein network organizing the phenotypic change of VSMC in the vascular injury diseases such as atherosclerosis and discovers OLR1 as a novel molecular link between the proliferative and inflammatory responses of VSMCs.
Collapse
|
15
|
Basak T, Varshney S, Akhtar S, Sengupta S. Understanding different facets of cardiovascular diseases based on model systems to human studies: a proteomic and metabolomic perspective. J Proteomics 2015; 127:50-60. [PMID: 25956427 DOI: 10.1016/j.jprot.2015.04.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 04/08/2015] [Accepted: 04/25/2015] [Indexed: 02/02/2023]
Abstract
UNLABELLED Cardiovascular disease has remained as the largest cause of morbidity and mortality worldwide. From dissecting the disease aetiology to identifying prognostic markers for better management of the disease is still a challenge for researchers. In the post human genome sequencing era much of the thrust has been focussed towards application of advanced genomic tools along with evaluation of traditional risk factors. With the advancement of next generation proteomics and metabolomics approaches it has now become possible to understand the protein interaction network & metabolic rewiring which lead to the perturbations of the disease phenotype. Further, elucidating different post translational modifications using advanced mass spectrometry based methods have provided an impetus towards in depth understanding of the proteome. The past decade has observed a plethora of studies where proteomics has been applied successfully to identify potential prognostic and diagnostic markers as well as to understand the disease mechanisms for various types of cardiovascular diseases. In this review, we attempted to document relevant proteomics based studies that have been undertaken either to identify potential biomarkers or have elucidated newer mechanistic insights into understanding the patho-physiology of cardiovascular disease, primarily coronary artery disease, cardiomyopathy, and myocardial ischemia. We have also provided a perspective on the potential of proteomics in combating this deadly disease. BIOLOGICAL SIGNIFICANCE This review has catalogued recent studies on proteomics and metabolomics involved in understanding several cardiovascular diseases (CVDs). A holistic systems biology based approach, of which proteomics and metabolomics are two very important components, would help in delineating various pathways associated with complex disorders like CVD. This would ultimately provide better mechanistic understanding of the disease biology leading to development of prognostic biomarkers. This article is part of a Special Issue entitled: Proteomics in India.
Collapse
Affiliation(s)
- Trayambak Basak
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110020, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-IGIB South Campus, New Delhi, India.
| | - Swati Varshney
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110020, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-IGIB South Campus, New Delhi, India
| | - Shamima Akhtar
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110020, India
| | - Shantanu Sengupta
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110020, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-IGIB South Campus, New Delhi, India.
| |
Collapse
|
16
|
Moxon JV, Liu D, Moran CS, Crossman DJ, Krishna SM, Yonglitthipagon P, Emeto TI, Morris DR, Padula MP, Mulvenna JP, Rush CM, Golledge J. Proteomic and genomic analyses suggest the association of apolipoprotein C1 with abdominal aortic aneurysm. Proteomics Clin Appl 2014; 8:762-72. [DOI: 10.1002/prca.201300119] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/23/2014] [Accepted: 01/27/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Joseph V. Moxon
- Vascular Biology Unit; Queensland Research Centre for Peripheral Vascular Disease; School of Medicine and Dentistry; James Cook University; Townsville Australia
| | - Dawei Liu
- Vascular Biology Unit; Queensland Research Centre for Peripheral Vascular Disease; School of Medicine and Dentistry; James Cook University; Townsville Australia
| | - Corey S. Moran
- Vascular Biology Unit; Queensland Research Centre for Peripheral Vascular Disease; School of Medicine and Dentistry; James Cook University; Townsville Australia
| | - David J. Crossman
- Faculty of Medical and Health Sciences; Department of Physiology; the University of Auckland; Auckland New Zealand
| | - Smriti M. Krishna
- Vascular Biology Unit; Queensland Research Centre for Peripheral Vascular Disease; School of Medicine and Dentistry; James Cook University; Townsville Australia
| | | | - Theophilus I. Emeto
- Vascular Biology Unit; Queensland Research Centre for Peripheral Vascular Disease; School of Medicine and Dentistry; James Cook University; Townsville Australia
- Microbiology and Immunology Department; School of Veterinary and Biomedical Sciences; James Cook University; Townsville Australia
| | - Dylan R. Morris
- Vascular Biology Unit; Queensland Research Centre for Peripheral Vascular Disease; School of Medicine and Dentistry; James Cook University; Townsville Australia
| | - Matthew P. Padula
- Proteomics Core Facility; University of Technology; Sydney Australia
| | - Jason P. Mulvenna
- Infectious Disease and Cancer; QIMR Berghofer Medical Research Institute; Brisbane Australia
| | - Catherine M. Rush
- Vascular Biology Unit; Queensland Research Centre for Peripheral Vascular Disease; School of Medicine and Dentistry; James Cook University; Townsville Australia
- Microbiology and Immunology Department; School of Veterinary and Biomedical Sciences; James Cook University; Townsville Australia
| | - Jonathan Golledge
- Vascular Biology Unit; Queensland Research Centre for Peripheral Vascular Disease; School of Medicine and Dentistry; James Cook University; Townsville Australia
- Department of Vascular and Endovascular Surgery; The Townsville Hospital; Townsville Australia
| |
Collapse
|
17
|
Shen X, Young R, Canty JM, Qu J. Quantitative proteomics in cardiovascular research: global and targeted strategies. Proteomics Clin Appl 2014; 8:488-505. [PMID: 24920501 DOI: 10.1002/prca.201400014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/02/2014] [Accepted: 06/06/2014] [Indexed: 11/05/2022]
Abstract
Extensive technical advances in the past decade have substantially expanded quantitative proteomics in cardiovascular research. This has great promise for elucidating the mechanisms of cardiovascular diseases and the discovery of cardiac biomarkers used for diagnosis and treatment evaluation. Global and targeted proteomics are the two major avenues of quantitative proteomics. While global approaches enable unbiased discovery of altered proteins via relative quantification at the proteome level, targeted techniques provide higher sensitivity and accuracy, and are capable of multiplexed absolute quantification in numerous clinical/biological samples. While promising, technical challenges need to be overcome to enable full utilization of these techniques in cardiovascular medicine. Here, we discuss recent advances in quantitative proteomics and summarize applications in cardiovascular research with an emphasis on biomarker discovery and elucidating molecular mechanisms of disease. We propose the integration of global and targeted strategies as a high-throughput pipeline for cardiovascular proteomics. Targeted approaches enable rapid, extensive validation of biomarker candidates discovered by global proteomics. These approaches provide a promising alternative to immunoassays and other low-throughput means currently used for limited validation.
Collapse
Affiliation(s)
- Xiaomeng Shen
- Department of Biochemistry, University at Buffalo, Buffalo, NY, USA; New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY, USA
| | | | | | | |
Collapse
|
18
|
Optimization of plasma sample pretreatment for quantitative analysis using iTRAQ labeling and LC-MALDI-TOF/TOF. PLoS One 2014; 9:e101694. [PMID: 24988083 PMCID: PMC4079693 DOI: 10.1371/journal.pone.0101694] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 06/11/2014] [Indexed: 01/19/2023] Open
Abstract
Shotgun proteomic methods involving iTRAQ (isobaric tags for relative and absolute quantitation) peptide labeling facilitate quantitative analyses of proteomes and searches for useful biomarkers. However, the plasma proteome's complexity and the highly dynamic plasma protein concentration range limit the ability of conventional approaches to analyze and identify a large number of proteins, including useful biomarkers. The goal of this paper is to elucidate the best approach for plasma sample pretreatment for MS- and iTRAQ-based analyses. Here, we systematically compared four approaches, which include centrifugal ultrafiltration, SCX chromatography with fractionation, affinity depletion, and plasma without fractionation, to reduce plasma sample complexity. We generated an optimized protocol for quantitative protein analysis using iTRAQ reagents and an UltrafleXtreme (Bruker Daltonics) MALDI TOF/TOF mass spectrometer. Moreover, we used a simple, rapid, efficient, but inexpensive sample pretreatment technique that generated an optimal opportunity for biomarker discovery. We discuss the results from the four sample pretreatment approaches and conclude that SCX chromatography without affinity depletion is the best plasma sample preparation pretreatment method for proteome analysis. Using this technique, we identified 1,780 unique proteins, including 1,427 that were quantified by iTRAQ with high reproducibility and accuracy.
Collapse
|
19
|
Corradini E, Vallur R, Raaijmakers LM, Feil S, Feil R, Heck AJR, Scholten A. Alterations in the cerebellar (Phospho)proteome of a cyclic guanosine monophosphate (cGMP)-dependent protein kinase knockout mouse. Mol Cell Proteomics 2014; 13:2004-16. [PMID: 24925903 DOI: 10.1074/mcp.m113.035154] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The cyclic nucleotide cyclic guanosine monophosphate (cGMP) plays an important role in learning and memory, but its signaling mechanisms in the mammalian brain are not fully understood. Using mass-spectrometry-based proteomics, we evaluated how the cerebellum adapts its (phospho)proteome in a knockout mouse model of cGMP-dependent protein kinase type I (cGKI). Our data reveal that a small subset of proteins in the cerebellum (∼3% of the quantified proteins) became substantially differentially expressed in the absence of cGKI. More changes were observed at the phosphoproteome level, with hundreds of sites being differentially phosphorylated between wild-type and knockout cerebellum. Most of these phosphorylated sites do not represent known cGKI substrates. An integrative computational network analysis of the data indicated that the differentially expressed proteins and proteins harboring differentially phosphorylated sites largely belong to a tight network in the Purkinje cells of the cerebellum involving important cGMP/cAMP signaling nodes (e.g. PDE5 and PKARIIβ) and Ca(2+) signaling (e.g. SERCA3). In this way, removal of cGKI could be linked to impaired cerebellar long-term depression at Purkinje cell synapses. In addition, we were able to identify a set of novel putative (phospho)proteins to be considered in this network. Overall, our data improve our understanding of cerebellar cGKI signaling and suggest novel players in cGKI-regulated synaptic plasticity.
Collapse
Affiliation(s)
- Eleonora Corradini
- From ‡Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands; §Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Raghavan Vallur
- ¶Interfakultäres Institut für Biochemie, Universität Tübingen, D-72074 Tübingen, Germany; ‖Graduate School for Cellular and Molecular Neuroscience, Universität Tübingen, D-72074 Tübingen, Germany; **German Center for Neurodegenerative diseases (DZNE), D-72076 Tübingen, Germany
| | - Linsey M Raaijmakers
- From ‡Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands; §Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Susanne Feil
- ¶Interfakultäres Institut für Biochemie, Universität Tübingen, D-72074 Tübingen, Germany
| | - Robert Feil
- ¶Interfakultäres Institut für Biochemie, Universität Tübingen, D-72074 Tübingen, Germany
| | - Albert J R Heck
- From ‡Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands; §Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands;
| | - Arjen Scholten
- From ‡Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands; §Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands;
| |
Collapse
|
20
|
Effects of hypertension and exercise on cardiac proteome remodelling. BIOMED RESEARCH INTERNATIONAL 2014; 2014:634132. [PMID: 24877123 PMCID: PMC4022191 DOI: 10.1155/2014/634132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/14/2014] [Indexed: 12/29/2022]
Abstract
Left ventricle hypertrophy is a common outcome of pressure overload stimulus closely associated with hypertension. This process is triggered by adverse molecular signalling, gene expression, and proteome alteration. Proteomic research has revealed that several molecular targets are associated with pathologic cardiac hypertrophy, including angiotensin II, endothelin-1 and isoproterenol. Several metabolic, contractile, and stress-related proteins are shown to be altered in cardiac hypertrophy derived by hypertension. On the other hand, exercise is a nonpharmacologic agent used for hypertension treatment, where cardiac hypertrophy induced by exercise training is characterized by improvement in cardiac function and resistance against ischemic insult. Despite the scarcity of proteomic research performed with exercise, healthy and pathologic heart proteomes are shown to be modulated in a completely different way. Hence, the altered proteome induced by exercise is mostly associated with cardioprotective aspects such as contractile and metabolic improvement and physiologic cardiac hypertrophy. The present review, therefore, describes relevant studies involving the molecular characteristics and alterations from hypertensive-induced and exercise-induced hypertrophy, as well as the main proteomic research performed in this field. Furthermore, proteomic research into the effect of hypertension on other target-demerged organs is examined.
Collapse
|
21
|
Recent advances in cardiovascular proteomics. J Proteomics 2012; 81:3-14. [PMID: 23153792 DOI: 10.1016/j.jprot.2012.10.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 10/10/2012] [Accepted: 10/31/2012] [Indexed: 01/08/2023]
Abstract
Cardiovascular diseases (CVDs) are the major source of global morbidity and death and more people die annually from CVDs than from any other cause. These diseases can occur quickly, as seen in acute myocardial infarction (AMI), or progress slowly over years as with chronic heart failure. Advances in mass spectrometry detection and analysis, together with improved isolation and enrichment techniques allowing for the separation of organelles and membrane proteins, now allow for the indepth analysis of the cardiac proteome. Here we outline current insights that have been provided through cardiovascular proteomics, and discuss studies that have developed innovative technologies which permit the examination of the protein complement in specific organelles including exosomes and secreted proteins. We highlight these foundational studies and illustrate how they are providing the technologies and tools which are now being applied to further study cardiovascular disease; provide new diagnostic markers and potentially new methods of cardiac patient management with identification of novel drug targets. This article is part of a Special Issue entitled: From protein structures to clinical applications.
Collapse
|
22
|
Kim YS, Jung H, Gil HW, Hong SY, Song HY. Proteomic analysis of changes in protein expression in serum from animals exposed to paraquat. Int J Mol Med 2012; 30:1521-7. [PMID: 23023206 DOI: 10.3892/ijmm.2012.1143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 08/13/2012] [Indexed: 11/06/2022] Open
Abstract
Paraquat (PQ) poisoning remains a major public health concern in many countries. Extensive research has focused on finding early diagnostic biomarkers of acute PQ poisoning. In order to investigate the characterization of diagnostic biomarkers in PQ poisoning, we utilized proteomic analysis using serum from rats exposed to PQ, and we identified 8 differentially expressed proteins from over 500 protein spots. The expression of apolipoprotein E (ApoE), preprohaptoglobin (Pphg), a precursor of haptoglobin (Hp), and complement component 3 (C3) proteins was greatly induced by PQ exposure while the expression of fibrinogen γ-chain (FGG) and Ac-158 was dramatically reduced. To further investigate the possibility of ApoE, Pphg and FGG as useful diagnostic biomarkers of PQ poisoning, western blot and qRT-PCR analyses were conducted using cell lines as well as rat and human sera. The expression levels of ApoE, Hp and FGG were significantly altered in the presence of PQ in both rat and human serum suggesting that these proteins may be appropriate candidate molecular biomarkers for the early diagnosis of acute PQ intoxication.
Collapse
Affiliation(s)
- Yong-Sik Kim
- Department of Microbiology, College of Medicine, Soon Chun Hyang University, Cheonan, Chungnam 330-090, Republic of Korea
| | | | | | | | | |
Collapse
|
23
|
Podder B, Kim YS, Zerin T, Song HY. Antioxidant effect of silymarin on paraquat-induced human lung adenocarcinoma A549 cell line. Food Chem Toxicol 2012; 50:3206-14. [DOI: 10.1016/j.fct.2012.06.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 06/04/2012] [Accepted: 06/07/2012] [Indexed: 12/31/2022]
|
24
|
|
25
|
Mirza SP. Quantitative Mass Spectrometry-Based Approaches in Cardiovascular Research. ACTA ACUST UNITED AC 2012; 5:477. [DOI: 10.1161/circgenetics.110.957753] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
26
|
Abstract
Hypertension is a major cardiovascular risk factor with a multifactorial pathogenesis, including genetic and environmental factors. In addition to hypothesis-driven strategies, unbiased approaches such as genomics, proteomics, and metabolomics are useful tools to help unravel the pathophysiology of hypertension and associated organ damage. During development of cardiovascular disease the key organs and tissues undergo extensive functional and structural changes that are characterized by alterations in the amount and type of proteins that are expressed. Proteomic approaches study the expression of large numbers of proteins in organs, tissues, cells, and body fluids. A number of different proteomic platforms are available, many of which combine two methods to separate proteins and peptides after an initial digestion step. Identification of these peptides and changes in their expression in parallel with disease processes or medical treatment will help to identify as yet unknown pathophysiological pathways. There is also potential to use proteomic signatures as biomarkers of cardiovascular disease that will contribute to population screening, diagnosis of diseases and their severity, and monitoring of therapeutic interventions.
Collapse
Affiliation(s)
- Christian Delles
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, UK.
| | | | | |
Collapse
|