1
|
Rodríguez JM, Jorge I, Martinez-Val A, Barrero-Rodríguez R, Magni R, Núñez E, Laguillo A, Devesa CA, López JA, Camafeita E, Vázquez J. iSanXoT: A standalone application for the integrative analysis of mass spectrometry-based quantitative proteomics data. Comput Struct Biotechnol J 2024; 23:452-459. [PMID: 38235360 PMCID: PMC10792623 DOI: 10.1016/j.csbj.2023.12.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/22/2023] [Accepted: 12/22/2023] [Indexed: 01/19/2024] Open
Abstract
Many bioinformatics tools are available for the quantitative analysis of proteomics experiments. Most of these tools use a dedicated statistical model to derive absolute quantitative protein values from mass spectrometry (MS) data. Here, we present iSanXoT, a standalone application that processes relative abundances between MS signals and then integrates them sequentially to upper levels using the previously published Generic Integration Algorithm (GIA). iSanXoT offers unique capabilities that complement conventional quantitative software applications, including statistical weighting and independent modeling of error distributions in each integration, aggregation of technical or biological replicates, quantification of posttranslational modifications, and analysis of coordinated protein behavior. iSanXoT is a standalone, user-friendly application that accepts output from popular proteomics pipelines and enables unrestricted creation of quantification workflows and fully customizable reports that can be reused across projects or shared among users. Numerous publications attest the successful application of diverse integrative workflows constructed using the GIA for the analysis of high-throughput quantitative proteomics experiments. iSanXoT has been tested with the main operating systems. Download links for the corresponding distributions are available at https://github.com/CNIC-Proteomics/iSanXoT/releases.
Collapse
Affiliation(s)
- Jose Manuel Rodríguez
- Proteomics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid 28029, Spain
| | - Inmaculada Jorge
- Laboratory of Cardiovascular Proteomics. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Ana Martinez-Val
- Laboratory of Cardiovascular Proteomics. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Rafael Barrero-Rodríguez
- Laboratory of Cardiovascular Proteomics. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Ricardo Magni
- Laboratory of Cardiovascular Proteomics. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Estefanía Núñez
- Laboratory of Cardiovascular Proteomics. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Andrea Laguillo
- Laboratory of Cardiovascular Proteomics. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Cristina A. Devesa
- Laboratory of Cardiovascular Proteomics. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Juan A. López
- Proteomics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid 28029, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Emilio Camafeita
- Proteomics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid 28029, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
2
|
Núñez E, Gómez-Serrano M, Calvo E, Bonzon-Kulichenko E, Trevisan-Herraz M, Rodríguez JM, García-Marqués F, Magni R, Lara-Pezzi E, Martín-Ventura JL, Camafeita E, Vázquez J. A Multiplexed Quantitative Proteomics Approach to the Human Plasma Protein Signature. Biomedicines 2024; 12:2118. [PMID: 39335631 PMCID: PMC11428418 DOI: 10.3390/biomedicines12092118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Despite the plasma proteome being able to provide a unique insight into the health and disease status of individuals, holding singular promise as a source of protein biomarkers that could be pivotal in the context of personalized medicine, only around 100 proteins covering a few human conditions have been approved as biomarkers by the US Food and Drug Administration (FDA) so far. Mass spectrometry (MS) currently has enormous potential for high-throughput analysis in clinical research; however, plasma proteomics remains challenging mainly due to the wide dynamic range of plasma protein abundances and the time-consuming procedures required. We applied a new MS-based multiplexed proteomics workflow to quantitate proteins, encompassing 67 FDA-approved biomarkers, in >1300 human plasma samples from a clinical cohort. Our results indicate that this workflow is suitable for large-scale clinical studies, showing good accuracy and reproducibility (coefficient of variation (CV) < 20 for 90% of the proteins). Furthermore, we identified plasma signature proteins (stable in time on an individual basis), stable proteins (exhibiting low biological variability and high temporal stability), and highly variable proteins (with low temporal stability) that can be used for personalized health monitoring and medicine.
Collapse
Affiliation(s)
- Estefanía Núñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; (E.N.); (E.C.); (E.B.-K.); (J.M.R.); (R.M.); (E.L.-P.)
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| | - María Gómez-Serrano
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany;
| | - Enrique Calvo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; (E.N.); (E.C.); (E.B.-K.); (J.M.R.); (R.M.); (E.L.-P.)
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| | - Elena Bonzon-Kulichenko
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; (E.N.); (E.C.); (E.B.-K.); (J.M.R.); (R.M.); (E.L.-P.)
| | - Marco Trevisan-Herraz
- International Center for Life, Newcastle University, Newcastle upon Tyne NE1 4EP, UK;
| | - José Manuel Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; (E.N.); (E.C.); (E.B.-K.); (J.M.R.); (R.M.); (E.L.-P.)
| | | | - Ricardo Magni
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; (E.N.); (E.C.); (E.B.-K.); (J.M.R.); (R.M.); (E.L.-P.)
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| | - Enrique Lara-Pezzi
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; (E.N.); (E.C.); (E.B.-K.); (J.M.R.); (R.M.); (E.L.-P.)
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| | - José Luis Martín-Ventura
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
- IIS-Fundación Jiménez-Díaz, 28015 Madrid, Spain
| | - Emilio Camafeita
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; (E.N.); (E.C.); (E.B.-K.); (J.M.R.); (R.M.); (E.L.-P.)
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; (E.N.); (E.C.); (E.B.-K.); (J.M.R.); (R.M.); (E.L.-P.)
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| |
Collapse
|
3
|
Vélez-López O, Carrasquillo-Carrión K, Cantres-Rosario YM, Machín-Martínez E, Álvarez-Ríos ME, Roche-Lima A, Tosado-Rodríguez EL, Meléndez LM. Analysis of Sigma-1 Receptor Antagonist BD1047 Effect on Upregulating Proteins in HIV-1-Infected Macrophages Exposed to Cocaine Using Quantitative Proteomics. Biomedicines 2024; 12:1934. [PMID: 39335448 PMCID: PMC11428496 DOI: 10.3390/biomedicines12091934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/08/2024] [Accepted: 08/10/2024] [Indexed: 09/30/2024] Open
Abstract
HIV-1 infects monocyte-derived macrophages (MDM) that migrate into the brain and secrete virus and neurotoxic molecules, including cathepsin B (CATB), causing cognitive dysfunction. Cocaine potentiates CATB secretion and neurotoxicity in HIV-infected MDM. Pretreatment with BD1047, a sigma-1 receptor antagonist, before cocaine exposure reduces HIV-1, CATB secretion, and neuronal apoptosis. We aimed to elucidate the intracellular pathways modulated by BD1047 in HIV-infected MDM exposed to cocaine. We hypothesized that the Sig1R antagonist BD1047, prior to cocaine, significantly deregulates proteins and pathways involved in HIV-1 replication and CATB secretion that lead to neurotoxicity. MDM culture lysates from HIV-1-infected women treated with BD1047 before cocaine were compared with untreated controls using TMT quantitative proteomics, bioinformatics, Lima statistics, and pathway analyses. Results demonstrate that pretreatment with BD1047 before cocaine dysregulated eighty (80) proteins when compared with the infected cocaine group. We found fifteen (15) proteins related to HIV-1 infection, CATB, and mitochondrial function. Upregulated proteins were related to oxidative phosphorylation (SLC25A-31), mitochondria (ATP5PD), ion transport (VDAC2-3), endoplasmic reticulum transport (PHB, TMED10, CANX), and cytoskeleton remodeling (TUB1A-C, ANXA1). BD1047 treatment protects HIV-1-infected MDM exposed to cocaine by upregulating proteins that reduce mitochondrial damage, ER transport, and exocytosis associated with CATB-induced neurotoxicity.
Collapse
Affiliation(s)
- Omar Vélez-López
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA;
| | - Kelvin Carrasquillo-Carrión
- Integrated Informatics, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00934, USA; (K.C.-C.); (A.R.-L.); (E.L.T.-R.)
| | - Yadira M. Cantres-Rosario
- Translational Proteomics, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00921, USA;
| | - Eraysy Machín-Martínez
- Department of Biology, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00921, USA; (E.M.-M.); (M.E.Á.-R.)
| | - Manuel E. Álvarez-Ríos
- Department of Biology, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00921, USA; (E.M.-M.); (M.E.Á.-R.)
| | - Abiel Roche-Lima
- Integrated Informatics, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00934, USA; (K.C.-C.); (A.R.-L.); (E.L.T.-R.)
| | - Eduardo L. Tosado-Rodríguez
- Integrated Informatics, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00934, USA; (K.C.-C.); (A.R.-L.); (E.L.T.-R.)
| | - Loyda M. Meléndez
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA;
- Translational Proteomics, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00921, USA;
| |
Collapse
|
4
|
Hawkins LM, Wang C, Chaput D, Batra M, Marsilia C, Awshah D, Suvorova ES. The Crk4-Cyc4 complex regulates G 2/M transition in Toxoplasma gondii. EMBO J 2024; 43:2094-2126. [PMID: 38600241 PMCID: PMC11148040 DOI: 10.1038/s44318-024-00095-4] [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: 01/10/2024] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/12/2024] Open
Abstract
A versatile division of apicomplexan parasites and a dearth of conserved regulators have hindered the progress of apicomplexan cell cycle studies. While most apicomplexans divide in a multinuclear fashion, Toxoplasma gondii tachyzoites divide in the traditional binary mode. We previously identified five Toxoplasma CDK-related kinases (Crk). Here, we investigated TgCrk4 and its cyclin partner TgCyc4. We demonstrated that TgCrk4 regulates conventional G2 phase processes, such as repression of chromosome rereplication and centrosome reduplication, and acts upstream of the spindle assembly checkpoint. The spatial TgCyc4 dynamics supported the TgCrk4-TgCyc4 complex role in the coordination of chromosome and centrosome cycles. We also identified a dominant TgCrk4-TgCyc4 complex interactor, TgiRD1 protein, related to DNA replication licensing factor CDT1 but played no role in licensing DNA replication in the G1 phase. Our results showed that TgiRD1 also plays a role in controlling chromosome and centrosome reduplication. Global phosphoproteome analyses identified TgCrk4 substrates, including TgORC4, TgCdc20, TgGCP2, and TgPP2ACA. Importantly, the phylogenetic and structural studies suggest the Crk4-Cyc4 complex is limited to a minor group of the binary dividing apicomplexans.
Collapse
Affiliation(s)
- Lauren M Hawkins
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Chengqi Wang
- College of Public Health, University of South Florida, Tampa, FL, 33612, USA
| | - Dale Chaput
- Proteomics Core, College of Arts and Sciences, University of South Florida, Tampa, FL, 33612, USA
| | - Mrinalini Batra
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Clem Marsilia
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Danya Awshah
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Elena S Suvorova
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.
| |
Collapse
|
5
|
Alvarez MS, Núñez E, Fuertes-Agudo M, Cucarella C, Fernandez-Velasco M, Boscá L, Vázquez J, Rossignol R, Martin-Sanz P, Casado M. Quantitative Proteomics Analysis Reveals That Cyclooxygenase-2 Modulates Mitochondrial Respiratory Chain Complex IV in Cardiomyocytes. Int J Mol Sci 2022; 23:13476. [PMID: 36362254 PMCID: PMC9655412 DOI: 10.3390/ijms232113476] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/24/2022] [Accepted: 10/30/2022] [Indexed: 10/10/2023] Open
Abstract
The biochemical mechanisms of cell injury and myocardial cell death after myocardial infarction remain unresolved. Cyclooxygenase 2 (COX-2), a key enzyme in prostanoid synthesis, is expressed in human ischemic myocardium and dilated cardiomyopathy, but it is absent in healthy hearts. To assess the role of COX-2 in cardiovascular physiopathology, we developed transgenic mice that constitutively express functional human COX-2 in cardiomyocytes under the control of the α-myosin heavy chain promoter. These animals had no apparent phenotype but were protected against ischemia-reperfusion injury in isolated hearts, with enhanced functional recovery and diminished cellular necrosis. To further explore the phenotype of this animal model, we carried out a differential proteome analysis of wild-type vs. transgenic cardiomyocytes. The results revealed a tissue-specific proteomic profile dominated by mitochondrial proteins. In particular, an increased expression of respiratory chain complex IV proteins was observed. This correlated with increased catalytic activity, enhanced respiratory capacity, and increased ATP levels in the heart of COX-2 transgenic mice. These data suggest a new link between COX-2 and mitochondria, which might contribute to the protective cardiac effects of COX-2 against ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Maria Soledad Alvarez
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain
| | - Estefanía Núñez
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Marina Fuertes-Agudo
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Carme Cucarella
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Maria Fernandez-Velasco
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, IDIPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Lisardo Boscá
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Rodrigue Rossignol
- Laboratoire Maladies Rares, CHU Pellegrin Place Amelie Rab, 33076 Bordeaux, France
| | - Paloma Martin-Sanz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Marta Casado
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| |
Collapse
|
6
|
The Plasmodium falciparum Nuclear Protein Phosphatase NIF4 Is Required for Efficient Merozoite Invasion and Regulates Artemisinin Sensitivity. mBio 2022; 13:e0189722. [PMID: 35938722 PMCID: PMC9426563 DOI: 10.1128/mbio.01897-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Artemisinin resistance in Plasmodium falciparum has been associated with a mutation in the NLI-interacting factor-like phosphatase PfNIF4, in addition to the mutations in the Kelch13 protein as the major determinant. We found that PfNIF4 was predominantly expressed at the schizont stage and localized in the nuclei of the parasite. To elucidate the functions of PfNIF4 in P. falciparum, we performed PfNIF4 knockdown (KD) using the inducible ribozyme system. PfNIF4 KD attenuated merozoite invasion and affected gametocytogenesis. PfNIF4 KD parasites also showed significantly increased in vitro susceptibility to artemisinins. Transcriptomic and proteomic analysis revealed that PfNIF4 KD led to the downregulation of gene categories involved in invasion and artemisinin resistance (e.g., mitochondrial function, membrane, and Kelch13 interactome) at the trophozoite and/or schizont stage. Consistent with PfNIF4 being a protein phosphatase, PfNIF4 KD resulted in an overall upregulation of the phosphoproteome of infected erythrocytes. Quantitative phosphoproteomic profiling identified a set of PfNIF4-regulated phosphoproteins with functional similarity to FCP1 substrates, particularly proteins involved in chromatin organization and transcriptional regulation. Specifically, we observed increased phosphorylation of Ser2/5 of the tandem repeats in the C-terminal domain (CTD) of RNA polymerase II (RNAPII) upon PfNIF4 KD. Furthermore, using the TurboID-based proteomic approach, we identified that PfNIF4 interacted with the RNAPII components, AP2-domain transcription factors, and chromatin-modifiers and binders. These findings suggest that PfNIF4 may act as the RNAPII CTD phosphatase, regulating the expression of general and parasite-specific cellular pathways during the blood-stage development.
Collapse
|
7
|
Nuñez E, Orera I, Carmona-Rodríguez L, Paño JR, Vázquez J, Corrales FJ. Mapping the Serum Proteome of COVID-19 Patients; Guidance for Severity Assessment. Biomedicines 2022; 10:1690. [PMID: 35884998 PMCID: PMC9313396 DOI: 10.3390/biomedicines10071690] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), whose outbreak in 2019 led to an ongoing pandemic with devastating consequences for the global economy and human health. According to the World Health Organization, COVID-19 has affected more than 481 million people worldwide, with 6 million confirmed deaths. The joint efforts of the scientific community have undoubtedly increased the pace of production of COVID-19 vaccines, but there is still so much uncharted ground to cover regarding the mechanisms of SARS-CoV-2 infection, replication and host response. These issues can be approached by proteomics with unprecedented capacity paving the way for the development of more efficient strategies for patient care. In this study, we present a deep proteome analysis that has been performed on a cohort of 72 COVID-19 patients aiming to identify serum proteins assessing the dynamics of the disease at different age ranges. A panel of 53 proteins that participate in several functions such as acute-phase response and inflammation, blood coagulation, cell adhesion, complement cascade, endocytosis, immune response, oxidative stress and tissue injury, have been correlated with patient severity, suggesting a molecular basis for their clinical stratification. Eighteen protein candidates were further validated by targeted proteomics in an independent cohort of 84 patients including a group of individuals that had satisfactorily resolved SARS-CoV-2 infection. Remarkably, all protein alterations were normalized 100 days after leaving the hospital, which further supports the reliability of the selected proteins as hallmarks of COVID-19 progression and grading. The optimized protein panel may prove its value for optimal severity assessment as well as in the follow up of COVID-19 patients.
Collapse
Affiliation(s)
- Estefanía Nuñez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
- Cardiovascular Proteomics Laboratory, Centro Nacional de Enfermedades Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Irene Orera
- Proteomics Research Core Facility, Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain;
| | | | - José Ramón Paño
- Division of Infectious Diseases, Hospital Clínico Universitario, IIS Aragón, Ciberinfec, 50009 Zaragoza, Spain;
| | - Jesús Vázquez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
- Cardiovascular Proteomics Laboratory, Centro Nacional de Enfermedades Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Fernando J. Corrales
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología (CSIC), 28049 Madrid, Spain;
| |
Collapse
|
8
|
Roldán-Montero R, Pérez-Sáez JM, Cerro-Pardo I, Oller J, Martinez-Lopez D, Nuñez E, Maller SM, Gutierrez-Muñoz C, Mendez-Barbero N, Escola-Gil JC, Michel JB, Mittelbrunn M, Vázquez J, Blanco-Colio LM, Rabinovich GA, Martin-Ventura JL. Galectin-1 prevents pathological vascular remodeling in atherosclerosis and abdominal aortic aneurysm. SCIENCE ADVANCES 2022; 8:eabm7322. [PMID: 35294231 PMCID: PMC8926342 DOI: 10.1126/sciadv.abm7322] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Pathological vascular remodeling is the underlying cause of atherosclerosis and abdominal aortic aneurysm (AAA). Here, we analyzed the role of galectin-1 (Gal-1), a β-galactoside-binding protein, as a therapeutic target for atherosclerosis and AAA. Mice lacking Gal-1 (Lgals1-/-) developed severe atherosclerosis induced by pAAV/D377Y-mPCSK9 adenovirus and displayed higher lipid levels and lower expression of contractile markers of vascular smooth muscle cells (VSMCs) in plaques than wild-type mice. Proteomic analysis of Lgals1-/- aortas showed changes in markers of VSMC phenotypic switch and altered composition of mitochondrial proteins. Mechanistically, Gal-1 silencing resulted in increased foam cell formation and mitochondrial dysfunction in VSMCs, while treatment with recombinant Gal-1 (rGal-1) prevented these effects. Furthermore, rGal-1 treatment attenuated atherosclerosis and elastase-induced AAA, leading to higher contractile VSMCs in aortic tissues. Gal-1 expression decreased in human atheroma and AAA compared to control tissue. Thus, Gal-1-driven circuits emerge as potential therapeutic strategies in atherosclerosis and AAA.
Collapse
Affiliation(s)
- Raquel Roldán-Montero
- IIS-Fundación Jiménez-Díaz-Autonoma University of Madrid (UAM). Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Juan M. Pérez-Sáez
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428ADN Buenos Aires, Argentina
| | - Isabel Cerro-Pardo
- IIS-Fundación Jiménez-Díaz-Autonoma University of Madrid (UAM). Madrid, Spain
| | - Jorge Oller
- Centro de Biología Molecular Severo Ochoa, Centro Superior de Investigaciones Científicas-UAM, Madrid, Spain
- Instituto de Investigación del Hospital 12 de Octubre, Madrid, Spain
| | | | - Estefania Nuñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Sebastian M. Maller
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428ADN Buenos Aires, Argentina
| | | | - Nerea Mendez-Barbero
- IIS-Fundación Jiménez-Díaz-Autonoma University of Madrid (UAM). Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | | | - Maria Mittelbrunn
- Centro de Biología Molecular Severo Ochoa, Centro Superior de Investigaciones Científicas-UAM, Madrid, Spain
- Instituto de Investigación del Hospital 12 de Octubre, Madrid, Spain
| | - Jesús Vázquez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Luis M. Blanco-Colio
- IIS-Fundación Jiménez-Díaz-Autonoma University of Madrid (UAM). Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Gabriel A. Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428ADN Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428AGE Buenos Aires, Argentina
- Corresponding author. (J.L.M.-V.); (G.A.R.)
| | - Jose L. Martin-Ventura
- IIS-Fundación Jiménez-Díaz-Autonoma University of Madrid (UAM). Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Corresponding author. (J.L.M.-V.); (G.A.R.)
| |
Collapse
|
9
|
Hawkins LM, Naumov AV, Batra M, Wang C, Chaput D, Suvorova ES. Novel CRK-Cyclin Complex Controls Spindle Assembly Checkpoint in Toxoplasma Endodyogeny. mBio 2021; 13:e0356121. [PMID: 35130726 PMCID: PMC8822342 DOI: 10.1128/mbio.03561-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/18/2022] [Indexed: 12/21/2022] Open
Abstract
Opportunistic parasites of the Apicomplexa phylum use a variety of division modes built on two types of cell cycles that incorporate two distinctive mechanisms of mitosis: uncoupled from and coupled to parasite budding. Parasites have evolved novel factors to regulate such unique replication mechanisms that are poorly understood. Here, we have combined genetics, quantitative fluorescence microscopy, and global proteomics approaches to examine endodyogeny in Toxoplasma gondii dividing by mitosis coupled to cytokinesis. In the current study, we focus on the steps controlled by the recently described atypical Cdk-related kinase T. gondii Crk6 (TgCrk6). While inspecting protein complexes, we found that this previously orphaned TgCrk6 kinase interacts with a parasite-specific atypical cyclin, TgCyc1. We built conditional expression models and examined primary cell cycle defects caused by the lack of TgCrk6 or TgCyc1. Quantitative microscopy assays revealed that tachyzoites deficient in either TgCrk6 or the cyclin partner TgCyc1 exhibit identical mitotic defects, suggesting cooperative action of the complex components. Further examination of the mitotic structures indicated that the TgCrk6/TgCyc1 complex regulates metaphase. This novel finding confirms a functional spindle assembly checkpoint (SAC) in T. gondii. Measuring global changes in protein expression and phosphorylation, we found evidence that canonical activities of the Toxoplasma SAC are intertwined with parasite-specific tasks. Analysis of phosphorylation motifs suggests that Toxoplasma metaphase is regulated by CDK, mitogen-activated kinase (MAPK), and Aurora kinases, while the TgCrk6/TgCyc1 complex specifically controls the centromere-associated network. IMPORTANCE The rate of Toxoplasma tachyzoite division directly correlates with the severity of the disease, toxoplasmosis, which affects humans and animals. Thus, a better understanding of the tachyzoite cell cycle would offer much-needed efficient tools to control the acute stage of infection. Although tachyzoites divide by binary division, the cell cycle architecture and regulation differ significantly from the conventional binary fission of their host cells. Unlike the unidirectional conventional cell cycle, the Toxoplasma budding cycle is braided and is regulated by multiple essential Cdk-related kinases (Crks) that emerged in the place of missing conventional cell cycle regulators. How these novel Crks control apicomplexan cell cycles is largely unknown. Here, we have discovered a novel parasite-specific complex, TgCrk6/TgCyc1, that orchestrates a major mitotic event, the spindle assembly checkpoint. We demonstrated that tachyzoites incorporated parasite-specific tasks in the canonical checkpoint functions.
Collapse
Affiliation(s)
- Lauren M. Hawkins
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Anatoli V. Naumov
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Mrinalini Batra
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Changqi Wang
- College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Dale Chaput
- Proteomics Core, College of Arts and Sciences, University of South Florida, Tampa, Florida, USA
| | - Elena S. Suvorova
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
10
|
López E, Marinaro F, de Pedro MDLÁ, Sánchez-Margallo FM, Gómez-Serrano M, Ponath V, Pogge von Strandmann E, Jorge I, Vázquez J, Fernández-Pereira LM, Crisóstomo V, Álvarez V, Casado JG. The Immunomodulatory Signature of Extracellular Vesicles From Cardiosphere-Derived Cells: A Proteomic and miRNA Profiling. Front Cell Dev Biol 2020; 8:321. [PMID: 32582685 PMCID: PMC7295954 DOI: 10.3389/fcell.2020.00321] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
Experimental data demonstrated that the regenerative potential and immunomodulatory capacity of cardiosphere-derived cells (CDCs) is mediated by paracrine mechanisms. In this process, extracellular vesicles derived from CDCs (EV-CDCs) are key mediators of their therapeutic effect. Considering the future applicability of these vesicles in human diseases, an accurate preclinical-to-clinical translation is needed, as well as an exhaustive molecular characterization of animal-derived therapeutic products. Based on that, the main goal of this study was to perform a comprehensive characterization of proteins and miRNAs in extracellular vesicles from porcine CDCs as a clinically relevant animal model. The analysis was performed by identification and quantification of proteins and miRNA expression profiles. Our results revealed the presence of clusters of immune-related and cardiac-related molecular biomarkers in EV-CDCs. Additionally, considering that priming stem cells with inflammatory stimuli may increase the therapeutic potential of released vesicles, here we studied the dynamic changes that occur in the extracellular vesicles from IFNγ-primed CDCs. These analyses detected statistically significant changes in several miRNAs and proteins. Notably, the increase in interleukin 6 (IL6) protein, as well as the increase in mir-125b (that targets IL6 receptor) was especially relevant. These results suggest a potential involvement of EV-CDCs in the regulation of the IL6/IL6R axis, with implications in inflammatory-mediated diseases.
Collapse
Affiliation(s)
- Esther López
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Federica Marinaro
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | | | - Francisco Miguel Sánchez-Margallo
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - María Gómez-Serrano
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Viviane Ponath
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany.,Clinic for Hematology, Oncology, and Immunology, Philipps University, Marburg, Germany
| | - Elke Pogge von Strandmann
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany.,Clinic for Hematology, Oncology, and Immunology, Philipps University, Marburg, Germany
| | - Inmaculada Jorge
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús Vázquez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Verónica Crisóstomo
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Verónica Álvarez
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Javier G Casado
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
11
|
Marinaro F, Macías-García B, Sánchez-Margallo FM, Blázquez R, Álvarez V, Matilla E, Hernández N, Gómez-Serrano M, Jorge I, Vázquez J, González-Fernández L, Pericuesta E, Gutiérrez-Adán A, Casado JG. Extracellular vesicles derived from endometrial human mesenchymal stem cells enhance embryo yield and quality in an aged murine model†. Biol Reprod 2020; 100:1180-1192. [PMID: 30596891 DOI: 10.1093/biolre/ioy263] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/22/2018] [Accepted: 12/26/2018] [Indexed: 12/21/2022] Open
Abstract
Advanced age is a risk factor undermining women's fertility. Hence, the optimization of assisted reproduction techniques is an interdisciplinary challenge that requires the improvement of in vitro culture systems. Here, we hypothesize that supplementation of embryo culture medium with extracellular vesicles from endometrial-derived mesenchymal stem cells (EV-endMSCs) may have a positive impact on the embryo competence of aged oocytes. In this work, 24 weeks old B6D2 female mice were used as egg donors and in vitro fertilization assays were performed using males from the same strain (8-12 weeks); the presumptive zygotes were incubated in the presence of 0, 10, 20, 40, or 80 μg/ml of EV-endMSCs. The results from the proteomic analysis of EV-endMSCs and the classification by Reactome pathways allowed us to identify proteins closely related with the fertilization process. Moreover, in our aged murine model, the supplementation of the embryo culture medium with EV-endMSCs improved the developmental competence of the embryos as well as the total blastomere count. Finally, gene expression analysis of murine blastocysts showed significant changes on core genes related to cellular response to oxidative stress, metabolism, placentation, and trophectoderm/inner cell mass formation. In summary, we demonstrate that EV-endMSCs increase the quality of the embryos, and according to proteomic and genomic analysis, presumably by modulating the expression of antioxidant enzymes and promoting pluripotent activity. Therefore, EV-endMSCs could be a valuable tool in human assisted reproduction improving the developmental competence of aged oocytes and increasing the odds of implantation and subsequent delivery.
Collapse
Affiliation(s)
- Federica Marinaro
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre (JUMISC), Cáceres, Spain
| | - Beatriz Macías-García
- Assisted Reproduction Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Francisco Miguel Sánchez-Margallo
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre (JUMISC), Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Rebeca Blázquez
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre (JUMISC), Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Verónica Álvarez
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre (JUMISC), Cáceres, Spain
| | - Elvira Matilla
- Assisted Reproduction Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Nuria Hernández
- Assisted Reproduction Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - María Gómez-Serrano
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Inmaculada Jorge
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús Vázquez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Lauro González-Fernández
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, Cáceres, Spain
| | | | | | - Javier G Casado
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre (JUMISC), Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
12
|
Trevisan-Herraz M, Bagwan N, García-Marqués F, Rodriguez JM, Jorge I, Ezkurdia I, Bonzon-Kulichenko E, Vázquez J. SanXoT: a modular and versatile package for the quantitative analysis of high-throughput proteomics experiments. Bioinformatics 2020; 35:1594-1596. [PMID: 30252043 PMCID: PMC6499250 DOI: 10.1093/bioinformatics/bty815] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 08/17/2018] [Accepted: 09/24/2018] [Indexed: 11/12/2022] Open
Abstract
Summary Mass spectrometry-based proteomics has had a formidable development in recent years, increasing the amount of data handled and the complexity of the statistical resources needed. Here we present SanXoT, an open-source, standalone software package for the statistical analysis of high-throughput, quantitative proteomics experiments. SanXoT is based on our previously developed weighted spectrum, peptide and protein statistical model and has been specifically designed to be modular, scalable and user-configurable. SanXoT allows limitless workflows that adapt to most experimental setups, including quantitative protein analysis in multiple experiments, systems biology, quantification of post-translational modifications and comparison and merging of experimental data from technical or biological replicates. Availability and implementation Download links for the SanXoT Software Package, source code and documentation are available at https://wikis.cnic.es/proteomica/index.php/SSP. Contact jvazquez@cnic.es or ebonzon@cnic.es Supplementary information Supplementary information is available at Bioinformatics online.
Collapse
Affiliation(s)
- Marco Trevisan-Herraz
- Vascular Pathophysiology Area, Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Navratan Bagwan
- Vascular Pathophysiology Area, Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Fernando García-Marqués
- Vascular Pathophysiology Area, Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Jose Manuel Rodriguez
- Vascular Pathophysiology Area, Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Inmaculada Jorge
- Vascular Pathophysiology Area, Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Iakes Ezkurdia
- Vascular Pathophysiology Area, Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Elena Bonzon-Kulichenko
- Vascular Pathophysiology Area, Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Jesús Vázquez
- Vascular Pathophysiology Area, Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
13
|
Marinaro F, Gómez-Serrano M, Jorge I, Silla-Castro JC, Vázquez J, Sánchez-Margallo FM, Blázquez R, López E, Álvarez V, Casado JG. Unraveling the Molecular Signature of Extracellular Vesicles From Endometrial-Derived Mesenchymal Stem Cells: Potential Modulatory Effects and Therapeutic Applications. Front Bioeng Biotechnol 2019; 7:431. [PMID: 31921832 PMCID: PMC6932983 DOI: 10.3389/fbioe.2019.00431] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022] Open
Abstract
Endometrial-derived Mesenchymal Stem Cells (endMSCs) are involved in the regeneration and remodeling of human endometrium, being considered one of the most promising candidates for stem cell-based therapies. Their therapeutic effects have been found to be mediated by extracellular vesicles (EV-endMSCs) with pro-angiogenic, anti-apoptotic, and immunomodulatory effects. Based on that, the main goal of this study was to characterize the proteome and microRNAome of these EV-endMSCs by proteomics and transcriptomics approaches. Additionally, we hypothesized that inflammatory priming of endMSCs may contribute to modify the therapeutic potential of these vesicles. High-throughput proteomics revealed that 617 proteins were functionally annotated as Extracellular exosome (GO:0070062), corresponding to the 70% of the EV-endMSC proteome. Bioinformatics analyses allowed us to identify that these proteins were involved in adaptive/innate immune response, complement activation, antigen processing/presentation, negative regulation of apoptosis, and different signaling pathways, among others. Of note, multiplexed quantitative proteomics and Systems Biology analyses showed that IFNγ priming significantly modulated the protein profile of these vesicles. As expected, proteins involved in antigen processing and presentation were significantly increased. Interestingly, immunomodulatory proteins, such as CSF1, ERAP1, or PYCARD were modified. Regarding miRNAs expression profile in EV-endMSCs, Next-Generation Sequencing (NGS) showed that the preferred site of microRNAome targeting was the nucleus (n = 371 microTargets), significantly affecting signal transduction (GO:0007165), cell proliferation (GO:0008283), and apoptotic processes (GO:0006915), among others. Interestingly, NGS analyses highlighted that several miRNAs, such as hsa-miR-150-5p or hsa-miR-196b-5p, were differentially expressed in IFNγ-primed EV-endMSCs. These miRNAs have a functional involvement in glucocorticoid receptor signaling, IL-6/8/12 signaling, and in the role of macrophages. In summary, these results allowed us to understand the complexity of the molecular networks in EV-endMSCs and their potential effects on target cells. To our knowledge, this is the first comprehensive study based on proteomic and genomic approaches to unravel the therapeutic potential of these extracellular vesicles, that may be used as immunomodulatory effectors in the treatment of inflammatory conditions.
Collapse
Affiliation(s)
- Federica Marinaro
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - María Gómez-Serrano
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain.,Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.,Center for Tumor Biology and Immunology, Institute of Molecular Biology and Tumor Research, Philipps University, Marburg, Germany
| | - Inmaculada Jorge
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain.,Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | - Jesús Vázquez
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain.,Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Francisco Miguel Sánchez-Margallo
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Rebeca Blázquez
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Esther López
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Verónica Álvarez
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Javier G Casado
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| |
Collapse
|
14
|
MAP: model-based analysis of proteomic data to detect proteins with significant abundance changes. Cell Discov 2019; 5:40. [PMID: 31636953 PMCID: PMC6796874 DOI: 10.1038/s41421-019-0107-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 05/14/2019] [Accepted: 06/24/2019] [Indexed: 12/14/2022] Open
Abstract
Isotope-labeling-based mass spectrometry (MS) is widely used in quantitative proteomic studies. With this technique, the relative abundance of thousands of proteins can be efficiently profiled in parallel, greatly facilitating the detection of proteins differentially expressed across samples. However, this task remains computationally challenging. Here we present a new approach, termed Model-based Analysis of Proteomic data (MAP), for this task. Unlike many existing methods, MAP does not require technical replicates to model technical and systematic errors, and instead utilizes a novel step-by-step regression analysis to directly assess the significance of observed protein abundance changes. We applied MAP to compare the proteomic profiles of undifferentiated and differentiated mouse embryonic stem cells (mESCs), and found it has superior performance compared with existing tools in detecting proteins differentially expressed during mESC differentiation. A web-based application of MAP is provided for online data processing at http://bioinfo.sibs.ac.cn/shaolab/MAP.
Collapse
|
15
|
Madugundu AK, Na CH, Nirujogi RS, Renuse S, Kim KP, Burns KH, Wilks C, Langmead B, Ellis SE, Collado‐Torres L, Halushka MK, Kim M, Pandey A. Integrated Transcriptomic and Proteomic Analysis of Primary Human Umbilical Vein Endothelial Cells. Proteomics 2019; 19:e1800315. [PMID: 30983154 PMCID: PMC6812510 DOI: 10.1002/pmic.201800315] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/17/2019] [Indexed: 01/11/2023]
Abstract
Understanding the molecular profile of every human cell type is essential for understanding its role in normal physiology and disease. Technological advancements in DNA sequencing, mass spectrometry, and computational methods allow us to carry out multiomics analyses although such approaches are not routine yet. Human umbilical vein endothelial cells (HUVECs) are a widely used model system to study pathological and physiological processes associated with the cardiovascular system. In this study, next-generation sequencing and high-resolution mass spectrometry to profile the transcriptome and proteome of primary HUVECs is employed. Analysis of 145 million paired-end reads from next-generation sequencing confirmed expression of 12 186 protein-coding genes (FPKM ≥0.1), 439 novel long non-coding RNAs, and revealed 6089 novel isoforms that were not annotated in GENCODE. Proteomics analysis identifies 6477 proteins including confirmation of N-termini for 1091 proteins, isoforms for 149 proteins, and 1034 phosphosites. A database search to specifically identify other post-translational modifications provide evidence for a number of modification sites on 117 proteins which include ubiquitylation, lysine acetylation, and mono-, di- and tri-methylation events. Evidence for 11 "missing proteins," which are proteins for which there was insufficient or no protein level evidence, is provided. Peptides supporting missing protein and novel events are validated by comparison of MS/MS fragmentation patterns with synthetic peptides. Finally, 245 variant peptides derived from 207 expressed proteins in addition to alternate translational start sites for seven proteins and evidence for novel proteoforms for five proteins resulting from alternative splicing are identified. Overall, it is believed that the integrated approach employed in this study is widely applicable to study any primary cell type for deeper molecular characterization.
Collapse
Affiliation(s)
- Anil K. Madugundu
- Center for Molecular MedicineNational Institute of Mental Health and NeurosciencesHosur RoadBangalore560029KarnatakaIndia
- Institute of BioinformaticsInternational Technology ParkBangalore560066KarnatakaIndia
- Manipal Academy of Higher EducationManipal576104KarnatakaIndia
- McKusick‐Nathans Institute of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Center for Individualized Medicine and Department of Laboratory Medicine and PathologyMayo ClinicRochesterMN55905USA
| | - Chan Hyun Na
- McKusick‐Nathans Institute of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMD21205USA
- NeurologyInstitute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Raja Sekhar Nirujogi
- McKusick‐Nathans Institute of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Santosh Renuse
- McKusick‐Nathans Institute of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Center for Individualized Medicine and Department of Laboratory Medicine and PathologyMayo ClinicRochesterMN55905USA
| | - Kwang Pyo Kim
- Department of Applied ChemistryKyung Hee UniversityYonginGyeonggi17104Republic of Korea
| | - Kathleen H. Burns
- McKusick‐Nathans Institute of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Departments of PathologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMD21205USA
- High Throughput Biology CenterJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Christopher Wilks
- Department of Computer ScienceJohns Hopkins UniversityBaltimoreMD21218USA
- Center for Computational BiologyJohns Hopkins UniversityBaltimoreMD21205USA
| | - Ben Langmead
- Department of Computer ScienceJohns Hopkins UniversityBaltimoreMD21218USA
- Center for Computational BiologyJohns Hopkins UniversityBaltimoreMD21205USA
| | - Shannon E. Ellis
- Center for Computational BiologyJohns Hopkins UniversityBaltimoreMD21205USA
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMD21205USA
| | - Leonardo Collado‐Torres
- Center for Computational BiologyJohns Hopkins UniversityBaltimoreMD21205USA
- Lieber Institute for Brain DevelopmentJohns Hopkins Medical CampusBaltimoreMD21205USA
| | - Marc K. Halushka
- Departments of PathologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Min‐Sik Kim
- Department of Applied ChemistryKyung Hee UniversityYonginGyeonggi17104Republic of Korea
- Department of New BiologyDGISTDaegu42988Republic of Korea
| | - Akhilesh Pandey
- Center for Molecular MedicineNational Institute of Mental Health and NeurosciencesHosur RoadBangalore560029KarnatakaIndia
- McKusick‐Nathans Institute of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Center for Individualized Medicine and Department of Laboratory Medicine and PathologyMayo ClinicRochesterMN55905USA
- NeurologyInstitute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Departments of PathologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Department of Biological ChemistryJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Department of OncologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
| |
Collapse
|
16
|
Smith JR, David LL, Appukuttan B, Wilmarth PA. Angiogenic and Immunologic Proteins Identified by Deep Proteomic Profiling of Human Retinal and Choroidal Vascular Endothelial Cells: Potential Targets for New Biologic Drugs. Am J Ophthalmol 2018; 193:197-229. [PMID: 29559410 PMCID: PMC6109601 DOI: 10.1016/j.ajo.2018.03.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 03/06/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE Diseases that involve retinal or choroidal vascular endothelial cells are leading causes of vision loss: age-related macular degeneration, retinal ischemic vasculopathies, and noninfectious posterior uveitis. Proteins differentially expressed by these endothelial cell populations are potential drug targets. We used deep proteomic profiling to define the molecular phenotype of human retinal and choroidal endothelial cells at the protein level. METHODS Retinal and choroidal vascular endothelial cells were separately isolated from 5 human eye pairs by selection on CD31. Total protein was extracted and digested, and peptide fractions were analyzed by reverse-phase liquid chromatography tandem mass spectrometry. Peptide sequences were assigned to fragment ion spectra, and proteins were inferred from openly accessible protein databases. Protein abundance was determined by spectral counting. Publicly available software packages were used to identify proteins that were differentially expressed between human retinal and choroidal endothelial cells, and to classify proteins that were highly abundant in each endothelial cell population. RESULTS Human retinal and/or choroidal vascular endothelial cells expressed 5042 nonredundant proteins. Setting the differential expression false discovery rate at 0.05, 498 proteins of 3454 quantifiable proteins (14.4%) with minimum mean spectral counts of 2.5 were differentially abundant in the 2 cell populations. Retinal and choroidal endothelial cells were enriched in angiogenic proteins, and retinal endothelial cells were also enriched in immunologic proteins. CONCLUSIONS This work describes the different protein expression profiles of human retinal and choroidal vascular endothelial cells, and provides multiple candidates for further study as novel treatments or drug targets for posterior eye diseases. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.
Collapse
Affiliation(s)
- Justine R Smith
- Flinders University, Adelaide, Australia; Oregon Health & Science University, Portland, Oregon, USA.
| | - Larry L David
- Flinders University, Adelaide, Australia; Oregon Health & Science University, Portland, Oregon, USA
| | - Binoy Appukuttan
- Flinders University, Adelaide, Australia; Oregon Health & Science University, Portland, Oregon, USA
| | - Phillip A Wilmarth
- Flinders University, Adelaide, Australia; Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
17
|
Molina-Sánchez P, Jorge I, Martinez-Pinna R, Blanco-Colio LM, Tarin C, Torres-Fonseca MM, Esteban M, Laustsen J, Ramos-Mozo P, Calvo E, Lopez JA, Ceniga MVD, Michel JB, Egido J, Andrés V, Vazquéz J, Meilhac O, Burillo E, Lindholt JS, Martin-Ventura JL. ApoA-I/HDL-C levels are inversely associated with abdominal aortic aneurysm progression. Thromb Haemost 2017; 113:1335-46. [DOI: 10.1160/th14-10-0874] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/21/2015] [Indexed: 12/18/2022]
Abstract
SummaryAbdominal aortic aneurysm (AAA) evolution is unpredictable, and there is no therapy except surgery for patients with an aortic size > 5 cm (large AAA). We aimed to identify new potential biomarkers that could facilitate prognosis and treatment of patients with AAA. A differential quantitative proteomic analysis of plasma proteins was performed in AAA patients at different stages of evolution [small AAA (aortic size=3�5cm) vs large AAA] using iTRAQ labelling, highthroughput nano-LC-MS/MS and a novel multi-layered statistical model. Among the proteins identified, ApoA-I was decreased in patients with large AAA compared to those with small AAA. These results were validated by ELISA on plasma samples from small (n=90) and large AAA (n=26) patients (150 ± 3 vs 133 ± 5 mg/dl, respectively, p< 0.001). ApoA-I levels strongly correlated with HDL-Cholesterol (HDL-C) concentration (r=0.9, p< 0.001) and showed a negative correlation with aortic size (r=-0.4, p< 0.01) and thrombus volume (r=-0.3, p< 0.01), which remained significant after adjusting for traditional risk factors. In a prospective study, HDL-C independently predicted aneurysmal growth rate in multiple linear regression analysis (n=122, p=0.008) and was inversely associated with need for surgical repair (Adjusted hazard ratio: 0.18, 95 % confidence interval: 0.04�0.74, p=0.018). In a nation-wide Danish registry, we found lower mean HDL-C concentration in large AAA patients (n=6,560) compared with patients with aorto-iliac occlusive disease (n=23,496) (0.89 ± 2.99 vs 1.59 ± 5.74 mmol/l, p< 0.001). Finally, reduced mean aortic AAA diameter was observed in AngII-infused mice treated with ApoA-I mimetic peptide compared with saline-injected controls. In conclusion, ApoAI/ HDL-C systemic levels are negatively associated with AAA evolution. Therapies targeting HDL functionality could halt AAA formation.
Collapse
|
18
|
Villar M, López V, Ayllón N, Cabezas-Cruz A, López JA, Vázquez J, Alberdi P, de la Fuente J. The intracellular bacterium Anaplasma phagocytophilum selectively manipulates the levels of vertebrate host proteins in the tick vector Ixodes scapularis. Parasit Vectors 2016; 9:467. [PMID: 27561965 PMCID: PMC5000436 DOI: 10.1186/s13071-016-1747-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/11/2016] [Indexed: 12/12/2022] Open
Abstract
Background The intracellular bacteria Anaplasma phagocytophilum are emerging zoonotic pathogens affecting human and animal health, and a good model for the study of tick-host-pathogen interactions. This tick-borne pathogen is transmitted by Ixodes scapularis in the United States where it causes human granulocytic anaplasmosis. Tick midguts and salivary glands play a major role during tick feeding and development, and in pathogen acquisition, multiplication and transmission. Vertebrate host proteins are found in tick midguts after feeding and have been described in the salivary glands of fed and unfed ticks, suggesting a role for these proteins during tick feeding and development. Furthermore, recent results suggested the hypothesis that pathogen infection affects tick metabolic processes to modify host protein digestion and persistence in the tick with possible implications for tick physiology and pathogen life-cycle. Methods To address this hypothesis, herein we used I. scapularis female ticks fed on uninfected and A. phagocytophilum-infected sheep to characterize host protein content in midguts and salivary glands by proteomic analysis of tick tissues. Results The results evidenced a clear difference in the host protein content between tick midguts and salivary glands in response to infection suggesting that A. phagocytophilum selectively manipulates the levels of vertebrate host proteins in ticks in a tissue-specific manner to facilitate pathogen infection, multiplication and transmission while preserving tick feeding and development. The mechanisms by which A. phagocytophilum manipulates the levels of vertebrate host proteins are not known, but the results obtained here suggested that it might include the modification of proteolytic pathways. Conclusions The results of this study provided evidence to support that A. phagocytophilum affect tick proteolytic pathways to selectively manipulate the levels of vertebrate host proteins in a tissue-specific manner to increase tick vector capacity. Investigating the biological relevance of host proteins in tick biology and pathogen infection and the mechanisms used by A. phagocytophilum to manipulate host protein content is essential to advance our knowledge of tick-host-pathogen molecular interactions. These results have implications for the identification of new targets for the development of vaccines for the control of tick-borne diseases. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1747-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Margarita Villar
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
| | - Vladimir López
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
| | - Nieves Ayllón
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
| | - Alejandro Cabezas-Cruz
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000, Lille, France
| | - Juan A López
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Pilar Alberdi
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
| | - José de la Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain. .,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, 74078, USA.
| |
Collapse
|
19
|
García-Marqués F, Trevisan-Herraz M, Martínez-Martínez S, Camafeita E, Jorge I, Lopez JA, Méndez-Barbero N, Méndez-Ferrer S, Del Pozo MA, Ibáñez B, Andrés V, Sánchez-Madrid F, Redondo JM, Bonzon-Kulichenko E, Vázquez J. A Novel Systems-Biology Algorithm for the Analysis of Coordinated Protein Responses Using Quantitative Proteomics. Mol Cell Proteomics 2016; 15:1740-60. [PMID: 26893027 DOI: 10.1074/mcp.m115.055905] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Indexed: 11/06/2022] Open
Abstract
The coordinated behavior of proteins is central to systems biology. However, the underlying mechanisms are poorly known and methods to analyze coordination by conventional quantitative proteomics are still lacking. We present the Systems Biology Triangle (SBT), a new algorithm that allows the study of protein coordination by pairwise quantitative proteomics. The Systems Biology Triangle detected statistically significant coordination in diverse biological models of very different nature and subjected to different kinds of perturbations. The Systems Biology Triangle also revealed with unprecedented molecular detail an array of coordinated, early protein responses in vascular smooth muscle cells treated at different times with angiotensin-II. These responses included activation of protein synthesis, folding, turnover, and muscle contraction - consistent with a differentiated phenotype-as well as the induction of migration and the repression of cell proliferation and secretion. Remarkably, the majority of the altered functional categories were protein complexes, interaction networks, or metabolic pathways. These changes could not be detected by other algorithms widely used by the proteomics community, and the vast majority of proteins involved have not been described before to be regulated by AngII. The unique capabilities of The Systems Biology Triangle to detect functional protein alterations produced by the coordinated action of proteins in pairwise quantitative proteomics experiments make this algorithm an attractive choice for the biological interpretation of results on a routine basis.
Collapse
Affiliation(s)
- Fernando García-Marqués
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Marco Trevisan-Herraz
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Sara Martínez-Martínez
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Emilio Camafeita
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Inmaculada Jorge
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Juan Antonio Lopez
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Nerea Méndez-Barbero
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Simón Méndez-Ferrer
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Miguel Angel Del Pozo
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Borja Ibáñez
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Vicente Andrés
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | | | - Juan Miguel Redondo
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Elena Bonzon-Kulichenko
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Jesús Vázquez
- From the ‡Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| |
Collapse
|
20
|
Chromatin remodeller SMARCA4 recruits topoisomerase 1 and suppresses transcription-associated genomic instability. Nat Commun 2016; 7:10549. [PMID: 26842758 PMCID: PMC4742980 DOI: 10.1038/ncomms10549] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/25/2015] [Indexed: 02/07/2023] Open
Abstract
Topoisomerase 1, an enzyme that relieves superhelical tension, is implicated in transcription-associated mutagenesis and genome instability-associated with neurodegenerative diseases as well as activation-induced cytidine deaminase. From proteomic analysis of TOP1-associated proteins, we identify SMARCA4, an ATP-dependent chromatin remodeller; FACT, a histone chaperone; and H3K4me3, a transcriptionally active chromatin marker. Here we show that SMARCA4 knockdown in a B-cell line decreases TOP1 recruitment to chromatin, and leads to increases in Igh/c-Myc chromosomal translocations, variable and switch region mutations and negative superhelicity, all of which are also observed in response to TOP1 knockdown. In contrast, FACT knockdown inhibits association of TOP1 with H3K4me3, and severely reduces DNA cleavage and Igh/c-Myc translocations, without significant effect on TOP1 recruitment to chromatin. We thus propose that SMARCA4 is involved in the TOP1 recruitment to general chromatin, whereas FACT is required for TOP1 binding to H3K4me3 at non-B DNA containing chromatin for the site-specific cleavage. Topoisomerase 1 (TOP1) relieves superhelical tension when DNA strands are unwound during transcription. Here, Husain et al. report that SMARCA4, an ATP-dependent chromatin remodeller, is associated with TOP1 and suppresses transcription-associated genomic instability.
Collapse
|
21
|
Zenón F, Jorge I, Cruz A, Suárez E, Segarra AC, Vázquez J, Meléndez LM, Serrano H. 18O proteomics reveal increased human apolipoprotein CIII in Hispanic HIV-1+ women with HAART that use cocaine. Proteomics Clin Appl 2015; 10:144-55. [PMID: 26255783 DOI: 10.1002/prca.201400204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 05/26/2015] [Accepted: 07/27/2015] [Indexed: 11/09/2022]
Abstract
PURPOSE Drug abuse is a major risk factor in the development and progression of HIV-1. This study defines the alterations in the plasma proteome of HIV-1-infected women that use cocaine. EXPERIMENTAL DESIGN Plasma samples from 12 HIV-seropositive Hispanic women under antiretroviral therapy were selected for this study. Six sample pairs were matched between nondrug users and cocaine users. After IgG and albumin depletion, SDS-PAGE, and in-gel digestion, peptides from nondrug users and cocaine users were labeled with (16) O and (18) O, respectively, and subjected to LC-MS/MS and quantitation using Proteome Discover and QuiXoT softwares and validated by ELISA. RESULTS A total of 1015 proteins were identified at 1% false discovery rates (FDR). Statistical analyses revealed 13 proteins with significant changes between the two groups, cocaine and noncocaine users (p < 0.05). The great majority pertained to protection defense function and the rest pertained to transport, homeostatic, regulation, and binding of ligands. Apolipoprotein CIII was increased in plasma of HIV+ Hispanic women positive for cocaine compared to HIV+ nondrug users (p ≤ 0.05). CONCLUSIONS AND CLINICAL RELEVANCE Increased human apolipoprotein CIII warrants that these patients be carefully monitored to avoid the increased risk of cardiovascular events associated with HIV, HAART, and cocaine use.
Collapse
Affiliation(s)
- Frances Zenón
- Department of Microbiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Inmaculada Jorge
- Laboratorio de Proteómica Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ailed Cruz
- Department of Biochemistry, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Erick Suárez
- Department of Biostatistics and Epidemiology, Graduate School of Public Health, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Annabell C Segarra
- Department of Physiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Jesús Vázquez
- Laboratorio de Proteómica Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Loyda M Meléndez
- Department of Microbiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Horacio Serrano
- Department of Internal Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| |
Collapse
|
22
|
Strippoli R, Loureiro J, Moreno V, Benedicto I, Pérez Lozano ML, Barreiro O, Pellinen T, Minguet S, Foronda M, Osteso MT, Calvo E, Vázquez J, López Cabrera M, del Pozo MA. Caveolin-1 deficiency induces a MEK-ERK1/2-Snail-1-dependent epithelial-mesenchymal transition and fibrosis during peritoneal dialysis. EMBO Mol Med 2015; 7:102-23. [PMID: 25550395 PMCID: PMC4309670 DOI: 10.15252/emmm.201404127] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Peritoneal dialysis (PD) is a form of renal replacement therapy whose repeated use can alter dialytic function through induction of epithelial–mesenchymal transition (EMT) and fibrosis, eventually leading to PD discontinuation. The peritoneum from Cav1−/− mice showed increased EMT, thickness, and fibrosis. Exposure of Cav1−/− mice to PD fluids further increased peritoneal membrane thickness, altered permeability, and increased the number of FSP-1/cytokeratin-positive cells invading the sub-mesothelial stroma. High-throughput quantitative proteomics revealed increased abundance of collagens, FN, and laminin, as well as proteins related to TGF-β activity in matrices derived from Cav1−/− cells. Lack of Cav1 was associated with hyperactivation of a MEK-ERK1/2-Snail-1 pathway that regulated the Smad2-3/Smad1-5-8 balance. Pharmacological blockade of MEK rescued E-cadherin and ZO-1 inter-cellular junction localization, reduced fibrosis, and restored peritoneal function in Cav1−/− mice. Moreover, treatment of human PD-patient-derived MCs with drugs increasing Cav1 levels, as well as ectopic Cav1 expression, induced re-acquisition of epithelial features. This study demonstrates a pivotal role of Cav1 in the balance of epithelial versus mesenchymal state and suggests targets for the prevention of fibrosis during PD.
Collapse
Affiliation(s)
- Raffaele Strippoli
- Department of Vascular biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain Department of Cellular Biotechnologies and Haematology, Sapienza University of Rome, Rome, Italy
| | - Jesús Loureiro
- Molecular Biology Unit, Hospital Universitario de la Princesa Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - Vanessa Moreno
- Department of Vascular biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Ignacio Benedicto
- Molecular Biology Unit, Hospital Universitario de la Princesa Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - María Luisa Pérez Lozano
- Molecular Biology Unit, Hospital Universitario de la Princesa Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - Olga Barreiro
- Department of Vascular biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Teijo Pellinen
- Department of Vascular biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Susana Minguet
- Department of Vascular biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Miguel Foronda
- Department of Vascular biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Maria Teresa Osteso
- Department of Vascular biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Enrique Calvo
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Vázquez
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel López Cabrera
- Molecular Biology Unit, Hospital Universitario de la Princesa Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - Miguel Angel del Pozo
- Department of Vascular biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
23
|
Ayllón N, Naranjo V, Hajdušek O, Villar M, Galindo RC, Kocan KM, Alberdi P, Šíma R, Cabezas-Cruz A, Rückert C, Bell-Sakyi L, Kazimírová M, Havlíková S, Klempa B, Kopáček P, de la Fuente J. Nuclease Tudor-SN Is Involved in Tick dsRNA-Mediated RNA Interference and Feeding but Not in Defense against Flaviviral or Anaplasma phagocytophilum Rickettsial Infection. PLoS One 2015; 10:e0133038. [PMID: 26186700 PMCID: PMC4506139 DOI: 10.1371/journal.pone.0133038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 06/23/2015] [Indexed: 11/18/2022] Open
Abstract
Tudor staphylococcal nuclease (Tudor-SN) and Argonaute (Ago) are conserved components of the basic RNA interference (RNAi) machinery with a variety of functions including immune response and gene regulation. The RNAi machinery has been characterized in tick vectors of human and animal diseases but information is not available on the role of Tudor-SN in tick RNAi and other cellular processes. Our hypothesis is that tick Tudor-SN is part of the RNAi machinery and may be involved in innate immune response and other cellular processes. To address this hypothesis, Ixodes scapularis and I. ricinus ticks and/or cell lines were used to annotate and characterize the role of Tudor-SN in dsRNA-mediated RNAi, immune response to infection with the rickettsia Anaplasma phagocytophilum and the flaviviruses TBEV or LGTV and tick feeding. The results showed that Tudor-SN is conserved in ticks and involved in dsRNA-mediated RNAi and tick feeding but not in defense against infection with the examined viral and rickettsial pathogens. The effect of Tudor-SN gene knockdown on tick feeding could be due to down-regulation of genes that are required for protein processing and blood digestion through a mechanism that may involve selective degradation of dsRNAs enriched in G:U pairs that form as a result of adenosine-to-inosine RNA editing. These results demonstrated that Tudor-SN plays a role in tick RNAi pathway and feeding but no strong evidence for a role in innate immune responses to pathogen infection was found.
Collapse
Affiliation(s)
- Nieves Ayllón
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
| | - Victoria Naranjo
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Ondrej Hajdušek
- Institute of Parasitology, Biology Centre, Academy of Sciences of the Czech Republic, Branišovská 31, 37005, České Budějovice, The Czech Republic
| | - Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
| | - Ruth C. Galindo
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Katherine M. Kocan
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Pilar Alberdi
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
| | - Radek Šíma
- Institute of Parasitology, Biology Centre, Academy of Sciences of the Czech Republic, Branišovská 31, 37005, České Budějovice, The Czech Republic
| | - Alejandro Cabezas-Cruz
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
- Center for Infection and Immunity of Lille (CIIL), INSERM U1019 –CNRS UMR 8204, Université Lille Nord de France, Institut Pasteur de Lille, Lille, France
| | - Claudia Rückert
- The Pirbright Institute, Ash Road, Pirbright, Woking, GU24 0NF, United Kingdom
| | - Lesley Bell-Sakyi
- The Pirbright Institute, Ash Road, Pirbright, Woking, GU24 0NF, United Kingdom
| | - Mária Kazimírová
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 84506, Bratislava, Slovakia
| | - Sabína Havlíková
- Institute of Virology, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia
| | - Boris Klempa
- Institute of Virology, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia
| | - Petr Kopáček
- Institute of Parasitology, Biology Centre, Academy of Sciences of the Czech Republic, Branišovská 31, 37005, České Budějovice, The Czech Republic
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
24
|
Ayllón N, Villar M, Galindo RC, Kocan KM, Šíma R, López JA, Vázquez J, Alberdi P, Cabezas-Cruz A, Kopáček P, de la Fuente J. Systems biology of tissue-specific response to Anaplasma phagocytophilum reveals differentiated apoptosis in the tick vector Ixodes scapularis. PLoS Genet 2015; 11:e1005120. [PMID: 25815810 PMCID: PMC4376793 DOI: 10.1371/journal.pgen.1005120] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 03/03/2015] [Indexed: 12/20/2022] Open
Abstract
Anaplasma phagocytophilum is an emerging pathogen that causes human granulocytic anaplasmosis. Infection with this zoonotic pathogen affects cell function in both vertebrate host and the tick vector, Ixodes scapularis. Global tissue-specific response and apoptosis signaling pathways were characterized in I. scapularis nymphs and adult female midguts and salivary glands infected with A. phagocytophilum using a systems biology approach combining transcriptomics and proteomics. Apoptosis was selected for pathway-focused analysis due to its role in bacterial infection of tick cells. The results showed tissue-specific differences in tick response to infection and revealed differentiated regulation of apoptosis pathways. The impact of bacterial infection was more pronounced in tick nymphs and midguts than in salivary glands, probably reflecting bacterial developmental cycle. All apoptosis pathways described in other organisms were identified in I. scapularis, except for the absence of the Perforin ortholog. Functional characterization using RNA interference showed that Porin knockdown significantly increases tick colonization by A. phagocytophilum. Infection with A. phagocytophilum produced complex tissue-specific alterations in transcript and protein levels. In tick nymphs, the results suggested a possible effect of bacterial infection on the inhibition of tick immune response. In tick midguts, the results suggested that A. phagocytophilum infection inhibited cell apoptosis to facilitate and establish infection through up-regulation of the JAK/STAT pathway. Bacterial infection inhibited the intrinsic apoptosis pathway in tick salivary glands by down-regulating Porin expression that resulted in the inhibition of Cytochrome c release as the anti-apoptotic mechanism to facilitate bacterial infection. However, tick salivary glands may promote apoptosis to limit bacterial infection through induction of the extrinsic apoptosis pathway. These dynamic changes in response to A. phagocytophilum in I. scapularis tissue-specific transcriptome and proteome demonstrated the complexity of the tick response to infection and will contribute to characterize gene regulation in ticks. The continuous human exploitation of environmental resources and the increase in human outdoor activities, which have allowed for the contact with arthropod vectors normally present in the field, has promoted the emergence and resurgence of vector-borne pathogens. Among these, Anaplasma phagocytophilum is an emerging bacterial pathogen transmitted to humans and other vertebrate hosts by ticks as they take a blood meal that causes human granulocytic anaplasmosis in the United States, Europe and Asia, with increasing numbers of affected people every year. Tick response to pathogen infection has been only partially characterized. In this study, global tissue-specific response and apoptosis signaling pathways were characterized in tick nymphs and adult female midguts and salivary glands infected with A. phagocytophilum using a systems biology approach combining transcriptomics and proteomics. The results demonstrated dramatic and complex tissue-specific response to A. phagocytophilum in the tick vector Ixodes scapularis, which reflected pathogen developmental cycle and the impact on tick apoptosis pathways. These dynamic changes in response to A. phagocytophilum in I. scapularis tissue-specific transcriptome and proteome demonstrated the complexity of the tick response to infection and contributes information on tick-pathogen interactions and for development of novel control strategies for pathogen infection and transmission.
Collapse
Affiliation(s)
- Nieves Ayllón
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ciudad Real, Spain
| | - Margarita Villar
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ciudad Real, Spain
| | - Ruth C. Galindo
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Katherine M. Kocan
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Radek Šíma
- Institute of Parasitology, Biology Centre, Academy of Sciences of the Czech Republic, České Budějovice, The Czech Republic
| | - Juan A. López
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Pilar Alberdi
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ciudad Real, Spain
| | - Alejandro Cabezas-Cruz
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ciudad Real, Spain
- Center for Infection and Immunity of Lille (CIIL), Université Lille Nord de France, Institut Pasteur de Lille, Lille, France
| | - Petr Kopáček
- Institute of Parasitology, Biology Centre, Academy of Sciences of the Czech Republic, České Budějovice, The Czech Republic
| | - José de la Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC, CSIC-UCLM-JCCM, Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
25
|
Fernandez-Sanz C, Ruiz-Meana M, Castellano J, Miro-Casas E, Nuñez E, Inserte J, Vázquez J, Garcia-Dorado D. Altered FoF1 ATP synthase and susceptibility to mitochondrial permeability transition pore during ischaemia and reperfusion in aging cardiomyocytes. Thromb Haemost 2015; 113:441-51. [PMID: 25631625 DOI: 10.1160/th14-10-0901] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/15/2015] [Indexed: 12/27/2022]
Abstract
Aging is a major determinant of the incidence and severity of ischaemic heart disease. Preclinical information suggests the existence of intrinsic cellular alterations that contribute to ischaemic susceptibility in senescent myocardium, by mechanisms not well established. We investigated the role of altered mitochondrial function in the adverse effect of aging. Isolated perfused hearts from old mice (> 20 months) displayed increased ischaemia-reperfusion injury as compared to hearts from adult mice (6 months) despite delayed onset of ischaemic rigor contracture. In cardiomyocytes from aging hearts there was a more rapid decline of mitochondrial membrane potential (Δψm) as compared to young ones, but ischaemic rigor shortening was also delayed. Transient recovery of Δψm observed during ischaemia, secondary to the reversal of mitochondrial FoF1 ATP synthase to ATPase mode, was markedly reduced in aging cardiomyocytes. Proteomic analysis demonstrated increased oxidation of different subunits of ATP synthase. Altered bionergetics in aging cells was associated with reduced mitochondrial calcium uptake and more severe cytosolic calcium overload during ischaemia-reperfusion. Despite attenuated ROS burst and mitochondrial calcium overload, mitochondrial permeability transition pore (mPTP) opening and cell death was increased in reperfused aged cells. In vitro studies demonstrated a significantly reduced calcium retention capacity in interfibrillar mitochondria from aging hearts. Our results identify altered FoF1 ATP synthase and increased sensitivity of mitochondria to undergo mPTP opening as important determinants of the reduced tolerance to ischaemia-reperfusion in aging hearts. Because ATP synthase has been proposed to conform mPTP, it is tempting to hypothesise that oxidation of ATP synthase underlie both phenomena.
Collapse
Affiliation(s)
| | - Marisol Ruiz-Meana
- Marisol Ruiz-Meana, Cardiologia, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron- Universitat Autonoma de Barcelona, Pg Vall d'Hebron 119-129, 08035 Barcelona, Spain, Tel.: +34 93 489 4037, Fax:+34 93 489 4032, E-mail:
| | | | | | | | | | | | | |
Collapse
|
26
|
Bonzon-Kulichenko E, Garcia-Marques F, Trevisan-Herraz M, Vázquez J. Revisiting peptide identification by high-accuracy mass spectrometry: problems associated with the use of narrow mass precursor windows. J Proteome Res 2014; 14:700-10. [PMID: 25494653 DOI: 10.1021/pr5007284] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Peptide identification is increasingly achieved through database searches in which mass precursor tolerance is set in the ppm range. This trend is driven by the high resolution and accuracy of modern mass spectrometers and the belief that the quality of peptide identification is fully controlled by estimating the false discovery rate (FDR) using the decoy-target approach. However, narrowing mass tolerance decreases the number of sequence candidates, and several authors have raised concerns that these search conditions can introduce inaccuracies. Here, we demonstrate that when scores that only depend on one sequence candidate are used, decoy-based estimates of the number of false positive identifications are accurate even with an average number of candidates of just 200, to the point that remarkably accurate FDR predictions can be made in completely different search conditions. However, when scores that are constructed taking information from additional sequence candidates are used together with low precursor mass tolerances, the proportion of peptides incorrectly identified may become significantly higher than the FDR estimated by the target-decoy approach. Our results suggest that with this kind of score the high mass accuracy of modern mass spectrometers should be exploited by using wide mass windows followed by postscoring mass filtering algorithms.
Collapse
Affiliation(s)
- Elena Bonzon-Kulichenko
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC) , Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | | | | | | |
Collapse
|
27
|
Defective sarcoplasmic reticulum-mitochondria calcium exchange in aged mouse myocardium. Cell Death Dis 2014; 5:e1573. [PMID: 25522267 PMCID: PMC4454162 DOI: 10.1038/cddis.2014.526] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 10/21/2014] [Accepted: 11/04/2014] [Indexed: 02/06/2023]
Abstract
Mitochondrial alterations are critically involved in increased vulnerability to disease during aging. We investigated the contribution of mitochondria–sarcoplasmic reticulum (SR) communication in cardiomyocyte functional alterations during aging. Heart function (echocardiography) and ATP/phosphocreatine (NMR spectroscopy) were preserved in hearts from old mice (>20 months) with respect to young mice (5–6 months). Mitochondrial membrane potential and resting O2 consumption were similar in mitochondria from young and old hearts. However, maximal ADP-stimulated O2 consumption was specifically reduced in interfibrillar mitochondria from aged hearts. Second generation proteomics disclosed an increased mitochondrial protein oxidation in advanced age. Because energy production and oxidative status are regulated by mitochondrial Ca2+, we investigated the effect of age on mitochondrial Ca2+ uptake. Although no age-dependent differences were found in Ca2+ uptake kinetics in isolated mitochondria, mitochondrial Ca2+ uptake secondary to SR Ca2+ release was significantly reduced in cardiomyocytes from old hearts, and this effect was associated with decreased NAD(P)H regeneration and increased mitochondrial ROS upon increased contractile activity. Immunofluorescence and proximity ligation assay identified the defective communication between mitochondrial voltage-dependent anion channel and SR ryanodine receptor (RyR) in cardiomyocytes from aged hearts associated with altered Ca2+ handling. Age-dependent alterations in SR Ca2+ transfer to mitochondria and in Ca2+ handling could be reproduced in cardiomyoctes from young hearts after interorganelle disruption with colchicine, at concentrations that had no effect in aged cardiomyocytes or isolated mitochondria. Thus, defective SR–mitochondria communication underlies inefficient interorganelle Ca2+ exchange that contributes to energy demand/supply mistmach and oxidative stress in the aged heart.
Collapse
|
28
|
Groult H, Ruiz-Cabello J, Lechuga-Vieco AV, Mateo J, Benito M, Bilbao I, Martínez-Alcázar MP, Lopez JA, Vázquez J, Herranz FF. Phosphatidylcholine-Coated Iron Oxide Nanomicelles for In Vivo Prolonged Circulation Time with an Antibiofouling Protein Corona. Chemistry 2014; 20:16662-71. [DOI: 10.1002/chem.201404221] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Indexed: 11/08/2022]
|
29
|
Chen D, Shah A, Nguyen H, Loo D, Inder KL, Hill MM. Online quantitative proteomics p-value calculator for permutation-based statistical testing of peptide ratios. J Proteome Res 2014; 13:4184-91. [PMID: 25058807 DOI: 10.1021/pr500525e] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The utility of high-throughput quantitative proteomics to identify differentially abundant proteins en-masse relies on suitable and accessible statistical methodology, which remains mostly an unmet need. We present a free web-based tool, called Quantitative Proteomics p-value Calculator (QPPC), designed for accessibility and usability by proteomics scientists and biologists. Being an online tool, there is no requirement for software installation. Furthermore, QPPC accepts generic peptide ratio data generated by any mass spectrometer and database search engine. Importantly, QPPC utilizes the permutation test that we recently found to be superior to other methods for analysis of peptide ratios because it does not assume normal distributions.1 QPPC assists the user in selecting significantly altered proteins based on numerical fold change, or standard deviation from the mean or median, together with the permutation p-value. Output is in the form of comma separated values files, along with graphical visualization using volcano plots and histograms. We evaluate the optimal parameters for use of QPPC, including the permutation level and the effect of outlier and contaminant peptides on p-value variability. The optimal parameters defined are deployed as default for the web-tool at http://qppc.di.uq.edu.au/ .
Collapse
Affiliation(s)
- David Chen
- School of Information and Communication Technology, Griffith University , 170 Kessels Road, Nathan, Brisbane, Queensland 4111, Australia
| | | | | | | | | | | |
Collapse
|
30
|
O18Quant: a semiautomatic strategy for quantitative analysis of high-resolution 16O/18O labeled data. BIOMED RESEARCH INTERNATIONAL 2014; 2014:971857. [PMID: 24901003 PMCID: PMC4037588 DOI: 10.1155/2014/971857] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/18/2014] [Indexed: 01/09/2023]
Abstract
Proteolytic 18O-labeling has been widely used in quantitative proteomics since it can uniformly label all peptides from different kinds of proteins. There have been multiple algorithms and tools developed over the last few years to analyze high-resolution proteolytic 16O/18O labeled mass spectra. We have developed a software package, O18Quant, which addresses two major issues in the previously developed algorithms. First, O18Quant uses a robust linear model (RLM) for peptide-to-protein ratio estimation. RLM can minimize the effect of outliers instead of iteratively removing them which is a common practice in other approaches. Second, the existing algorithms lack applicable implementation. We address this by implementing O18Quant using C# under Microsoft.net framework and R. O18Quant automatically calculates the peptide/protein relative ratio and provides a friendly graphical user interface (GUI) which allows the user to manually validate the quantification results at scan, peptide, and protein levels. The intuitive GUI of O18Quant can greatly enhance the user's visualization and understanding of the data analysis. O18Quant can be downloaded for free as part of the software suite ProteomicsTools.
Collapse
|
31
|
Jorge I, Burillo E, Mesa R, Baila-Rueda L, Moreno M, Trevisan-Herraz M, Silla-Castro JC, Camafeita E, Ortega-Muñoz M, Bonzon-Kulichenko E, Calvo I, Cenarro A, Civeira F, Vázquez J. The human HDL proteome displays high inter-individual variability and is altered dynamically in response to angioplasty-induced atheroma plaque rupture. J Proteomics 2014; 106:61-73. [PMID: 24747125 DOI: 10.1016/j.jprot.2014.04.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/03/2014] [Accepted: 04/07/2014] [Indexed: 12/28/2022]
Abstract
Recent findings support potential roles for HDL in cardiovascular pathophysiology not related to lipid metabolism. We address whether HDL proteome is dynamically altered in atheroma plaque rupture. We used immunoaffinity purification of HDL samples from coronary artery disease patients before and after percutaneous transluminal coronary angioplasty (PTCA), a model of atheroma plaque disruption. Samples were analyzed by quantitative proteomics using stable isotope labeling and results were subjected to statistical analysis of protein variance using a novel algorithm. We observed high protein variability in HDL composition between individuals, indicating that HDL protein composition is highly patient-specific. However, intra-individual protein variances remained at low levels, confirming the reproducibility of the method used for HDL isolation and protein quantification. A systems biology analysis of HDL protein alterations induced by PTCA revealed an increase in two protein clusters that included several apolipoproteins, fibrinogen-like protein 1 and other intracellular proteins, and a decrease in antithrombin-III, annexin A1 and several immunoglobulins. Our results support the concept of HDL as dynamic platforms that donate and receive a variety of molecules and provide an improved methodology to use HDL proteome for the systematic analysis of differences among individuals and the search for cardiovascular biomarkers. Biological significance The HDL proteome is an interesting model of clinical relevance and has been previously described to be dynamically altered in response to pathophysiological conditions and cardiovascular diseases. Our study suggests that interindividual variability of HDL proteome is higher than previously thought and provided the detection of a set of proteins that changed their abundance in response to plaque rupture, supporting the concept of HDL as dynamic platforms that donate and receive a variety of molecules.
Collapse
Affiliation(s)
- Inmaculada Jorge
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Elena Burillo
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain; Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria de Aragón, I+CS, Zaragoza, Spain
| | - Raquel Mesa
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Lucía Baila-Rueda
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria de Aragón, I+CS, Zaragoza, Spain
| | - Margoth Moreno
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Marco Trevisan-Herraz
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Juan Carlos Silla-Castro
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Emilio Camafeita
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | - Elena Bonzon-Kulichenko
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Isabel Calvo
- Hospital Universitario Miguel Servet, Servicio de Cardiología, Zaragoza, Spain
| | - Ana Cenarro
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria de Aragón, I+CS, Zaragoza, Spain
| | - Fernando Civeira
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria de Aragón, I+CS, Zaragoza, Spain
| | - Jesús Vázquez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.
| |
Collapse
|
32
|
Navarro P, Trevisan-Herraz M, Bonzon-Kulichenko E, Núñez E, Martínez-Acedo P, Pérez-Hernández D, Jorge I, Mesa R, Calvo E, Carrascal M, Hernáez ML, García F, Bárcena JA, Ashman K, Abian J, Gil C, Redondo JM, Vázquez J. General statistical framework for quantitative proteomics by stable isotope labeling. J Proteome Res 2014; 13:1234-47. [PMID: 24512137 DOI: 10.1021/pr4006958] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The combination of stable isotope labeling (SIL) with mass spectrometry (MS) allows comparison of the abundance of thousands of proteins in complex mixtures. However, interpretation of the large data sets generated by these techniques remains a challenge because appropriate statistical standards are lacking. Here, we present a generally applicable model that accurately explains the behavior of data obtained using current SIL approaches, including (18)O, iTRAQ, and SILAC labeling, and different MS instruments. The model decomposes the total technical variance into the spectral, peptide, and protein variance components, and its general validity was demonstrated by confronting 48 experimental distributions against 18 different null hypotheses. In addition to its general applicability, the performance of the algorithm was at least similar than that of other existing methods. The model also provides a general framework to integrate quantitative and error information fully, allowing a comparative analysis of the results obtained from different SIL experiments. The model was applied to the global analysis of protein alterations induced by low H₂O₂ concentrations in yeast, demonstrating the increased statistical power that may be achieved by rigorous data integration. Our results highlight the importance of establishing an adequate and validated statistical framework for the analysis of high-throughput data.
Collapse
Affiliation(s)
- Pedro Navarro
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM , 28049 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ischemic preconditioning protects cardiomyocyte mitochondria through mechanisms independent of cytosol. J Mol Cell Cardiol 2014; 68:79-88. [PMID: 24434643 DOI: 10.1016/j.yjmcc.2014.01.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/03/2014] [Indexed: 12/22/2022]
Abstract
Mitochondria play a central role in the protection conferred by ischemic preconditioning (IP) by not fully elucidated mechanisms. We investigated whether IP protects mitochondria against ischemia-reperfusion (IR) injury through mechanisms independent of cytosolic signaling. In isolated rat hearts, sublethal IR increased superoxide production and reduced complex-I- and II-mediated respiration in subsarcolemmal (SS), but not interfibrillar (IF) mitochondria. This effect of IR on mitochondrial respiration was significantly attenuated by IP. Similar results were obtained in isolated cardiac mitochondria subjected to in vitro IR. The reduction in SS mitochondrial respiration in the heart and in vitro model was paralleled by an increase in oxidized cysteine residues, which was also prevented by IP. IP was also protective in mitochondria submitted to lethal IR. The protective effect of IP against respiratory failure was unaffected by inhibition of mitochondrial KATP channels or mitochondrial permeability transition. However, IP protection was lost in mitochondria from genetically-modified animals in which connexin-43, a protein present in SS but not IF mitochondria, was replaced by connexin-32. Our results demonstrate the existence of a protective mitochondrial mechanism or "mitochondrial preconditioning" independent of cytosol that confers protection against IR-induced respiratory failure and oxidative damage, and requires connexin-43.
Collapse
|
34
|
Quantitative proteomics analysis of high-density lipoproteins by stable 18O-isotope labeling. Methods Mol Biol 2013; 1000:139-56. [PMID: 23585090 DOI: 10.1007/978-1-62703-405-0_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
For the large-scale study of dynamic proteomes, quantitative proteomic approaches based on stable isotope labeling and mass spectrometry (MS) have been developed as a high-throughput, reproducible, and robust alternative to conventional gel-based techniques. In this chapter, we describe in detail a quantitative proteomic strategy based on HDL isolation by affinity chromatography, in-gel trypsin digestion of protein extracts, peptide (18)O labeling, separation by off-gel isoelectric focusing, and peptide analysis on a linear ion trap mass spectrometer, followed by the application of a robust multilayered statistical model. This protocol is of universal applicability and has been successfully applied to the global characterization of the HDL proteome with some specific considerations for this particle, paving the way to the in-depth study of the protein cargo of HDL and its implication in cardiovascular diseases.
Collapse
|
35
|
Hernandez-Fernaud JR, Reid SE, Neilson LJ, Zanivan S. Quantitative mass spectrometry-based proteomics in angiogenesis. Proteomics Clin Appl 2013; 7:464-76. [PMID: 23161605 DOI: 10.1002/prca.201200055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 10/13/2012] [Accepted: 10/25/2012] [Indexed: 12/29/2022]
Abstract
The process of new blood vessel formation from pre-existing ones is called angiogenesis. Beyond playing a critical role in the physiological development of the vascular system, angiogenesis is a well-recognised hallmark of cancer. Unbiased system-wide approaches are required to complement the current knowledge, and intimately understand the molecular mechanisms regulating this process in physiological and pathological conditions. In this review we describe the cellular and molecular dynamics regulating the physiological growth of vessels and their deregulation in cancer, survey in vitro and in vivo models currently exploited to investigate various aspects of angiogenesis and describe state-of-the-art and most widespread methods and technologies in MS shotgun proteomics. Finally, we focus on current applications of MS to better understand endothelial cell behaviour and propose how modern proteomics can impact on angiogenesis research.
Collapse
|
36
|
Mendes M, Pérez-Hernandez D, Vázquez J, Coelho AV, Cunha C. Proteomic changes in HEK-293 cells induced by hepatitis delta virus replication. J Proteomics 2013; 89:24-38. [PMID: 23770296 DOI: 10.1016/j.jprot.2013.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/21/2013] [Accepted: 06/04/2013] [Indexed: 02/07/2023]
Abstract
UNLABELLED Hepatitis delta virus (HDV) infection greatly increases the risk of hepatocellular carcinoma in hepatitis B virus chronically infected patients. HDV is highly dependent on host factors for accomplishment of the replication cycle. However, these factors are largely unknown and the mechanisms involved in the pathogenicity of the virus still remain elusive. Here, we made use of the HEK-293 cell line, which was engineered in order to mimic HDV replication. Five different proteomes were analyzed and compared using a MS-based quantitative proteomics approach by (18)O/(16)O stable isotope labeling. About 3000 proteins were quantified and 89 found to be differentially expressed as a consequence HDV RNA replication. The down-regulation of p53 , HSPE, and ELAV as well as the up-regulation of Transportin 1 , EIF3D, and Cofilin 1 were validated by Western blot. A systems biology approach was additionally used to analyze altered pathways and networks. The G2/M DNA damage checkpoint and pyruvate metabolism were among the most affected pathways, and Cancer was the most likely disease associated to HDV replication. Western blot analysis allowed identifying 14-3-3 σ interactor as down-regulated protein acting in the G2/M cell cycle control checkpoint. This evidence supports deregulation of G2/M checkpoint as a possible mechanism involved in the promotion of HDV associated hepatocellular carcinoma. BIOLOGICAL SIGNIFICANCE This manuscript provides a description of changes observed in the cellular proteome that arise as result of expression of the hepatitis delta virus (HDV) antigen as well as virus genome replication. Using a systems biology approach cancer was found to be the most probable disease associated with HDV replication. Additionally, results show that HDV alters the regulation of G2/M cell cycle control checkpoint. Taken together, our data provide new insights into probable mechanisms associated with the increased incidence of hepatocellular carcinoma observed in HDV infected patients.
Collapse
Affiliation(s)
- Marta Mendes
- Unidade de Microbiologia Médica, Centro de Malária e outras Doenças Tropicais, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisboa, Portugal
| | | | | | | | | |
Collapse
|
37
|
Isern J, Martín-Antonio B, Ghazanfari R, Martín A, López J, del Toro R, Sánchez-Aguilera A, Arranz L, Martín-Pérez D, Suárez-Lledó M, Marín P, Van Pel M, Fibbe W, Vázquez J, Scheding S, Urbano-Ispizúa Á, Méndez-Ferrer S. Self-Renewing Human Bone Marrow Mesenspheres Promote Hematopoietic Stem Cell Expansion. Cell Rep 2013; 3:1714-24. [DOI: 10.1016/j.celrep.2013.03.041] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 02/28/2013] [Accepted: 03/27/2013] [Indexed: 12/11/2022] Open
|
38
|
Perez-Hernandez D, Gutiérrez-Vázquez C, Jorge I, López-Martín S, Ursa A, Sánchez-Madrid F, Vázquez J, Yáñez-Mó M. The intracellular interactome of tetraspanin-enriched microdomains reveals their function as sorting machineries toward exosomes. J Biol Chem 2013; 288:11649-61. [PMID: 23463506 DOI: 10.1074/jbc.m112.445304] [Citation(s) in RCA: 379] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Extracellular vesicles are emerging as a potent mechanism of intercellular communication because they can systemically exchange genetic and protein material between cells. Tetraspanin molecules are commonly used as protein markers of extracellular vesicles, although their role in the unexplored mechanisms of cargo selection into exosomes has not been addressed. For that purpose, we have characterized the intracellular tetraspanin-enriched microdomain (TEM) interactome by high throughput mass spectrometry, in both human lymphoblasts and their derived exosomes, revealing a clear pattern of interaction networks. Proteins interacting with TEM receptors cytoplasmic regions presented a considerable degree of overlap, although some highly specific CD81 tetraspanin ligands, such as Rac GTPase, were detected. Quantitative proteomics showed that TEM ligands account for a great proportion of the exosome proteome and that a selective repertoire of CD81-associated molecules, including Rac, is not correctly routed to exosomes in cells from CD81-deficient animals. Our data provide evidence that insertion into TEM may be necessary for protein inclusion into the exosome structure.
Collapse
Affiliation(s)
- Daniel Perez-Hernandez
- Laboratory of Protein Chemistry and Proteomics, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, E-28049 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Rodríguez-Suárez E, Whetton AD. The application of quantification techniques in proteomics for biomedical research. MASS SPECTROMETRY REVIEWS 2013; 32:1-26. [PMID: 22847841 DOI: 10.1002/mas.21347] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 02/09/2012] [Accepted: 02/10/2012] [Indexed: 06/01/2023]
Abstract
The systematic analysis of biological processes requires an understanding of the quantitative expression patterns of proteins, their interacting partners and their subcellular localization. This information was formerly difficult to accrue as the relative quantification of proteins relied on antibody-based methods and other approaches with low throughput. The advent of soft ionization techniques in mass spectrometry plus advances in separation technologies has aligned protein systems biology with messenger RNA, DNA, and microarray technologies to provide data on systems as opposed to singular protein entities. Another aspect of quantitative proteomics that increases its importance for the coming few years is the significant technical developments underway both for high pressure liquid chromatography and mass spectrum devices. Hence, robustness, reproducibility and mass accuracy are still improving with every new generation of instruments. Nonetheless, the methods employed require validation and comparison to design fit for purpose experiments in advanced protein analyses. This review considers the newly developed systematic protein investigation methods and their value from the standpoint that relative or absolute protein quantification is required de rigueur in biomedical research.
Collapse
|
40
|
Tejera E, Rocha-Perugini V, López-Martín S, Pérez-Hernández D, Bachir AI, Horwitz AR, Vázquez J, Sánchez-Madrid F, Yáñez-Mo M. CD81 regulates cell migration through its association with Rac GTPase. Mol Biol Cell 2012; 24:261-73. [PMID: 23264468 PMCID: PMC3564539 DOI: 10.1091/mbc.e12-09-0642] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Data presented here provide evidence for a new direct interaction of the GTPase Rac with the C-terminal cytoplasmic domain of tetraspanin CD81. Tetraspanin-enriched, microdomain-dependent compartmentalization is a novel regulatory mechanism of Rac activity turnover, which provides a novel mechanism for regulation of cell motility by tetraspanins. CD81 is a member of the tetraspanin family that has been described to have a key role in cell migration of tumor and immune cells. To unravel the mechanisms of CD81-regulated cell migration, we performed proteomic analyses that revealed an interaction of the tetraspanin C-terminal domain with the small GTPase Rac. Direct interaction was confirmed biochemically. Moreover, microscopy cross-correlation analysis demonstrated the in situ integration of both molecules into the same molecular complex. Pull-down experiments revealed that CD81-Rac interaction was direct and independent of Rac activation status. Knockdown of CD81 resulted in enhanced protrusion rate, altered focal adhesion formation, and decreased cell migration, correlating with increased active Rac. Reexpression of wild-type CD81, but not its truncated form lacking the C-terminal cytoplasmic domain, rescued these effects. The phenotype of CD81 knockdown cells was mimicked by treatment with a soluble peptide with the C-terminal sequence of the tetraspanin. Our data show that the interaction of Rac with the C-terminal cytoplasmic domain of CD81 is a novel regulatory mechanism of the GTPase activity turnover. Furthermore, they provide a novel mechanism for tetraspanin-dependent regulation of cell motility and open new avenues for tetraspanin-targeted reagents by the use of cell-permeable peptides.
Collapse
Affiliation(s)
- Emilio Tejera
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria La Princesa, 2006 Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Mascaraque V, Hernáez ML, Jiménez-Sánchez M, Hansen R, Gil C, Martín H, Cid VJ, Molina M. Phosphoproteomic analysis of protein kinase C signaling in Saccharomyces cerevisiae reveals Slt2 mitogen-activated protein kinase (MAPK)-dependent phosphorylation of eisosome core components. Mol Cell Proteomics 2012; 12:557-74. [PMID: 23221999 DOI: 10.1074/mcp.m112.020438] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The cell wall integrity (CWI) pathway of the model organism Saccharomyces cerevisiae has been thoroughly studied as a paradigm of the mitogen-activated protein kinase (MAPK) pathway. It consists of a classic MAPK module comprising the Bck1 MAPK kinase kinase, two redundant MAPK kinases (Mkk1 and Mkk2), and the Slt2 MAPK. This module is activated under a variety of stimuli related to cell wall homeostasis by Pkc1, the only member of the protein kinase C family in budding yeast. Quantitative phosphoproteomics based on stable isotope labeling of amino acids in cell culture is a powerful tool for globally studying protein phosphorylation. Here we report an analysis of the yeast phosphoproteome upon overexpression of a PKC1 hyperactive allele that specifically activates CWI MAPK signaling in the absence of external stimuli. We found 82 phosphopeptides originating from 43 proteins that showed enhanced phosphorylation in these conditions. The MAPK S/T-P target motif was significantly overrepresented in these phosphopeptides. Hyperphosphorylated proteins provide putative novel targets of the Pkc1-cell wall integrity pathway involved in diverse functions such as the control of gene expression, protein synthesis, cytoskeleton maintenance, DNA repair, and metabolism. Remarkably, five components of the plasma-membrane-associated protein complex known as eisosomes were found among the up-regulated proteins. We show here that Pkc1-induced phosphorylation of the eisosome core components Pil1 and Lsp1 was not exerted directly by Pkc1, but involved signaling through the Slt2 MAPK module.
Collapse
Affiliation(s)
- Victoria Mascaraque
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid and Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Plaza de Ramón y Cajal s/n, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Zainal-Abidin Z, Veith PD, Dashper SG, Zhu Y, Catmull DV, Chen YY, Heryanto DC, Chen D, Pyke JS, Tan K, Mitchell HL, Reynolds EC. Differential proteomic analysis of a polymicrobial biofilm. J Proteome Res 2012; 11:4449-64. [PMID: 22808953 DOI: 10.1021/pr300201c] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia exist in a polymicrobial biofilm associated with chronic periodontitis. The aim of this study was to culture these three species as a polymicrobial biofilm and to determine proteins important for bacterial interactions. In a flow cell all three species attached and grew as a biofilm; however, after 90 h of culture P. gingivalis and T. denticola were closely associated and dominated the polymicrobial biofilm. For comparison, planktonic cultures of P. gingivalis and T. denticola were grown separately in continuous culture. Whole cell lysates were subjected to SDS-PAGE, followed by in-gel proteolytic H₂¹⁶O/H₂¹⁸O labeling. From two replicates, 135 and 174 P. gingivalis proteins and 134 and 194 T. denticola proteins were quantified by LC-MALDI TOF/TOF MS. The results suggest a change of strategy in iron acquisition by P. gingivalis due to large increases in the abundance of HusA and HusB in the polymicrobial biofilm while HmuY and other iron/haem transport systems decreased. Significant changes in the abundance of peptidases and enzymes involved in glutamate and glycine catabolism suggest syntrophy. These data indicate an intimate association between P. gingivalis and T. denticola in a biofilm that may play a role in disease pathogenesis.
Collapse
Affiliation(s)
- Zamirah Zainal-Abidin
- Oral Health CRC, Melbourne Dental School and the Bio21 Institute, The University of Melbourne, Victoria 3010, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Cunningham R, Ma D, Li L. Mass Spectrometry-based Proteomics and Peptidomics for Systems Biology and Biomarker Discovery. FRONTIERS IN BIOLOGY 2012; 7:313-335. [PMID: 24504115 PMCID: PMC3913178 DOI: 10.1007/s11515-012-1218-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The scientific community has shown great interest in the field of mass spectrometry-based proteomics and peptidomics for its applications in biology. Proteomics technologies have evolved to produce large datasets of proteins or peptides involved in various biological and disease progression processes producing testable hypothesis for complex biological questions. This review provides an introduction and insight to relevant topics in proteomics and peptidomics including biological material selection, sample preparation, separation techniques, peptide fragmentation, post-translation modifications, quantification, bioinformatics, and biomarker discovery and validation. In addition, current literature and remaining challenges and emerging technologies for proteomics and peptidomics are presented.
Collapse
Affiliation(s)
- Robert Cunningham
- Department of Chemistry, University of Wisconsin-Madison, 777, Highland Avenue, Madison, WI 53705-2222, USA
| | - Di Ma
- School of Pharmacy, University of Wisconsin-Madison, 777, Highland Avenue, Madison, WI 53705-2222, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, 777, Highland Avenue, Madison, WI 53705-2222, USA
- School of Pharmacy, University of Wisconsin-Madison, 777, Highland Avenue, Madison, WI 53705-2222, USA
| |
Collapse
|
44
|
Gautier V, Mouton-Barbosa E, Bouyssié D, Delcourt N, Beau M, Girard JP, Cayrol C, Burlet-Schiltz O, Monsarrat B, Gonzalez de Peredo A. Label-free quantification and shotgun analysis of complex proteomes by one-dimensional SDS-PAGE/NanoLC-MS: evaluation for the large scale analysis of inflammatory human endothelial cells. Mol Cell Proteomics 2012; 11:527-39. [PMID: 22518033 PMCID: PMC3412980 DOI: 10.1074/mcp.m111.015230] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 04/10/2012] [Indexed: 12/21/2022] Open
Abstract
To perform differential studies of complex protein mixtures, strategies for reproducible and accurate quantification are needed. Here, we evaluated a quantitative proteomic workflow based on nanoLC-MS/MS analysis on an LTQ-Orbitrap-VELOS mass spectrometer and label-free quantification using the MFPaQ software. In such label-free quantitative studies, a compromise has to be found between two requirements: repeatability of sample processing and MS measurements, allowing an accurate quantification, and high proteomic coverage of the sample, allowing quantification of minor species. The latter is generally achieved through sample fractionation, which may induce experimental bias during the label-free comparison of samples processed, and analyzed independently. In this work, we wanted to evaluate the performances of MS intensity-based label-free quantification when a complex protein sample is fractionated by one-dimensional SDS-PAGE. We first tested the efficiency of the analysis without protein fractionation and could achieve quite good quantitative repeatability in single-run analysis (median coefficient of variation of 5%, 99% proteins with coefficient of variation <48%). We show that sample fractionation by one-dimensional SDS-PAGE is associated with a moderate decrease of quantitative measurement repeatability while largely improving the depth of proteomic coverage. We then applied the method for a large scale proteomic study of the human endothelial cell response to inflammatory cytokines, such as TNFα, interferon γ, and IL1β, which allowed us to finely decipher at the proteomic level the biological pathways involved in endothelial cell response to proinflammatory cytokines.
Collapse
Affiliation(s)
- Violette Gautier
- From ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France, and
- §Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France
| | - Emmanuelle Mouton-Barbosa
- §Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France
| | - David Bouyssié
- From ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France, and
- §Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France
| | - Nicolas Delcourt
- §Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France
| | - Mathilde Beau
- From ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France, and
- §Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France
| | - Jean-Philippe Girard
- From ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France, and
- §Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France
| | - Corinne Cayrol
- From ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France, and
- §Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France
| | - Odile Burlet-Schiltz
- From ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France, and
- §Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France
| | - Bernard Monsarrat
- From ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France, and
- §Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France
| | - Anne Gonzalez de Peredo
- From ‡Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France, and
- §Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, F-31077 Toulouse, France
| |
Collapse
|
45
|
Gordón-Alonso M, Sala-Valdés M, Rocha-Perugini V, Pérez-Hernández D, López-Martín S, Ursa A, Alvarez S, Kolesnikova TV, Vázquez J, Sánchez-Madrid F, Yáñez-Mó M. EWI-2 association with α-actinin regulates T cell immune synapses and HIV viral infection. THE JOURNAL OF IMMUNOLOGY 2012; 189:689-700. [PMID: 22689882 DOI: 10.4049/jimmunol.1103708] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
EWI motif-containing protein 2 (EWI-2) is a member of the Ig superfamily that links tetraspanin-enriched microdomains to the actin cytoskeleton. We found that EWI-2 colocalizes with CD3 and CD81 at the central supramolecular activation cluster of the T cell immune synapse. Silencing of the endogenous expression or overexpression of a cytoplasmic truncated mutant of EWI-2 in T cells increases IL-2 secretion upon Ag stimulation. Mass spectrometry experiments of pull-downs with the C-term intracellular domain of EWI-2 revealed the specific association of EWI-2 with the actin-binding protein α-actinin; this association was regulated by PIP2. α-Actinin regulates the immune synapse formation and is required for efficient T cell activation. We extended these observations to virological synapses induced by HIV and found that silencing of either EWI-2 or α-actinin-4 increased cell infectivity. Our data suggest that the EWI-2-α-actinin complex is involved in the regulation of the actin cytoskeleton at T cell immune and virological synapses, providing a link between membrane microdomains and the formation of polarized membrane structures involved in T cell recognition.
Collapse
|
46
|
Martínez-Acedo P, Núñez E, Gómez FJS, Moreno M, Ramos E, Izquierdo-Álvarez A, Miró-Casas E, Mesa R, Rodriguez P, Martínez-Ruiz A, Dorado DG, Lamas S, Vázquez J. A novel strategy for global analysis of the dynamic thiol redox proteome. Mol Cell Proteomics 2012; 11:800-13. [PMID: 22647871 DOI: 10.1074/mcp.m111.016469] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Nitroxidative stress in cells occurs mainly through the action of reactive nitrogen and oxygen species (RNOS) on protein thiol groups. Reactive nitrogen and oxygen species-mediated protein modifications are associated with pathophysiological states, but can also convey physiological signals. Identification of Cys residues that are modified by oxidative stimuli still poses technical challenges and these changes have never been statistically analyzed from a proteome-wide perspective. Here we show that GELSILOX, a method that combines a robust proteomics protocol with a new computational approach that analyzes variance at the peptide level, allows a simultaneous analysis of dynamic alterations in the redox state of Cys sites and of protein abundance. GELSILOX permits the characterization of the major endothelial redox targets of hydrogen peroxide in endothelial cells and reveals that hypoxia induces a significant increase in the status of oxidized thiols. GELSILOX also detected thiols that are redox-modified by ischemia-reperfusion in heart mitochondria and demonstrated that these alterations are abolished in ischemia-preconditioned animals.
Collapse
Affiliation(s)
- Pablo Martínez-Acedo
- Centro de Biología Molecular Severo Ochoa, Nicolás Cabrera 1, 28049 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Starkey JM, Tilton RG. Proteomics and systems biology for understanding diabetic nephropathy. J Cardiovasc Transl Res 2012; 5:479-90. [PMID: 22581264 DOI: 10.1007/s12265-012-9372-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 05/01/2012] [Indexed: 01/07/2023]
Abstract
Like many diseases, diabetic nephropathy is defined in a histopathological context and studied using reductionist approaches that attempt to ameliorate structural changes. Novel technologies in mass spectrometry-based proteomics have the ability to provide a deeper understanding of the disease beyond classical histopathology, redefine the characteristics of the disease state, and identify novel approaches to reduce renal failure. The goal is to translate these new definitions into improved patient outcomes through diagnostic, prognostic, and therapeutic tools. Here, we review progress made in studying the proteomics of diabetic nephropathy and provide an introduction to the informatics tools used in the analysis of systems biology data, while pointing out statistical issues for consideration. Novel bioinformatics methods may increase biomarker identification, and other tools, including selective reaction monitoring, may hasten clinical validation.
Collapse
Affiliation(s)
- Jonathan M Starkey
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555-1060, USA
| | | |
Collapse
|
48
|
Characterization of the tick-pathogen interface by quantitative proteomics. Ticks Tick Borne Dis 2012; 3:154-8. [PMID: 22647712 DOI: 10.1016/j.ttbdis.2012.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 02/03/2012] [Accepted: 02/09/2012] [Indexed: 11/23/2022]
Abstract
Ticks are vectors of pathogens that affect human and animal health worldwide. Ticks and the pathogens they transmit have co-evolved molecular interactions involving genetic traits of both the tick and the pathogen that mediate their development and survival. Proteomics and genomics studies of infected ticks are required to understand tick-pathogen interactions and identify potential vaccine antigens to control tick infestations and pathogen transmission. In this paper, the application of quantitative proteomics to characterize differential protein expression in ticks and cultured tick cells in response to pathogen infection is reviewed. Analyses using (a) two-dimensional differential in gel electrophoresis (DIGE) labeling and (b) protein one-step in gel digestion, peptide iTRAQ labeling, and isoelectric focusing fractionation, both followed by peptide and protein identifications by mass spectrometry resulted in the identification of host, pathogen, and tick proteins differentially expressed in response to infection. Although at its infancy, these results showed that quantitative proteomics is a powerful approach to characterize the tick-pathogen interface and demonstrated pathogen and tick-specific differences in protein expression in ticks and cultured tick cells in response to pathogen infection.
Collapse
|
49
|
Bonzon-Kulichenko E, Martínez-Martínez S, Trevisan-Herraz M, Navarro P, Redondo JM, Vázquez J. Quantitative in-depth analysis of the dynamic secretome of activated Jurkat T-cells. J Proteomics 2011; 75:561-71. [PMID: 21920478 DOI: 10.1016/j.jprot.2011.08.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 07/22/2011] [Accepted: 08/24/2011] [Indexed: 10/17/2022]
|
50
|
Gao Y, Gopee NV, Howard PC, Yu LR. Proteomic analysis of early response lymph node proteins in mice treated with titanium dioxide nanoparticles. J Proteomics 2011; 74:2745-59. [PMID: 21884834 PMCID: PMC3215788 DOI: 10.1016/j.jprot.2011.08.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 08/09/2011] [Accepted: 08/15/2011] [Indexed: 10/17/2022]
Abstract
Human exposure to nanoparticles is inevitable from natural and anthropogenic sources. Titanium dioxide (TiO2) nanoparticles are increasingly being used in pharmaceutical and cosmetic products. Previous studies revealed that TiO2 levels were significantly increased in tissues (e.g., lymph nodes) after mice were injected with nanosized TiO2. To identify early response lymph node proteins to TiO2 nanoparticles, groups of mice were intradermally injected with a low dose of DeGussa P25 TiO2 nanoparticles or vehicle alone. The proteomes of lymph nodes at 24 h were quantitatively analyzed using trypsin-catalyzed 16O/18O labeling in conjunction with two-dimensional liquid chromatography separation and tandem mass spectrometry (2DLC-MS/MS). A total of 33 proteins were significantly changed (over 1.3-fold, p<0.05) in the mice treated with TiO2 nanoparticles, which accounted for approximately 1% of the total proteins identified. The differentially expressed proteins mainly involve the immune response (e.g., inflammation), lipid and fatty acid metabolism, mRNA processing, and nucleosome assembly. Regulation of functionally distinct classes of proteins could be mediated by estrogen receptor (ESR1), PPARγ, and c-Myc signalings, etc. The differentially expressed proteins identified in this experiment could represent early response proteins to TiO2 nanoparticle treatment in mouse lymph nodes.
Collapse
Affiliation(s)
- Yuan Gao
- Center of Excellence for Proteomics, Division of Systems Biology, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA
| | - Neera V. Gopee
- Division of Veterinary Services, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA
| | - Paul C. Howard
- Division of Biochemical Toxicology and Office of Scientific Coordination, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA
| | - Li-Rong Yu
- Center of Excellence for Proteomics, Division of Systems Biology, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA
| |
Collapse
|