1
|
Alexovič M, Uličná C, Sabo J, Davalieva K. Human peripheral blood mononuclear cells as a valuable source of disease-related biomarkers: Evidence from comparative proteomics studies. Proteomics Clin Appl 2024; 18:e2300072. [PMID: 37933719 DOI: 10.1002/prca.202300072] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/08/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023]
Abstract
PURPOSE The discovery of specific and sensitive disease-associated biomarkers for early diagnostic purposes of many diseases is still highly challenging due to various complex molecular mechanisms triggered, high variability of disease-related interactions, and an overlap of manifestations among diseases. Human peripheral blood mononuclear cells (PBMCs) contain protein signatures corresponding to essential immunological interplay. Certain diseases stimulate PBMCs and contribute towards modulation of their proteome which can be effectively identified and evaluated via the comparative proteomics approach. EXPERIMENTAL DESIGN In this review, we made a detailed survey of the PBMCS-derived protein biomarker candidates for a variety of diseases, published in the last 15 years. Articles were preselected to include only comparative proteomics studies. RESULTS PBMC-derived biomarkers were investigated for cancer, glomerular, neurodegenerative/neurodevelopmental, psychiatric, chronic inflammatory, autoimmune, endocrinal, infectious, and other diseases. A detailed review of these studies encompassed the proteomics platforms, proposed candidate biomarkers, their immune cell type specificity, and potential clinical application. CONCLUSIONS Overall, PBMCs have shown a solid potential in giving early diagnostic and prognostic biomarkers for many diseases. The future of PBMC biomarker research should reveal its full potential through well-designed comparative studies and extensive testing of the most promising protein biomarkers identified so far.
Collapse
Affiliation(s)
- Michal Alexovič
- Department of Medical and Clinical Biophysics, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovakia
| | - Csilla Uličná
- Department of Medical and Clinical Biophysics, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovakia
| | - Ján Sabo
- Department of Medical and Clinical Biophysics, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovakia
| | - Katarina Davalieva
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, Skopje, North Macedonia
| |
Collapse
|
2
|
Álvarez I, Tirado-Herranz A, Alvarez-Palomo B, Osete JR, Edel MJ. Proteomic Analysis of Human iPSC-Derived Neural Stem Cells and Motor Neurons Identifies Proteasome Structural Alterations. Cells 2023; 12:2800. [PMID: 38132120 PMCID: PMC10742145 DOI: 10.3390/cells12242800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Proteins targeted by the ubiquitin proteasome system (UPS) are identified for degradation by the proteasome, which has been implicated in the development of neurodegenerative diseases. Major histocompatibility complex (MHC) molecules present peptides broken down by the proteasome and are involved in neuronal plasticity, regulating the synapse number and axon regeneration in the central or peripheral nervous system during development and in brain diseases. The mechanisms governing these effects are mostly unknown, but evidence from different compartments of the cerebral cortex indicates the presence of immune-like MHC receptors in the central nervous system. METHODS We used human induced pluripotent stem cells (iPSCs) differentiated into neural stem cells and then into motor neurons as a developmental model to better understand the structure of the proteasome in developing motor neurons. We performed a proteomic analysis of starting human skin fibroblasts, their matching iPSCs, differentiated neural stem cells and motor neurons that highlighted significant differences in the constitutive proteasome and immunoproteasome subunits during development toward motor neurons from iPSCs. RESULTS The proteomic analysis showed that the catalytic proteasome subunits expressed in fibroblasts differed from those in the neural stem cells and motor neurons. Western blot analysis confirmed the proteomic data, particularly the decreased expression of the β5i (PSMB8) subunit immunoproteasome in MNs compared to HFFs and increased β5 (PSMB5) in MNs compared to HFFs. CONCLUSION The constitutive proteasome subunits are upregulated in iPSCs and NSCs from HFFs. Immunoproteasome subunit β5i expression is higher in MNs than NSCs; however, overall, there is more of a constitutive proteasome structure in MNs when comparing HFFs to MNs. The proteasome composition may have implications for motor neuron development and neurodevelopmental diseases that warrant further investigation.
Collapse
Affiliation(s)
- Iñaki Álvarez
- Departament de Biologia Cellular, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Fisiologia i Immunologia, 08193 Barcelona, Spain; (I.Á.); (A.T.-H.)
| | - Adrián Tirado-Herranz
- Departament de Biologia Cellular, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Fisiologia i Immunologia, 08193 Barcelona, Spain; (I.Á.); (A.T.-H.)
| | - Belén Alvarez-Palomo
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain;
| | - Jordi Requena Osete
- Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, 4956 Oslo, Norway
| | - Michael J. Edel
- Department of Anatomy and Embryology, Faculty of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Discipline of Medical Sciences and Genetics, School of Biomedical Sciences, University of Western Australia, Perth 6009, Australia
| |
Collapse
|
3
|
Nakayama M, Marchi H, Dmitrieva AM, Chakraborty A, Merl-Pham J, Hennen E, Le Gleut R, Ruppert C, Guenther A, Kahnert K, Behr J, Hilgendorff A, Hauck SM, Adler H, Staab-Weijnitz CA. Quantitative proteomics of differentiated primary bronchial epithelial cells from chronic obstructive pulmonary disease and control identifies potential novel host factors post-influenza A virus infection. Front Microbiol 2023; 13:957830. [PMID: 36713229 PMCID: PMC9875134 DOI: 10.3389/fmicb.2022.957830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) collectively refers to chronic and progressive lung diseases that cause irreversible limitations in airflow. Patients with COPD are at high risk for severe respiratory symptoms upon influenza virus infection. Airway epithelial cells provide the first-line antiviral defense, but whether or not their susceptibility and response to influenza virus infection changes in COPD have not been elucidated. Therefore, this study aimed to compare the susceptibility of COPD- and control-derived airway epithelium to the influenza virus and assess protein changes during influenza virus infection by quantitative proteomics. Materials and methods The presence of human- and avian-type influenza A virus receptor was assessed in control and COPD lung sections as well as in fully differentiated primary human bronchial epithelial cells (phBECs) by lectin- or antibody-based histochemical staining. PhBECs were from COPD lungs, including cells from moderate- and severe-stage diseases, and from age-, sex-, smoking, and history-matched control lung specimens. Protein profiles pre- and post-influenza virus infection in vitro were directly compared using quantitative proteomics, and selected findings were validated by qRT-PCR and immunoblotting. Results The human-type influenza receptor was more abundant in human airways than the avian-type influenza receptor, a property that was retained in vitro when differentiating phBECs at the air-liquid interface. Proteomics of phBECs pre- and post-influenza A virus infection with A/Puerto Rico/8/34 (PR8) revealed no significant differences between COPD and control phBECs in terms of flu receptor expression, cell type composition, virus replication, or protein profile pre- and post-infection. Independent of health state, a robust antiviral response to influenza virus infection was observed, as well as upregulation of several novel influenza virus-regulated proteins, including PLSCR1, HLA-F, CMTR1, DTX3L, and SHFL. Conclusion COPD- and control-derived phBECs did not differ in cell type composition, susceptibility to influenza virus infection, and proteomes pre- and post-infection. Finally, we identified novel influenza A virus-regulated proteins in bronchial epithelial cells that might serve as potential targets to modulate the pathogenicity of infection and acute exacerbations.
Collapse
Affiliation(s)
- Misako Nakayama
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M BioArchive, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany,Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Hannah Marchi
- Core Facility Statistical Consulting, Helmholtz Zentrum München, Munich, Germany,Faculty of Business Administration and Economics, Bielefeld University, Bielefeld, Germany
| | - Anna M. Dmitrieva
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Ashesh Chakraborty
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M BioArchive, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Juliane Merl-Pham
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Neuherberg, Germany
| | - Elisabeth Hennen
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M BioArchive, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Ronan Le Gleut
- Core Facility Statistical Consulting, Helmholtz Zentrum München, Munich, Germany
| | - Clemens Ruppert
- Department of Internal Medicine, Medizinische Klinik II, Member of the German Center of Lung Research (DZL), Giessen, Germany
| | - Andreas Guenther
- Department of Internal Medicine, Medizinische Klinik II, Member of the German Center of Lung Research (DZL), Giessen, Germany
| | - Kathrin Kahnert
- Department of Medicine V, Ludwig Maximilian University (LMU) Munich, Member of the German Center of Lung Research, University Hospital, Munich, Germany
| | - Jürgen Behr
- Department of Medicine V, Ludwig Maximilian University (LMU) Munich, Member of the German Center of Lung Research, University Hospital, Munich, Germany
| | - Anne Hilgendorff
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M BioArchive, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Stefanie M. Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Neuherberg, Germany
| | - Heiko Adler
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany,Institute of Asthma and Allergy Prevention, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany,*Correspondence: Heiko Adler,
| | - Claudia A. Staab-Weijnitz
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M BioArchive, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany,Claudia A. Staab-Weijnitz, ; https://orcid.org/0000-0002-1211-7834
| |
Collapse
|
4
|
Orth MF, Surdez D, Faehling T, Ehlers AC, Marchetto A, Grossetête S, Volckmann R, Zwijnenburg DA, Gerke JS, Zaidi S, Alonso J, Sastre A, Baulande S, Sill M, Cidre-Aranaz F, Ohmura S, Kirchner T, Hauck SM, Reischl E, Gymrek M, Pfister SM, Strauch K, Koster J, Delattre O, Grünewald TGP. Systematic multi-omics cell line profiling uncovers principles of Ewing sarcoma fusion oncogene-mediated gene regulation. Cell Rep 2022; 41:111761. [PMID: 36476851 DOI: 10.1016/j.celrep.2022.111761] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 08/25/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Ewing sarcoma (EwS) is characterized by EWSR1-ETS fusion transcription factors converting polymorphic GGAA microsatellites (mSats) into potent neo-enhancers. Although the paucity of additional mutations makes EwS a genuine model to study principles of cooperation between dominant fusion oncogenes and neo-enhancers, this is impeded by the limited number of well-characterized models. Here we present the Ewing Sarcoma Cell Line Atlas (ESCLA), comprising whole-genome, DNA methylation, transcriptome, proteome, and chromatin immunoprecipitation sequencing (ChIP-seq) data of 18 cell lines with inducible EWSR1-ETS knockdown. The ESCLA shows hundreds of EWSR1-ETS-targets, the nature of EWSR1-ETS-preferred GGAA mSats, and putative indirect modes of EWSR1-ETS-mediated gene regulation, converging in the duality of a specific but plastic EwS signature. We identify heterogeneously regulated EWSR1-ETS-targets as potential prognostic EwS biomarkers. Our freely available ESCLA (http://r2platform.com/escla/) is a rich resource for EwS research and highlights the power of comprehensive datasets to unravel principles of heterogeneous gene regulation by chimeric transcription factors.
Collapse
Affiliation(s)
- Martin F Orth
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, 80337 Munich, Germany
| | - Didier Surdez
- INSERM Unit 830 "Genetics and Biology of Cancers," Institut Curie Research Center, 75005 Paris, France; Balgrist University Hospital, Faculty of Medicine, University of Zürich, 8008 Zürich, Switzerland
| | - Tobias Faehling
- Hopp Children's Cancer Center (KiTZ), 69120 Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Anna C Ehlers
- Hopp Children's Cancer Center (KiTZ), 69120 Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Aruna Marchetto
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, 80337 Munich, Germany
| | - Sandrine Grossetête
- INSERM Unit 830 "Genetics and Biology of Cancers," Institut Curie Research Center, 75005 Paris, France
| | - Richard Volckmann
- Department of Oncogenomics, Amsterdam University Medical Centers (AUMC), 1105 Amsterdam, the Netherlands
| | - Danny A Zwijnenburg
- Department of Oncogenomics, Amsterdam University Medical Centers (AUMC), 1105 Amsterdam, the Netherlands
| | - Julia S Gerke
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, 80337 Munich, Germany
| | - Sakina Zaidi
- INSERM Unit 830 "Genetics and Biology of Cancers," Institut Curie Research Center, 75005 Paris, France
| | - Javier Alonso
- Unidad de Tumores Sólidos Infantiles, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CB06/07/1009, CIBERER-ISCIII), 28029 Madrid, Spain
| | - Ana Sastre
- Unidad Hemato-oncología Pediátrica, Hospital Infantil Universitario La Paz, 28029 Madrid, Spain
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, 75005 Paris, France
| | - Martin Sill
- Hopp Children's Cancer Center (KiTZ), 69120 Heidelberg, Germany; Division of Pediatric Neuro-Oncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Florencia Cidre-Aranaz
- Hopp Children's Cancer Center (KiTZ), 69120 Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Shunya Ohmura
- Hopp Children's Cancer Center (KiTZ), 69120 Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Thomas Kirchner
- Institute of Pathology, Faculty of Medicine, LMU Munich, 80337 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, 80337 Munich, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Eva Reischl
- Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Melissa Gymrek
- Division of Genetics, Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA; Department of Computer Science and Engineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Stefan M Pfister
- Hopp Children's Cancer Center (KiTZ), 69120 Heidelberg, Germany; Division of Pediatric Neuro-Oncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Department of Pediatric Hematology & Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Konstantin Strauch
- Institute of Medical Biometry, Epidemiology, and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Jan Koster
- Department of Oncogenomics, Amsterdam University Medical Centers (AUMC), 1105 Amsterdam, the Netherlands
| | - Olivier Delattre
- INSERM Unit 830 "Genetics and Biology of Cancers," Institut Curie Research Center, 75005 Paris, France
| | - Thomas G P Grünewald
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, 80337 Munich, Germany; Hopp Children's Cancer Center (KiTZ), 69120 Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| |
Collapse
|
5
|
Zauhar R, Biber J, Jabri Y, Kim M, Hu J, Kaplan L, Pfaller AM, Schäfer N, Enzmann V, Schlötzer-Schrehardt U, Straub T, Hauck SM, Gamlin PD, McFerrin MB, Messinger J, Strang CE, Curcio CA, Dana N, Pauly D, Grosche A, Li M, Stambolian D. As in Real Estate, Location Matters: Cellular Expression of Complement Varies Between Macular and Peripheral Regions of the Retina and Supporting Tissues. Front Immunol 2022; 13:895519. [PMID: 35784369 PMCID: PMC9240314 DOI: 10.3389/fimmu.2022.895519] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/11/2022] [Indexed: 01/02/2023] Open
Abstract
The cellular events that dictate the initiation of the complement pathway in ocular degeneration, such as age-related macular degeneration (AMD), is poorly understood. Using gene expression analysis (single cell and bulk), mass spectrometry, and immunohistochemistry, we dissected the role of multiple retinal and choroidal cell types in determining the complement homeostasis. Our scRNA-seq data show that the cellular response to early AMD is more robust in the choroid, particularly in fibroblasts, pericytes and endothelial cells. In late AMD, complement changes were more prominent in the retina especially with the expression of the classical pathway initiators. Notably, we found a spatial preference for these differences. Overall, this study provides insights into the heterogeneity of cellular responses for complement expression and the cooperation of neighboring cells to complete the pathway in healthy and AMD eyes. Further, our findings provide new cellular targets for therapies directed at complement.
Collapse
Affiliation(s)
- Randy Zauhar
- Department of Chemistry and Biochemistry, The University of the Sciences in Philadelphia, Philadelphia, PA, United States
| | - Josef Biber
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Yassin Jabri
- Department of Ophthalmology, University Hospital Regensburg, Regensburg, Germany
| | - Mijin Kim
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jian Hu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Lew Kaplan
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Anna M. Pfaller
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Nicole Schäfer
- Department of Ophthalmology, University Hospital Regensburg, Regensburg, Germany
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, Regensburg, Germany
| | - Volker Enzmann
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Tobias Straub
- Bioinformatics Unit, Biomedical Center, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Stefanie M. Hauck
- Metabolomics and Proteomics Core and Research Unit Protein Science, Helmholtz-Zentrum München, Neuherberg, Germany
| | - Paul D. Gamlin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Michael B. McFerrin
- Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jeffrey Messinger
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Christianne E. Strang
- Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Nicholas Dana
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Diana Pauly
- Department of Ophthalmology, University Hospital Regensburg, Regensburg, Germany
- Experimental Ophthalmology, University of Marburg, Marburg, Germany
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Dwight Stambolian
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
6
|
Suomi T, Elo LL. Statistical and machine learning methods to study human CD4+ T cell proteome profiles. Immunol Lett 2022; 245:8-17. [DOI: 10.1016/j.imlet.2022.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 11/05/2022]
|
7
|
Alexovič M, Lindner JR, Bober P, Longuespée R, Sabo J, Davalieva K. Human peripheral blood mononuclear cells: A review of recent proteomic applications. Proteomics 2022; 22:e2200026. [PMID: 35348286 DOI: 10.1002/pmic.202200026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 11/07/2022]
Abstract
Human peripheral blood mononuclear cells (PBMCs) represent a sentinel blood sample which reacts to different pathophysiological stimuli in the form of immunological responses/immunophenotypic changes. The study of molecular content of PBMCs can provide better understanding of immune processes giving the possibility of monitoring the health conditions of the host organism. Proteomic analysis of PBMCs can achieve mentioned goal as important immune-related biomarkers are easily accessible for analysis. PBMCs have been gaining attention in different research areas including preclinical or clinical investigations. In this review, recent applications of proteomic analysis of PBMCs are described and discussed. Approaches are divided based on different proteomic workflows such as in-gel, in-solution and on-filter modes. The effect of various diseases such as autoimmune, cancer, neurodegenerative, viral, metabolic, and various immune stimulations such as radiation, vaccine, corticosteroids over PBMCs proteome, are described with emphasis on promising protein biomarker candidates. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Michal Alexovič
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P.J. Šafárik in Košice, Košice, Slovakia
| | - Joshua Raoul Lindner
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Bober
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P.J. Šafárik in Košice, Košice, Slovakia
| | - Rémi Longuespée
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Ján Sabo
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P.J. Šafárik in Košice, Košice, Slovakia
| | - Katarina Davalieva
- Research Centre for Genetic Engineering and Biotechnology, "Georgi D Efremov", Macedonian Academy of Sciences and Arts, USA
| |
Collapse
|
8
|
MS4A15 drives ferroptosis resistance through calcium-restricted lipid remodeling. Cell Death Differ 2022; 29:670-686. [PMID: 34663908 PMCID: PMC8901757 DOI: 10.1038/s41418-021-00883-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 01/07/2023] Open
Abstract
Ferroptosis is an iron-dependent form of cell death driven by biochemical processes that promote oxidation within the lipid compartment. Calcium (Ca2+) is a signaling molecule in diverse cellular processes such as migration, neurotransmission, and cell death. Here, we uncover a crucial link between ferroptosis and Ca2+ through the identification of the novel tetraspanin MS4A15. MS4A15 localizes to the endoplasmic reticulum, where it blocks ferroptosis by depleting luminal Ca2+ stores and reprogramming membrane phospholipids to ferroptosis-resistant species. Specifically, prolonged Ca2+ depletion inhibits lipid elongation and desaturation, driving lipid droplet dispersion and formation of shorter, more saturated ether lipids that protect phospholipids from ferroptotic reactive species. We further demonstrate that increasing luminal Ca2+ levels can preferentially sensitize refractory cancer cell lines. In summary, MS4A15 regulation of anti-ferroptotic lipid reservoirs provides a key resistance mechanism that is distinct from antioxidant and lipid detoxification pathways. Manipulating Ca2+ homeostasis offers a compelling strategy to balance cellular lipids and cell survival in ferroptosis-associated diseases.
Collapse
|
9
|
Konigsberg IR, Borie R, Walts AD, Cardwell J, Rojas M, Metzger F, Hauck SM, Fingerlin TE, Yang IV, Schwartz DA. Molecular Signatures of Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2021; 65:430-441. [PMID: 34038697 PMCID: PMC8525208 DOI: 10.1165/rcmb.2020-0546oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/24/2021] [Indexed: 11/24/2022] Open
Abstract
Molecular patterns and pathways in idiopathic pulmonary fibrosis (IPF) have been extensively investigated, but few studies have assimilated multiomic platforms to provide an integrative understanding of molecular patterns that are relevant in IPF. Herein, we combine the coding and noncoding transcriptomes, DNA methylomes, and proteomes from IPF and healthy lung tissue to identify molecules and pathways associated with this disease. RNA sequencing, Illumina MethylationEPIC array, and liquid chromatography-mass spectrometry proteomic data were collected on lung tissue from 24 subjects with IPF and 14 control subjects. Significant differential features were identified by using linear models adjusting for age and sex, inflation, and bias when appropriate. Data Integration Analysis for Biomarker Discovery Using a Latent Component Method for Omics Studies was used for integrative multiomic analysis. We identified 4,643 differentially expressed transcripts aligning to 3,439 genes, 998 differentially abundant proteins, 2,500 differentially methylated regions, and 1,269 differentially expressed long noncoding RNAs (lncRNAs) that were significant after correcting for multiple tests (false discovery rate < 0.05). Unsupervised hierarchical clustering using 20 coding mRNA, protein, methylation, and lncRNA features with the highest loadings on the top latent variable from the four data sets demonstrates perfect separation of IPF and control lungs. Our analysis confirmed previously validated molecules and pathways known to be dysregulated in disease and implicated novel molecular features as potential drivers and modifiers of disease. For example, 4 proteins, 18 differentially methylated regions, and 10 lncRNAs were found to have strong correlations (|r| > 0.8) with MMP7 (matrix metalloproteinase 7). Therefore, by using a system biology approach, we have identified novel molecular relationships in IPF.
Collapse
Affiliation(s)
- Iain R. Konigsberg
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - Raphael Borie
- Department of Medicine, Bichat Hospital, Paris, France
| | - Avram D. Walts
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - Jonathan Cardwell
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - Mauricio Rojas
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Fabian Metzger
- Research Unit for Protein Science, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; and
| | - Stefanie M. Hauck
- Research Unit for Protein Science, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; and
| | - Tasha E. Fingerlin
- Department of Immunology and Genomic Medicine and Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado
| | - Ivana V. Yang
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - David A. Schwartz
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| |
Collapse
|
10
|
Schuster M, Tewary G, Bao X, Subedi P, Hauck SM, Olsen AK, Eide DM, Trott KR, Götz S, Atkinson MJ, Rosemann M. In vitro cellular and proteome assays identify Wnt pathway and CDKN2A-regulated senescence affected in mesenchymal stem cells from mice after a chronic LD gamma irradiation in utero. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2021; 60:397-410. [PMID: 34287697 PMCID: PMC8310520 DOI: 10.1007/s00411-021-00925-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 07/07/2021] [Indexed: 06/13/2023]
Abstract
Reliable data on the effects of chronic prenatal exposure to low dose (LD) of ionizing radiation in humans are missing. There are concerns about adverse long-term effects that may persist throughout postnatal life of the offspring. Due to their slow cell cycle kinetics and life-long residence time in the organism, mesenchymal stem cells (MSCs) are more susceptible to low level genotoxic stress caused by extrinsic multiple LD events. The aim of this study was to investigate the effect of chronic, prenatal LD gamma irradiation to the biology of MSCs later in life. C3H mice were exposed in utero to chronic prenatal irradiation of 10 mGy/day over a period of 3 weeks. Two years later, MSCs were isolated from the bone marrow and analyzed in vitro for their radiosensitivity, for cellular senescence and for DNA double-strand break recognition after a second acute gamma-irradiation. In addition to these cellular assays, changes in protein expression were measured using HPLC-MS/MS and dysregulated molecular signaling pathways identified using bioinformatics. We observed radiation-induced proteomic changes in MSCs from the offspring of in utero irradiated mice (leading to ~ 9.4% of all detected proteins being either up- or downregulated) as compared to non-irradiated controls. The proteomic changes map to regulation pathways involved in the extracellular matrix, the response to oxidative stress, and the Wnt signaling pathway. In addition, chronic prenatal LD irradiation lead to an increased rate of in vitro radiation-induced senescence later in life and to an increased number of residual DNA double-strand breaks after 4 Gy irradiation, indicating a remarkable interaction of in vivo radiation in combination with a second acute dose of in vitro radiation. This study provides the first insight into a molecular mechanism of persistent MSC damage response by ionizing radiation exposure during prenatal time and will help to predict therapeutic safety and efficacy with respect to a clinical application of stem cells.
Collapse
Affiliation(s)
- Martina Schuster
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Gargi Tewary
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Xuanwen Bao
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Prabal Subedi
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), 80939, Munich, Germany
| | - Ann Karin Olsen
- Department of Molecular Biology/Domain for Infection Control and Environmental Health, Norwegian Institute of Public Health, Lovisenberggt. 8, 0456, Oslo, Norway
| | - Dag Markus Eide
- Department of Molecular Biology/Domain for Infection Control and Environmental Health, Norwegian Institute of Public Health, Lovisenberggt. 8, 0456, Oslo, Norway
| | - Klaus Rüdiger Trott
- Chair of Radiation Biology, Technical University Munich (TUM), 80333, Munich, Germany
| | - Sebastian Götz
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Michael J Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
- Chair of Radiation Biology, Technical University Munich (TUM), 80333, Munich, Germany
| | - Michael Rosemann
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany.
- Medical Graduate School, Technical University Munich (TUM), 80333, Munich, Germany.
| |
Collapse
|
11
|
Miljanovic N, Hauck SM, van Dijk RM, Di Liberto V, Rezaei A, Potschka H. Proteomic signature of the Dravet syndrome in the genetic Scn1a-A1783V mouse model. Neurobiol Dis 2021; 157:105423. [PMID: 34144125 DOI: 10.1016/j.nbd.2021.105423] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 05/14/2021] [Accepted: 06/08/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Dravet syndrome is a rare, severe pediatric epileptic encephalopathy associated with intellectual and motor disabilities. Proteomic profiling in a mouse model of Dravet syndrome can provide information about the molecular consequences of the genetic deficiency and about pathophysiological mechanisms developing during the disease course. METHODS A knock-in mouse model of Dravet syndrome with Scn1a haploinsufficiency was used for whole proteome, seizure, and behavioral analysis. Hippocampal tissue was dissected from two- (prior to epilepsy manifestation) and four- (following epilepsy manifestation) week-old male mice and analyzed using LC-MS/MS with label-free quantification. Proteomic data sets were subjected to bioinformatic analysis including pathway enrichment analysis. The differential expression of selected proteins was confirmed by immunohistochemical staining. RESULTS The findings confirmed an increased susceptibility to hyperthermia-associated seizures, the development of spontaneous seizures, and behavioral alterations in the novel Scn1a-A1873V mouse model of Dravet syndrome. As expected, proteomic analysis demonstrated more pronounced alterations following epilepsy manifestation. In particular, proteins involved in neurotransmitter dynamics, receptor and ion channel function, synaptic plasticity, astrogliosis, neoangiogenesis, and nitric oxide signaling showed a pronounced regulation in Dravet mice. Pathway enrichment analysis identified several significantly regulated pathways at the later time point, with pathways linked to synaptic transmission and glutamatergic signaling dominating the list. CONCLUSION In conclusion, the whole proteome analysis in a mouse model of Dravet syndrome demonstrated complex molecular alterations in the hippocampus. Some of these alterations may have an impact on excitability or may serve a compensatory function, which, however, needs to be further confirmed by future investigations. The proteomic data indicate that, due to the molecular consequences of the genetic deficiency, the pathophysiological mechanisms may become more complex during the course of the disease. As a result, the management of Dravet syndrome may need to consider further molecular and cellular alterations. Ensuing functional follow-up studies, this data set may provide valuable guidance for the future development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Nina Miljanovic
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Germany
| | - R Maarten van Dijk
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Valentina Di Liberto
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Ali Rezaei
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany.
| |
Collapse
|
12
|
Sagmeister S, Merl-Pham J, Petrera A, Deeg CA, Hauck SM. High glucose treatment promotes extracellular matrix proteome remodeling in Mller glial cells. PeerJ 2021; 9:e11316. [PMID: 34046254 PMCID: PMC8139267 DOI: 10.7717/peerj.11316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/30/2021] [Indexed: 11/20/2022] Open
Abstract
Background The underlying pathomechanisms in diabetic retinopathy (DR) remain incompletely understood. The aim of this study was to add to the current knowledge about the particular role of retinal Mller glial cells (RMG) in the initial processes of DR. Methods Applying a quantitative proteomic workflow, we investigated changes of primary porcine RMG under short term high glucose treatment as well as glycolysis inhibition treatment. Results We revealed significant changes in RMG proteome primarily in proteins building the extracellular matrix (ECM) indicating fundamental remodeling processes of ECM as novel rapid response to high glucose treatment. Among others, Osteopontin (SPP1) as well as its interacting integrins were significantly downregulated and organotypic retinal explant culture confirmed the selective loss of SPP1 in RMG upon treatment. Since SPP1 in the retina has been described neuroprotective for photoreceptors and functions against experimentally induced cell swelling, its rapid loss under diabetic conditions may point to a direct involvement of RMG to the early neurodegenerative processes driving DR. Data are available via ProteomeXchange with identifier PXD015879.
Collapse
Affiliation(s)
- Sandra Sagmeister
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Martinsried, Germany.,Research Unit Protein Science and Metabolomics and Proteomics Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Munich, Germany
| | - Juliane Merl-Pham
- Research Unit Protein Science and Metabolomics and Proteomics Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Munich, Germany
| | - Agnese Petrera
- Research Unit Protein Science and Metabolomics and Proteomics Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Munich, Germany
| | - Cornelia A Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Martinsried, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Munich, Germany
| |
Collapse
|
13
|
Bühler L, Maida A, Vogl ES, Georgiadi A, Takacs A, Kluth O, Schürmann A, Feuchtinger A, von Toerne C, Tsokanos FF, Klepac K, Wolff G, Sakurai M, Ekim Üstünel B, Nawroth P, Herzig S. Lipocalin 13 enhances insulin secretion but is dispensable for systemic metabolic control. Life Sci Alliance 2021; 4:4/4/e202000898. [PMID: 33536239 PMCID: PMC7898469 DOI: 10.26508/lsa.202000898] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/12/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Thorough preclinical evaluation reveals a negligible role of lipocalin 13 in systemic glucose and lipid metabolism. Members of the lipocalin protein family serve as biomarkers for kidney disease and acute phase inflammatory reactions, and are under preclinical development for the diagnosis and therapy of allergies. However, none of the lipocalin family members has made the step into clinical development, mostly due to their complex biological activity and the lack of in-depth mechanistic knowledge. Here, we show that the hepatokine lipocalin 13 (LCN13) triggers glucose-dependent insulin secretion and cell proliferation of primary mouse islets. However, inhibition of endogenous LCN13 expression in lean mice did not alter glucose and lipid homeostasis. Enhanced hepatic secretion of LCN13 in either diet-induced or genetic obesity led to no discernible impact on systemic glucose and lipid metabolism, neither in preventive nor therapeutic setting. Of note, loss or forced LCN13 hepatic secretion did not trigger any compensatory regulation of related lipocalin family members. Together, these data are in stark contrast to the suggested gluco-regulatory and therapeutic role of LCN13 in obesity, and imply complex regulatory steps in LCN13 biology at the organismic level mitigating its principal insulinotropic effects.
Collapse
Affiliation(s)
- Lea Bühler
- Institute for Diabetes and Cancer (IDC), Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Adriano Maida
- Institute for Diabetes and Cancer (IDC), Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Elena Sophie Vogl
- Institute for Diabetes and Cancer (IDC), Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Anastasia Georgiadi
- Institute for Diabetes and Cancer (IDC), Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Andrea Takacs
- Institute for Diabetes and Cancer (IDC), Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Oliver Kluth
- German Center for Diabetes Research (DZD), Neuherberg, Germany.,Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), Neuherberg, Germany.,Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany.,Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christine von Toerne
- Research Unit Protein Science, Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Foivos-Filippos Tsokanos
- Institute for Diabetes and Cancer (IDC), Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Katarina Klepac
- Institute for Diabetes and Cancer (IDC), Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Gretchen Wolff
- Institute for Diabetes and Cancer (IDC), Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Minako Sakurai
- Institute for Diabetes and Cancer (IDC), Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Bilgen Ekim Üstünel
- Institute for Diabetes and Cancer (IDC), Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Peter Nawroth
- Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Centre Munich, German Research Center for Environmental Health, Neuherberg, Germany .,Joint Heidelberg-IDC Transnational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,Chair Molecular Metabolic Control, Medical Faculty, Technical University Munich, Munich, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
14
|
Mathai SK, Cardwell J, Metzger F, Powers J, Walts AD, Kropski JA, Eickelberg O, Hauck SM, Yang IV, Schwartz DA. Preclinical Pulmonary Fibrosis Circulating Protein Biomarkers. Am J Respir Crit Care Med 2021; 202:1720-1724. [PMID: 32755483 PMCID: PMC7737579 DOI: 10.1164/rccm.202003-0724le] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Susan K Mathai
- University of Colorado School of Medicine Aurora, Colorado.,Baylor University Medical Center at Dallas Dallas, Texas
| | | | | | - Julia Powers
- University of Colorado School of Medicine Aurora, Colorado
| | - Avram D Walts
- University of Colorado School of Medicine Aurora, Colorado
| | | | | | | | - Ivana V Yang
- University of Colorado School of Medicine Aurora, Colorado
| | | |
Collapse
|
15
|
Giese IM, Schilloks MC, Degroote RL, Weigand M, Renner S, Wolf E, Hauck SM, Deeg CA. Chronic Hyperglycemia Drives Functional Impairment of Lymphocytes in Diabetic INSC94Y Transgenic Pigs. Front Immunol 2021; 11:607473. [PMID: 33552065 PMCID: PMC7862560 DOI: 10.3389/fimmu.2020.607473] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
People with diabetes mellitus have an increased risk for infections, however, there is still a critical gap in precise knowledge about altered immune mechanisms in this disease. Since diabetic INSC94Y transgenic pigs exhibit elevated blood glucose and a stable diabetic phenotype soon after birth, they provide a favorable model to explore functional alterations of immune cells in an early stage of diabetes mellitus in vivo. Hence, we investigated peripheral blood mononuclear cells (PBMC) of these diabetic pigs compared to non-diabetic wild-type littermates. We found a 5-fold decreased proliferative response of T cells in INSC94Y tg pigs to polyclonal T cell mitogen phytohemagglutinin (PHA). Using label-free LC-MS/MS, a total of 3,487 proteins were quantified, and distinct changes in protein abundances in CD4+ T cells of early-stage diabetic pigs were detectable. Additionally, we found significant increases in mitochondrial oxygen consumption rate (OCR) and higher basal glycolytic activity in PBMC of diabetic INSC94Y tg pigs, indicating an altered metabolic immune cell phenotype. Thus, our study provides new insights into molecular mechanisms of dysregulated immune cells triggered by permanent hyperglycemia.
Collapse
Affiliation(s)
- Isabella-Maria Giese
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | | | - Roxane L. Degroote
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Maria Weigand
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Simone Renner
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Stefanie M. Hauck
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Protein Science, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Munich, Germany
| | - Cornelia A. Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| |
Collapse
|
16
|
Yang L, Kraft VAN, Pfeiffer S, Merl‐Pham J, Bao X, An Y, Hauck SM, Schick JA. Nonsense-mediated decay factor SMG7 sensitizes cells to TNFα-induced apoptosis via CYLD tumor suppressor and the noncoding oncogene Pvt1. Mol Oncol 2020; 14:2420-2435. [PMID: 32602581 PMCID: PMC7530794 DOI: 10.1002/1878-0261.12754] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Nonsense-mediated decay (NMD) proteins are responsible for the surveillance and degradation of aberrant RNAs. Suppressor with morphogenetic effect on genitalia 7 (SMG7) is an NMD complex protein and a regulator of tumor necrosis factor (TNF)-induced extrinsic apoptosis; however, this unique function has not been explored in detail. In this study, we show that loss of Smg7 leads to unrestricted expression of long noncoding RNAs (lncRNAs) in addition to NMD targets. Functional analysis of Smg7-/- cells showed downregulation of the tumor suppressor cylindromatosis (CYLD) and diminished caspase activity, thereby switching cells to nuclear factor-κB (NF-κB)-mediated protection. This positive relationship between SMG7 and CYLD was found to be widely conserved in human cancer cell lines and renal carcinoma samples from The Cancer Genome Atlas. In addition to CYLD suppression, upregulation of lncRNAs Pvt1 and Adapt33 rendered cells resistant to TNF, while pharmacologic inhibition of NF-κB in Pvt1-overexpressing TNF-resistant cells and Smg7-deficient spheroids re-established TNF-induced lethality. Thus, loss of SMG7 decouples regulation of two separate oncogenic factors with cumulative downstream effects on the NF-κB pathway. The data highlight a novel and specific regulation of oncogenic factors by SMG7 and pinpoint a composite tumor suppressor role in response to TNF.
Collapse
Affiliation(s)
- Limeng Yang
- Genetics and Cellular Engineering GroupInstitute of Molecular Toxicology and PharmacologyHelmholtz Zentrum Munich GmbHGerman Research Center for Environmental HealthNeuherbergGermany
| | - Vanessa A. N. Kraft
- Genetics and Cellular Engineering GroupInstitute of Molecular Toxicology and PharmacologyHelmholtz Zentrum Munich GmbHGerman Research Center for Environmental HealthNeuherbergGermany
| | - Susanne Pfeiffer
- Genetics and Cellular Engineering GroupInstitute of Molecular Toxicology and PharmacologyHelmholtz Zentrum Munich GmbHGerman Research Center for Environmental HealthNeuherbergGermany
| | - Juliane Merl‐Pham
- Research Unit Protein ScienceHelmholtz Zentrum Munich GmbHGerman Research Center for Environmental HealthNeuherbergGermany
| | - Xuanwen Bao
- Institute of Radiation BiologyHelmholtz Zentrum Munich GmbHGerman Research Center for Environmental HealthNeuherbergGermany
| | - Yu An
- Department of Chinese MedicineNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Stefanie M. Hauck
- Research Unit Protein ScienceHelmholtz Zentrum Munich GmbHGerman Research Center for Environmental HealthNeuherbergGermany
| | - Joel A. Schick
- Genetics and Cellular Engineering GroupInstitute of Molecular Toxicology and PharmacologyHelmholtz Zentrum Munich GmbHGerman Research Center for Environmental HealthNeuherbergGermany
| |
Collapse
|
17
|
Henke C, Töllner K, van Dijk RM, Miljanovic N, Cordes T, Twele F, Bröer S, Ziesak V, Rohde M, Hauck SM, Vogel C, Welzel L, Schumann T, Willmes DM, Kurzbach A, El-Agroudy NN, Bornstein SR, Schneider SA, Jordan J, Potschka H, Metallo CM, Köhling R, Birkenfeld AL, Löscher W. Disruption of the sodium-dependent citrate transporter SLC13A5 in mice causes alterations in brain citrate levels and neuronal network excitability in the hippocampus. Neurobiol Dis 2020; 143:105018. [PMID: 32682952 DOI: 10.1016/j.nbd.2020.105018] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/08/2020] [Accepted: 07/11/2020] [Indexed: 12/28/2022] Open
Abstract
In addition to tissues such as liver, the plasma membrane sodium-dependent citrate transporter, NaCT (SLC13A5), is highly expressed in brain neurons, but its function is not understood. Loss-of-function mutations in the human SLC13A5 gene have been associated with severe neonatal encephalopathy and pharmacoresistant seizures. The molecular mechanisms of these neurological alterations are not clear. We performed a detailed examination of a Slc13a5 deletion mouse model including video-EEG monitoring, behavioral tests, and electrophysiologic, proteomic, and metabolomic analyses of brain and cerebrospinal fluid. The experiments revealed an increased propensity for epileptic seizures, proepileptogenic neuronal excitability changes in the hippocampus, and significant citrate alterations in the CSF and brain tissue of Slc13a5 deficient mice, which may underlie the neurological abnormalities. These data demonstrate that SLC13A5 is involved in brain citrate regulation and suggest that abnormalities in this regulation can induce seizures. The present study is the first to (i) establish the Slc13a5-knockout mouse model as a helpful tool to study the neuronal functions of NaCT and characterize the molecular mechanisms by which functional deficiency of this citrate transporter causes epilepsy and impairs neuronal function; (ii) evaluate all hypotheses that have previously been suggested on theoretical grounds to explain the neurological phenotype of SLC13A5 mutations; and (iii) indicate that alterations in brain citrate levels result in neuronal network excitability and increased seizure propensity.
Collapse
Affiliation(s)
- Christine Henke
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Germany; Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - R Maarten van Dijk
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Nina Miljanovic
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Thekla Cordes
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Sonja Bröer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Vanessa Ziesak
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
| | - Marco Rohde
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Center Munich, Neuherberg, Germany
| | - Charlotte Vogel
- Department of Biometry, Epidemiology and Information Processing, University of Veterinary Medicine Hannover, Germany
| | - Lisa Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Tina Schumann
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Germany; Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Diana M Willmes
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Germany; Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Anica Kurzbach
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Germany; Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Nermeen N El-Agroudy
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Germany; Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Germany
| | | | - Jens Jordan
- Institute for Aerospace Medicine, German Aerospace Center (DLR) and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA
| | - Rüdiger Köhling
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
| | - Andreas L Birkenfeld
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Germany; Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
18
|
The Archaeal Proteome Project advances knowledge about archaeal cell biology through comprehensive proteomics. Nat Commun 2020; 11:3145. [PMID: 32561711 PMCID: PMC7305310 DOI: 10.1038/s41467-020-16784-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/18/2020] [Indexed: 11/08/2022] Open
Abstract
While many aspects of archaeal cell biology remain relatively unexplored, systems biology approaches like mass spectrometry (MS) based proteomics offer an opportunity for rapid advances. Unfortunately, the enormous amount of MS data generated often remains incompletely analyzed due to a lack of sophisticated bioinformatic tools and field-specific biological expertise for data interpretation. Here we present the initiation of the Archaeal Proteome Project (ArcPP), a community-based effort to comprehensively analyze archaeal proteomes. Starting with the model archaeon Haloferax volcanii, we reanalyze MS datasets from various strains and culture conditions. Optimized peptide spectrum matching, with strict control of false discovery rates, facilitates identifying > 72% of the reference proteome, with a median protein sequence coverage of 51%. These analyses, together with expert knowledge in diverse aspects of cell biology, provide meaningful insights into processes such as N-terminal protein maturation, N-glycosylation, and metabolism. Altogether, ArcPP serves as an invaluable blueprint for comprehensive prokaryotic proteomics.
Collapse
|
19
|
Sun L, Xi S, He G, Li Z, Gang X, Sun C, Guo W, Wang G. Two to Tango: Dialogue between Adaptive and Innate Immunity in Type 1 Diabetes. J Diabetes Res 2020; 2020:4106518. [PMID: 32802890 PMCID: PMC7415089 DOI: 10.1155/2020/4106518] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/18/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a long-term and chronic autoimmune disorder, in which the immune system attacks the pancreatic β-cells. Both adaptive and innate immune systems are involved in T1DM development. Both B-cells and T-cells, including CD4 + and CD8 + T-cells, as well as other T-cell subsets, could affect onset of autoimmunity. Furthermore, cells involved in innate immunity, including the macrophages, dendritic cells, and natural killer (NK) cells, could also accelerate or decelerate T1DM development. In this review, the crosstalk and function of immune cells in the pathogenesis of T1DM, as well as the corresponding therapeutic interventions, are discussed.
Collapse
Affiliation(s)
- Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Shugang Xi
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Guangyu He
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Zhuo Li
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Chenglin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Weiying Guo
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021 Jilin, China
| |
Collapse
|
20
|
Pauly D, Agarwal D, Dana N, Schäfer N, Biber J, Wunderlich KA, Jabri Y, Straub T, Zhang NR, Gautam AK, Weber BHF, Hauck SM, Kim M, Curcio CA, Stambolian D, Li M, Grosche A. Cell-Type-Specific Complement Expression in the Healthy and Diseased Retina. Cell Rep 2019; 29:2835-2848.e4. [PMID: 31775049 PMCID: PMC6911814 DOI: 10.1016/j.celrep.2019.10.084] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/24/2019] [Accepted: 10/22/2019] [Indexed: 02/06/2023] Open
Abstract
Complement dysregulation is a feature of many retinal diseases, yet mechanistic understanding at the cellular level is limited. Given this knowledge gap about which retinal cells express complement, we performed single-cell RNA sequencing on ~92,000 mouse retinal cells and validated our results in five major purified retinal cell types. We found evidence for a distributed cell-type-specific complement expression across 11 cell types. Notably, Müller cells are the major contributor of complement activators c1s, c3, c4, and cfb. Retinal pigment epithelium (RPE) mainly expresses cfh and the terminal complement components, whereas cfi and cfp transcripts are most abundant in neurons. Aging enhances c1s, cfb, cfp, and cfi expression, while cfh expression decreases. Transient retinal ischemia increases complement expression in microglia, Müller cells, and RPE. In summary, we report a unique complement expression signature for murine retinal cell types suggesting a well-orchestrated regulation of local complement expression in the retinal microenvironment. Overshooting complement activity contributes to retinal degeneration. Pauly et al. demonstrate a distinct complement expression profile of retinal cell types that changes with aging and during retinal degeneration. This prompts the intriguing concept of a local retinal complement activation possibly independent of the systemic components typically produced by the liver.
Collapse
Affiliation(s)
- Diana Pauly
- Experimental Ophthalmology, University Hospital Regensburg, Regensburg 93053, Germany.
| | - Divyansh Agarwal
- Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas Dana
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicole Schäfer
- Experimental Ophthalmology, University Hospital Regensburg, Regensburg 93053, Germany
| | - Josef Biber
- Department of Physiological Genomics, Biomedical Center, Ludwig Maximilians University Munich, Planegg-Martinsried 82152, Germany
| | - Kirsten A Wunderlich
- Department of Physiological Genomics, Biomedical Center, Ludwig Maximilians University Munich, Planegg-Martinsried 82152, Germany
| | - Yassin Jabri
- Experimental Ophthalmology, University Hospital Regensburg, Regensburg 93053, Germany
| | - Tobias Straub
- Core Facility Bioinformatics, Biomedical Center, Ludwig Maximilians University Munich, Planegg-Martinsried 82152, Germany
| | - Nancy R Zhang
- Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Avneesh K Gautam
- Department of Medicine, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bernhard H F Weber
- Institute of Human Genetics, University of Regensburg, Regensburg 93053, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Center Munich, Research Center for Environmental Health (GmbH), Munich 80939, Germany
| | - Mijin Kim
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christine A Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | - Dwight Stambolian
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Antje Grosche
- Department of Physiological Genomics, Biomedical Center, Ludwig Maximilians University Munich, Planegg-Martinsried 82152, Germany.
| |
Collapse
|
21
|
Purcell AW, Sechi S, DiLorenzo TP. The Evolving Landscape of Autoantigen Discovery and Characterization in Type 1 Diabetes. Diabetes 2019; 68:879-886. [PMID: 31010879 PMCID: PMC6477901 DOI: 10.2337/dbi18-0066] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/29/2019] [Indexed: 12/20/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that is caused, in part, by T cell-mediated destruction of insulin-producing β-cells. High risk for disease, in those with genetic susceptibility, is predicted by the presence of two or more autoantibodies against insulin, the 65-kDa form of glutamic acid decarboxylase (GAD65), insulinoma-associated protein 2 (IA-2), and zinc transporter 8 (ZnT8). Despite this knowledge, we still do not know what leads to the breakdown of tolerance to these autoantigens, and we have an incomplete understanding of T1D etiology and pathophysiology. Several new autoantibodies have recently been discovered using innovative technologies, but neither their potential utility in monitoring disease development and treatment nor their role in the pathophysiology and etiology of T1D has been explored. Moreover, neoantigen generation (through posttranslational modification, the formation of hybrid peptides containing two distinct regions of an antigen or antigens, alternative open reading frame usage, and translation of RNA splicing variants) has been reported, and autoreactive T cells that target these neoantigens have been identified. Collectively, these new studies provide a conceptual framework to understand the breakdown of self-tolerance, if such modifications occur in a tissue- or disease-specific context. A recent workshop sponsored by the National Institute of Diabetes and Digestive and Kidney Diseases brought together investigators who are using new methods and technologies to identify autoantigens and characterize immune responses toward these proteins. Researchers with diverse expertise shared ideas and identified resources to accelerate antigen discovery and the detection of autoimmune responses in T1D. The application of this knowledge will direct strategies for the identification of improved biomarkers for disease progression and treatment response monitoring and, ultimately, will form the foundation for novel antigen-specific therapeutics. This Perspective highlights the key issues that were addressed at the workshop and identifies areas for future investigation.
Collapse
Affiliation(s)
- Anthony W Purcell
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Salvatore Sechi
- Division of Diabetes, Endocrinology, and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Teresa P DiLorenzo
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY
- Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
22
|
Niersmann C, Hauck SM, Kannenberg JM, Röhrig K, von Toerne C, Roden M, Herder C, Carstensen-Kirberg M. Omentin-regulated proteins combine a pro-inflammatory phenotype with an anti-inflammatory counterregulation in human adipocytes: A proteomics analysis. Diabetes Metab Res Rev 2019; 35:e3074. [PMID: 30198166 DOI: 10.1002/dmrr.3074] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/09/2018] [Accepted: 09/03/2018] [Indexed: 12/17/2022]
Abstract
AIMS Experimental and epidemiological studies reported controversial data on the role of omentin in type 2 diabetes and cardiovascular diseases. This study aimed to characterise the impact of omentin on the secretome of human adipocytes to analyse the enrichment of these proteins in metabolic and cellular signalling pathways underlying its physiological function. MATERIAL/METHODS Differentiated primary human adipocytes were treated without or with 500 or 2000 ng/mL omentin for 24 hours. The secretome was analysed by liquid chromatography coupled tandem-mass spectrometry. Differences in protein secretion between untreated and omentin-treated adipocytes were compared using a paired t-test. Other potential upstream regulators and the overrepresentation in canonical pathways of omentin-stimulated proteins were analysed using Ingenuity Pathway Analysis. RESULTS The supernatant of adipocytes contained 3493 proteins, of which 140 were differentially secreted by both concentrations of omentin compared with untreated adipocytes. Among the most strongly increased proteins, tumour necrosis factor-inducible gene 6 protein (TNFAIP6) was increased by 140-fold in the supernatant. Omentin-regulated proteins were overrepresented in seven canonical pathways including eukaryotic initiation factor 2 signalling, complement system, and inhibition of matrix metalloproteases. We further identified 25 other potential upstream activators of omentin-regulated proteins, mainly pro-inflammatory cytokines and transcription regulators including NFκB. CONCLUSIONS In differentiated human adipocytes, the release of the anti-inflammatory TNFAIP6 might be part of a counterregulatory response to the pro-inflammatory action of omentin. Omentin-regulated proteins were overrepresented in pathways indicating cellular stress, a pro-inflammatory environment and a crosstalk with other organs. Other potential activators of omentin-regulated proteins point towards a central role of NFκB activation in the omentin-induced secretory process.
Collapse
Affiliation(s)
- Corinna Niersmann
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Stefanie M Hauck
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), München-Neuherberg, Germany
| | - Julia M Kannenberg
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Karin Röhrig
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Christine von Toerne
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), München-Neuherberg, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Maren Carstensen-Kirberg
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
23
|
Müller D, Telieps T, Eugster A, Weinzierl C, Jolink M, Ziegler AG, Bonifacio E. Novel minor HLA DR associated antigens in type 1 diabetes. Clin Immunol 2018; 194:87-91. [PMID: 29990590 DOI: 10.1016/j.clim.2018.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/04/2018] [Accepted: 07/01/2018] [Indexed: 12/12/2022]
Abstract
Type 1 diabetes is an autoimmune disease leading to insulin deficiency. Autoantibodies to beta cell proteins are already present in the asymptomatic phase of type 1 diabetes. Recent findings have suggested a number of additional minor autoantigens in patients with type 1 diabetes. We have established luciferase immunoprecipitation systems (LIPS) for anti-MTIF3, anti-PPIL2, anti-NUP50 and anti-MLH1 and analyzed samples from 500 patients with type 1 diabetes at onset of clinical disease and 200 healthy individuals who had a family history of type 1 diabetes but no evidence of beta cell autoantibodies. We show significantly higher frequencies of anti-MTIF3, anti-PPIL2 and anti-MLH1 in recent onset type 1 diabetes patients in comparison to controls. In addition, antibodies to NUP50 were associated with HLA-DRB1*03 and antibodies to MLH1 were associated with HLA-DRB1*04 genotypes.
Collapse
Affiliation(s)
- Denise Müller
- DFG Research Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Tanja Telieps
- Helmholtz Zentrum München, Institute for Diabetes and Obesity, Neuherberg, Germany
| | - Anne Eugster
- DFG Research Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christina Weinzierl
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, Neuherberg, Germany
| | - Manja Jolink
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, Neuherberg, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, Neuherberg, Germany; Forschergruppe Diabetes e.V, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ezio Bonifacio
- DFG Research Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Helmholtz Zentrum München, Institute for Diabetes and Obesity, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Paul Langerhans Institute Dresden, German Center for Diabetes Research (DZD), Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|