1
|
Cristóbal-Lecina E, El-Maiss J, Figueras E, Singh AC, Krishnamoorthy S, Østerbye T, Pascual García C, Andreu D. Acid-Modulated Peptide Synthesis for Application on Oxide Biosensor Interfaces. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:3092. [PMID: 38132988 PMCID: PMC10746054 DOI: 10.3390/nano13243092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
In this paper we report an acid-modulated strategy for novel peptide microarray production on biosensor interfaces. We initially selected a controlled pore glass (CPG) as a support for solid-phase peptide synthesis (SPPS) to implement a chemistry that can be performed at the interface of multiple field effect transistor (FET) sensors, eventually to generate label-free peptide microarrays for protein screening. Our chemistry uses a temporary protection of the N-terminal amino function of each amino acid building block with a tert-butyloxycarbonyl (Boc) group that can be removed after each SPPS cycle, in combination with semi-permanent protection of the side chains of trifunctional amino acid residues. Such a protection scheme with a well-proven record of application in conventional, batchwise SPPS has been fine-tuned for optimal performance on CPG and, from there, translated to SPR chips that allow layer-by-layer monitoring of amino acid coupling. Our results validate this acid-modulated synthesis as a feasible approach for producing peptides in high yields and purity on flat glass surfaces, such as those in bio-FETs.
Collapse
Affiliation(s)
- Edgar Cristóbal-Lecina
- Proteomics and Protein Chemistry Unit, Department of Medicine and Life Sciences, Pompeu Fabra University, 08003 Barcelona, Spain;
| | - Janwa El-Maiss
- MRT Department, Luxembourg Institute of Science and Technology, L-4420 Belvaux, Luxembourg; (J.E.-M.); (A.C.S.); (S.K.)
| | - Eduard Figueras
- Proteomics and Protein Chemistry Unit, Department of Medicine and Life Sciences, Pompeu Fabra University, 08003 Barcelona, Spain;
| | - Aruna Chandra Singh
- MRT Department, Luxembourg Institute of Science and Technology, L-4420 Belvaux, Luxembourg; (J.E.-M.); (A.C.S.); (S.K.)
| | - Sivashankar Krishnamoorthy
- MRT Department, Luxembourg Institute of Science and Technology, L-4420 Belvaux, Luxembourg; (J.E.-M.); (A.C.S.); (S.K.)
| | - Thomas Østerbye
- Department of Immunology and Microbiology, University of Copenhagen, 2200 København, Denmark;
| | - César Pascual García
- MRT Department, Luxembourg Institute of Science and Technology, L-4420 Belvaux, Luxembourg; (J.E.-M.); (A.C.S.); (S.K.)
| | - David Andreu
- Proteomics and Protein Chemistry Unit, Department of Medicine and Life Sciences, Pompeu Fabra University, 08003 Barcelona, Spain;
| |
Collapse
|
2
|
Dopico XC, Mandolesi M, Hedestam GBK. Untangling immunoglobulin genotype-function associations. Immunol Lett 2023:S0165-2478(23)00073-1. [PMID: 37209913 DOI: 10.1016/j.imlet.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/19/2023] [Accepted: 05/12/2023] [Indexed: 05/22/2023]
Abstract
Immunoglobulin (IG) genes, encoding B cell receptors (BCRs), are fundamental components of the mammalian immune system, which evolved to recognize the diverse antigenic universe present in nature. To handle these myriad inputs, BCRs are generated through combinatorial recombination of a set of highly polymorphic germline genes, resulting in a vast repertoire of antigen receptors that initiate responses to pathogens and regulate commensals. Following antigen recognition and B cell activation, memory B cells and plasma cells form, allowing for the development of anamnestic antibody (Ab) responses. How inherited variation in IG genes impacts host traits, disease susceptibility, and Ab recall responses is a topic of great interest. Here, we consider approaches to translate emerging knowledge about IG genetic diversity and expressed repertoires to inform our understanding of Ab function in health and disease etiology. As our understanding of IG genetics grows, so will our need for tools to decipher preferences for IG gene or allele usage in different contexts, to better understand antibody responses at the population level.
Collapse
Affiliation(s)
- Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 17177, Sweden.
| | - Marco Mandolesi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 17177, Sweden
| | | |
Collapse
|
3
|
Hogan JM, Lee PS, Wong SC, West SM, Morishige WH, Bee C, Tapia GC, Rajpal A, Strop P, Dollinger G. Residue-Level Characterization of Antibody Binding Epitopes Using Carbene Chemical Footprinting. Anal Chem 2023; 95:3922-3931. [PMID: 36791402 DOI: 10.1021/acs.analchem.2c03091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Characterization of antibody binding epitopes is an important factor in therapeutic drug discovery, as the binding site determines and drives antibody pharmacology and pharmacokinetics. Here, we present a novel application of carbene chemical footprinting with mass spectrometry for identification of antibody binding epitopes at the single-residue level. Two different photoactivated diazirine reagents provide complementary labeling information allowing structural refinement of the antibody binding interface. We applied this technique to map the epitopes of multiple MICA and CTLA-4 antibodies and validated the findings with X-ray crystallography and yeast surface display epitope mapping. The characterized epitopes were used to understand biolayer interferometry-derived competitive binding results at the structural level. We show that carbene footprinting provides fast and high-resolution epitope information critical in the antibody selection process and enables mechanistic understanding of function to accelerate the drug discovery process.
Collapse
Affiliation(s)
- Jason M Hogan
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Peter S Lee
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Susan C Wong
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Sean M West
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Winse H Morishige
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Christine Bee
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Gamze Camdere Tapia
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Arvind Rajpal
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Pavel Strop
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Gavin Dollinger
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| |
Collapse
|
4
|
Balakrishnan D, El Maiss J, Olthuis W, Pascual García C. Miniaturized Control of Acidity in Multiplexed Microreactors. ACS OMEGA 2023; 8:7587-7594. [PMID: 36872992 PMCID: PMC9979314 DOI: 10.1021/acsomega.2c06897] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
The control of acidity drives the assembly of biopolymers that are essential for a wide range of applications. Its miniaturization can increase the speed and the possibilities of combinatorial throughput for their manipulation, similar to the way that the miniaturization of transistors allows logical operations in microelectronics with a high throughput. Here, we present a device containing multiplexed microreactors, each one enabling independent electrochemical control of acidity in ∼2.5 nL volumes, with a large acidity range from pH 3 to 7 and an accuracy of at least 0.4 pH units. The attained pH within each microreactor (with footprints of ∼0.3 mm2 for each spot) was kept constant for long retention times (∼10 min) and over repeated cycles of >100. The acidity is driven by redox proton exchange reactions, which can be driven at different rates influencing the efficiency of the device in order to achieve more charge exchange (larger acidity range) or better reversibility. The achieved performance in acidity control, miniaturization, and the possibility to multiplex paves the way for the control of combinatorial chemistry through pH- and acidity-controlled reactions.
Collapse
Affiliation(s)
- Divya Balakrishnan
- Luxembourg
Institute of Science and Technology (LIST), 41 Rue du Brill, L-4422Belvaux, Luxembourg
| | - Janwa El Maiss
- Luxembourg
Institute of Science and Technology (LIST), 41 Rue du Brill, L-4422Belvaux, Luxembourg
| | - Wouter Olthuis
- MESA+
Institute, University of Twente, Drienerlolaan 5, 7522 NBEnschede, Netherlands
| | - César Pascual García
- Luxembourg
Institute of Science and Technology (LIST), 41 Rue du Brill, L-4422Belvaux, Luxembourg
| |
Collapse
|
5
|
Abstract
The diversity of the antigen-specific humoral immune response reflects the interaction of the immune system with pathogens and autoantigens. Peptide microarray analysis opens up new perspectives for the use of antibodies as diagnostic biomarkers and provides unique access to a more differentiated view on humoral responses to disease. This review focuses on the latest applications of peptide microarrays for the serologic medical diagnosis of autoimmunity, infectious diseases (including COVID-19), and cancer.
Collapse
Affiliation(s)
- Carsten Grötzinger
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
6
|
Jung J, Lee LE, Kim H, Kim JE, Jang SH, Roh JS, Lee B, Robinson WH, Sohn DH, Pyun JC, Song JJ. Extracellular histones aggravate autoimmune arthritis by lytic cell death. Front Immunol 2022; 13:961197. [PMID: 36032105 PMCID: PMC9410568 DOI: 10.3389/fimmu.2022.961197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 07/21/2022] [Indexed: 12/03/2022] Open
Abstract
Although recent studies have demonstrated a proinflammatory effect of extracellular histones in sepsis via endothelial cytotoxicity, little is known about their contribution to autoimmune arthritis. Therefore, we investigated the role of extracellular histones in autoimmune arthritis and their cytotoxic effect on synoviocytes and macrophages. We measured histones in the synovial fluid of patients with rheumatoid arthritis (RA) and evaluated arthritis severity in a serum-transfer arthritis (STA) mouse model with intraperitoneal histone injection. Histone-induced cytotoxicity was measured using SYTOX green staining in the synoviocyte cell line MH7A and macrophages differentiated from the monocytic cell line THP-1, and the production of damage-associated molecular patterns (DAMPs) was measured by HMGB1 and ATP. Furthermore, we performed RNA-seq analysis of THP-1 cells stimulated with H2B-α1 peptide or with its citrullinated form. The levels of histones were elevated in RA synovial fluid, and histones aggravated arthritis in the STA model. Histones induced cytotoxicity and DAMP production in synoviocytes and macrophages. Chondroitin sulfate reduced histone-induced cytotoxicity, while lipopolysaccharides aggravated cytotoxicity. Moreover, the cytotoxicity decreased when the arginines in H2B-α1 were replaced with citrullines, which demonstrated its electrostatic nature. In transcriptome analysis, H2B-α1 changed the gene expression pattern of THP-1 cells involving chemokines, interleukin-1, -4, -10, -13, and toll-like receptor (TLR) signaling pathways. Extracellular histones were increased in RA synovial fluid and aggravated synovitis in STA. They induced lytic cell death through electrostatic interaction with synoviocytes and macrophages, leading to the secretion of DAMPs. These findings suggest that histones play a central role in autoimmune arthritis.
Collapse
Affiliation(s)
- Jaeyong Jung
- Department of Materials Science and Engineering, Yonsei University, Seoul, South Korea
| | - Lucy Eunju Lee
- Division of Rheumatology, Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang, South Korea
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Hanna Kim
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji Eun Kim
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Hoon Jang
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong Seong Roh
- Department of Herbal Prescription, College of Korean Medicine, Daegu Haany University, Gyeongsan, South Korea
| | - Beomgu Lee
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, South Korea
| | - William H. Robinson
- VA Palo Alto Health Care System, Palo Alto, CA, United States
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, South Korea
- *Correspondence: Jason Jungsik Song, ; Dong Hyun Sohn, ; Jae-Chul Pyun,
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, Seoul, South Korea
- *Correspondence: Jason Jungsik Song, ; Dong Hyun Sohn, ; Jae-Chul Pyun,
| | - Jason Jungsik Song
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Jason Jungsik Song, ; Dong Hyun Sohn, ; Jae-Chul Pyun,
| |
Collapse
|
7
|
Paris G, Heidepriem J, Tsouka A, Liu Y, Mattes DS, Pinzón Martín S, Dallabernardina P, Mende M, Lindner C, Wawrzinek R, Rademacher C, Seeberger PH, Breitling F, Bischoff FR, Wolf T, Loeffler FF. Automated Laser-Transfer Synthesis of High-Density Microarrays for Infectious Disease Screening. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2200359. [PMID: 35429012 DOI: 10.1002/adma.202200359] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/23/2022] [Indexed: 06/14/2023]
Abstract
Laser-induced forward transfer (LIFT) is a rapid laser-patterning technique for high-throughput combinatorial synthesis directly on glass slides. A lack of automation and precision limits LIFT applications to simple proof-of-concept syntheses of fewer than 100 compounds. Here, an automated synthesis instrument is reported that combines laser transfer and robotics for parallel synthesis in a microarray format with up to 10 000 individual reactions cm- 2 . An optimized pipeline for amide bond formation is the basis for preparing complex peptide microarrays with thousands of different sequences in high yield with high reproducibility. The resulting peptide arrays are of higher quality than commercial peptide arrays. More than 4800 15-residue peptides resembling the entire Ebola virus proteome on a microarray are synthesized to study the antibody response of an Ebola virus infection survivor. Known and unknown epitopes that serve now as a basis for Ebola diagnostic development are identified. The versatility and precision of the synthesizer is demonstrated by in situ synthesis of fluorescent molecules via Schiff base reaction and multi-step patterning of precisely definable amounts of fluorophores. This automated laser transfer synthesis approach opens new avenues for high-throughput chemical synthesis and biological screening.
Collapse
Affiliation(s)
- Grigori Paris
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476, Potsdam, Germany
- Department of System Dynamics and Friction Physics, Institute of Mechanics, Technical University of Berlin, Str. des 17. Juni 135, 10623, Berlin, Germany
| | - Jasmin Heidepriem
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476, Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195, Berlin, Germany
| | - Alexandra Tsouka
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476, Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195, Berlin, Germany
| | - Yuxin Liu
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476, Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195, Berlin, Germany
| | - Daniela S Mattes
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafe, Germany
| | - Sandra Pinzón Martín
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476, Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195, Berlin, Germany
| | - Pietro Dallabernardina
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476, Potsdam, Germany
| | - Marco Mende
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476, Potsdam, Germany
| | - Celina Lindner
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476, Potsdam, Germany
| | - Robert Wawrzinek
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476, Potsdam, Germany
| | - Christoph Rademacher
- Department of Pharmaceutical Sciences, University of Vienna, Althanstr. 14, Vienna, 1090, Austria
- Department of Microbiology and Immunobiology, Max F. Perutz Laboratories GmbH, Dr.-Bohr-Gasse 9, Vienna, 1030, Austria
| | - Peter H Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476, Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195, Berlin, Germany
| | - Frank Breitling
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafe, Germany
| | - Frank Ralf Bischoff
- Department of Functional Genome Analysis, German Cancer Research Center, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Timo Wolf
- Infectious Diseases Unit, Department of Medicine, Goethe University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Felix F Loeffler
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Muehlenberg 1, 14476, Potsdam, Germany
| |
Collapse
|
8
|
Yiu G, Rasmussen TK, Tsai BL, Diep VK, Haddon DJ, Tsoi J, Miller GD, Comin-Anduix B, Deleuran B, Crooks GM, Utz PJ. High Interferon Signature Leads to Increased STAT1/3/5 Phosphorylation in PBMCs From SLE Patients by Single Cell Mass Cytometry. Front Immunol 2022; 13:833636. [PMID: 35185925 PMCID: PMC8851522 DOI: 10.3389/fimmu.2022.833636] [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: 12/11/2021] [Accepted: 01/11/2022] [Indexed: 12/02/2022] Open
Abstract
The establishment of an “interferon (IFN) signature” to subset SLE patients on disease severity has led to therapeutics targeting IFNα. Here, we investigate IFN signaling in SLE using multiplexed protein arrays and single cell cytometry by time of flight (CyTOF). First, the IFN signature for SLE patients (n=81) from the Stanford Lupus Registry is determined using fluidigm qPCR measuring 44 previously determined IFN-inducible transcripts. IFN-high (IFN-H) patients have increased SLE criteria and renal/CNS/immunologic involvement, and increased autoantibody reactivity against spliceosome-associated antigens. CyTOF analysis is performed on non-stimulated and stimulated (IFNα, IFNγ, IL-21) PBMCs from SLE patients (n=25) and HCs (n=9) in a panel identifying changes in phosphorylation of intracellular signaling proteins (pTOF). Another panel is utilized to detect changes in intracellular cytokine (ICTOF) production in non-stimulated and stimulated (PMA/ionomycin) PBMCs from SLE patients (n=31) and HCs (n=17). Bioinformatic analysis by MetaCyto and OMIQ reveal phenotypic changes in immune cell subsets between IFN-H and IFN-low (IFN-L) patients. Most notably, IFN-H patients exhibit increased STAT1/3/5 phosphorylation downstream of cytokine stimulation and increased phosphorylation of non-canonical STAT proteins. These results suggest that IFN signaling in SLE modulates STAT phosphorylation, potentially uncovering possible targets for future therapeutic approaches.
Collapse
Affiliation(s)
- Gloria Yiu
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States.,Department of Rheumatology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tue Kruse Rasmussen
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States.,Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | - Brandon L Tsai
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Vivian K Diep
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States
| | - David J Haddon
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States
| | - Jennifer Tsoi
- Department of Surgery David Geffen School of Medicine, Johnson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Gopika D Miller
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States
| | - Begoña Comin-Anduix
- Department of Surgery David Geffen School of Medicine, Johnson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), Los Angeles, CA, United States.,Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| | - Bent Deleuran
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | - Gay M Crooks
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), Los Angeles, CA, United States.,Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Eli and Edythe Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles (UCLA), Los Angeles, CA, United States.,Division of Pediatric Hematology-Oncology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Paul J Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States.,Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
9
|
Li S, Song G, Bai Y, Song N, Zhao J, Liu J, Hu C. Applications of Protein Microarrays in Biomarker Discovery for Autoimmune Diseases. Front Immunol 2021; 12:645632. [PMID: 34012435 PMCID: PMC8126629 DOI: 10.3389/fimmu.2021.645632] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/13/2021] [Indexed: 01/18/2023] Open
Abstract
Dysregulated autoantibodies and cytokines were deemed to provide important cues for potential illnesses, such as various carcinomas and autoimmune diseases. Increasing biotechnological approaches have been applied to screen and identify the specific alterations of these biomolecules as distinctive biomarkers in diseases, especially autoimmune diseases. As a versatile and robust platform, protein microarray technology allows researchers to easily profile dysregulated autoantibodies and cytokines associated with autoimmune diseases using various biological specimens, mainly serum samples. Here, we summarize the applications of protein microarrays in biomarker discovery for autoimmune diseases. In addition, the key issues in the process of using this approach are presented for improving future studies.
Collapse
Affiliation(s)
- Siting Li
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Guang Song
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yina Bai
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Ning Song
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Jian Liu
- Department of Rheumatology, Aerospace Center Hospital, Aerospace, Clinical Medical College, Peking University, Beijing, China
| | - Chaojun Hu
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| |
Collapse
|
10
|
Guttieres D, Sinskey AJ, Springs SL. Models to inform neutralizing antibody therapy strategies during pandemics: the case of SARS-CoV-2. Antib Ther 2021; 4:60-71. [PMID: 33928236 PMCID: PMC8022923 DOI: 10.1093/abt/tbab006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/09/2021] [Accepted: 03/19/2021] [Indexed: 12/13/2022] Open
Abstract
Background Neutralizing antibodies (nAbs) against SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) can play an important role in reducing impacts of the COVID-19 pandemic, complementing ongoing public health efforts such as diagnostics and vaccination. Rapidly designing, manufacturing and distributing nAbs requires significant planning across the product value chain and an understanding of the opportunities, challenges and risks throughout. Methods A systems framework comprised of four critical components is presented to aid in developing effective end-to-end nAbs strategies in the context of a pandemic: (1) product design and optimization, (2) epidemiology, (3) demand and (4) supply. Quantitative models are used to estimate product demand using available epidemiological data, simulate biomanufacturing operations from typical bioprocess parameters and calculate antibody production costs to meet clinical needs under various realistic scenarios. Results In a US-based case study during the 9-month period from March 15 to December 15, 2020, the projected number of SARS-CoV-2 infections was 15.73 million. The estimated product volume needed to meet therapeutic demand for the maximum number of clinically eligible patients ranged between 6.3 and 31.5 tons for 0.5 and 2.5 g dose sizes, respectively. The relative production scale and cost needed to meet demand are calculated for different centralized and distributed manufacturing scenarios. Conclusions Meeting demand for anti-SARS-CoV-2 nAbs requires significant manufacturing capacity and planning for appropriate administration in clinical settings. MIT Center for Biomedical Innovation’s data-driven tools presented can help inform time-critical decisions by providing insight into important operational and policy considerations for making nAbs broadly accessible, while considering time and resource constraints.
Collapse
Affiliation(s)
- Donovan Guttieres
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anthony J Sinskey
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stacy L Springs
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
11
|
Schulte C, Khayenko V, Nordblom NF, Tippel F, Peck V, Gupta AJ, Maric HM. High-throughput determination of protein affinities using unmodified peptide libraries in nanomolar scale. iScience 2021; 24:101898. [PMID: 33364586 PMCID: PMC7753147 DOI: 10.1016/j.isci.2020.101898] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/13/2020] [Accepted: 12/01/2020] [Indexed: 12/02/2022] Open
Abstract
Protein-protein interactions (PPIs) are of fundamental importance for our understanding of physiology and pathology. PPIs involving short, linear motifs play a major role in immunological recognition, signaling, and regulation and provide attractive starting points for pharmaceutical intervention. Yet, state-of-the-art protein-peptide affinity determination approaches exhibit limited throughput and sensitivity, often resulting from ligand immobilization, labeling, or synthesis. Here, we introduce a high-throughput method for in-solution analysis of protein-peptide interactions using a phenomenon called temperature related intensity change (TRIC). We use TRIC for the identification and fine-mapping of low- and high-affinity protein interaction sites and the definition of sequence binding requirements. Validation is achieved by microarray-based studies using wild-type and mutated recombinant protein and the native protein within tissue lysates. On-chip neutralization and strong correlation with structural data establish TRIC as a quasi-label-free method to determine binding affinities of unmodified peptide libraries with large dynamic range.
Collapse
Affiliation(s)
- Clemens Schulte
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Vladimir Khayenko
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Noah Frieder Nordblom
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Franziska Tippel
- Nanotemper Technologies GmbH, Flößergasse 4, 81369 Munich, Germany
| | - Violetta Peck
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Amit Jean Gupta
- Nanotemper Technologies GmbH, Flößergasse 4, 81369 Munich, Germany
| | - Hans Michael Maric
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| |
Collapse
|
12
|
Anderson SE, Longbotham JE, O'Kane PT, Ugur FS, Fujimori DG, Mrksich M. Exploring the Ligand Preferences of the PHD1 Domain of Histone Demethylase KDM5A Reveals Tolerance for Modifications of the Q5 Residue of Histone 3. ACS Chem Biol 2021; 16:205-213. [PMID: 33314922 PMCID: PMC8168426 DOI: 10.1021/acschembio.0c00891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Understanding the ligand preferences of epigenetic reader domains enables identification of modification states of chromatin with which these domains associate and can yield insight into recruitment and catalysis of chromatin-acting complexes. However, thorough exploration of the ligand preferences of reader domains is hindered by the limitations of traditional protein-ligand binding assays. Here, we evaluate the binding preferences of the PHD1 domain of histone demethylase KDM5A using the protein interaction by SAMDI (PI-SAMDI) assay, which measures protein-ligand binding in a high-throughput and sensitive manner via binding-induced enhancement in the activity of a reporter enzyme, in combination with fluorescence polarization. The PI-SAMDI assay was validated by confirming its ability to accurately profile the relative binding affinity of a set of well-characterized histone 3 (H3) ligands of PHD1. The assay was then used to assess the affinity of PHD1 for 361 H3 mutant ligands, a select number of which were further characterized by fluorescence polarization. Together, these experiments revealed PHD1's tolerance for H3Q5 mutations, including an unexpected tolerance for aromatic residues in this position. Motivated by this finding, we further demonstrate a high-affinity interaction between PHD1 and recently identified Q5-serotonylated H3. This work yields interesting insights into permissible PHD1-H3 interactions and demonstrates the value of interfacing PI-SAMDI and fluorescence polarization in investigations of protein-ligand binding.
Collapse
Affiliation(s)
- Sarah E Anderson
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - James E Longbotham
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California 94158, United States
| | - Patrick T O'Kane
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Fatima S Ugur
- Chemistry and Chemical Biology Graduate Program, University of California San Francisco, San Francisco, California 94158, United States
| | - Danica Galonić Fujimori
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California 94158, United States
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, United States
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, California 94158, United States
| | - Milan Mrksich
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Department of Cell and Developmental Biology, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
13
|
Ladner JT, Henson SN, Boyle AS, Engelbrektson AL, Fink ZW, Rahee F, D'ambrozio J, Schaecher KE, Stone M, Dong W, Dadwal S, Yu J, Caligiuri MA, Cieplak P, Bjørås M, Fenstad MH, Nordbø SA, Kainov DE, Muranaka N, Chee MS, Shiryaev SA, Altin JA. Epitope-resolved profiling of the SARS-CoV-2 antibody response identifies cross-reactivity with endemic human coronaviruses. CELL REPORTS MEDICINE 2021; 2:100189. [PMID: 33495758 PMCID: PMC7816965 DOI: 10.1016/j.xcrm.2020.100189] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/11/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022]
Abstract
The SARS-CoV-2 proteome shares regions of conservation with endemic human coronaviruses (CoVs), but it remains unknown to what extent these may be cross-recognized by the antibody response. Here, we study cross-reactivity using a highly multiplexed peptide assay (PepSeq) to generate an epitope-resolved view of IgG reactivity across all human CoVs in both COVID-19 convalescent and negative donors. PepSeq resolves epitopes across the SARS-CoV-2 Spike and Nucleocapsid proteins that are commonly targeted in convalescent donors, including several sites also recognized in some uninfected controls. By comparing patterns of homologous reactivity between CoVs and using targeted antibody-depletion experiments, we demonstrate that SARS-CoV-2 elicits antibodies that cross-recognize pandemic and endemic CoV antigens at two Spike S2 subunit epitopes. We further show that these cross-reactive antibodies preferentially bind endemic homologs. Our findings highlight sites at which the SARS-CoV-2 response appears to be shaped by previous CoV exposures and which have the potential to raise broadly neutralizing responses. PepSeq enables fully in vitro, highly multiplexed peptide-based antibody assays Epitope mapping shows preexisting antibody reactivity to SARS-CoV-2 antigens Antibodies cross-recognize endemic and pandemic antigens in the Spike S2 subunit Cross-reactive antibodies raised by SARS-CoV-2 preferentially bind endemic homologs
Collapse
Affiliation(s)
- Jason T Ladner
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Sierra N Henson
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Annalee S Boyle
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Anna L Engelbrektson
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Zane W Fink
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Fatima Rahee
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | | | | | - Mars Stone
- Vitalant Research Institute and Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Wenjuan Dong
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Sanjeet Dadwal
- Division of Infectious Diseases, City of Hope National Medical Center, Duarte, CA, USA
| | - Jianhua Yu
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Michael A Caligiuri
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Piotr Cieplak
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Magnar Bjørås
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mona H Fenstad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Immunology and Transfusion Medicine, St. Olavs Hospital, Trondheim, Norway
| | - Svein A Nordbø
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Medical Microbiology, St. Olavs Hospital, Trondheim, Norway
| | - Denis E Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | - Sergey A Shiryaev
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - John A Altin
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| |
Collapse
|
14
|
Heiss K, Heidepriem J, Fischer N, Weber LK, Dahlke C, Jaenisch T, Loeffler FF. Rapid Response to Pandemic Threats: Immunogenic Epitope Detection of Pandemic Pathogens for Diagnostics and Vaccine Development Using Peptide Microarrays. J Proteome Res 2020; 19:4339-4354. [PMID: 32892628 PMCID: PMC7640972 DOI: 10.1021/acs.jproteome.0c00484] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Indexed: 12/18/2022]
Abstract
Emergence and re-emergence of pathogens bearing the risk of becoming a pandemic threat are on the rise. Increased travel and trade, growing population density, changes in urbanization, and climate have a critical impact on infectious disease spread. Currently, the world is confronted with the emergence of a novel coronavirus SARS-CoV-2, responsible for yet more than 800 000 deaths globally. Outbreaks caused by viruses, such as SARS-CoV-2, HIV, Ebola, influenza, and Zika, have increased over the past decade, underlining the need for a rapid development of diagnostics and vaccines. Hence, the rational identification of biomarkers for diagnostic measures on the one hand, and antigenic targets for vaccine development on the other, are of utmost importance. Peptide microarrays can display large numbers of putative target proteins translated into overlapping linear (and cyclic) peptides for a multiplexed, high-throughput antibody analysis. This enabled for example the identification of discriminant/diagnostic epitopes in Zika or influenza and mapping epitope evolution in natural infections versus vaccinations. In this review, we highlight synthesis platforms that facilitate fast and flexible generation of high-density peptide microarrays. We further outline the multifaceted applications of these peptide array platforms for the development of serological tests and vaccines to quickly encounter pandemic threats.
Collapse
Affiliation(s)
- Kirsten Heiss
- PEPperPRINT
GmbH, Rischerstrasse
12, 69123 Heidelberg, Germany
| | - Jasmin Heidepriem
- Max
Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Am Muehlenberg 1, 14476 Potsdam, Germany
| | - Nico Fischer
- Section
Clinical Tropical Medicine, Department of Infectious Diseases, Heidelberg University Hospital, INF 324, 69120 Heidelberg, Germany
| | - Laura K. Weber
- PEPperPRINT
GmbH, Rischerstrasse
12, 69123 Heidelberg, Germany
- Institute
of Microstructure Technology, Karlsruhe
Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Christine Dahlke
- Division
of Infectious Diseases, First Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department
of Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
- German
Center for Infection Research, Partner Site
Hamburg-Lübeck-Borstel-Riems, 38124 Braunschweig, Germany
| | - Thomas Jaenisch
- Heidelberg
Institute of Global Health (HIGH), Heidelberg
University Hospital, Im Neuenheimer Feld 130, 69120 Heidelberg, Germany
- Center
for Global Health, Colorado School of Public Health, University of Colorado, Aurora, Colorado 80045, United States
- Department
of Epidemiology, Colorado School of Public Health, University of Colorado, Aurora, Colorado 80045, United States
| | - Felix F. Loeffler
- Max
Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Am Muehlenberg 1, 14476 Potsdam, Germany
| |
Collapse
|
15
|
Xie Z, Gan T, Fang L, Zhou X. Recent progress in creating complex and multiplexed surface-grafted macromolecular architectures. SOFT MATTER 2020; 16:8736-8759. [PMID: 32969442 DOI: 10.1039/d0sm01043j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Surface-grafted macromolecules, including polymers, DNA, peptides, etc., are versatile modifications to tailor the interfacial functions in a wide range of fields. In this review, we aim to provide an overview of the most recent progress in engineering surface-grafted chains for the creation of complex and multiplexed surface architectures over micro- to macro-scopic areas. A brief introduction to surface grafting is given first. Then the fabrication of complex surface architectures is summarized with a focus on controlled chain conformations, grafting densities and three-dimensional structures. Furthermore, recent advances are highlighted for the generation of multiplexed arrays with designed chemical composition in both horizontal and vertical dimensions. The applications of such complicated macromolecular architectures are then briefly discussed. Finally, some perspective outlooks for future studies and challenges are suggested. We hope that this review will be helpful to those just entering this field and those in the field requiring quick access to useful reference information about the progress in the properties, processing, performance, and applications of functional surface-grafted architectures.
Collapse
Affiliation(s)
- Zhuang Xie
- School of Materials Science and Engineering, and Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Xingangxi Road No. 135, Guangzhou, Guangdong Province 510275, P. R. China.
| | - Tiansheng Gan
- College of Chemistry and Environmental Engineering, Shenzhen University, Nanhai Avenue 3688, Shenzhen, Guangdong Province 518055, P. R. China.
| | - Lvye Fang
- School of Materials Science and Engineering, and Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Xingangxi Road No. 135, Guangzhou, Guangdong Province 510275, P. R. China.
| | - Xuechang Zhou
- College of Chemistry and Environmental Engineering, Shenzhen University, Nanhai Avenue 3688, Shenzhen, Guangdong Province 518055, P. R. China.
| |
Collapse
|
16
|
Immunological fingerprint of 4CMenB recombinant antigens via protein microarray reveals key immunosignatures correlating with bactericidal activity. Nat Commun 2020; 11:4994. [PMID: 33020485 PMCID: PMC7536418 DOI: 10.1038/s41467-020-18791-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 09/10/2020] [Indexed: 11/08/2022] Open
Abstract
Serogroup B meningococcus (MenB) is a leading cause of meningitis and sepsis across the world and vaccination is the most effective way to protect against this disease. 4CMenB is a multi-component vaccine against MenB, which is now licensed for use in subjects >2 months of age in several countries. In this study, we describe the development and use of an ad hoc protein microarray to study the immune response induced by the three major 4CMenB antigenic components (fHbp, NHBA and NadA) in individual sera from vaccinated infants, adolescents and adults. The resulting 4CMenB protein antigen fingerprinting allowed the identification of specific human antibody repertoire correlating with the bactericidal response elicited in each subject. This work represents an example of epitope mapping of the immune response induced by a multicomponent vaccine in different age groups with the identification of protective signatures. It shows the high flexibility of this microarray based methodology in terms of high-throughput information and minimal volume of biological samples needed. 4CMenB is an approved multi-component vaccine against Serogroup B meningococcus. Here the authors develop a protein microarray for three major 4CMenB antigenic components (fHbp, NHBA and NadA) and describe antibody repertoires in sera from vaccinated infants, adolescents and adults correlating with bactericidal response.
Collapse
|
17
|
Ladner JT, Henson SN, Boyle AS, Engelbrektson AL, Fink ZW, Rahee F, D’ambrozio J, Schaecher KE, Stone M, Dong W, Dadwal S, Yu J, Caligiuri MA, Cieplak P, Bjørås M, Fenstad MH, Nordbø SA, Kainov DE, Muranaka N, Chee MS, Shiryaev SA, Altin JA. Epitope-resolved profiling of the SARS-CoV-2 antibody response identifies cross-reactivity with an endemic human CoV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.27.222943. [PMID: 32743570 PMCID: PMC7386487 DOI: 10.1101/2020.07.27.222943] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A high-resolution understanding of the antibody response to SARS-CoV-2 is important for the design of effective diagnostics, vaccines and therapeutics. However, SARS-CoV-2 antibody epitopes remain largely uncharacterized, and it is unknown whether and how the response may cross-react with related viruses. Here, we use a multiplexed peptide assay ('PepSeq') to generate an epitope-resolved view of reactivity across all human coronaviruses. PepSeq accurately detects SARS-CoV-2 exposure and resolves epitopes across the Spike and Nucleocapsid proteins. Two of these represent recurrent reactivities to conserved, functionally-important sites in the Spike S2 subunit, regions that we show are also targeted for the endemic coronaviruses in pre-pandemic controls. At one of these sites, we demonstrate that the SARS-CoV-2 response strongly and recurrently cross-reacts with the endemic virus hCoV-OC43. Our analyses reveal new diagnostic and therapeutic targets, including a site at which SARS-CoV-2 may recruit common pre-existing antibodies and with the potential for broadly-neutralizing responses.
Collapse
Affiliation(s)
- Jason T Ladner
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Sierra N Henson
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Annalee S Boyle
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Anna L Engelbrektson
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Zane W Fink
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Fatima Rahee
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | | | | | - Mars Stone
- Vitalant Research Institute, San Francisco, CA, USA
| | - Wenjuan Dong
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Sanjeet Dadwal
- Division of Infectious Diseases, City of Hope National Medical Center, Duarte, CA, USA
| | - Jianhua Yu
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Michael A Caligiuri
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Piotr Cieplak
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Magnar Bjørås
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mona H Fenstad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Immunology and Transfusion Medicine, St. Olavs Hospital, Trondheim, Norway
| | - Svein A Nordbø
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Medical Microbiology, St. Olavs Hospital, Trondheim, Norway
| | - Denis E Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | - Sergey A Shiryaev
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - John A Altin
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| |
Collapse
|
18
|
Implications of antibody-dependent enhancement of infection for SARS-CoV-2 countermeasures. Nat Biotechnol 2020; 38:789-791. [PMID: 32504046 DOI: 10.1038/s41587-020-0577-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
19
|
Nguyen HQ, Roy J, Harink B, Damle NP, Latorraca NR, Baxter BC, Brower K, Longwell SA, Kortemme T, Thorn KS, Cyert MS, Fordyce PM. Quantitative mapping of protein-peptide affinity landscapes using spectrally encoded beads. eLife 2019; 8:e40499. [PMID: 31282865 PMCID: PMC6728138 DOI: 10.7554/elife.40499] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 07/03/2019] [Indexed: 12/22/2022] Open
Abstract
Transient, regulated binding of globular protein domains to Short Linear Motifs (SLiMs) in disordered regions of other proteins drives cellular signaling. Mapping the energy landscapes of these interactions is essential for deciphering and perturbing signaling networks but is challenging due to their weak affinities. We present a powerful technology (MRBLE-pep) that simultaneously quantifies protein binding to a library of peptides directly synthesized on beads containing unique spectral codes. Using MRBLE-pep, we systematically probe binding of calcineurin (CN), a conserved protein phosphatase essential for the immune response and target of immunosuppressants, to the PxIxIT SLiM. We discover that flanking residues and post-translational modifications critically contribute to PxIxIT-CN affinity and identify CN-binding peptides based on multiple scaffolds with a wide range of affinities. The quantitative biophysical data provided by this approach will improve computational modeling efforts, elucidate a broad range of weak protein-SLiM interactions, and revolutionize our understanding of signaling networks.
Collapse
Affiliation(s)
- Huy Quoc Nguyen
- Department of GeneticsStanford UniversityStanfordUnited States
| | - Jagoree Roy
- Department of BiologyStanford UniversityStanfordUnited States
| | - Björn Harink
- Department of GeneticsStanford UniversityStanfordUnited States
| | - Nikhil P Damle
- Department of BiologyStanford UniversityStanfordUnited States
| | | | - Brian C Baxter
- Department of Biochemistry and BiophysicsUniversity of California, San FranciscoSan FranciscoUnited States
| | - Kara Brower
- Department of BioengineeringStanford UniversityStanfordUnited States
| | - Scott A Longwell
- Department of BioengineeringStanford UniversityStanfordUnited States
| | - Tanja Kortemme
- Department of Bioengineering and Therapeutic SciencesUniversity of California, San FranciscoSan FranciscoUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Kurt S Thorn
- Department of Biochemistry and BiophysicsUniversity of California, San FranciscoSan FranciscoUnited States
| | - Martha S Cyert
- Department of BiologyStanford UniversityStanfordUnited States
| | - Polly Morrell Fordyce
- Department of GeneticsStanford UniversityStanfordUnited States
- Department of BioengineeringStanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
- ChEM-H InstituteStanford UniversityStanfordUnited States
| |
Collapse
|
20
|
Mattes DS, Jung N, Weber LK, Bräse S, Breitling F. Miniaturized and Automated Synthesis of Biomolecules-Overview and Perspectives. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1806656. [PMID: 31033052 DOI: 10.1002/adma.201806656] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 02/02/2019] [Indexed: 06/09/2023]
Abstract
Chemical synthesis is performed by reacting different chemical building blocks with defined stoichiometry, while meeting additional conditions, such as temperature and reaction time. Such a procedure is especially suited for automation and miniaturization. Life sciences lead the way to synthesizing millions of different oligonucleotides in extremely miniaturized reaction sites, e.g., pinpointing active genes in whole genomes, while chemistry advances different types of automation. Recent progress in matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) imaging could match miniaturized chemical synthesis with a powerful analytical tool to validate the outcome of many different synthesis pathways beyond applications in the life sciences. Thereby, due to the radical miniaturization of chemical synthesis, thousands of molecules can be synthesized. This in turn should allow ambitious research, e.g., finding novel synthesis routes or directly screening for photocatalysts. Herein, different technologies are discussed that might be involved in this endeavor. A special emphasis is given to the obstacles that need to be tackled when depositing tiny amounts of materials to many different extremely miniaturized reaction sites.
Collapse
Affiliation(s)
- Daniela S Mattes
- Institute of Microstructure Technology (IMT), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Nicole Jung
- Institute of Organic Chemistry (IOC), Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, 76131, Karlsruhe, Germany
| | - Laura K Weber
- Institute of Microstructure Technology (IMT), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Stefan Bräse
- Institute of Organic Chemistry (IOC), Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, 76131, Karlsruhe, Germany
| | - Frank Breitling
- Institute of Microstructure Technology (IMT), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
21
|
Saini G, Trenchevska O, Howell LJ, Boyd JG, Smith DP, Jain V, Linford MR. Performance Comparison of Three Chemical Vapor Deposited Aminosilanes in Peptide Synthesis: Effects of Silane on Peptide Stability and Purity. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:11925-11932. [PMID: 30208711 DOI: 10.1021/acs.langmuir.8b01298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Silicon oxide substrates underwent gas-phase functionalization with various aminosilanes, and the resulting surfaces were evaluated for their suitability as a solid support for solid phase peptide synthesis (SPPS). APTES (3-aminopropyltriethoxysilane), APDEMS (3-aminopropyldiethoxymethylsilane), and APDIPES (3-aminopropyldiisopropylethoxysilane) were individually applied to thermal oxide-terminated silicon substrates via gas-phase deposition. Coated surfaces were characterized by spectroscopic ellipsometry (SE), contact angle goniometry, X-ray photoelectron spectroscopy (XPS), atomic force microscopy (AFM), and spectrophotometry. Model oligopeptides with 16 residues were synthesized on the amino surfaces, and the chemical stabilities of the resulting surfaces were evaluated against a stringent side chain deprotection (SCD) step, which contained trifluoroacetic acid (TFA) and trifluoromethanesulfonic acid (TFMSA). Functionalized surface thickness loss during SCD was most acute for APDIPES and the observed relative stability order was APTES > APDEMS > APDIPES. Amino surfaces were evaluated for compatibility with stepwise peptide synthesis where complete deprotection and coupling cycles are paramount. Model trimer syntheses indicated that routine capping of unreacted amines with acetic anhydride significantly increased purity as measured by MALDI-MS. An inverse correlation between the amine loading density and peptide purity was observed. In general, peptide purity was highest for the lowest amine density APDIPES surface.
Collapse
Affiliation(s)
- Gaurav Saini
- HealthTell Inc. , Chandler , Arizona 85226 , United States
| | | | - Loren J Howell
- HealthTell Inc. , Chandler , Arizona 85226 , United States
| | - James G Boyd
- HealthTell Inc. , Chandler , Arizona 85226 , United States
| | - David P Smith
- HealthTell Inc. , Chandler , Arizona 85226 , United States
| | - Varun Jain
- Department of Chemistry and Biochemistry , Brigham Young University , Provo , Utah 84602 , United States
| | - Matthew R Linford
- Department of Chemistry and Biochemistry , Brigham Young University , Provo , Utah 84602 , United States
| |
Collapse
|
22
|
Guiding recombinant antivenom development by omics technologies. N Biotechnol 2018; 45:19-27. [DOI: 10.1016/j.nbt.2017.05.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/16/2017] [Indexed: 11/23/2022]
|
23
|
Li X, Li H, Hu Q, Lin J, Zhang Q, Li Y, Li J, Chen T, Zhang Q, Qiu Y. Detection of epitopes in systemic lupus erythematosus using peptide microarray. Mol Med Rep 2018. [PMID: 29532871 PMCID: PMC5928640 DOI: 10.3892/mmr.2018.8710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a common autoimmune disease, which features the secretion of antibodies directed against autoantigens in vivo. In the present study, a peptide microarray was developed to detect the epitopes recognized by autoantibodies in patients with SLE for an effective method of diagnosis. SLE-associated epitopes in 14 autoantigens were predicted using the antigenic epitope prediction software DNA star. Peptides were synthesized based on the predicted antigenic epitopes and immobilized on a slide surface and developed into a peptide microarray. Using this peptide microarray the autoantibodies in 120 patients with SLE and 110 healthy subjects were detected. A total of 73 potential antigenic epitopes in 14 autoantigens were predicted and screened. The peptide microarray based on the 73 epitopes was used to detect the autoantibodies in patients with SLE. A total of 14 epitopes with potential diagnostic values were screened out. The sensitivity and specificity of the 14 epitopes for the diagnosis of SLE were 71.6 and 85.8%, respectively. An optimal set of epitopes for SLE diagnosis was obtained. As individual patients had a specific autoantibody spectrum it was possible to detect autoantibodies in SLE and perform the diagnosis of SLE using the peptide microarray.
Collapse
Affiliation(s)
- Xin Li
- Clinical Laboratory of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Haixia Li
- Clinical Laboratory of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiongdan Hu
- Department of Nephrology, The Traditional Chinese Medicine Hospital, Affiliated to Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jinfei Lin
- South China Institute of Microbial Ecology and Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiong Zhang
- Clinical Laboratory of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yao Li
- Clinical Laboratory of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Juan Li
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Tao Chen
- South China Institute of Microbial Ecology and Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiong Zhang
- Department of Nephrology, The Traditional Chinese Medicine Hospital, Affiliated to Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yurong Qiu
- Clinical Laboratory of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
24
|
Schwaminger S, Blank‐Shim SA, Borkowska‐Panek M, Anand P, Fraga‐García P, Fink K, Wenzel W, Berensmeier S. Experimental characterization and simulation of amino acid and peptide interactions with inorganic materials. Eng Life Sci 2018; 18:84-100. [PMID: 32624891 PMCID: PMC6999452 DOI: 10.1002/elsc.201700019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 06/02/2017] [Accepted: 07/03/2017] [Indexed: 02/06/2023] Open
Abstract
Inspired by nature, many applications and new materials benefit from the interplay of inorganic materials and biomolecules. A fundamental understanding of complex organic-inorganic interactions would improve the controlled production of nanomaterials and biosensors to the development of biocompatible implants for the human body. Although widely exploited in applications, the interaction of amino acids and peptides with most inorganic surfaces is not fully understood. To date, precisely characterizing complex surfaces of inorganic materials and analyzing surface-biomolecule interactions remain challenging both experimentally and computationally. This article reviews several approaches to characterizing biomolecule-surface interactions and illustrates the advantages and disadvantages of the methods presented. First, we explain how the adsorption mechanism of amino acids/peptides to inorganic surfaces can be determined and how thermodynamic and kinetic process constants can be obtained. Second, we demonstrate how this data can be used to develop models for peptide-surface interactions. The understanding and simulation of such interactions constitute a basis for developing molecules with high affinity binding domains in proteins for bioprocess engineering and future biomedical technologies.
Collapse
Affiliation(s)
| | | | | | - Priya Anand
- Institute of NanotechnologyKarlsruhe Institute of TechnologyKarlsruheGermany
| | - Paula Fraga‐García
- Bioseparation Engineering GroupTechnical University of MunichMünchenGermany
| | - Karin Fink
- Institute of NanotechnologyKarlsruhe Institute of TechnologyKarlsruheGermany
| | - Wolfgang Wenzel
- Institute of NanotechnologyKarlsruhe Institute of TechnologyKarlsruheGermany
| | - Sonja Berensmeier
- Bioseparation Engineering GroupTechnical University of MunichMünchenGermany
| |
Collapse
|
25
|
Martin LJ, Akhavan B, Bilek MMM. Electric fields control the orientation of peptides irreversibly immobilized on radical-functionalized surfaces. Nat Commun 2018; 9:357. [PMID: 29367659 PMCID: PMC5783936 DOI: 10.1038/s41467-017-02545-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/07/2017] [Indexed: 01/12/2023] Open
Abstract
Surface functionalization of an implantable device with bioactive molecules can overcome adverse biological responses by promoting specific local tissue integration. Bioactive peptides have advantages over larger protein molecules due to their robustness and sterilizability. Their relatively small size presents opportunities to control the peptide orientation on approach to a surface to achieve favourable presentation of bioactive motifs. Here we demonstrate control of the orientation of surface-bound peptides by tuning electric fields at the surface during immobilization. Guided by computational simulations, a peptide with a linear conformation in solution is designed. Electric fields are used to control the peptide approach towards a radical-functionalized surface. Spontaneous, irreversible immobilization is achieved when the peptide makes contact with the surface. Our findings show that control of both peptide orientation and surface concentration is achieved simply by varying the solution pH or by applying an electric field as delivered by a small battery.
Collapse
Affiliation(s)
- Lewis J Martin
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia
| | - Behnam Akhavan
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia.
- School of Aerospace, Mechanical and Mechatronic Engineering, University of Sydney, Sydney, NSW, 2006, Australia.
| | - Marcela M M Bilek
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia.
- School of Aerospace, Mechanical and Mechatronic Engineering, University of Sydney, Sydney, NSW, 2006, Australia.
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia.
- University of Sydney Nano Institute, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
26
|
Affiliation(s)
- Lindsey C. Szymczak
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Hsin-Yu Kuo
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Milan Mrksich
- Institute of Chemical Biology and Nanomedicine, Hunan University, Changsha 410082, China
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
27
|
A Simple Platform for the Rapid Development of Antimicrobials. Sci Rep 2017; 7:17610. [PMID: 29242618 PMCID: PMC5730575 DOI: 10.1038/s41598-017-17941-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/29/2017] [Indexed: 11/09/2022] Open
Abstract
Recent infectious outbreaks highlight the need for platform technologies that can be quickly deployed to develop therapeutics needed to contain the outbreak. We present a simple concept for rapid development of new antimicrobials. The goal was to produce in as little as one week thousands of doses of an intervention for a new pathogen. We tested the feasibility of a system based on antimicrobial synbodies. The system involves creating an array of 100 peptides that have been selected for broad capability to bind and/or kill viruses and bacteria. The peptides are pre-screened for low cell toxicity prior to large scale synthesis. Any pathogen is then assayed on the chip to find peptides that bind or kill it. Peptides are combined in pairs as synbodies and further screened for activity and toxicity. The lead synbody can be quickly produced in large scale, with completion of the entire process in one week.
Collapse
|
28
|
Cha SW, Bonissone S, Na S, Pevzner PA, Bafna V. The Antibody Repertoire of Colorectal Cancer. Mol Cell Proteomics 2017; 16:2111-2124. [PMID: 29046389 PMCID: PMC5724175 DOI: 10.1074/mcp.ra117.000397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Indexed: 12/31/2022] Open
Abstract
Immunotherapy is becoming increasingly important in the fight against cancers, using and manipulating the body's immune response to treat tumors. Understanding the immune repertoire-the collection of immunological proteins-of treated and untreated cells is possible at the genomic, but technically difficult at the protein level. Standard protein databases do not include the highly divergent sequences of somatic rearranged immunoglobulin genes, and may lead to miss identifications in a mass spectrometry search. We introduce a novel proteogenomic approach, AbScan, to identify these highly variable antibody peptides, by developing a customized antibody database construction method using RNA-seq reads aligned to immunoglobulin (Ig) genes.AbScan starts by filtering transcript (RNA-seq) reads that match the template for Ig genes. The retained reads are used to construct a repertoire graph using the "split" de Bruijn graph: a graph structure that improves on the standard de Bruijn graph to capture the high diversity of Ig genes in a compact manner. AbScan corrects for sequencing errors, and converts the graph to a format suitable for searching with MS/MS search tools. We used AbScan to create an antibody database from 90 RNA-seq colorectal tumor samples. Next, we used proteogenomic analysis to search MS/MS spectra of matched colorectal samples from the Clinical Proteomic Tumor Analysis Consortium (CPTAC) against the AbScan generated database. AbScan identified 1,940 distinct antibody peptides. Correlating with previously identified Single Amino-Acid Variants (SAAVs) in the tumor samples, we identified 163 pairs (antibody peptide, SAAV) with significant cooccurrence pattern in the 90 samples. The presence of coexpressed antibody and mutated peptides was correlated with survival time of the individuals. Our results suggest that AbScan (https://github.com/csw407/AbScan.git) is an effective tool for a proteomic exploration of the immune response in cancers.
Collapse
Affiliation(s)
- Seong Won Cha
- From the ‡Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, California
| | | | - Seungjin Na
- ¶Department of Computer Science and Engineering, University of California, San Diego, La Jolla, California 92037
| | - Pavel A Pevzner
- ¶Department of Computer Science and Engineering, University of California, San Diego, La Jolla, California 92037
| | - Vineet Bafna
- ¶Department of Computer Science and Engineering, University of California, San Diego, La Jolla, California 92037
| |
Collapse
|
29
|
Blank-Shim SA, Schwaminger SP, Borkowska-Panek M, Anand P, Yamin P, Fraga-García P, Fink K, Wenzel W, Berensmeier S. Binding patterns of homo-peptides on bare magnetic nanoparticles: insights into environmental dependence. Sci Rep 2017; 7:14047. [PMID: 29070786 PMCID: PMC5656586 DOI: 10.1038/s41598-017-13928-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 10/04/2017] [Indexed: 12/30/2022] Open
Abstract
Magnetic nanoparticles (MNP) are intensively investigated for applications in nanomedicine, catalysis and biotechnology, where their interaction with peptides and proteins plays an important role. However, the characterisation of the interaction of individual amino acids with MNP remains challenging. Here, we classify the affinity of 20 amino acid homo-hexamers to unmodified iron oxide nanoparticles using peptide arrays in a variety of conditions as a basis to identify and rationally design selectively binding peptides. The choice of buffer system is shown to strongly influence the availability of peptide binding sites on the MNP surface. We find that under certain buffer conditions peptides of different charges can bind the MNP and that the relative strength of the interactions can be modulated by changing the buffer. We further present a model for the competition between the buffer and the MNP's electrostatically binding to the adsorption sites. Thereby, we demonstrate that the charge distribution on the surface can be used to correlate the binding of positively and negatively charged peptides to the MNP. This analysis enables us to engineer the binding of MNP on peptides and contribute to better understand the bio-nano interactions, a step towards the design of affinity tags for advanced biomaterials.
Collapse
Affiliation(s)
- Silvia A Blank-Shim
- Bioseparation Engineering Group, Department of Mechanical Engineering, Technical University of Munich, 85748, Garching b. München, Germany
| | - Sebastian P Schwaminger
- Bioseparation Engineering Group, Department of Mechanical Engineering, Technical University of Munich, 85748, Garching b. München, Germany
| | - Monika Borkowska-Panek
- Institute of Nanotechnology, Karlsruhe Institute of Technology, 76344, Eggenstein-Leopoldshafen, Germany
| | - Priya Anand
- Institute of Nanotechnology, Karlsruhe Institute of Technology, 76344, Eggenstein-Leopoldshafen, Germany
| | - Peyman Yamin
- Institute of Nanotechnology, Karlsruhe Institute of Technology, 76344, Eggenstein-Leopoldshafen, Germany
| | - Paula Fraga-García
- Bioseparation Engineering Group, Department of Mechanical Engineering, Technical University of Munich, 85748, Garching b. München, Germany
| | - Karin Fink
- Institute of Nanotechnology, Karlsruhe Institute of Technology, 76344, Eggenstein-Leopoldshafen, Germany
| | - Wolfgang Wenzel
- Institute of Nanotechnology, Karlsruhe Institute of Technology, 76344, Eggenstein-Leopoldshafen, Germany.
| | - Sonja Berensmeier
- Bioseparation Engineering Group, Department of Mechanical Engineering, Technical University of Munich, 85748, Garching b. München, Germany.
| |
Collapse
|
30
|
Yang H, Liang N, Wang M, Fei Y, Sun J, Li Z, Xu Y, Guo C, Cao Z, Li S, Jiao Y. Long noncoding RNA MALAT-1 is a novel inflammatory regulator in human systemic lupus erythematosus. Oncotarget 2017; 8:77400-77406. [PMID: 29100395 PMCID: PMC5652787 DOI: 10.18632/oncotarget.20490] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/24/2017] [Indexed: 11/29/2022] Open
Abstract
Despite growing evidence that Long noncoding RNAs (lncRNAs) can regulate gene expression and widely take part in autoimmune and inflammatory diseases, our knowledge of systemic lupus erythematosus (SLE)-related lincRNAs remains limited. In this study, we aimed to explore the contribution of the lncRNA metastasis associated lung adenocarcinoma transcript 1 (MALAT1) to the pathogenesis of SLE. PBMCs were obtained from SLE patients and healthy donors. The expression levels of MALAT-1 were measured by quantitative PCR. Small interfering RNA (siRNA) was then used to knock down the expression of MALAT1 in order to determine the role of MALAT1 in the expression levels of IL-21 and SIRT1 signaling pathway in primary monocytes of SLE patients. Here, we found MALAT-1 expression was abnormally increased in SLE patients and predominantly expressed in human monocytes. Additionally, silencing MALAT-1 significantly reduced the expression of IL-21 in primary monocytes of SLE patients. Furthermore, MALAT-1 exerts its detrimental effects by regulating SIRT1 signaling. Our results demonstrate that MALAT-1 is the key regulatory factor in the pathogenesis of SLE and provides potentially novel target for therapeutic intervention.
Collapse
Affiliation(s)
- Huaxia Yang
- Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, The Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Min Wang
- Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, The Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunyun Fei
- Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, The Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian Sun
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiyuan Li
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Xu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhili Cao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuchen Jiao
- National Key Laboratory of Molecular Oncology, Chinese Academy of Medical Sciences Cancer Hospital, Beijing, China
| |
Collapse
|
31
|
Cornett EM, Dickson BM, Rothbart SB. Analysis of Histone Antibody Specificity with Peptide Microarrays. J Vis Exp 2017. [PMID: 28809825 DOI: 10.3791/55912] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Post-translational modifications (PTMs) on histone proteins are widely studied for their roles in regulating chromatin structure and gene expression. The mass production and distribution of antibodies specific to histone PTMs has greatly facilitated research on these marks. As histone PTM antibodies are key reagents for many chromatin biochemistry applications, rigorous analysis of antibody specificity is necessary for accurate data interpretation and continued progress in the field. This protocol describes an integrated pipeline for the design, fabrication and use of peptide microarrays for profiling the specificity of histone antibodies. The design and analysis aspects of this procedure are facilitated by ArrayNinja, an open-source and interactive software package we recently developed to streamline the customization of microarray print formats. This pipeline has been used to screen a large number of commercially available and widely used histone PTM antibodies, and data generated from these experiments are freely available through an online and expanding Histone Antibody Specificity Database. Beyond histones, the general methodology described herein can be applied broadly to the analysis of PTM-specific antibodies.
Collapse
|
32
|
Yiu G, Rasmussen TK, Ajami B, Haddon DJ, Chu AD, Tangsombatvisit S, Haynes WA, Diep V, Steinman L, Faix J, Utz PJ. Development of Th17-Associated Interstitial Kidney Inflammation in Lupus-Prone Mice Lacking the Gene Encoding STAT-1. Arthritis Rheumatol 2017; 68:1233-44. [PMID: 26636548 DOI: 10.1002/art.39535] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 11/24/2015] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Type I interferon (IFN) signaling is a central pathogenic pathway in systemic lupus erythematosus (SLE), and therapeutics targeting type I IFN signaling are in development. Multiple proteins with overlapping functions play a role in IFN signaling, but the signaling events downstream of receptor engagement are unclear. This study was undertaken to investigate the roles of the type I and type II IFN signaling components IFN-α/β/ω receptor 2 (IFNAR-2), IFN regulatory factor 9 (IRF-9), and STAT-1 in a mouse model of SLE. METHODS We used immunohistochemical staining and highly multiplexed assays to characterize pathologic changes in histology, autoantibody production, cytokine/chemokine profiles, and STAT phosphorylation in order to investigate the individual roles of IFNAR-2, IRF-9, and STAT-1 in MRL/lpr mice. RESULTS We found that STAT-1(-/-) mice, but not IRF-9(-/-) or IFNAR-2(-/-) mice, developed interstitial nephritis characterized by infiltration with retinoic acid receptor-related orphan nuclear receptor γt-positive lymphocytes, macrophages, and eosinophils. Despite pronounced interstitial kidney disease and abnormal kidney function, STAT-1(-/-) mice had decreased proteinuria, glomerulonephritis, and autoantibody production. Phosphospecific flow cytometry revealed shunting of STAT phosphorylation from STAT-1 to STAT-3/4. CONCLUSION We describe unique contributions of STAT-1 to pathology in different kidney compartments in a mouse model, and provide potentially novel insight into tubulointerstitial nephritis, a poorly understood complication that predicts end-stage kidney disease in SLE patients.
Collapse
Affiliation(s)
- Gloria Yiu
- Stanford University School of Medicine, Stanford, California
| | - Tue K Rasmussen
- Stanford University School of Medicine, Stanford, California, and Aarhus University, Aarhus, Denmark
| | - Bahareh Ajami
- Stanford University School of Medicine, Stanford, California
| | - David J Haddon
- Stanford University School of Medicine, Stanford, California
| | - Alvina D Chu
- Stanford University School of Medicine, Stanford, California
| | | | | | - Vivian Diep
- Stanford University School of Medicine, Stanford, California
| | - Larry Steinman
- Stanford University School of Medicine and Institute for Immunity, Transplantation, and Infection, Stanford, California
| | - James Faix
- Stanford University School of Medicine, Stanford, California
| | - Paul J Utz
- Stanford University School of Medicine and Institute for Immunity, Transplantation, and Infection, Stanford, California
| |
Collapse
|
33
|
Zandian A, Forsström B, Häggmark-Månberg A, Schwenk JM, Uhlén M, Nilsson P, Ayoglu B. Whole-Proteome Peptide Microarrays for Profiling Autoantibody Repertoires within Multiple Sclerosis and Narcolepsy. J Proteome Res 2017; 16:1300-1314. [DOI: 10.1021/acs.jproteome.6b00916] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Arash Zandian
- Affinity Proteomics, SciLifeLab,
School of Biotechnology, KTH - Royal Institute of Technology, SE-171 21 Solna, Sweden
| | - Björn Forsström
- Affinity Proteomics, SciLifeLab,
School of Biotechnology, KTH - Royal Institute of Technology, SE-171 21 Solna, Sweden
| | - Anna Häggmark-Månberg
- Affinity Proteomics, SciLifeLab,
School of Biotechnology, KTH - Royal Institute of Technology, SE-171 21 Solna, Sweden
| | - Jochen M. Schwenk
- Affinity Proteomics, SciLifeLab,
School of Biotechnology, KTH - Royal Institute of Technology, SE-171 21 Solna, Sweden
| | - Mathias Uhlén
- Affinity Proteomics, SciLifeLab,
School of Biotechnology, KTH - Royal Institute of Technology, SE-171 21 Solna, Sweden
| | - Peter Nilsson
- Affinity Proteomics, SciLifeLab,
School of Biotechnology, KTH - Royal Institute of Technology, SE-171 21 Solna, Sweden
| | - Burcu Ayoglu
- Affinity Proteomics, SciLifeLab,
School of Biotechnology, KTH - Royal Institute of Technology, SE-171 21 Solna, Sweden
| |
Collapse
|
34
|
Lee JR, Haddon DJ, Gupta N, Price JV, Credo GM, Diep VK, Kim K, Hall DA, Baechler EC, Petri M, Varma M, Utz PJ, Wang SX. High-Resolution Analysis of Antibodies to Post-Translational Modifications Using Peptide Nanosensor Microarrays. ACS NANO 2016; 10:10652-10660. [PMID: 27636738 PMCID: PMC5367622 DOI: 10.1021/acsnano.6b03786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Autoantibodies are a hallmark of autoimmune diseases such as lupus and have the potential to be used as biomarkers for diverse diseases, including immunodeficiency, infectious disease, and cancer. More precise detection of antibodies to specific targets is needed to improve diagnosis of such diseases. Here, we report the development of reusable peptide microarrays, based on giant magnetoresistive (GMR) nanosensors optimized for sensitively detecting magnetic nanoparticle labels, for the detection of antibodies with a resolution of a single post-translationally modified amino acid. We have also developed a chemical regeneration scheme to perform multiplex assays with a high level of reproducibility, resulting in greatly reduced experimental costs. In addition, we show that peptides synthesized directly on the nanosensors are approximately two times more sensitive than directly spotted peptides. Reusable peptide nanosensor microarrays enable precise detection of autoantibodies with high resolution and sensitivity and show promise for investigating antibody-mediated immune responses to autoantigens, vaccines, and pathogen-derived antigens as well as other fundamental peptide-protein interactions.
Collapse
Affiliation(s)
- Jung-Rok Lee
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - D. James Haddon
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, California 94305, United States
| | - Nidhi Gupta
- Intel Corporation, Santa Clara, California 95052, United States
| | - Jordan V. Price
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, California 94305, United States
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, Berkeley, California 94720, United States
| | - Grace M. Credo
- Intel Corporation, Santa Clara, California 95052, United States
| | - Vivian K. Diep
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, California 94305, United States
| | - Kyunglok Kim
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - Drew A. Hall
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
- Department of Electrical and Computer Engineering, University of California, San Diego, California 92093, United States
| | - Emily C. Baechler
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, United States
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Madoo Varma
- Intel Corporation, Santa Clara, California 95052, United States
| | - Paul J. Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, California 94305, United States
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Shan X. Wang
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
- Corresponding Author.
| |
Collapse
|
35
|
Autoantibodies against Modified Histone Peptides in SLE Patients Are Associated with Disease Activity and Lupus Nephritis. PLoS One 2016; 11:e0165373. [PMID: 27780265 PMCID: PMC5079581 DOI: 10.1371/journal.pone.0165373] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/11/2016] [Indexed: 11/29/2022] Open
Abstract
Persistent exposure of the immune system to death cell debris leads to autoantibodies against chromatin in patients with systemic lupus erythematosus (SLE). Deposition of anti-chromatin/chromatin complexes can instigate inflammation in multiple organs including the kidney. Previously we identified specific cell death-associated histone modifications as targets of autoantibodies in SLE. In this study we addressed, in a large cohort of SLE patients and controls, the question whether plasma reactivities with specific histone peptides associated with serology and clinical features. Plasma from SLE patients with and without lupus nephritis, disease controls, and healthy controls, were tested in ELISA with histone H4 peptide acetylated at lysines 8, 12 and 16 (H4pac), H2B peptide acetylated at lysine 12 (H2Bpac), H3 peptide trimethylated at lysine 27 (H3pme), and their unmodified equivalents. SLE patients displayed a higher reactivity with the modified equivalent of each peptide. Reactivity with H4pac showed both a high sensitivity (89%) and specificity (91%) for SLE, while H2Bpac exhibited a high specificity (96%) but lower sensitivity (69%). Reactivity with H3pme appeared not specific for SLE. Anti-H4pac and anti-H2Bpac reactivity demonstrated a high correlation with disease activity. Moreover, patients reacting with multiple modified histone peptides exhibited higher SLEDAI and lower C3 levels. SLE patients with renal involvement showed higher reactivity with H2B/H2Bpac and a more pronounced reactivity with the modified equivalent of H3pme and H2Bpac. In conclusion, reactivity with H4pac and H2Bpac is specific for SLE patients and correlates with disease activity, whereas reactivity with H2Bpac is in particular associated with lupus nephritis.
Collapse
|
36
|
Zhao C, He B, Wang G, Ma Y, Yang W. Hierarchical PEG-Based 3D Patterns Grafting from Polymer Substrate by Surface Initiated Visible Light Photolithography. Macromol Rapid Commun 2016; 37:1611-1617. [DOI: 10.1002/marc.201600307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/20/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Changwen Zhao
- State Key Laboratory of Chemical Resource Engineering; Beijing University of Chemical Technology; Beijing 100029 China
- Beijing Laboratory of Biomedical Materials; Beijing University of Chemical Technology; Beijing 100029 China
| | - Bin He
- State Key Laboratory of Chemical Resource Engineering; Beijing University of Chemical Technology; Beijing 100029 China
- Beijing Laboratory of Biomedical Materials; Beijing University of Chemical Technology; Beijing 100029 China
| | - Guan Wang
- State Key Laboratory of Chemical Resource Engineering; Beijing University of Chemical Technology; Beijing 100029 China
- Beijing Laboratory of Biomedical Materials; Beijing University of Chemical Technology; Beijing 100029 China
| | - Yuhong Ma
- Key Laboratory of Carbon Fiber and Functional Polymers; Ministry of Education; Beijing University of Chemical Technology; Beijing 100029 China
| | - Wantai Yang
- State Key Laboratory of Chemical Resource Engineering; Beijing University of Chemical Technology; Beijing 100029 China
- Beijing Laboratory of Biomedical Materials; Beijing University of Chemical Technology; Beijing 100029 China
| |
Collapse
|
37
|
High-flexibility combinatorial peptide synthesis with laser-based transfer of monomers in solid matrix material. Nat Commun 2016; 7:11844. [PMID: 27296868 PMCID: PMC4911634 DOI: 10.1038/ncomms11844] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/06/2016] [Indexed: 01/05/2023] Open
Abstract
Laser writing is used to structure surfaces in many different ways in materials and life sciences. However, combinatorial patterning applications are still limited. Here we present a method for cost-efficient combinatorial synthesis of very-high-density peptide arrays with natural and synthetic monomers. A laser automatically transfers nanometre-thin solid material spots from different donor slides to an acceptor. Each donor bears a thin polymer film, embedding one type of monomer. Coupling occurs in a separate heating step, where the matrix becomes viscous and building blocks diffuse and couple to the acceptor surface. Furthermore, we can consecutively deposit two material layers of activation reagents and amino acids. Subsequent heat-induced mixing facilitates an in situ activation and coupling of the monomers. This allows us to incorporate building blocks with click chemistry compatibility or a large variety of commercially available non-activated, for example, posttranslationally modified building blocks into the array's peptides with >17,000 spots per cm2. Peptide arrays are used in areas such as measuring protein-protein interactions, but achieving high density in synthesis is challenging. Here, the authors report a method for the combinatorial synthesis of high density peptides arrays by laser driven sequential transfer of monomers onto acceptor surfaces.
Collapse
|
38
|
Multiplex giant magnetoresistive biosensor microarrays identify interferon-associated autoantibodies in systemic lupus erythematosus. Sci Rep 2016; 6:27623. [PMID: 27279139 PMCID: PMC4899742 DOI: 10.1038/srep27623] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/20/2016] [Indexed: 01/05/2023] Open
Abstract
High titer, class-switched autoantibodies are a hallmark of systemic lupus erythematosus (SLE). Dysregulation of the interferon (IFN) pathway is observed in individuals with active SLE, although the association of specific autoantibodies with chemokine score, a combined measurement of three IFN-regulated chemokines, is not known. To identify autoantibodies associated with chemokine score, we developed giant magnetoresistive (GMR) biosensor microarrays, which allow the parallel measurement of multiple serum antibodies to autoantigens and peptides. We used the microarrays to analyze serum samples from SLE patients and found individuals with high chemokine scores had significantly greater reactivity to 13 autoantigens than individuals with low chemokine scores. Our findings demonstrate that multiple autoantibodies, including antibodies to U1-70K and modified histone H2B tails, are associated with IFN dysregulation in SLE. Further, they show the microarrays are capable of identifying autoantibodies associated with relevant clinical manifestations of SLE, with potential for use as biomarkers in clinical practice.
Collapse
|
39
|
Abstract
Autoantibodies are a key component for the diagnosis, prognosis and monitoring of various diseases. In order to discover novel autoantibody targets, highly multiplexed assays based on antigen arrays hold a great potential and provide possibilities to analyze hundreds of body fluid samples for their reactivity pattern against thousands of antigens in parallel. Here, we provide an overview of the available technologies for producing antigen arrays, highlight some of the technical and methodological considerations and discuss their applications as discovery tools. Together with recent studies utilizing antigen arrays, we give an overview on how the different types of antigen arrays have and will continue to deliver novel insights into autoimmune diseases among several others.
Collapse
|
40
|
Kumar V, Rayan NA, Muratani M, Lim S, Elanggovan B, Xin L, Lu T, Makhija H, Poschmann J, Lufkin T, Ng HH, Prabhakar S. Comprehensive benchmarking reveals H2BK20 acetylation as a distinctive signature of cell-state-specific enhancers and promoters. Genome Res 2016; 26:612-23. [PMID: 26957309 PMCID: PMC4864461 DOI: 10.1101/gr.201038.115] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 03/07/2016] [Indexed: 12/12/2022]
Abstract
Although over 35 different histone acetylation marks have been described, the overwhelming majority of regulatory genomics studies focus exclusively on H3K27ac and H3K9ac. In order to identify novel epigenomic traits of regulatory elements, we constructed a benchmark set of validated enhancers by performing 140 enhancer assays in human T cells. We tested 40 chromatin signatures on this unbiased enhancer set and identified H2BK20ac, a little-studied histone modification, as the most predictive mark of active enhancers. Notably, we detected a novel class of functionally distinct enhancers enriched in H2BK20ac but lacking H3K27ac, which was present in all examined cell lines and also in embryonic forebrain tissue. H2BK20ac was also unique in highlighting cell-type-specific promoters. In contrast, other acetylation marks were present in all active promoters, regardless of cell-type specificity. In stimulated microglial cells, H2BK20ac was more correlated with cell-state-specific expression changes than H3K27ac, with TGF-beta signaling decoupling the two acetylation marks at a subset of regulatory elements. In summary, our study reveals a previously unknown connection between histone acetylation and cell-type-specific gene regulation and indicates that H2BK20ac profiling can be used to uncover new dimensions of gene regulation.
Collapse
Affiliation(s)
- Vibhor Kumar
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Nirmala Arul Rayan
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Masafumi Muratani
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan; Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Stefan Lim
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Bavani Elanggovan
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Lixia Xin
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Tess Lu
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Harshyaa Makhija
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Jeremie Poschmann
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Thomas Lufkin
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore 138672, Singapore; Department of Biology, Clarkson University, Potsdam, New York 13699, USA
| | - Huck Hui Ng
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore 138672, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Shyam Prabhakar
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| |
Collapse
|
41
|
Choung RS, Marietta EV, Van Dyke CT, Brantner TL, Rajasekaran J, Pasricha PJ, Wang T, Bei K, Krishna K, Krishnamurthy HK, Snyder MR, Jayaraman V, Murray JA. Determination of B-Cell Epitopes in Patients with Celiac Disease: Peptide Microarrays. PLoS One 2016; 11:e0147777. [PMID: 26824466 PMCID: PMC4732949 DOI: 10.1371/journal.pone.0147777] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 01/07/2016] [Indexed: 12/12/2022] Open
Abstract
Background Most antibodies recognize conformational or discontinuous epitopes that have a specific 3-dimensional shape; however, determination of discontinuous B-cell epitopes is a major challenge in bioscience. Moreover, the current methods for identifying peptide epitopes often involve laborious, high-cost peptide screening programs. Here, we present a novel microarray method for identifying discontinuous B-cell epitopes in celiac disease (CD) by using a silicon-based peptide array and computational methods. Methods Using a novel silicon-based microarray platform with a multi-pillar chip, overlapping 12-mer peptide sequences of all native and deamidated gliadins, which are known to trigger CD, were synthesized in situ and used to identify peptide epitopes. Results Using a computational algorithm that considered disease specificity of peptide sequences, 2 distinct epitope sets were identified. Further, by combining the most discriminative 3-mer gliadin sequences with randomly interpolated3- or 6-mer peptide sequences, novel discontinuous epitopes were identified and further optimized to maximize disease discrimination. The final discontinuous epitope sets were tested in a confirmatory cohort of CD patients and controls, yielding 99% sensitivity and 100% specificity. Conclusions These novel sets of epitopes derived from gliadin have a high degree of accuracy in differentiating CD from controls, compared with standard serologic tests. The method of ultra-high-density peptide microarray described here would be broadly useful to develop high-fidelity diagnostic tests and explore pathogenesis.
Collapse
Affiliation(s)
- Rok Seon Choung
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States of America
| | - Eric V. Marietta
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States of America
| | - Carol T. Van Dyke
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States of America
| | - Tricia L. Brantner
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States of America
| | | | - Pankaj J. Pasricha
- Center for Neurogastroenterology, Johns Hopkins University, Baltimore, MD, United States of America
| | - Tianhao Wang
- Vibrant Sciences LLC, San Carlos, CA, United States of America
| | - Kang Bei
- Vibrant Sciences LLC, San Carlos, CA, United States of America
| | - Karthik Krishna
- Vibrant Sciences LLC, San Carlos, CA, United States of America
| | | | - Melissa R. Snyder
- Division of Clinical Biochemistry and Immunology, Mayo Clinic, Rochester, MN, United States of America
| | | | - Joseph A. Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States of America
- * E-mail:
| |
Collapse
|
42
|
Hinchliffe TE, Lin ZT, Wu T. Protein arrays for biomarker discovery in lupus. Proteomics Clin Appl 2016; 10:625-34. [PMID: 26684273 DOI: 10.1002/prca.201500060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 11/05/2015] [Accepted: 12/09/2015] [Indexed: 11/08/2022]
Abstract
Lupus is one of the most common autoimmune diseases, yet many mechanisms of its pathogenesis are not fully known. Over the last few years, advances in protein array technology have accelerated rapidly, resulting in many promising insights toward the discovery of novel lupus biomarkers that may become useful in disease diagnosis and management. Still, only two types of analytical protein arrays thus far, being antibody and antigen arrays, have found notable usage toward lupus biomarker discovery. In this review, we summarize current protein array technologies being used for biomarker discoveries in lupus and associated biomarker findings, as well as protein arrays that are likely to be used for lupus biomarker discovery in the near future.
Collapse
Affiliation(s)
- Taylor E Hinchliffe
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Zuan-Tao Lin
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| |
Collapse
|
43
|
Abstract
The diversity of the antigen-specific humoral immune response reflects the interaction of the immune system with pathogens and autoantigens. Peptide microarray analysis opens up new perspectives for the use of antibodies as diagnostic biomarkers and provides unique access to a more differentiated serological diagnosis. This review focusses on latest applications of peptide microarrays for the serologic medical diagnosis of autoimmunity, infectious diseases, and cancer.
Collapse
|
44
|
Wang W, Wang Z, Bu X, Li R, Zhou M, Hu Z. Discovering of Tumor-targeting Peptides using Bi-functional Microarray. Adv Healthc Mater 2015; 4:2802-8. [PMID: 26548577 DOI: 10.1002/adhm.201500724] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Indexed: 01/06/2023]
Abstract
A bi-functional microarray for in situ peptide screening is presented herein, from which an affinity peptide towards EpCAM is screened out for tumor cell capture.
Collapse
Affiliation(s)
- Weizhi Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety; National Center for Nanoscience and Technology of China; Beijing 100190 China
| | - Zihua Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety; National Center for Nanoscience and Technology of China; Beijing 100190 China
| | - Xiangli Bu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety; National Center for Nanoscience and Technology of China; Beijing 100190 China
| | - Ren Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety; National Center for Nanoscience and Technology of China; Beijing 100190 China
| | - Mingxing Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety; National Center for Nanoscience and Technology of China; Beijing 100190 China
| | - Zhiyuan Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety; National Center for Nanoscience and Technology of China; Beijing 100190 China
| |
Collapse
|
45
|
Rother N, van der Vlag J. Disturbed T Cell Signaling and Altered Th17 and Regulatory T Cell Subsets in the Pathogenesis of Systemic Lupus Erythematosus. Front Immunol 2015; 6:610. [PMID: 26648939 PMCID: PMC4663269 DOI: 10.3389/fimmu.2015.00610] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/16/2015] [Indexed: 12/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the presence of autoantibodies against nuclear components. Circulating immune complexes of chromatin and autoantibodies deposit in various tissues leading to inflammation and tissue damage. It has been well documented that autoimmunity in SLE depends on autoreactive T cells. In this review, we summarize the literature that addresses the roles of T cell signaling, and Th17 and regulatory T cells (Tregs) in the development of SLE. T cell receptor (TCR) signaling appears to be aberrant in T cells of patients with SLE. In particular, defects in the TCRζ chain, Syk kinase, and calcium signaling molecules have been associated with SLE, which leads to hyperresponsive autoreactive T cells. Furthermore, in patients with SLE increased numbers of autoreactive Th17 cells have been documented, and Th17 cells appear to be responsible for tissue inflammation and damage. In addition, reduced numbers of Tregs as well as Tregs with an impaired regulatory function have been associated with SLE. The altered balance between the number of Tregs and Th17 cells in SLE may result from changes in the cytokine milieu that favors the development of Th17 cells over Tregs.
Collapse
Affiliation(s)
- Nils Rother
- Department of Nephrology, Radboud University Medical Center, Radboud Institute of Molecular Life Sciences , Nijmegen , Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud University Medical Center, Radboud Institute of Molecular Life Sciences , Nijmegen , Netherlands
| |
Collapse
|
46
|
Secondary Structure Determination of Peptides and Proteins After Immobilization. Methods Mol Biol 2015; 1352:35-50. [PMID: 26490466 DOI: 10.1007/978-1-4939-3037-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The presentation of immobilized peptides and other small biomolecules attached to surfaces can be greatly affected by the attachment chemistry and linking moieties, resulting in altered activity and specificity. For this reason, it is critical to understand how the various aspects of surface immobilization-underlying substrate properties, tether structure, and site of linkage-affect the secondary and quaternary structures of the immobilized species. Here, we present methods for attaching cysteine-containing peptides to quartz surfaces and determining the secondary structure of surface-immobilized peptides. We specifically show that, even when covalently immobilized, changes in peptide conformation can still occur, with measurement occurring in real time.
Collapse
|
47
|
Haddon DJ, Jarrell JA, Diep VK, Wand HE, Price JV, Tangsombatvisit S, Credo GM, Mackey S, Dekker CL, Baechler EC, Liu CL, Varma M, Utz PJ. Mapping epitopes of U1-70K autoantibodies at single-amino acid resolution. Autoimmunity 2015; 48:513-23. [PMID: 26333287 DOI: 10.3109/08916934.2015.1077233] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The mechanisms underlying development of ribonucleoprotein (RNP) autoantibodies are unclear. The U1-70K protein is the predominant target of RNP autoantibodies, and the RNA binding domain has been shown to be the immunodominant autoantigenic region of U1-70K, although the specific epitopes are not known. To precisely map U1-70K epitopes, we developed silicon-based peptide microarrays with >5700 features, corresponding to 843 unique peptides derived from the U1-70K protein. The microarrays feature overlapping peptides, with single-amino acid resolution in length and location, spanning amino acids 110-170 within the U1-70K RNA binding domain. We evaluated the serum IgG of a cohort of patients with systemic lupus erythematosus (SLE; n = 26) using the microarrays, and identified multiple reactive epitopes, including peptides 116-121 and 143-148. Indirect peptide ELISA analysis of the sera of patients with SLE (n = 88) revealed that ∼14% of patients had serum IgG reactivity to 116-121, while reactivity to 143-148 appeared to be limited to a single patient. SLE patients with serum reactivity to 116-121 had significantly lower SLE Disease Activity Index (SLEDAI) scores at the time of sampling, compared to non-reactive patients. Minimal reactivity to the peptides was observed in the sera of healthy controls (n = 92). Competitive ELISA showed antibodies to 116-121 bind a common epitope in U1-70K (68-72) and the matrix protein M1 of human influenza B viruses. Institutional Review Boards approved this study. Knowledge of the precise epitopes of U1-70K autoantibodies may provide insight into the mechanisms of development of anti-RNP, identify potential clinical biomarkers and inform ongoing clinical trails of peptide-based therapeutics.
Collapse
Affiliation(s)
- David James Haddon
- a Department of Medicine , Division of Immunology & Rheumatology, Stanford University School of Medicine , Stanford , CA , USA
| | - Justin Ansel Jarrell
- a Department of Medicine , Division of Immunology & Rheumatology, Stanford University School of Medicine , Stanford , CA , USA
| | - Vivian K Diep
- a Department of Medicine , Division of Immunology & Rheumatology, Stanford University School of Medicine , Stanford , CA , USA
| | - Hannah E Wand
- a Department of Medicine , Division of Immunology & Rheumatology, Stanford University School of Medicine , Stanford , CA , USA
| | - Jordan V Price
- b Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis , University of California at Berkeley , Berkeley , CA , USA
| | - Stephanie Tangsombatvisit
- a Department of Medicine , Division of Immunology & Rheumatology, Stanford University School of Medicine , Stanford , CA , USA
| | | | - Sally Mackey
- d Department of Pediatrics, Division of Infectious Diseases , Stanford University School of Medicine , Stanford , CA , USA , and
| | - Cornelia L Dekker
- d Department of Pediatrics, Division of Infectious Diseases , Stanford University School of Medicine , Stanford , CA , USA , and
| | - Emily C Baechler
- e Department of Medicine , Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School , Minneapolis , MN , USA
| | - Chih Long Liu
- a Department of Medicine , Division of Immunology & Rheumatology, Stanford University School of Medicine , Stanford , CA , USA
| | - Madoo Varma
- c Intel Corporation , Santa Clara , CA , USA
| | - Paul J Utz
- a Department of Medicine , Division of Immunology & Rheumatology, Stanford University School of Medicine , Stanford , CA , USA
| |
Collapse
|
48
|
Achyuthan KE, Wheeler DR. Easy parallel screening of reagent stability, quality control, and metrology in solid phase peptide synthesis (SPPS) and peptide couplings for microarrays. J Pept Sci 2015; 21:751-7. [PMID: 26310933 DOI: 10.1002/psc.2806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/04/2015] [Accepted: 07/06/2015] [Indexed: 12/21/2022]
Abstract
Evaluating the stability of coupling reagents, quality control (QC), and surface functionalization metrology are all critical to the production of high quality peptide microarrays. We describe a broadly applicable screening technique for evaluating the fidelity of solid phase peptide synthesis (SPPS), the stability of activation/coupling reagents, and a microarray surface metrology tool. This technique was used to assess the stability of the activation reagent 1-{[1-(Cyano-2-ethoxy-2-oxo-ethylidenaminooxy)dimethylamino-morpholinomethylene]}methaneaminiumHexafluorophosphate (COMU) (Sigma-Aldrich, St. Louis, MO, USA) by SPPS of Leu-Enkephalin (YGGFL) or the coupling of commercially synthesized YGGFL peptides to (3-aminopropyl)triethyoxysilane-modified glass surfaces. Coupling efficiency was quantitated by fluorescence signaling based on immunoreactivity of the YGGFL motif. It was concluded that COMU solutions should be prepared fresh and used within 5 h when stored at ~23 °C and not beyond 24 h if stored refrigerated, both in closed containers. Caveats to gauging COMU stability by absorption spectroscopy are discussed. Commercial YGGFL peptides needed independent QC, due to immunoreactivity variations for the same sequence synthesized by different vendors. This technique is useful in evaluating the stability of other activation/coupling reagents besides COMU and as a metrology tool for SPPS and peptide microarrays.
Collapse
Affiliation(s)
- Komandoor E Achyuthan
- Biological/Chemical/Physical Microsensors Department, Sandia National Laboratories, 1515 Eubank Blvd., Albuquerque, NM, 87185, USA
| | - David R Wheeler
- Special Technologies Department, Sandia National Laboratories, 1515 Eubank Blvd., Albuquerque, NM, 87185, USA
| |
Collapse
|
49
|
Furman D, Davis MM. New approaches to understanding the immune response to vaccination and infection. Vaccine 2015; 33:5271-81. [PMID: 26232539 DOI: 10.1016/j.vaccine.2015.06.117] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/26/2015] [Accepted: 06/29/2015] [Indexed: 02/06/2023]
Abstract
The immune system is a network of specialized cell types and tissues that communicates via cytokines and direct contact, to orchestrate specific types of defensive responses. Until recently, we could only study immune responses in a piecemeal, highly focused fashion, on major components like antibodies to the pathogen. But recent advances in technology and in our understanding of the many components of the system, innate and adaptive, have made possible a broader approach, where both the multiple responding cells and cytokines in the blood are measured. This systems immunology approach to a vaccine response or an infection gives us a more holistic picture of the different parts of the immune system that are mobilized and should allow us a much better understanding of the pathways and mechanisms of such responses, as well as to predict vaccine efficacy in different populations well in advance of efficacy studies. Here we summarize the different technologies and methods and discuss how they can inform us about the differences between diseases and vaccines, and how they can greatly accelerate vaccine development.
Collapse
Affiliation(s)
- David Furman
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, United States; Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, United States; Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, United States; Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA, United States.
| |
Collapse
|
50
|
Haddon DJ, Diep VK, Price JV, Limb C, Utz PJ, Balboni I. Autoantigen microarrays reveal autoantibodies associated with proliferative nephritis and active disease in pediatric systemic lupus erythematosus. Arthritis Res Ther 2015; 17:162. [PMID: 26081107 PMCID: PMC4493823 DOI: 10.1186/s13075-015-0682-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 06/11/2015] [Indexed: 12/31/2022] Open
Abstract
Introduction Pediatric systemic lupus erythematosus (pSLE) patients often initially present with more active and severe disease than adults, including a higher frequency of lupus nephritis. Specific autoantibodies, including anti-C1q, anti-DNA and anti-alpha-actinin, have been associated with kidney involvement in SLE, and DNA antibodies are capable of initiating early-stage lupus nephritis in severe combined immunodeficiency (SCID) mice. Over 100 different autoantibodies have been described in SLE patients, highlighting the need for comprehensive autoantibody profiling. Knowledge of the antibodies associated with pSLE and proliferative nephritis will increase the understanding of SLE pathogenesis, and may aid in monitoring patients for renal flare. Methods We used autoantigen microarrays composed of 140 recombinant or purified antigens to compare the serum autoantibody profiles of new-onset pSLE patients (n = 45) to healthy controls (n = 17). We also compared pSLE patients with biopsy-confirmed class III or IV proliferative nephritis (n = 23) and without significant renal involvement (n = 18). We performed ELISA with selected autoantigens to validate the microarray findings. We created a multiple logistic regression model, based on the ELISA and clinical information, to predict whether a patient had proliferative nephritis, and used a validation cohort (n = 23) and longitudinal samples (88 patient visits) to test its accuracy. Results Fifty autoantibodies were at significantly higher levels in the sera of pSLE patients compared to healthy controls, including anti-B cell-activating factor (BAFF). High levels of anti-BAFF were associated with active disease. Thirteen serum autoantibodies were present at significantly higher levels in pSLE patients with proliferative nephritis than those without, and we confirmed five autoantigens (dsDNA, C1q, collagens IV and X and aggrecan) by ELISA. Our model, based on ELISA measurements and clinical variables, correctly identified patients with proliferative nephritis with 91 % accuracy. Conclusions Autoantigen microarrays are an ideal platform for identifying autoantibodies associated with both pSLE and specific clinical manifestations of pSLE. Using multiple regression analysis to integrate autoantibody and clinical data permits accurate prediction of clinical manifestations with complex etiologies in pSLE. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0682-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- D James Haddon
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA.
| | - Vivian K Diep
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA.
| | - Jordan V Price
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California at Berkeley, 142 Life Sciences Addition #3200, Berkeley, CA, 94720, USA.
| | - Cindy Limb
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA.
| | - Paul J Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA.
| | - Imelda Balboni
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, Stanford University School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA.
| |
Collapse
|