1
|
Alirezaylavasani A, Skeie LG, Egner IM, Chopra A, Dahl TB, Prebensen C, Vaage JT, Halvorsen B, Lund-Johansen F, Tonby K, Reikvam DH, Stiksrud B, Holter JC, Dyrhol-Riise AM, Munthe LA, Kared H. Vaccine responses and hybrid immunity in people living with HIV after SARS-CoV-2 breakthrough infections. NPJ Vaccines 2024; 9:185. [PMID: 39384763 PMCID: PMC11464709 DOI: 10.1038/s41541-024-00972-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 09/19/2024] [Indexed: 10/11/2024] Open
Abstract
The COVID-19 pandemic posed a challenge for people living with HIV (PLWH), particularly immune non-responders (INR) with compromised CD4 T-cell reconstitution following antiretroviral therapy (CD4 count <350 cells per mm3). Their diminished vaccine responses raised concerns about their vulnerability to SARS-CoV-2 breakthrough infections (BTI). Our in-depth study here revealed chronic inflammation in PLWH and a limited anti-Spike IgG response after vaccination in INR. Nevertheless, the imprinting of Spike-specific B cells by vaccination significantly enhanced the humoral responses after BTI. Notably, the magnitude of cellular CD4 response in all PLWH was comparable to that in healthy donors (HD). However, the polyfunctionality and phenotype of Spike-specific CD8 T cells in INR differed from controls. The findings highlight the need for additional boosters with variant vaccines, and for monitoring ART adherence and the durability of both humoral and cellular anti-SARS-CoV-2 immunity in INR.
Collapse
Affiliation(s)
- Amin Alirezaylavasani
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Linda Gail Skeie
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Ingrid Marie Egner
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Adity Chopra
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Tuva Børresdatter Dahl
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Christian Prebensen
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - John Torgils Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Kristian Tonby
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dag Henrik Reikvam
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgitte Stiksrud
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Jan Cato Holter
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| |
Collapse
|
2
|
Kared H, Alirezaylavasani A, Lund KP, Chopra A, Tietze L, de Matos Kasahara T, Goll GL, Grødeland G, Kaarbø M, Reisæter AV, Hovd M, Heldal K, Vaage JT, Lund-Johansen F, Midtvedt K, Åsberg A, Munthe LA. Hybrid and SARS-CoV-2-vaccine immunity in kidney transplant recipients. EBioMedicine 2023; 97:104833. [PMID: 37844534 PMCID: PMC10585642 DOI: 10.1016/j.ebiom.2023.104833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Kidney transplant recipients (KTR) are at high risk for severe COVID-19 and have demonstrated poor response to vaccination, making it unclear whether successive vaccinations offer immunity and protection. METHODS We conducted a serologically guided interventional study where KTR patients that failed to seroconvert were revaccinated and also monitored seroconversion of KTR following the Norwegian vaccination program. We analysed IgG anti-RBD Spike responses from dose 2 (n = 432) up to after the 6th (n = 37) mRNA vaccine dose. The frequency and phenotype of Spike-specific T and B cell responses were assessed in the interventional cohort after 3-4 vaccine doses (n = 30). Additionally, we evaluated the Specific T and B cell response to breakthrough infection (n = 32), measured inflammatory cytokines and broadly cross-neutralizing antibodies, and defined the incidence of COVID-19-related hospitalizations and deaths. The Norwegian KTR cohort has a male dominance (2323 males, 1297 females), PBMC were collected from 114 male and 78 female donors. FINDINGS After vaccine dose 3, most KTR developed Spike-specific T cell responses but had significantly reduced Spike-binding B cells and few memory cells. The B cell response included a cross-reactive subset that could bind Omicron VOC, which expanded after breakthrough infection (BTI) and gave rise to a memory IgG+ B cell response. After BTI, KTR had increased Spike-specific T cells, emergent non-Spike T and B cell responses, and a systemic inflammatory signature. Late seroconversion occurred after doses 5-6, but 38% (14/37) of KTR had no detectable immunity even after multiple vaccine doses. INTERPRETATION Boosting vaccination can induce Spike-specific immunity that may expand in breakthrough infections highlighting the benefit of vaccination to protect this vulnerable population. FUNDING CEPI and internal funds.
Collapse
Affiliation(s)
- Hassen Kared
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway.
| | - Amin Alirezaylavasani
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Katrine Persgård Lund
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Adity Chopra
- Department of Immunology, Oslo University Hospital, Oslo, Norway; ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lisa Tietze
- Department of Immunology, Oslo University Hospital, Oslo, Norway; ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Guro Løvik Goll
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Gunnveig Grødeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Mari Kaarbø
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Anna Varberg Reisæter
- Department of Transplantation Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Markus Hovd
- Department of Transplantation Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway; Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Kristian Heldal
- Department of Transplantation Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - John Torgils Vaage
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway; ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Karsten Midtvedt
- Department of Transplantation Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Anders Åsberg
- Department of Transplantation Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway; Norwegian Renal Registry, Oslo University Hospital-Rikshospitalet, Oslo, Norway; Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
3
|
Meyer S, Blaas I, Bollineni RC, Delic-Sarac M, Tran TT, Knetter C, Dai KZ, Madssen TS, Vaage JT, Gustavsen A, Yang W, Nissen-Meyer LSH, Douvlataniotis K, Laos M, Nielsen MM, Thiede B, Søraas A, Lund-Johansen F, Rustad EH, Olweus J. Prevalent and immunodominant CD8 T cell epitopes are conserved in SARS-CoV-2 variants. Cell Rep 2023; 42:111995. [PMID: 36656713 PMCID: PMC9826989 DOI: 10.1016/j.celrep.2023.111995] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/16/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023] Open
Abstract
The emergence of SARS-CoV-2 variants of concern (VOC) is driven by mutations that mediate escape from neutralizing antibodies. There is also evidence that mutations can cause loss of T cell epitopes. However, studies on viral escape from T cell immunity have been hampered by uncertain estimates of epitope prevalence. Here, we map and quantify CD8 T cell responses to SARS-CoV-2-specific minimal epitopes in blood drawn from April to June 2020 from 83 COVID-19 convalescents. Among 37 HLA ligands eluted from five prevalent alleles and an additional 86 predicted binders, we identify 29 epitopes with an immunoprevalence ranging from 3% to 100% among individuals expressing the relevant HLA allele. Mutations in VOC are reported in 10.3% of the epitopes, while 20.6% of the non-immunogenic peptides are mutated in VOC. The nine most prevalent epitopes are conserved in VOC. Thus, comprehensive mapping of epitope prevalence does not provide evidence that mutations in VOC are driven by escape of T cell immunity.
Collapse
Affiliation(s)
- Saskia Meyer
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0379 Oslo, Norway,Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Isaac Blaas
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0379 Oslo, Norway,Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Ravi Chand Bollineni
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0379 Oslo, Norway,Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Marina Delic-Sarac
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0379 Oslo, Norway,Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Trung T. Tran
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | - Cathrine Knetter
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0379 Oslo, Norway,Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Ke-Zheng Dai
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | | | - John T. Vaage
- Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway,Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | - Alice Gustavsen
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | - Weiwen Yang
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0379 Oslo, Norway,Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | | | - Karolos Douvlataniotis
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0379 Oslo, Norway,Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Maarja Laos
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0379 Oslo, Norway,Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway,Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Morten Milek Nielsen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0379 Oslo, Norway,Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, 0371 Oslo, Norway
| | - Arne Søraas
- Department of Microbiology, Oslo University Hospital, 0424 Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway,ImmunoLingo Convergence Center, University of Oslo, 0372 Oslo, Norway
| | - Even H. Rustad
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0379 Oslo, Norway,Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway,Corresponding author
| | - Johanna Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, 0379 Oslo, Norway,Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway,Corresponding author
| |
Collapse
|
4
|
Titers of antibodies against ancestral SARS-CoV-2 correlate with levels of neutralizing antibodies to multiple variants. NPJ Vaccines 2022; 7:174. [PMID: 36585405 PMCID: PMC9801350 DOI: 10.1038/s41541-022-00586-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/24/2022] [Indexed: 12/31/2022] Open
Abstract
Diagnostic assays currently used to monitor the efficacy of COVID-19 vaccines measure levels of antibodies to the receptor-binding domain of ancestral SARS-CoV-2 (RBDwt). However, the predictive value for protection against new variants of concern (VOCs) has not been firmly established. Here, we used bead-based arrays and flow cytometry to measure binding of antibodies to spike proteins and receptor-binding domains (RBDs) from VOCs in 12,000 serum samples. Effects of sera on RBD-ACE2 interactions were measured as a proxy for neutralizing antibodies. The samples were obtained from healthy individuals or patients on immunosuppressive therapy who had received two to four doses of COVID-19 vaccines and from COVID-19 convalescents. The results show that anti-RBDwt titers correlate with the levels of binding- and neutralizing antibodies against the Alpha, Beta, Gamma, Delta, Epsilon and Omicron variants. The benefit of multiplexed analysis lies in the ability to measure a wide range of anti-RBD titers using a single dilution of serum for each assay. The reactivity patterns also yield an internal reference for neutralizing activity and binding antibody units per milliliter (BAU/ml). Results obtained with sera from vaccinated healthy individuals and patients confirmed and extended results from previous studies on time-dependent waning of antibody levels and effects of immunosuppressive agents. We conclude that anti-RBDwt titers correlate with levels of neutralizing antibodies against VOCs and propose that our method may be implemented to enhance the precision and throughput of immunomonitoring.
Collapse
|
5
|
Kared H, Wolf AS, Alirezaylavasani A, Ravussin A, Solum G, Tran TT, Lund-Johansen F, Vaage JT, Nissen-Meyer LS, Nygaard UC, Hungnes O, Robertson AH, Næss LM, Trogstad L, Magnus P, Munthe LA, Mjaaland S. Immune responses in Omicron SARS-CoV-2 breakthrough infection in vaccinated adults. Nat Commun 2022; 13:4165. [PMID: 35851055 PMCID: PMC9293966 DOI: 10.1038/s41467-022-31888-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023] Open
Abstract
The SARS-CoV-2 Omicron variant has more than 15 mutations in the receptor binding domain of the Spike protein enabling increased transmissibility and viral escape from antibodies in vaccinated individuals. It is unclear how vaccine immunity protects against Omicron infection. Here we show that vaccinated participants at a super-spreader event have robust recall response of humoral and pre-existing cellular immunity induced by the vaccines, and an emergent de novo T cell response to non-Spike antigens. Individuals with Omicron SARS-CoV-2 breakthrough infections have significantly increased activated SARS-CoV-2 wild type Spike-specific cytotoxic T cells, activated follicular helper (TFH) cells, functional T cell responses, boosted humoral responses, and rapid release of Spike and RBD-specific IgG+ B cell plasmablasts and memory B cells into circulation. Omicron breakthrough infection affords significantly increased de novo memory T cell responses to non-Spike viral antigens. Concerted T and B cell responses may provide durable and broad immunity.
Collapse
Affiliation(s)
- Hassen Kared
- KG Jebsen Centre for B cell malignancy, Institute of Clinical medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
| | - Asia-Sophia Wolf
- Division of Infection Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Amin Alirezaylavasani
- KG Jebsen Centre for B cell malignancy, Institute of Clinical medicine, University of Oslo, Oslo, Norway
| | - Anthony Ravussin
- Division of Infection Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Guri Solum
- Division of Infection Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Trung The Tran
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical medicine, University of Oslo, Oslo, Norway
| | | | | | - Unni C Nygaard
- Division of Infection Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Olav Hungnes
- Division of Infection Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Anna H Robertson
- Division of Infection Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Lisbeth Meyer Næss
- Division of Infection Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Lill Trogstad
- Division of Infection Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Per Magnus
- Center for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Ludvig A Munthe
- KG Jebsen Centre for B cell malignancy, Institute of Clinical medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
| | - Siri Mjaaland
- Division of Infection Control, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
6
|
Riise J, Meyer S, Blaas I, Chopra A, Tran TT, Delic-Sarac M, Hestdalen ML, Brodin E, Rustad EH, Dai KZ, Vaage JT, Nissen-Meyer LSH, Sund F, Wader KF, Bjornevik AT, Meyer PA, Nygaard GO, König M, Smeland S, Lund-Johansen F, Olweus J, Kolstad A. Rituximab-treated lymphoma patients develop strong CD8 T-cell responses following COVID-19 vaccination. Br J Haematol 2022; 197:697-708. [PMID: 35254660 PMCID: PMC9111866 DOI: 10.1111/bjh.18149] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/25/2022] [Accepted: 03/05/2022] [Indexed: 11/29/2022]
Abstract
B‐cell depletion induced by anti‐cluster of differentiation 20 (CD20) monoclonal antibody (mAb) therapy of patients with lymphoma is expected to impair humoral responses to severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2) vaccination, but effects on CD8 T‐cell responses are unknown. Here, we investigated humoral and CD8 T‐cell responses following two vaccinations in patients with lymphoma undergoing anti‐CD20‐mAb therapy as single agent or in combination with chemotherapy or other anti‐neoplastic agents during the last 9 months prior to inclusion, and in healthy age‐matched blood donors. Antibody measurements showed that seven of 110 patients had antibodies to the receptor‐binding domain of the SARS‐CoV‐2 Spike protein 3–6 weeks after the second dose of vaccination. Peripheral blood CD8 T‐cell responses against prevalent human leucocyte antigen (HLA) class I SARS‐CoV‐2 epitopes were determined by peptide‐HLA multimer analysis. Strong CD8 T‐cell responses were observed in samples from 20/29 patients (69%) and 12/16 (75%) controls, with similar median response magnitudes in the groups and some of the strongest responses observed in patients. We conclude that despite the absence of humoral immune responses in fully SARS‐CoV‐2‐vaccinated, anti‐CD20‐treated patients with lymphoma, their CD8 T‐cell responses reach similar frequencies and magnitudes as for controls. Patients with lymphoma on B‐cell depleting therapies are thus likely to benefit from current coronavirus disease 2019 (COVID‐19) vaccines, and development of vaccines aimed at eliciting T‐cell responses to non‐Spike epitopes might provide improved protection.
Collapse
Affiliation(s)
- Jon Riise
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Saskia Meyer
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Isaac Blaas
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Adity Chopra
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Trung T Tran
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Marina Delic-Sarac
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Malu Lian Hestdalen
- Department of Hematology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Ellen Brodin
- Hematological Research Group, Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Even Holth Rustad
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Hematological Research Group, Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Ke-Zheng Dai
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - John Torgils Vaage
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Immunology, Oslo University Hospital, Oslo, Norway
| | | | - Fredrik Sund
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway
| | - Karin F Wader
- Department of Oncology, St Olav University Hospital, Trondheim, Norway
| | - Anne T Bjornevik
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Peter A Meyer
- Department of Oncology and Hematology, Stavanger University Hospital, Stavanger, Norway
| | - Gro O Nygaard
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Marton König
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Sigbjørn Smeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway.,ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Johanna Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Arne Kolstad
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
7
|
Landeira-Viñuela A, Díez P, Juanes-Velasco P, Lécrevisse Q, Orfao A, De Las Rivas J, Fuentes M. Deepening into Intracellular Signaling Landscape through Integrative Spatial Proteomics and Transcriptomics in a Lymphoma Model. Biomolecules 2021; 11:1776. [PMID: 34944421 PMCID: PMC8699084 DOI: 10.3390/biom11121776] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/11/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Human Proteome Project (HPP) presents a systematic characterization of the protein landscape under different conditions using several complementary-omic techniques (LC-MS/MS proteomics, affinity proteomics, transcriptomics, etc.). In the present study, using a B-cell lymphoma cell line as a model, comprehensive integration of RNA-Seq transcriptomics, MS/MS, and antibody-based affinity proteomics (combined with size-exclusion chromatography) (SEC-MAP) were performed to uncover correlations that could provide insights into protein dynamics at the intracellular level. Here, 5672 unique proteins were systematically identified by MS/MS analysis and subcellular protein extraction strategies (neXtProt release 2020-21, MS/MS data are available via ProteomeXchange with identifier PXD003939). Moreover, RNA deep sequencing analysis of this lymphoma B-cell line identified 19,518 expressed genes and 5707 protein coding genes (mapped to neXtProt). Among these data sets, 162 relevant proteins (targeted by 206 antibodies) were systematically analyzed by the SEC-MAP approach, providing information about PTMs, isoforms, protein complexes, and subcellular localization. Finally, a bioinformatic pipeline has been designed and developed for orthogonal integration of these high-content proteomics and transcriptomics datasets, which might be useful for comprehensive and global characterization of intracellular protein profiles.
Collapse
Affiliation(s)
- Alicia Landeira-Viñuela
- Department of Medicine and General Cytometry Service-Nucleus, USAL/IBSAL, 37000 Salamanca, Spain; (A.L.-V.); (P.D.); (P.J.-V.); (Q.L.); (A.O.)
| | - Paula Díez
- Department of Medicine and General Cytometry Service-Nucleus, USAL/IBSAL, 37000 Salamanca, Spain; (A.L.-V.); (P.D.); (P.J.-V.); (Q.L.); (A.O.)
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain
| | - Pablo Juanes-Velasco
- Department of Medicine and General Cytometry Service-Nucleus, USAL/IBSAL, 37000 Salamanca, Spain; (A.L.-V.); (P.D.); (P.J.-V.); (Q.L.); (A.O.)
| | - Quentin Lécrevisse
- Department of Medicine and General Cytometry Service-Nucleus, USAL/IBSAL, 37000 Salamanca, Spain; (A.L.-V.); (P.D.); (P.J.-V.); (Q.L.); (A.O.)
| | - Alberto Orfao
- Department of Medicine and General Cytometry Service-Nucleus, USAL/IBSAL, 37000 Salamanca, Spain; (A.L.-V.); (P.D.); (P.J.-V.); (Q.L.); (A.O.)
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain;
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service-Nucleus, USAL/IBSAL, 37000 Salamanca, Spain; (A.L.-V.); (P.D.); (P.J.-V.); (Q.L.); (A.O.)
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
8
|
Huovinen T, Lindenburg L, Minter R, Hollfelder F. Multiplexed Affinity Characterization of Protein Binders Directly from a Crude Cell Lysate by Covalent Capture on Suspension Bead Arrays. Anal Chem 2021; 93:2166-2173. [PMID: 33397084 PMCID: PMC7861142 DOI: 10.1021/acs.analchem.0c03992] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
![]()
The precise determination of affinity and specificity is a crucial step in the
development of new protein reagents for therapy and diagnostics. Paradoxically, the
selection of protein binders, e.g., antibody fragments, from large combinatorial
repertoires is a rapid process compared to the subsequent characterization of selected
clones. Here we demonstrate the use of suspension bead arrays (SBA) in combination with
flow cytometry to facilitate the post-selection analysis of binder affinities. The array
is designed to capture the proteins of interest (POIs) covalently on the surface of
superparamagnetic color-coded microbeads directly from expression cell lysate, based on
SpyTag-SpyCatcher coupling by isopeptide bond formation. This concept was validated by
analyzing the affinities of a typical phage display output, i.e., clones consisting of
single-chain variable fragment antibodies (scFvs), as SpyCatcher fusions in 12- and
24-plex SBA formats using a standard three-laser flow cytometer. We demonstrate that the
equilibrium dissociation constants (Kd) obtained from
multiplexed SBA assays correlate well with experiments performed on a larger scale,
while the antigen consumption was reduced >100-fold compared to the conventional
96-well plate format. Protein screening and characterization by SBAs is a rapid and
reagent-saving analytical format for combinatorial protein engineering to address
specificity maturation and cross-reactivity profiling of antibodies.
Collapse
Affiliation(s)
- Tuomas Huovinen
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, CB2 1GA Cambridge, U.K
| | - Laurens Lindenburg
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, CB2 1GA Cambridge, U.K
| | - Ralph Minter
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Milstein Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Florian Hollfelder
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, CB2 1GA Cambridge, U.K
| |
Collapse
|
9
|
Holter JC, Pischke SE, de Boer E, Lind A, Jenum S, Holten AR, Tonby K, Barratt-Due A, Sokolova M, Schjalm C, Chaban V, Kolderup A, Tran T, Tollefsrud Gjølberg T, Skeie LG, Hesstvedt L, Ormåsen V, Fevang B, Austad C, Müller KE, Fladeby C, Holberg-Petersen M, Halvorsen B, Müller F, Aukrust P, Dudman S, Ueland T, Andersen JT, Lund-Johansen F, Heggelund L, Dyrhol-Riise AM, Mollnes TE. Systemic complement activation is associated with respiratory failure in COVID-19 hospitalized patients. Proc Natl Acad Sci U S A 2020; 117:25018-25025. [PMID: 32943538 PMCID: PMC7547220 DOI: 10.1073/pnas.2010540117] [Citation(s) in RCA: 261] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Respiratory failure in the acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic is hypothesized to be driven by an overreacting innate immune response, where the complement system is a key player. In this prospective cohort study of 39 hospitalized coronavirus disease COVID-19 patients, we describe systemic complement activation and its association with development of respiratory failure. Clinical data and biological samples were obtained at admission, days 3 to 5, and days 7 to 10. Respiratory failure was defined as PO2/FiO2 ratio of ≤40 kPa. Complement activation products covering the classical/lectin (C4d), alternative (C3bBbP) and common pathway (C3bc, C5a, and sC5b-9), the lectin pathway recognition molecule MBL, and antibody serology were analyzed by enzyme-immunoassays; viral load by PCR. Controls comprised healthy blood donors. Consistently increased systemic complement activation was observed in the majority of COVID-19 patients during hospital stay. At admission, sC5b-9 and C4d were significantly higher in patients with than without respiratory failure (P = 0.008 and P = 0.034). Logistic regression showed increasing odds of respiratory failure with sC5b-9 (odds ratio 31.9, 95% CI 1.4 to 746, P = 0.03) and need for oxygen therapy with C4d (11.7, 1.1 to 130, P = 0.045). Admission sC5b-9 and C4d correlated significantly to ferritin (r = 0.64, P < 0.001; r = 0.69, P < 0.001). C4d, sC5b-9, and C5a correlated with antiviral antibodies, but not with viral load. Systemic complement activation is associated with respiratory failure in COVID-19 patients and provides a rationale for investigating complement inhibitors in future clinical trials.
Collapse
Affiliation(s)
- Jan C Holter
- Department of Microbiology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
| | - Soeren E Pischke
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway;
- Division of Emergencies and Critical Care, Oslo University Hospital, 0424 Oslo, Norway
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | - Eline de Boer
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | - Andreas Lind
- Department of Microbiology, Oslo University Hospital, 0424 Oslo, Norway
| | - Synne Jenum
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Aleksander R Holten
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Acute Medicine, Oslo University Hospital, 0424 Oslo, Norway
| | - Kristian Tonby
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Andreas Barratt-Due
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Division of Emergencies and Critical Care, Oslo University Hospital, 0424 Oslo, Norway
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | - Marina Sokolova
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | - Camilla Schjalm
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | - Viktoriia Chaban
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | - Anette Kolderup
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Pharmacology, University of Oslo, 0315 Oslo, Norway
| | - Trung Tran
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | - Torleif Tollefsrud Gjølberg
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
- Department of Pharmacology, University of Oslo, 0315 Oslo, Norway
- Department of Ophthalmology, Oslo University Hospital, 0424 Oslo, Norway
| | - Linda G Skeie
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Liv Hesstvedt
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Vidar Ormåsen
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Børre Fevang
- Research Institute of Internal Medicine, Oslo University Hospital, 0424 Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Cathrine Austad
- Department of Internal Medicine, Vestre Viken Hospital Trust, 3004 Drammen, Norway
| | - Karl Erik Müller
- Department of Internal Medicine, Vestre Viken Hospital Trust, 3004 Drammen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, 5007 Bergen, Norway
| | - Cathrine Fladeby
- Department of Microbiology, Oslo University Hospital, 0424 Oslo, Norway
| | | | - Bente Halvorsen
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, 0424 Oslo, Norway
| | - Fredrik Müller
- Department of Microbiology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
| | - Pål Aukrust
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, 0424 Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
- Faculty of Health Sciences, K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9037 Tromsø, Norway
| | - Susanne Dudman
- Department of Microbiology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
| | - Thor Ueland
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, 0424 Oslo, Norway
- Faculty of Health Sciences, K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9037 Tromsø, Norway
| | - Jan Terje Andersen
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
- ImmunoLingo Convergence Centre, University of Oslo, 0315 Oslo, Norway
| | - Lars Heggelund
- Department of Internal Medicine, Vestre Viken Hospital Trust, 3004 Drammen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, 5007 Bergen, Norway
| | - Anne M Dyrhol-Riise
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Tom E Mollnes
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- Department of Immunology, Oslo University Hospital, 0424 Oslo, Norway
- Faculty of Health Sciences, K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9037 Tromsø, Norway
- Research Laboratory, Nordland Hospital Bodø, 8092 Bodø, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| |
Collapse
|
10
|
Kalina T, Lundsten K, Engel P. Relevance of Antibody Validation for Flow Cytometry. Cytometry A 2019; 97:126-136. [DOI: 10.1002/cyto.a.23895] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 07/10/2019] [Accepted: 08/22/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Tomas Kalina
- CLIP‐Childhood Leukemia Investigation Prague, Department of Pediatric Hematology and Oncology, 2nd Medical SchoolCharles University and University Hospital Motol Prague Czech Republic
| | | | - Pablo Engel
- Department of Biomedical SciencesFaculty of Medicine and Health Sciences, University of Barcelona Barcelona Spain
| |
Collapse
|
11
|
de la Rosa Carrillo D, Sikorski K, Khnykin D, Wu W, Lund-Johansen F. High-resolution antibody array analysis of proteins from primary human keratinocytes and leukocytes. PLoS One 2018; 13:e0209271. [PMID: 30589857 PMCID: PMC6307719 DOI: 10.1371/journal.pone.0209271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 12/03/2018] [Indexed: 11/19/2022] Open
Abstract
Antibody array analysis of labeled proteomes has high throughput and is simple to perform, but validation remains challenging. Here, we used differential detergent fractionation and size exclusion chromatography in sequence for high-resolution separation of biotinylated proteins from human primary keratinocytes and leukocytes. Ninety-six sample fractions from each cell type were analyzed with microsphere-based antibody arrays and flow cytometry (microsphere affinity proteomics; MAP). Monomeric proteins and multi-molecular complexes in the cytosol, cytoplasmic organelles, membranes and nuclei were resolved as discrete peaks of antibody reactivity across the fractions. The fractionation also provided a two-dimensional matrix for assessment of specificity. Thus, antibody reactivity peaks were considered to represent specific binding if the position in the matrix was in agreement with published information about i) subcellular location, ii) size of the intended target, and iii) cell type-dependent variation in protein expression. Similarities in the reactivity patterns of either different antibodies to the same protein or antibodies to similar proteins were used as additional supporting evidence. This approach provided validation of several hundred proteins and identification of monomeric proteins and protein complexes. High-resolution MAP solves many of the problems associated with obtaining specificity with immobilized antibodies and a protein label. Thus, laboratories with access to chromatography and flow cytometry can perform large-scale protein analysis on a daily basis. This opens new possibilities for cell biology research in dermatology and validation of antibodies.
Collapse
Affiliation(s)
- Daniel de la Rosa Carrillo
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Dermatology, Oslo University Hospital, Oslo, Norway
- * E-mail:
| | - Krzysztof Sikorski
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| | - Denis Khnykin
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Weiwei Wu
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| |
Collapse
|
12
|
Dagher M, Kleinman M, Ng A, Juncker D. Ensemble multicolour FRET model enables barcoding at extreme FRET levels. NATURE NANOTECHNOLOGY 2018; 13:925-932. [PMID: 30061659 DOI: 10.1038/s41565-018-0205-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 06/17/2018] [Indexed: 05/02/2023]
Abstract
Quantitative models of Förster resonance energy transfer (FRET)-pioneered by Förster-define our understanding of FRET and underpin its widespread use. However, multicolour FRET (mFRET), which arises between multiple, stochastically distributed fluorophores, lacks a mechanistic model and remains intractable. mFRET notably arises in fluorescently barcoded microparticles, resulting in a complex, non-orthogonal fluorescence response that impedes their encoding and decoding. Here, we introduce an ensemble mFRET (emFRET) model, and apply it to guide barcoding into regimes with extreme FRET. We further introduce a facile, proportional multicolour labelling method using oligonucleotides as homogeneous linkers. A total of 580 barcodes were rapidly designed and validated using four dyes-with FRET efficiencies reaching 76%-and used for multiplexed immunoassays with cytometric readout and fully automated decoding. The emFRET model helps to expand the barcoding capacity of barcoded microparticles using common organic dyes and will benefit other applications subject to stochastic mFRET.
Collapse
Affiliation(s)
- Milad Dagher
- Biomedical Engineering Department, McGill University, Montreal, Quebec, Canada
- McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada
| | - Michael Kleinman
- Biomedical Engineering Department, McGill University, Montreal, Quebec, Canada
- McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada
| | - Andy Ng
- Biomedical Engineering Department, McGill University, Montreal, Quebec, Canada
- McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada
| | - David Juncker
- Biomedical Engineering Department, McGill University, Montreal, Quebec, Canada.
- McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada.
- Neurology and Neurosurgery Department, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
13
|
Strønen E, Toebes M, Kelderman S, van Buuren MM, Yang W, van Rooij N, Donia M, Böschen ML, Lund-Johansen F, Olweus J, Schumacher TN. Targeting of cancer neoantigens with donor-derived T cell receptor repertoires. Science 2016; 352:1337-41. [PMID: 27198675 DOI: 10.1126/science.aaf2288] [Citation(s) in RCA: 338] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 05/04/2016] [Indexed: 12/14/2022]
Abstract
Accumulating evidence suggests that clinically efficacious cancer immunotherapies are driven by T cell reactivity against DNA mutation-derived neoantigens. However, among the large number of predicted neoantigens, only a minority is recognized by autologous patient T cells, and strategies to broaden neoantigen-specific T cell responses are therefore attractive. We found that naïve T cell repertoires of healthy blood donors provide a source of neoantigen-specific T cells, responding to 11 of 57 predicted human leukocyte antigen (HLA)-A*02:01-binding epitopes from three patients. Many of the T cell reactivities involved epitopes that in vivo were neglected by patient autologous tumor-infiltrating lymphocytes. Finally, T cells redirected with T cell receptors identified from donor-derived T cells efficiently recognized patient-derived melanoma cells harboring the relevant mutations, providing a rationale for the use of such "outsourced" immune responses in cancer immunotherapy.
Collapse
Affiliation(s)
- Erlend Strønen
- Department of Cancer Immunology, Oslo University Hospital Radiumhospitalet, Oslo, Norway. K. G. Jebsen Centers for Cancer Immunotherapy and for Inflammation Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Mireille Toebes
- Division of Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Sander Kelderman
- Division of Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Marit M van Buuren
- Division of Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Weiwen Yang
- Department of Cancer Immunology, Oslo University Hospital Radiumhospitalet, Oslo, Norway. K. G. Jebsen Centers for Cancer Immunotherapy and for Inflammation Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nienke van Rooij
- Division of Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Marco Donia
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Maxi-Lu Böschen
- Department of Cancer Immunology, Oslo University Hospital Radiumhospitalet, Oslo, Norway. K. G. Jebsen Centers for Cancer Immunotherapy and for Inflammation Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- K. G. Jebsen Centers for Cancer Immunotherapy and for Inflammation Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway. Department of Immunology and Transfusion Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Johanna Olweus
- Department of Cancer Immunology, Oslo University Hospital Radiumhospitalet, Oslo, Norway. K. G. Jebsen Centers for Cancer Immunotherapy and for Inflammation Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Ton N Schumacher
- Division of Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands.
| |
Collapse
|
14
|
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
|
15
|
Kanderova V, Kuzilkova D, Stuchly J, Vaskova M, Brdicka T, Fiser K, Hrusak O, Lund-Johansen F, Kalina T. High-resolution Antibody Array Analysis of Childhood Acute Leukemia Cells. Mol Cell Proteomics 2016; 15:1246-61. [PMID: 26785729 DOI: 10.1074/mcp.m115.054593] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Indexed: 11/06/2022] Open
Abstract
Acute leukemia is a disease pathologically manifested at both genomic and proteomic levels. Molecular genetic technologies are currently widely used in clinical research. In contrast, sensitive and high-throughput proteomic techniques for performing protein analyses in patient samples are still lacking. Here, we used a technology based on size exclusion chromatography followed by immunoprecipitation of target proteins with an antibody bead array (Size Exclusion Chromatography-Microsphere-based Affinity Proteomics, SEC-MAP) to detect hundreds of proteins from a single sample. In addition, we developed semi-automatic bioinformatics tools to adapt this technology for high-content proteomic screening of pediatric acute leukemia patients.To confirm the utility of SEC-MAP in leukemia immunophenotyping, we tested 31 leukemia diagnostic markers in parallel by SEC-MAP and flow cytometry. We identified 28 antibodies suitable for both techniques. Eighteen of them provided excellent quantitative correlation between SEC-MAP and flow cytometry (p< 0.05). Next, SEC-MAP was applied to examine 57 diagnostic samples from patients with acute leukemia. In this assay, we used 632 different antibodies and detected 501 targets. Of those, 47 targets were differentially expressed between at least two of the three acute leukemia subgroups. The CD markers correlated with immunophenotypic categories as expected. From non-CD markers, we found DBN1, PAX5, or PTK2 overexpressed in B-cell precursor acute lymphoblastic leukemias, LAT, SH2D1A, or STAT5A overexpressed in T-cell acute lymphoblastic leukemias, and HCK, GLUD1, or SYK overexpressed in acute myeloid leukemias. In addition, OPAL1 overexpression corresponded to ETV6-RUNX1 chromosomal translocation.In summary, we demonstrated that SEC-MAP technology is a powerful tool for detecting hundreds of proteins in clinical samples obtained from pediatric acute leukemia patients. It provides information about protein size and reveals differences in protein expression between particular leukemia subgroups. Forty-seven of SEC-MAP identified targets were validated by other conventional method in this study.
Collapse
Affiliation(s)
- Veronika Kanderova
- From the ‡CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, 2 Faculty of Medicine, Charles University in Prague, V Uvalu 84, 15006 Prague 5, Czech Republic
| | - Daniela Kuzilkova
- From the ‡CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, 2 Faculty of Medicine, Charles University in Prague, V Uvalu 84, 15006 Prague 5, Czech Republic
| | - Jan Stuchly
- From the ‡CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, 2 Faculty of Medicine, Charles University in Prague, V Uvalu 84, 15006 Prague 5, Czech Republic
| | - Martina Vaskova
- From the ‡CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, 2 Faculty of Medicine, Charles University in Prague, V Uvalu 84, 15006 Prague 5, Czech Republic
| | - Tomas Brdicka
- §Institute of Molecular Genetics, Academy of Sciences of the Czech Republic; Videnska 1083, 14220 Prague, Czech Republic
| | - Karel Fiser
- From the ‡CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, 2 Faculty of Medicine, Charles University in Prague, V Uvalu 84, 15006 Prague 5, Czech Republic
| | - Ondrej Hrusak
- From the ‡CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, 2 Faculty of Medicine, Charles University in Prague, V Uvalu 84, 15006 Prague 5, Czech Republic
| | - Fridtjof Lund-Johansen
- ¶Department of Immunology, Oslo University Hospital, Rikshospitalet; Sognsvannsveien 20, 0372 Oslo, Norway
| | - Tomas Kalina
- From the ‡CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, 2 Faculty of Medicine, Charles University in Prague, V Uvalu 84, 15006 Prague 5, Czech Republic;
| |
Collapse
|
16
|
|
17
|
Almagro JC, Gilliland GL, Breden F, Scott JK, Sok D, Pauthner M, Reichert JM, Helguera G, Andrabi R, Mabry R, Bléry M, Voss JE, Laurén J, Abuqayyas L, Barghorn S, Ben-Jacob E, Crowe JE, Huston JS, Johnston SA, Krauland E, Lund-Johansen F, Marasco WA, Parren PWHI, Xu KY. Antibody engineering and therapeutics, The Annual Meeting of the Antibody Society: December 8-12, 2013, Huntington Beach, CA. MAbs 2014; 6:577-618. [PMID: 24589717 PMCID: PMC4011904 DOI: 10.4161/mabs.28421] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The 24th Antibody Engineering & Therapeutics meeting brought together a broad range of participants who were updated on the latest advances in antibody research and development. Organized by IBC Life Sciences, the gathering is the annual meeting of The Antibody Society, which serves as the scientific sponsor. Preconference workshops on 3D modeling and delineation of clonal lineages were featured, and the conference included sessions on a wide variety of topics relevant to researchers, including systems biology; antibody deep sequencing and repertoires; the effects of antibody gene variation and usage on antibody response; directed evolution; knowledge-based design; antibodies in a complex environment; polyreactive antibodies and polyspecificity; the interface between antibody therapy and cellular immunity in cancer; antibodies in cardiometabolic medicine; antibody pharmacokinetics, distribution and off-target toxicity; optimizing antibody formats for immunotherapy; polyclonals, oligoclonals and bispecifics; antibody discovery platforms; and antibody-drug conjugates.
Collapse
Affiliation(s)
| | | | | | | | - Devin Sok
- The Scripps Research Institute; La Jolla, CA USA
| | | | | | - Gustavo Helguera
- CONICET; Laboratorio Biotecnología Farmacéutica; Instituto de Biología y Medicina Experimental, IBYME; Ciudad Autónoma de Buenos Aires, Argentina
| | | | | | | | - James E Voss
- The Scripps Research Institute; La Jolla, CA USA
| | - Juha Laurén
- Regeneron Pharmaceuticals, Inc.; Tarrytown, NY USA
| | | | | | | | - James E Crowe
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | | | | - Kai Y Xu
- University of Maryland; Baltimore, MD USA
| |
Collapse
|
18
|
González-González M, Bartolome R, Jara-Acevedo R, Casado-Vela J, Dasilva N, Matarraz S, García J, Alcazar JA, Sayagues JM, Orfao A, Fuentes M. Evaluation of homo- and hetero-functionally activated glass surfaces for optimized antibody arrays. Anal Biochem 2014; 450:37-45. [PMID: 24440232 DOI: 10.1016/j.ab.2014.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 01/07/2014] [Indexed: 11/17/2022]
Abstract
Antibody arrays hold great promise for biomedical applications, but they are typically manufactured using chemically functionalized surfaces that still require optimization. Here, we describe novel hetero-functionally activated glass surfaces favoring oriented antibody binding for improved performance in protein microarray applications. Antibody arrays manufactured in our facility using the functionalization chemistries described here proved to be reproducible and stable and also showed good signal intensities. As a proof-of-principle of the glass surface functionalization protocols described in this article, we built antibody-based arrays functionalized with different chemistries that enabled the simultaneous detection of 71 human leukocyte membrane differentiation antigens commonly found in peripheral blood mononuclear cells. Such detection is specific and semi-quantitative and can be performed in a single assay under native conditions. In summary, the protocol described here, based on the use of antibody array technology, enabled the concurrent detection of a set of membrane proteins under native conditions in a specific, selective, and semi-quantitative manner and in a single assay.
Collapse
Affiliation(s)
- María González-González
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC)-IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, 37007 Salamanca, Spain
| | - Raquel Bartolome
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC)-IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, 37007 Salamanca, Spain
| | | | - Juan Casado-Vela
- Spanish National Research Council (CSIC)-Spanish National Biotechnology Centre (CNB), 28049 Madrid, Spain
| | - Noelia Dasilva
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC)-IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, 37007 Salamanca, Spain
| | - Sergio Matarraz
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC)-IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, 37007 Salamanca, Spain
| | - Jacinto García
- Servicio de Cirugía, Hospital Clínico Universitario de Salamanca-IBSAL, 37007 Salamanca, Spain
| | - J A Alcazar
- Servicio de Cirugía, Hospital Clínico Universitario de Salamanca-IBSAL, 37007 Salamanca, Spain
| | - J M Sayagues
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC)-IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, 37007 Salamanca, Spain
| | - Alberto Orfao
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC)-IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, 37007 Salamanca, Spain.
| | - Manuel Fuentes
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC)-IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
19
|
Wilson R. Sensitivity and specificity: twin goals of proteomics assays. Can they be combined? Expert Rev Proteomics 2014; 10:135-49. [DOI: 10.1586/epr.13.7] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
20
|
Ayoglu B, Häggmark A, Neiman M, Igel U, Uhlén M, Schwenk JM, Nilsson P. Systematic antibody and antigen-based proteomic profiling with microarrays. Expert Rev Mol Diagn 2014; 11:219-34. [DOI: 10.1586/erm.10.110] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Juncker D, Bergeron S, Laforte V, Li H. Cross-reactivity in antibody microarrays and multiplexed sandwich assays: shedding light on the dark side of multiplexing. Curr Opin Chem Biol 2013; 18:29-37. [PMID: 24534750 DOI: 10.1016/j.cbpa.2013.11.012] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 11/22/2013] [Accepted: 11/26/2013] [Indexed: 11/25/2022]
Abstract
Immunoassays are indispensable for research and clinical analysis, and following the emergence of the omics paradigm, multiplexing of immunoassays is more needed than ever. Cross-reactivity (CR) in multiplexed immunoassays has been unexpectedly difficult to mitigate, preventing scaling up of multiplexing, limiting assay performance, and resulting in inaccurate and even false results, and wrong conclusions. Here, we review CR and its consequences in single and dual antibody single-plex and multiplex assays. We establish a distinction between sample-driven and reagent-driven CR, and describe how it affects the performance of antibody microarrays. Next, we review and evaluate various platforms aimed at mitigating CR, including SOMAmers and protein fractionation-bead assays, as well as dual Ab methods including (i) conventional multiplex assays, (ii) proximity ligation assays, (iii) immuno-mass spectrometry, (iv) sequential multiplex analyte capture, (v) antibody colocalization microarrays and (vi) force discrimination assays.
Collapse
Affiliation(s)
- David Juncker
- McGill University & Genome Quebec Innovation Centre, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada H3A 0G1; Department of Biomedical Engineering, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada H3A 0G1; Department of Neurology and Neurosurgery, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada H3A 2B4.
| | - Sébastien Bergeron
- McGill University & Genome Quebec Innovation Centre, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada H3A 0G1; Department of Biomedical Engineering, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada H3A 0G1
| | - Veronique Laforte
- McGill University & Genome Quebec Innovation Centre, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada H3A 0G1; Department of Biomedical Engineering, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada H3A 0G1; Department of Neurology and Neurosurgery, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada H3A 2B4
| | - Huiyan Li
- McGill University & Genome Quebec Innovation Centre, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada H3A 0G1; Department of Biomedical Engineering, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada H3A 0G1
| |
Collapse
|
22
|
Neiman M, Fredolini C, Johansson H, Lehtiö J, Nygren PÅ, Uhlén M, Nilsson P, Schwenk JM. Selectivity analysis of single binder assays used in plasma protein profiling. Proteomics 2013; 13:3406-10. [PMID: 24151238 PMCID: PMC4265267 DOI: 10.1002/pmic.201300030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 09/24/2013] [Accepted: 10/04/2013] [Indexed: 11/29/2022]
Abstract
The increasing availability of antibodies toward human proteins enables broad explorations of the proteomic landscape in cells, tissues, and body fluids. This includes assays with antibody suspension bead arrays that generate protein profiles of plasma samples by flow cytometer analysis. However, antibody selectivity is context dependent so it is necessary to corroborate on-target detection over off-target binding. To address this, we describe a concept to directly verify interactions from antibody-coupled beads by analysis of their eluates by Western blots and MS. We demonstrate selective antibody binding in complex samples with antibodies toward a set of chosen proteins with different abundance in plasma and serum, and illustrate the need to adjust sample and bead concentrations accordingly. The presented approach will serve as an important tool for resolving differential protein profiles from antibody arrays within plasma biomarker discoveries.
Collapse
Affiliation(s)
- Maja Neiman
- Science for Life Laboratory, School of Biotechnology, KTH-Royal Institute of Technology, Solna, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Kristensen AR, Foster LJ. High throughput strategies for probing the different organizational levels of protein interaction networks. MOLECULAR BIOSYSTEMS 2013; 9:2201-12. [DOI: 10.1039/c3mb70135b] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
24
|
Guo SL, Chen PC, Chen MS, Cheng YC, Lin JM, Lee HC, Chen CS. A fast universal immobilization of immunoglobulin G at 4 °C for the development of array-based immunoassays. PLoS One 2012; 7:e51370. [PMID: 23236488 PMCID: PMC3517563 DOI: 10.1371/journal.pone.0051370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 11/07/2012] [Indexed: 11/19/2022] Open
Abstract
To maintain the antibody activity and enhance performance of array-based immunoassays, protein G was used to allow a shorter duration of immunoglobulin G immobilization at 4 °C, with the antibody placed in the appropriate orientation. The multiplexed detection of six pain-related message molecules (PRMMs) was used as examples for the development of array-based immunoassays: substance P, calcitonin gene-related peptide, nerve growth factor, brain-derived neurotrophic factor, tumor necrosis factor-α, and β-endorphin. Protein G- and non-protein G-coated slides were tested. Compared to non-protein G immunoassays, protein G shortened the antibody immobilization time at 4 °C from overnight to 2 hours. Only protein G-facilitated immunoassays succeeded in simultaneously detecting all six PRMMs with high specificity. Dose-response curves showed that the limits of detection of the protein G-multiplexed immunoassays for the PRMMs was approximately 164, 167, 120, 60, 80, and 92 pg/ml, respectively. Thus, protein G effectively shortens the duration of antibody immobilization at 4 °C, allowing the use of sensitive array-based immunoassays for the simultaneous detection of PRMMs.
Collapse
Affiliation(s)
- Shu-Lin Guo
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan
- Department of Anesthesiology, Cathay General Hospital, Taipei, Taiwan
| | - Po-Chung Chen
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan
| | - Ming-Shuo Chen
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan
| | - Yu-Che Cheng
- Department of Medical Research, Cathay General Hospital, Taipei, Taiwan
- Institute of Biomedical Engineering, National Central University, Jhongli, Taiwan
| | - Jun-Mu Lin
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan
| | - Hoong-Chien Lee
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan
- * E-mail: (HL); (CC)
| | - Chien-Sheng Chen
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan
- * E-mail: (HL); (CC)
| |
Collapse
|
25
|
Gonzalez-Gonzalez M, Jara-Acevedo R, Matarraz S, Jara-Acevedo M, Paradinas S, Sayagües J, Orfao A, Fuentes M. Nanotechniques in proteomics: Protein microarrays and novel detection platforms. Eur J Pharm Sci 2012; 45:499-506. [DOI: 10.1016/j.ejps.2011.07.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 07/07/2011] [Accepted: 07/14/2011] [Indexed: 11/16/2022]
|
26
|
Dasilva N, Díez P, Matarraz S, González-González M, Paradinas S, Orfao A, Fuentes M. Biomarker discovery by novel sensors based on nanoproteomics approaches. SENSORS 2012; 12:2284-308. [PMID: 22438764 PMCID: PMC3304166 DOI: 10.3390/s120202284] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 01/20/2012] [Accepted: 02/14/2012] [Indexed: 12/23/2022]
Abstract
During the last years, proteomics has facilitated biomarker discovery by coupling high-throughput techniques with novel nanosensors. In the present review, we focus on the study of label-based and label-free detection systems, as well as nanotechnology approaches, indicating their advantages and applications in biomarker discovery. In addition, several disease biomarkers are shown in order to display the clinical importance of the improvement of sensitivity and selectivity by using nanoproteomics approaches as novel sensors.
Collapse
Affiliation(s)
- Noelia Dasilva
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC), Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca 37007, Spain; E-Mails: (N.D.); (P.D.); (S.M.); (M.G.-G.); (A.O.)
| | - Paula Díez
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC), Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca 37007, Spain; E-Mails: (N.D.); (P.D.); (S.M.); (M.G.-G.); (A.O.)
| | - Sergio Matarraz
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC), Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca 37007, Spain; E-Mails: (N.D.); (P.D.); (S.M.); (M.G.-G.); (A.O.)
| | - María González-González
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC), Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca 37007, Spain; E-Mails: (N.D.); (P.D.); (S.M.); (M.G.-G.); (A.O.)
| | - Sara Paradinas
- Departamento de Química Analítica, Facultad de Ciencias Químicas, University of Salamanca, Salamanca 37008, Spain; E-Mail:
| | - Alberto Orfao
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC), Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca 37007, Spain; E-Mails: (N.D.); (P.D.); (S.M.); (M.G.-G.); (A.O.)
| | - Manuel Fuentes
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC), Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca 37007, Spain; E-Mails: (N.D.); (P.D.); (S.M.); (M.G.-G.); (A.O.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +34-923-294-811; Fax: +34-923-294-743
| |
Collapse
|
27
|
Ghafari H, Hanley QS. Analysis of layered assays and volume microarrays in stratified media. Analyst 2012; 137:5520-6. [DOI: 10.1039/c2an35767d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
28
|
Stuchlý J, Kanderová V, Fišer K, Černá D, Holm A, Wu W, Hrušák O, Lund-Johansen F, Kalina T. An automated analysis of highly complex flow cytometry-based proteomic data. Cytometry A 2011; 81:120-9. [DOI: 10.1002/cyto.a.22011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 10/19/2011] [Accepted: 11/28/2011] [Indexed: 01/08/2023]
|
29
|
Slaastad H, Wu W, Goullart L, Kanderova V, Tjønnfjord G, Stuchly J, Kalina T, Holm A, Lund-Johansen F. Multiplexed immuno-precipitation with 1725 commercially available antibodies to cellular proteins. Proteomics 2011; 11:4578-82. [PMID: 21956872 DOI: 10.1002/pmic.201000744] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 08/29/2011] [Accepted: 08/31/2011] [Indexed: 11/10/2022]
Abstract
Antibody array analysis of complex samples requires capture reagents with exceptional specificity. The frequency of antibodies with label-based detection may be as low as 5%. Here, however, we show that as many as 25% of commercially available antibodies are useful when biotinylated cellular proteins are fractionated by size exclusion chromatography (SEC) first. A microsphere multiplex with 1725 antibodies to cellular proteins was added to 24 SEC fractions, labelled with streptavidin and analyzed by flow cytometry (microsphere-based affinity proteomics, MAP) The SEC-MAP approach resolved different targets captured by each antibody as reactivity peaks across the separation range of the SEC column (10-670kDa). Complex reactivity profiles demonstrated that most antibodies bound more than one target. However, specific binding was readily detected as reactivity peaks common for different antibodies to the same protein. We optimized sample preparation and found that amine-reactive biotin rarely inhibited antibody binding when the biotin to lysine ratio was kept below 1:1 during labelling. Moreover, several epitopes that were inaccessible to antibodies in native proteins were unmasked after heat denaturation with 0.1% of SDS. The SEC-MAP format should allow researchers to build multiplexed assays with antibodies purchased for use in e.g. Western blotting.
Collapse
Affiliation(s)
- Heidi Slaastad
- Department of Immunology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Pontén F, Schwenk JM, Asplund A, Edqvist PHD. The Human Protein Atlas as a proteomic resource for biomarker discovery. J Intern Med 2011; 270:428-46. [PMID: 21752111 DOI: 10.1111/j.1365-2796.2011.02427.x] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The analysis of tissue-specific expression at both the gene and protein levels is vital for understanding human biology and disease. Antibody-based proteomics provides a strategy for the systematic generation of antibodies against all human proteins to combine with protein profiling in tissues and cells using tissue microarrays, immunohistochemistry and immunofluorescence. The Human Protein Atlas project was launched in 2003 with the aim of creating a map of protein expression patterns in normal cells, tissues and cancer. At present, 11,200 unique proteins corresponding to over 50% of all human protein-encoding genes have been analysed. All protein expression data, including underlying high-resolution images, are published on the free and publically available Human Protein Atlas portal (http://www.proteinatlas.org). This database provides an important source of information for numerous biomedical research projects, including biomarker discovery efforts. Moreover, the global analysis of how our genome is expressed at the protein level has provided basic knowledge on the ubiquitous expression of a large proportion of our proteins and revealed the paucity of cell- and tissue-type-specific proteins.
Collapse
Affiliation(s)
- F Pontén
- Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | | | | | | |
Collapse
|
31
|
Häggmark A, Neiman M, Drobin K, Zwahlen M, Uhlén M, Nilsson P, Schwenk JM. Classification of protein profiles from antibody microarrays using heat and detergent treatment. N Biotechnol 2011; 29:564-70. [PMID: 22023822 DOI: 10.1016/j.nbt.2011.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 10/09/2011] [Accepted: 10/09/2011] [Indexed: 01/16/2023]
Abstract
Antibody microarrays offer new opportunities for exploring the proteome and to identify biomarker candidates in human serum and plasma. Here, we have investigated the effect of heat and detergents on an antibody-based suspension bead array (SBA) assay using polyclonal antibodies and biotinylated plasma samples. With protein profiles from more than 2300 antibodies generated in 384-plex antibody SBAs, three major classes of heat and detergent susceptibility could be described. The results show that washing of the beads with SDS (rather than Tween) after target binding lowered intensity levels of basically all profiles and that about 50% of the profiles appeared to be lowered to a similar extent by heating of the sample. About 33% of the profiles appeared to be insensitive to heat treatment while another 17% showed a positive influence of heat to yield elevated profiles. The results suggest that the classification of antibodies is driven by the molecular properties of the antibody-antigen interaction and can generally not be predicted based on protein class or Western blot data. The experimental scheme presented here can be used to systematically categorize antibodies and thereby combine antibodies with similar properties into targeted arrays for analysis of plasma and serum.
Collapse
Affiliation(s)
- Anna Häggmark
- Science for Life Laboratory Stockholm, School of Biotechnology, KTH - Royal Institute of Technology, Box 1031, SE-171 21 Solna, Sweden
| | | | | | | | | | | | | |
Collapse
|
32
|
Holm A, Wu W, Lund-Johansen F. Antibody array analysis of labelled proteomes: how should we control specificity? N Biotechnol 2011; 29:578-85. [PMID: 21840428 DOI: 10.1016/j.nbt.2011.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 07/26/2011] [Accepted: 07/27/2011] [Indexed: 11/15/2022]
Abstract
Researchers who use protein binders in multiplexed assays can be divided into two camps. One believes that arrays with proteome-wide coverage will become a reality once we have developed binders for all proteins. The sceptics claim that detection with immobilized protein binders and sample labelling will not provide the required specificity. In this article, we review the evidence showing that antibody array analysis of labelled samples can provide meaningful data and discuss the issues raised by the sceptics. We argue that direct the evidence for monospecificity has yet to be published. This will require assays designed to resolve the proteins captured by each binder. One option is to combine array measurement with protein separation. We have developed an assay where labelled sample proteins are separated by size exclusion chromatography (SEC) before contact with microsphere-based arrays (Size-MAP; size exclusion chromatography-resolved microsphere-based affinity proteomics). The effect is an 'antibody array Western blot' where reactivity of immobilized binders is resolved against the size of the proteins in the sample. We show that Size-MAP is useful to discriminate monospecific- and polyreactive antibodies and for automatic detection of reacting with the same target. The possibility to test specificity directly in array-based measurement should be useful to select the best binders and to determine whether the DNA microarray for the proteome is a realistic goal or not.
Collapse
Affiliation(s)
- Anders Holm
- Department of Immunology, Clinic of Specialized Medicine and Surgery Oslo University Hospital, Rikshospitalet, N-0027 Oslo, Norway
| | | | | |
Collapse
|
33
|
Deswal S, Schulze AK, Höfer T, Schamel WWA. Quantitative analysis of protein phosphorylations and interactions by multi-colour IP-FCM as an input for kinetic modelling of signalling networks. PLoS One 2011; 6:e22928. [PMID: 21829558 PMCID: PMC3146539 DOI: 10.1371/journal.pone.0022928] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 07/01/2011] [Indexed: 11/22/2022] Open
Abstract
Background To understand complex biological signalling mechanisms, mathematical modelling of signal transduction pathways has been applied successfully in last few years. However, precise quantitative measurements of signal transduction events such as activation-dependent phosphorylation of proteins, remains one bottleneck to this success. Methodology/Principal Findings We use multi-colour immunoprecipitation measured by flow cytometry (IP-FCM) for studying signal transduction events to unrivalled precision. In this method, antibody-coupled latex beads capture the protein of interest from cellular lysates and are then stained with differently fluorescent-labelled antibodies to quantify the amount of the immunoprecipitated protein, of an interaction partner and of phosphorylation sites. The fluorescence signals are measured by FCM. Combining this procedure with beads containing defined amounts of a fluorophore allows retrieving absolute numbers of stained proteins, and not only relative values. Using IP-FCM we derived multidimensional data on the membrane-proximal T-cell antigen receptor (TCR-CD3) signalling network, including the recruitment of the kinase ZAP70 to the TCR-CD3 and subsequent ZAP70 activation by phosphorylation in the murine T-cell hybridoma and primary murine T cells. Counter-intuitively, these data showed that cell stimulation by pervanadate led to a transient decrease of the phospho-ZAP70/ZAP70 ratio at the TCR. A mechanistic mathematical model of the underlying processes demonstrated that an initial massive recruitment of non-phosphorylated ZAP70 was responsible for this behaviour. Further, the model predicted a temporal order of multisite phosphorylation of ZAP70 (with Y319 phosphorylation preceding phosphorylation at Y493) that we subsequently verified experimentally. Conclusions/Significance The quantitative data sets generated by IP-FCM are one order of magnitude more precise than Western blot data. This accuracy allowed us to gain unequalled insight into the dynamics of the TCR-CD3-ZAP70 signalling network.
Collapse
Affiliation(s)
- Sumit Deswal
- Max Planck Institute of Immunobiology and Epigenetics, and Faculty of Biology, Biology III, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, Freiburg, Germany
| | - Anna K. Schulze
- Research Group Modeling of Biological Systems, German Cancer Research Center and BioQuant Center, Heidelberg, Germany
| | - Thomas Höfer
- Research Group Modeling of Biological Systems, German Cancer Research Center and BioQuant Center, Heidelberg, Germany
| | - Wolfgang W. A. Schamel
- Max Planck Institute of Immunobiology and Epigenetics, and Faculty of Biology, Biology III, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Centre of Chronic Immunodeficiency (CCI), University Medical Center Freiburg, and University of Freiburg, Freiburg, Germany
- * E-mail:
| |
Collapse
|
34
|
Next-generation plasma profiling: affinity array potential. Bioanalysis 2011; 3:1543-6. [DOI: 10.4155/bio.11.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
35
|
Matarraz S, González-González M, Jara M, Orfao A, Fuentes M. New technologies in cancer. Protein microarrays for biomarker discovery. Clin Transl Oncol 2011; 13:156-61. [DOI: 10.1007/s12094-011-0635-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
36
|
Sevecka M, Wolf-Yadlin A, MacBeath G. Lysate microarrays enable high-throughput, quantitative investigations of cellular signaling. Mol Cell Proteomics 2011; 10:M110.005363. [PMID: 21296872 DOI: 10.1074/mcp.m110.005363] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lysate microarrays (reverse-phase protein arrays) hold great promise as a tool for systems-level investigations of signaling and multiplexed analyses of disease biomarkers. To date, however, widespread use of this technology has been limited by questions concerning data quality and the specificity of detection reagents. To address these concerns, we developed a strategy to identify high-quality reagents for use with lysate microarrays. In total, we tested 383 antibodies for their ability to quantify changes in protein abundance or modification in 20 biological contexts across 17 cell lines. Antibodies yielding significant differences in signal were further evaluated by immunoblotting and 82 passed our rigorous criteria. The large-scale data set from our screen revealed that cell fate decisions are encoded not just by the identities of proteins that are activated, but by differences in their signaling dynamics as well. Overall, our list of validated antibodies and associated protocols establish lysate microarrays as a robust tool for systems biology.
Collapse
Affiliation(s)
- Mark Sevecka
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St., Cambridge, Massachusetts 02138, USA
| | | | | |
Collapse
|
37
|
Mustafa SA, Hoheisel JD, Alhamdani MSS. Secretome profiling with antibody microarrays. MOLECULAR BIOSYSTEMS 2011; 7:1795-801. [DOI: 10.1039/c1mb05071k] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|