1
|
Siedlik JA, Deckert JA, Dunbar AJ, Bhatta A, Gigliotti NM, Chan MA, Benedict SH, Bubak M, Vardiman JP, Gallagher PM. Acute high-intensity exercise enhances T cell proliferation compared to moderate-intensity exercise. Appl Physiol Nutr Metab 2025; 50:1-12. [PMID: 39947139 DOI: 10.1139/apnm-2024-0420] [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] [Indexed: 03/12/2025]
Abstract
Conventional belief is that high-intensity (HI) exercise inhibits immune function; however, recent work challenges this position. The purpose of this was to quantify changes in T cell proliferative capacity following either a HI or moderate-intensity (MI) exercise. Sixteen males were randomly selected to a HI or MI exercise group. Blood was obtained baseline and immediately, 1, 4, and 6 h post-exercise for analyses of CD3+ T cell proliferation (co-stimulation via phytohaemagglutinin or CD3 + CD28). The proliferative response increased in T cells in the HI group and remained significantly elevated up to 6 h post-exercise in both co-stimulation conditions. In contrast, the MI group saw no change proliferative ability following exercise. Analyses of serum stress hormones, and immunomodulatory cytokines failed to reveal any correlated variations that could clarify the T cell findings. We suggest the increase in proliferative capacity following HI exercise is indicative of an exercise-induced activation that provides for enhanced functional responses to stimuli. Moreover, this study shows that HI exercise increases T cell processes, effectively priming them for activation in response to stimuli. This study is registered with ClinicalTrials.gov (NCT06638684).
Collapse
Affiliation(s)
- Jacob A Siedlik
- Department of Biology, University of West Florida, Pensacola, FL, USA
- Florida Institute for Human and Machine Cognition, Pensacola, FL, USA
| | - Jake A Deckert
- University of Washington School of Medicine, Spokane, WA, USA
- Department of Human Physiology, Gonzaga University, Spokane, WA, USA
| | - Amanda J Dunbar
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Anuja Bhatta
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Nicole M Gigliotti
- Department of Pediatrics, Children's Mercy Hospitals & Clinics, Kansas City, MO, USA
| | - Marcia A Chan
- Department of Pediatrics, Children's Mercy Hospitals & Clinics, Kansas City, MO, USA
| | - Stephen H Benedict
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Matthew Bubak
- Department of Health, Sport, and Exercise Sciences, University of Kansas, Lawrence, KS, USA
| | - John P Vardiman
- Department of Nutrition, Kansas State University, Manhattan, KS, USA
| | - Philip M Gallagher
- Department of Health, Sport, and Exercise Sciences, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
2
|
Kamada H, Takahashi D, Shimizu M, Uchida M, Watanabe Y, Nakajima F, Miyata S, Satake M. A novel immunocomplex capture fluorescence assay (ICFA) using fluorescent beads and transfected cells for specific identification of human neutrophil antigen (HNA)-1a and -1b antibodies. Transfusion 2024; 64:906-918. [PMID: 38530740 DOI: 10.1111/trf.17813] [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: 04/13/2023] [Revised: 11/29/2023] [Accepted: 03/14/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND To identify specific human neutrophil antigen (HNA) antibodies, assays using neutrophils such as monoclonal antibody-specific immobilization of granulocyte antigens (MAIGA) are recommended. However, these assays are limited by labor-intensive neutrophil preparation and varying antigen expression levels. METHODS We evaluated a newly developed immunocomplex capture fluorescence assay (ICFA) for identifying HNA-1 antibodies and compared it to MAIGA and LABScreen Multi (LABM), which utilizes recombinant HNA-coated Luminex beads. For ICFA, HNA-1a or HNA-1b transfected cells replaced neutrophils. Cells incubated with serum were lysed, and immune complexes were captured using five CD16 monoclonal antibody-conjugated Luminex beads. Nine antisera with known specificity and 26 samples suspected of containing HNA antibodies were analyzed by ICFA and MAIGA using neutrophils or transfected cells (ICFA-N or ICFA-T, and MAIGA-N or MAIGA-T, respectively). RESULTS ICFA-T and MAIGA-N accurately determined the specificity of all antibodies in the nine antiserum samples. The ICFA-T detection limit was 2048-fold for anti-HNA-1a and 256-fold for anti-HNA-1b; the limits of MAIGA-T, MAIGA-N, and LABM were 32-, 4 ~ 64-, and 128-fold for anti-HNA-1a and 64-, 16 ~ 64-, and 32-fold for anti-HNA-1b, respectively. Twelve and 7 of the remaining 26 samples tested negative and positive, respectively, in both ICFA-T and MAIGA-N. Antibody specificity against HNA-1a or HNA-1b determined using ICFA-T agreed with that determined using MAIGA-N and LABM. Another seven samples tested positive in ICFA-T but negative in MAIGA-N. CONCLUSION The novel ICFA is highly sensitive and exhibits specificity similar to MAIGA and LABM for detecting HNA-1 antibodies.
Collapse
Affiliation(s)
- Hiromi Kamada
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Daisuke Takahashi
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Marie Shimizu
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Miyuki Uchida
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Yoshihisa Watanabe
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | | | - Shigeki Miyata
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Masahiro Satake
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| |
Collapse
|
3
|
Azarmanesh D, Pearlman J, Carbone ET, DiNatale JC, Bertone-Johnson ER. Construct Validation of the Dietary Inflammatory Index (DII) among Young College-Aged Women. Nutrients 2023; 15:4553. [PMID: 37960206 PMCID: PMC10647813 DOI: 10.3390/nu15214553] [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: 10/09/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
The Dietary Inflammatory Index (DII) is designed to assess the inflammatory potential of the diet. While previous research has utilized DII among college-aged women, no study to date has validated it in this population. We conducted a construct validation of DII among 393 healthy women aged 18-31 years against a robust panel of 14 inflammatory biomarkers, including CRP, IL-1β, IL-4, IL-6, IL-10, and TNF-α, which were used in the development of DII. Three linear regression models were constructed: (1) an age-adjusted model, (2) the most parsimonious model based on likelihood ratio tests, and (3) a fully adjusted model for age, race, body mass index, waist circumference, physical activity, smoking status, and nonsteroidal anti-inflammatory drug use. DII was derived from the Harvard food frequency questionnaire and categorized into quartiles. Consistent with our hypothesis, DII was negatively and significantly associated with back-transformed IL-10 levels, confirming that a more pro-inflammatory diet was associated with lower levels of an anti-inflammatory cytokine (Model 3: Q4 vs. Q1 β = 0.62; 95% CI: 0.42, 0.93; p-trend = 0.04). While validated in other populations, DII may not be a suitable tool for assessing the inflammatory potential of the diet among college-aged women.
Collapse
Affiliation(s)
- Deniz Azarmanesh
- Department of Human Nutrition and Hospitality Management, The University of Alabama, Tuscaloosa, AL 35487, USA;
| | - Jessica Pearlman
- Institute for Social Science Research, University of Massachusetts Amherst, Amherst, MA 01003, USA;
| | - Elena T. Carbone
- Department of Nutrition and Commonwealth Honors College, University of Massachusetts Amherst, Amherst, MA 01003, USA;
| | - Janie C. DiNatale
- Department of Human Nutrition and Hospitality Management, The University of Alabama, Tuscaloosa, AL 35487, USA;
| | | |
Collapse
|
4
|
Ramirez MF, Honigberg M, Wang D, Parekh JK, Bielawski K, Courchesne P, Larson MD, Levy D, Murabito JM, Ho JE, Lau ES. Protein Biomarkers of Early Menopause and Incident Cardiovascular Disease. J Am Heart Assoc 2023; 12:e028849. [PMID: 37548169 PMCID: PMC10492938 DOI: 10.1161/jaha.122.028849] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/20/2023] [Indexed: 08/08/2023]
Abstract
Background Premature and early menopause are independently associated with greater risk of cardiovascular disease (CVD). However, mechanisms linking age of menopause with CVD remain poorly characterized. Methods and Results We measured 71 circulating CVD protein biomarkers in 1565 postmenopausal women enrolled in the FHS (Framingham Heart Study). We examined the association of early menopause with biomarkers and tested whether early menopause modified the association of biomarkers with incident cardiovascular outcomes (heart failure, major CVD, and all-cause death) using multivariable-adjusted linear regression and Cox models, respectively. Among 1565 postmenopausal women included (mean age 62 years), 395 (25%) had a history of early menopause. Of 71 biomarkers examined, we identified 7 biomarkers that were significantly associated with early menopause, of which 5 were higher in women with early menopause including adrenomedullin and resistin, and 2 were higher in women without early menopause including insulin growth factor-1 and CNTN1 (contactin-1) (Benjamini-Hochberg adjusted P<0.1 for all). Early menopause also modified the association of specific biomarkers with incident cardiovascular outcomes including adrenomedullin (Pint<0.05). Conclusions Early menopause is associated with circulating levels of CVD protein biomarkers and appears to modify the association between select biomarkers with incident cardiovascular outcomes. Identified biomarkers reflect several distinct biological pathways, including inflammation, adiposity, and neurohormonal regulation. Further investigation of these pathways may provide mechanistic insights into the pathogenesis, prevention, and treatment of early menopause-associated CVD.
Collapse
Affiliation(s)
- Mariana F. Ramirez
- CardioVascular Institute and Division of Cardiology, Department of MedicineBeth Israel Deaconess Medical CenterBostonMAUSA
| | - Michael Honigberg
- Cardiovascular Research Center and Division of Cardiology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Dongyu Wang
- CardioVascular Institute and Division of Cardiology, Department of MedicineBeth Israel Deaconess Medical CenterBostonMAUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMAUSA
| | - Juhi K. Parekh
- CardioVascular Institute and Division of Cardiology, Department of MedicineBeth Israel Deaconess Medical CenterBostonMAUSA
| | - Kamila Bielawski
- Cardiovascular Research Center and Division of Cardiology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Paul Courchesne
- Framingham Heart StudyFraminghamMAUSA
- Population Sciences Branch, Division of Intramural ResearchNational Heart, Lung, and Blood InstituteFraminghamMAUSA
| | | | - Daniel Levy
- Framingham Heart StudyFraminghamMAUSA
- Population Sciences Branch, Division of Intramural ResearchNational Heart, Lung, and Blood InstituteFraminghamMAUSA
| | - Joanne M. Murabito
- Framingham Heart StudyFraminghamMAUSA
- Department of Medicine, Section of General Internal MedicineBoston University School of Medicine and Boston Medical CenterBostonMAUSA
| | - Jennifer E. Ho
- CardioVascular Institute and Division of Cardiology, Department of MedicineBeth Israel Deaconess Medical CenterBostonMAUSA
| | - Emily S. Lau
- Cardiovascular Research Center and Division of Cardiology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| |
Collapse
|
5
|
Schneider MM, Scheidt T, Priddey AJ, Xu CK, Hu M, Meisl G, Devenish SRA, Dobson CM, Kosmoliaptsis V, Knowles TPJ. Microfluidic antibody affinity profiling of alloantibody-HLA interactions in human serum. Biosens Bioelectron 2023; 228:115196. [PMID: 36921387 DOI: 10.1016/j.bios.2023.115196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/17/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023]
Abstract
Antibody profiling is a fundamental component of understanding the humoral response in a wide range of disease areas. Most currently used approaches operate by capturing antibodies onto functionalised surfaces. Such measurements of surface binding are governed by an overall antibody titre, while the two fundamental molecular parameters, antibody affinity and antibody concentration, are challenging to determine individually from such approaches. Here, by applying microfluidic diffusional sizing (MDS), we show how we can overcome this challenge and demonstrate reliable quantification of alloantibody binding affinity and concentration of alloantibodies binding to Human Leukocyte Antigens (HLA), an extensively used clinical biomarker in organ transplantation, both in buffer and in crude human serum. Capitalising on the ability to vary both serum and HLA concentrations during MDS, we show that both affinity and concentration of HLA-specific antibodies can be determined directly in serum when neither of these parameters is known. Finally, we provide proof of principle in clinical transplant patient sera that our assay enables differentiation of alloantibody reactivity against HLA proteins of highly similar structure, providing information not attainable through currently available techniques. These results outline a path towards detection and in-depth profiling of humoral immunity and may enable further insights into the clinical relevance of antibody reactivity in clinical transplantation and beyond.
Collapse
Affiliation(s)
- Matthias M Schneider
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Tom Scheidt
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Ashley J Priddey
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Catherine K Xu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Mengsha Hu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Sean R A Devenish
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Rd, Cambridge, CB1 8DH, UK
| | - Christopher M Dobson
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Vasilis Kosmoliaptsis
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK; NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK; NIHR Cambridge Biomedical Research Centre, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK; Cavendish Laboratory, Department of Physics, University of Cambridge, JJ Thomson Ave, Cambridge, CB3 0HE, UK.
| |
Collapse
|
6
|
Ozulumba T, Montalbine AN, Ortiz-Cárdenas JE, Pompano RR. New tools for immunologists: models of lymph node function from cells to tissues. Front Immunol 2023; 14:1183286. [PMID: 37234163 PMCID: PMC10206051 DOI: 10.3389/fimmu.2023.1183286] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The lymph node is a highly structured organ that mediates the body's adaptive immune response to antigens and other foreign particles. Central to its function is the distinct spatial assortment of lymphocytes and stromal cells, as well as chemokines that drive the signaling cascades which underpin immune responses. Investigations of lymph node biology were historically explored in vivo in animal models, using technologies that were breakthroughs in their time such as immunofluorescence with monoclonal antibodies, genetic reporters, in vivo two-photon imaging, and, more recently spatial biology techniques. However, new approaches are needed to enable tests of cell behavior and spatiotemporal dynamics under well controlled experimental perturbation, particularly for human immunity. This review presents a suite of technologies, comprising in vitro, ex vivo and in silico models, developed to study the lymph node or its components. We discuss the use of these tools to model cell behaviors in increasing order of complexity, from cell motility, to cell-cell interactions, to organ-level functions such as vaccination. Next, we identify current challenges regarding cell sourcing and culture, real time measurements of lymph node behavior in vivo and tool development for analysis and control of engineered cultures. Finally, we propose new research directions and offer our perspective on the future of this rapidly growing field. We anticipate that this review will be especially beneficial to immunologists looking to expand their toolkit for probing lymph node structure and function.
Collapse
Affiliation(s)
- Tochukwu Ozulumba
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Jennifer E. Ortiz-Cárdenas
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia (UVA) Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
7
|
Takvorian KS, Wang D, Courchesne P, Vasan RS, Benjamin EJ, Cheng S, Larson MG, Levy D, Ho JE. The Association of Protein Biomarkers With Incident Heart Failure With Preserved and Reduced Ejection Fraction. Circ Heart Fail 2023; 16:e009446. [PMID: 36475777 PMCID: PMC9937440 DOI: 10.1161/circheartfailure.121.009446] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 08/25/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) and heart failure with reduced ejection fraction (HFrEF) are distinct clinical entities, yet there is scant evidence for associations of proteomic signatures with future development of HFpEF versus HFrEF. METHODS We evaluated the association of 71 protein biomarkers with incident HFpEF versus HFrEF (left ventricular ejection fraction ≥ versus <50%) among Framingham Heart Study participants using multivariable Cox models. RESULTS Among 7038 participants (mean age 49 years; 54% women), 5 biomarkers were associated with increased risk of incident HFpEF (false discovery rate q<0.05): NT-proBNP (N-terminal pro-B-type natriuretic peptide; hazard ratio [HR], 2.13; 95% CI, 1.52-2.99; P<0.001), growth differentiation factor-15 (HR, 1.67; 95% CI, 1.32-2.12; P<0.001), adrenomedullin (HR, 1.58; 95% CI, 1.23-2.04; P<0.001), uncarboxylated matrix Gla protein (HR, 1.55; 95% CI 1.23-1.95; P<0.001), and C-reactive protein (HR, 1.46; 95% CI, 1.17-1.83; P=0.001). Fourteen biomarkers were associated with incident HFrEF (multivariable P<0.001, q<0.05 for all). Of these, 3 biomarkers were associated with both HF subtypes (NT-proBNP, growth differentiation factor-15, and C-reactive protein). When compared directly, myeloperoxidase, resistin, and paraoxanase-1 were more strongly associated with HFrEF than HFpEF. CONCLUSIONS We identified 5 protein biomarkers of new-onset HFpEF representing pathways of inflammation, cardiac stress, and vascular stiffness, which partly overlapped with HFrEF. We found 14 biomarkers associated with new-onset HFrEF, with some distinct associations including myeloperoxidase, resistin, and paraoxanase-1. Taken together, these findings provide insights into similarities and differences in the development of HF subtypes. REGISTRATION URL: https://clinicaltrials.gov/ct2/show/NCT00005121; Unique identifier: NCT0005121.
Collapse
Affiliation(s)
| | - Dongyu Wang
- Cardiovascular Institute and Department of Medicine, Beth Israel Deaconness Medical Center, Boston, MA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Paul Courchesne
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Ramachandran S. Vasan
- Department of Medicine and Boston University School of Medicine, Boston, MA
- Cardiology and Preventive Medicine Sections, Boston University School of Medicine, Boston, MA
- The Framingham Heart Study, Framingham, MA
- Department of Epidemiology and Boston University School of Public Health, Boston, MA
| | - Emelia J. Benjamin
- Cardiology and Preventive Medicine Sections, Boston University School of Medicine, Boston, MA
- The Framingham Heart Study, Framingham, MA
- Department of Epidemiology and Boston University School of Public Health, Boston, MA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai, Los Angeles, CA
| | - Martin G. Larson
- The Framingham Heart Study, Framingham, MA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Daniel Levy
- The Framingham Heart Study, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda MD
| | - Jennifer E. Ho
- Cardiovascular Institute and Department of Medicine, Beth Israel Deaconness Medical Center, Boston, MA
- Division of Cardiology, Department of Medicine, Beth Israel Deaconness Medical Center, Boston, MA
| |
Collapse
|
8
|
Liu J, Allo B, Winnik MA. Development of Multiplexed Bead-Based Immunoassays for Profiling Soluble Cytokines and CD163 Using Mass Cytometry. ACS MEASUREMENT SCIENCE AU 2022; 2:629-640. [PMID: 36573076 PMCID: PMC9783081 DOI: 10.1021/acsmeasuresciau.2c00038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 06/17/2023]
Abstract
Bead-based immunoassays are multiparametric analysis allowing for the simultaneous quantification of a large number of biomarkers within a single sample. Mass cytometry is an emerging cytometric technique that offers a high multiplexing capacity in a high-throughput setting but has not yet been applied to bead-based assays. In this study, we developed a multiplex bead-based immunoassay of cytokines and CD163 designed for mass cytometry (MC). A set of 11 types of lanthanide-encoded microbeads were synthesized by two-stage dispersion polymerization as classifier candidates for the assay. These beads were then decorated with different Abs on the surface to capture the target cytokines in solution. Gold nanoparticles were employed as reporters to identify the binding of target cytokines on the classifier surface. As a proof-of-concept study, we first developed four-plex and nine-plex assays of mixtures of cytokines in standard solutions. The MC signal intensities of these immunoassays were responsive to the concentration differences in the standard solutions with high detection sensitivities at low analyte concentrations. Finally, we examined a sample of peripheral blood mononuclear cells (PBMCs) with the nine-plex assay, comparing an unstimulated sample with a sample stimulated to promote cytokine secretion.
Collapse
Affiliation(s)
- Jieyi Liu
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Bedilu Allo
- Fluidigm
Canada (now Standard BioTools Canada), 1380 Rodick Road, Markham, Ontario L3R 4G5, Canada
| | - Mitchell A. Winnik
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Department
of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
9
|
Leupold L, Sigutova V, Gerasimova E, Regensburger M, Zundler S, Zunke F, Xiang W, Winner B, Prots I. The Quest for Anti-α-Synuclein Antibody Specificity-Lessons Learnt From Flow Cytometry Analysis. Front Neurol 2022; 13:869103. [PMID: 35911883 PMCID: PMC9334871 DOI: 10.3389/fneur.2022.869103] [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: 02/03/2022] [Accepted: 06/09/2022] [Indexed: 11/18/2022] Open
Abstract
The accumulation of alpha-synuclein (aSyn) is the hallmark of a group of neurodegenerative conditions termed synucleopathies. Physiological functions of aSyn, including those outside of the CNS, remain elusive. However, a reliable and reproducible evaluation of aSyn protein expression in different cell types and especially in low-expressing cells is impeded by the existence of a huge variety of poorly characterized anti-aSyn antibodies and a lack of a routinely used sensitive detection methods. Here, we developed a robust flow cytometry-based workflow for aSyn detection and antibody validation. We test our workflow using three commercially available antibodies (MJFR1, LB509, and 2A7) in a variety of human cell types, including induced pluripotent stem cells, T lymphocytes, and fibroblasts, and provide a cell- and antibody-specific map for aSyn expression. Strikingly, we demonstrate a previously unobserved unspecificity of the LB509 antibody, while the MJFR1 clone revealed specific aSyn binding however with low sensitivity. On the other hand, we identified an aSyn-specific antibody clone 2A7 with an optimal sensitivity for detecting aSyn in a range of cell types, including those with low aSyn expression. We further utilize our workflow to demonstrate the ability of the 2A7 antibody to distinguish between physiological differences in aSyn expression in neuronal and non-neuronal cells from the cortical organoids, and in neural progenitors and midbrain dopaminergic neurons from healthy controls and in patients with Parkinson's disease who have aSyn gene locus duplication. Our results provide a proof of principle for the use of high-throughput flow cytometry-based analysis of aSyn and highlight the necessity of rigorous aSyn antibody validation to facilitate the research of aSyn physiology and pathology.
Collapse
Affiliation(s)
- Lukas Leupold
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Veronika Sigutova
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Elizaveta Gerasimova
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Regensburger
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, Translational Research Center (TRC), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Iryna Prots
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
10
|
Deuchler SK, Schubert R, Singh P, Chedid A, Kenikstul N, Scott J, Kohnen T, Ackermann H, Koch F. Vitreous cytokine levels following the administration of a single 0.19 mg fluocinolone acetonide (ILUVIEN®) implant in patients with refractory diabetic macular edema (DME)-results from the ILUVIT study. Graefes Arch Clin Exp Ophthalmol 2022; 260:2537-2547. [PMID: 35239010 PMCID: PMC9325820 DOI: 10.1007/s00417-022-05564-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/29/2021] [Accepted: 01/11/2022] [Indexed: 11/26/2022] Open
Abstract
Purpose To investigate the changes in vitreous inflammatory and angiogenic cytokine levels, primarily interleukin-(IL)-6, following intravitreal injection of the 0.19 mg fluocinolone acetonide (FAc, ILUVIEN®) implant in patients with diabetic macular edema. Methods A single-center phase IV study involving 12 patients’ eyes with diabetic macular edema. Vitreous fluid samples were obtained prior to intravitreal injection of the fluocinolone acetonide implant and then again over a 6-month period. Vitreous samples were examined using a cytometric bead array to measure IL-6, IL-8, IP-10, MCP-1, VEGF, and CD54. PIGF and PEDF were measured using an enzyme-linked immunosorbent assay. Changes in the cytokine and chemokine expression patterns were analyzed. Clinical parameters such as BCVA and center point thickness (CPT) were also examined. Results There were mean reductions in all parameters between baseline and month 6. Significant changes (p < 0.05 versus baseline) were observed in the expression of IL-6, IP-10, MCP-1, and CD54 following the administration of fluocinolone acetonide implant. VEGF and PIGF increased at month 1 before declining at month 6, though this trend was not significant. CPT decreased rapidly between screening and the first follow-up visit, and this decrease was sustained. BCVA remained relatively stable throughout. Conclusion This study demonstrated changes in vitreous inflammatory and angiogenic cytokine levels following intravitreal injection of the FAc implant in patients with diabetic macular edema. Data show that the fluocinolone acetonide implant led to rapid and sustained reductions of some inflammatory cytokines with improvement of the overall clinical picture. Supplementary Information The online version contains supplementary material available at 10.1007/s00417-022-05564-2.
Collapse
Affiliation(s)
- Svenja K Deuchler
- Augenzentrum Frankfurt, Georg-Baumgarten-Str. 3, Frankfurt am Main, 60549, Germany.
| | - Ralf Schubert
- Pneumological-Immunological Laboratory, Goethe University Hospital, Frankfurt am Main, Germany
| | - Pankaj Singh
- Department of Ophthalmology, Goethe University Hospital, Frankfurt am Main, Germany
| | - Adonis Chedid
- Department of Ophthalmology, Goethe University Hospital, Frankfurt am Main, Germany
| | - Ninel Kenikstul
- Department of Ophthalmology, Goethe University Hospital, Frankfurt am Main, Germany
| | - Julia Scott
- Augenzentrum Frankfurt, Georg-Baumgarten-Str. 3, Frankfurt am Main, 60549, Germany
| | - Thomas Kohnen
- Department of Ophthalmology, Goethe University Hospital, Frankfurt am Main, Germany
| | - Hanns Ackermann
- Institute of Biostatistics, Goethe University Hospital, Frankfurt am Main, Germany
| | - Frank Koch
- Augenzentrum Frankfurt, Georg-Baumgarten-Str. 3, Frankfurt am Main, 60549, Germany
- Department of Ophthalmology, Goethe University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
11
|
Anselm V, Sommersdorf C, Carrasco-Triguero M, Katavolos P, Planatscher H, Steinhilber A, Joos T, Poetz O. Matrix and Sampling Effects on Quantification of Protein Biomarkers of Drug-Induced Liver Injury. J Proteome Res 2021; 20:4985-4994. [PMID: 34554759 DOI: 10.1021/acs.jproteome.1c00478] [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: 11/30/2022]
Abstract
Macrophage colony stimulating factor 1 receptor (MCSF1R), osteopontin (OPN), high-mobility group protein B1 (HMGB1), glutamate dehydrogenase (GLDH), keratin 18 (K18), and caspase-cleaved keratin 18 (ccK18) are considered promising mechanistic biomarkers for the diagnosis of drug-induced liver injury. Here, we aim to elucidate the impact of the sample matrix and handling on the quantification of these emerging protein biomarkers. We investigated effects such as time from collection to centrifugation during serum (± gel) or EDTA plasma preparation on two assay platforms: immunoaffinity liquid chromatography mass spectrometric assays and sandwich immunoassays. Furthermore, we measured GLDH activity with an enzymatic activity assay. Matrix effects were observed particularly for HMGB1 and MCSF1R. HMGB1 levels were higher in serum than in plasma, whereas higher concentrations of MCSF1R were observed in plasma than in serum. A comparison of sample collection to centrifugation time ranging from 15 to 60 min demonstrated increasing levels of HMGB1 in serum, while MCSF1R, OPN, GLDH, and ccK18 concentrations remained stable. Additionally, there was a poor correlation in HMGB1 and ccK18 levels between serum and plasma. Considering the observed matrix effects, we recommend plasma as a matrix of choice and cross-study comparison studies to be limited to those using the same matrix.
Collapse
Affiliation(s)
| | | | | | - Paula Katavolos
- Genentech, San Francisco, California 94080, United States.,Bristol-Myers Squibb, New Brunswick, New Jersey 08901, United States (at Genentech during the conduct of this study)
| | | | | | - Thomas Joos
- SIGNATOPE GmbH, Reutlingen 72770, Germany.,NMI Natural and Medical Sciences Institute at the University of Tuebingen, Reutlingen 72770, Germany
| | - Oliver Poetz
- SIGNATOPE GmbH, Reutlingen 72770, Germany.,NMI Natural and Medical Sciences Institute at the University of Tuebingen, Reutlingen 72770, Germany
| |
Collapse
|
12
|
Kim Y, Lu S, Ho JE, Hwang SJ, Yao C, Huan T, Levy D, Ma J. Proteins as Mediators of the Association Between Diet Quality and Incident Cardiovascular Disease and All-Cause Mortality: The Framingham Heart Study. J Am Heart Assoc 2021; 10:e021245. [PMID: 34482708 PMCID: PMC8649513 DOI: 10.1161/jaha.121.021245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Biological mechanisms underlying the association of a healthy diet with chronic diseases remain unclear. Targeted proteomics may facilitate the understanding of mechanisms linking diet to chronic diseases. Methods and Results We examined 6360 participants (mean age 50 years; 54% women) in the Framingham Heart Study. The associations between diet and 71 cardiovascular disease (CVD)‐related proteins were examined using 3 diet quality scores: the Alternate Healthy Eating Index, the modified Mediterranean‐style Diet Score, and the modified Dietary Approaches to Stop Hypertension diet score. A mediation analysis was conducted to examine which proteins mediated the associations of diet with incident CVD and all‐cause mortality. Thirty of the 71 proteins were associated with at least 1 diet quality score (P<0.0007) after adjustment for multiple covariates in all study participants and confirmed by an internal validation analysis. Gene ontology analysis identified inflammation‐related pathways such as regulation of cell killing and neuroinflammatory response (Bonferroni corrected P<0.05). During a median follow‐up of 13 years, we documented 512 deaths and 488 incident CVD events. Higher diet quality scores were associated with lower risk of CVD (P≤0.03) and mortality (P≤0.004). After adjusting for multiple potential confounders, 4 proteins (B2M [beta‐2‐microglobulin], GDF15 [growth differentiation factor 15], sICAM1 [soluble intercellular adhesion molecule 1], and UCMGP [uncarboxylated matrix Gla‐protein]) mediated the association between at least 1 diet quality score and all‐cause mortality (median proportion of mediation ranged from 8.6% to 25.9%). We also observed that GDF15 mediated the association of the Alternate Healthy Eating Index with CVD (median proportion of mediation: 8.6%). Conclusions Diet quality is associated with new‐onset CVD and mortality and with circulating CVD‐related proteins. Several proteins appear to mediate the association of diet with these outcomes.
Collapse
Affiliation(s)
- Youjin Kim
- Nutrition Epidemiology and Data Science Friedman School of Nutrition Science and Policy Tufts University Boston MA
| | - Sophia Lu
- Health Sciences Sargent CollegeBoston University Boston MA
| | - Jennifer E Ho
- Division of Cardiology Department of Medicine and Cardiovascular Research Center Massachusetts General Hospital Boston MA
| | - Shih-Jen Hwang
- Population Sciences Branch National Heart, Lung, and Blood InstituteNIH Bethesda MD.,Framingham Heart Study Framingham MA
| | - Chen Yao
- Population Sciences Branch National Heart, Lung, and Blood InstituteNIH Bethesda MD.,Framingham Heart Study Framingham MA
| | - Tianxiao Huan
- Department of Ophthalmology and Visual Sciences University of Massachusetts Medical School Worcester MA
| | - Daniel Levy
- Population Sciences Branch National Heart, Lung, and Blood InstituteNIH Bethesda MD.,Framingham Heart Study Framingham MA
| | - Jiantao Ma
- Nutrition Epidemiology and Data Science Friedman School of Nutrition Science and Policy Tufts University Boston MA
| |
Collapse
|
13
|
Deuchler S, Schubert R, Singh P, Chedid A, Brui N, Kenikstul N, Kohnen T, Ackermann H, Koch F. Vitreous expression of cytokines and growth factors in patients with diabetic retinopathy-An investigation of their expression based on clinical diabetic retinopathy grade. PLoS One 2021; 16:e0248439. [PMID: 34010297 PMCID: PMC8133486 DOI: 10.1371/journal.pone.0248439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/25/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetic retinopathy (DR) is an inflammatory condition that affects the posterior of the eye; yet, there are limited published data on techniques measuring the expression of growth and inflammatory factors (GIF) from the posterior segment. The purpose of the current study was two-fold: to sample the vitreous fluid from the eyes of patients with DR and assess the expression of GIF. As DR is an inflammatory disease, the second objective of this study was to determine the relationship between the status of DR and the expression of vitreous GIF. This non-randomized clinical trial was approved by BfARM for the analysis and evaluation of 12 eyes from patients with diabetic macular edema. Vitreous sampling was performed before treatment with fluocinolone acetonide and the extracted vitreous material was examined for the determination of GIF including interleukins 6 (IL-6) and 8 (IL-8), interferon gamma-inducible protein (IP-10), monocyte chemoattractant protein-1 (MCP-1), placental growth factor (PIGF), pigment epithelium-derived factor (PEDF), VEGF (vascular endothelial growth factor) and intercellular adhesion molecule (CD54). These were linearly compared with the grade of inflammation in the vitreous assessed via DR score and ART. Additionally, all eyes were grouped based on their diabetic retinopathy status. All cytokine levels, except MCP-1 and PEDF, were numerically higher in DME patients with proliferative DR than those with non-proliferative DR. DR grade was found to linearly correlate with the expression of CD54 (p = 0.02, rho = 0.64), IL-8 (p = 0.03, rho = 0.64) and PIGF (p = 0.007, rho = 0.76). A correlation was found between ART and CD54 (p = 0.02, rho = 0.66) and also between ART and IL-8 (p = 0.04, rho = 0.60). A trend was found between ART and PIGF (p = 0.08, rho 0.52). For IL-6, there appeared to be a trend with DR grade (p = 0.14, rho = 0.45) and ART (p = 0.09, rho = 0.51). Proliferative DR was shown to be associated with a significant higher expression of CD54, IL-8 and PIGF, thus suggesting that they are potentially important in defining and monitoring the effectiveness of a patients' therapy. Vitreous probes may be helpful in deciding which therapy to administer (i.e. anti-VEGF or corticosteroid or both) based on the expression of GIF. Registry EudraCT number: 2016-004488-38; DRKS-ID: DRKS00014915.
Collapse
Affiliation(s)
- Svenja Deuchler
- Department of Ophthalmology, Goethe University Frankfurt, Frankfurt am Main, Germany
- * E-mail:
| | - Ralf Schubert
- Pneumological-immunological Laboratory, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Pankaj Singh
- Department of Ophthalmology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Adonis Chedid
- Department of Ophthalmology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Natallia Brui
- Department of Ophthalmology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ninel Kenikstul
- Department of Ophthalmology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Thomas Kohnen
- Department of Ophthalmology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Hanns Ackermann
- Institute of Biostatistics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Frank Koch
- Department of Ophthalmology, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
14
|
Dobaño C, Vidal M, Santano R, Jiménez A, Chi J, Barrios D, Ruiz-Olalla G, Rodrigo Melero N, Carolis C, Parras D, Serra P, Martínez de Aguirre P, Carmona-Torre F, Reina G, Santamaria P, Mayor A, García-Basteiro AL, Izquierdo L, Aguilar R, Moncunill G. Highly Sensitive and Specific Multiplex Antibody Assays To Quantify Immunoglobulins M, A, and G against SARS-CoV-2 Antigens. J Clin Microbiol 2021; 59:e01731-20. [PMID: 33127841 PMCID: PMC8111153 DOI: 10.1128/jcm.01731-20] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
Reliable serological tests are required to determine the prevalence of antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and to characterize immunity to the disease in order to address key knowledge gaps in the coronavirus disease 2019 (COVID-19) pandemic. Quantitative suspension array technology (qSAT) assays based on the xMAP Luminex platform overcome the limitations of rapid diagnostic tests and enzyme-linked immunosorbent assays (ELISAs) with their higher precision, dynamic range, throughput, miniaturization, cost-efficiency, and multiplexing capacity. We developed three qSAT assays for IgM, IgA, and IgG against a panel of eight SARS-CoV-2 antigens, including spike protein (S), nucleocapsid protein (N), and membrane protein (M) constructs. The assays were optimized to minimize the processing time and maximize the signal-to-noise ratio. We evaluated their performances using 128 prepandemic plasma samples (negative controls) and 104 plasma samples from individuals with SARS-CoV-2 diagnosis (positive controls), of whom 5 were asymptomatic, 51 had mild symptoms, and 48 were hospitalized. Preexisting IgG antibodies recognizing N, M, and S proteins were detected in negative controls, which is suggestive of cross-reactivity to common-cold coronaviruses. The best-performing antibody/antigen signatures had specificities of 100% and sensitivities of 95.78% at ≥14 days and 95.65% at ≥21 days since the onset of symptoms, with areas under the curve (AUCs) of 0.977 and 0.999, respectively. Combining multiple markers as assessed by qSAT assays has the highest efficiency, breadth, and versatility to accurately detect low-level antibody responses for obtaining reliable data on the prevalence of exposure to novel pathogens in a population. Our assays will allow gaining insights into antibody correlates of immunity and their kinetics, required for vaccine development to combat the COVID-19 pandemic.
Collapse
Affiliation(s)
- Carlota Dobaño
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Rebeca Santano
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Jordi Chi
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Diana Barrios
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Gemma Ruiz-Olalla
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Natalia Rodrigo Melero
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Carlo Carolis
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Daniel Parras
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Pau Serra
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | - Francisco Carmona-Torre
- Infectious Diseases Division, Clínica Universidad de Navarra, Pamplona, Spain
- Clinical Microbiology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gabriel Reina
- Clínica Universidad de Navarra, Navarra Institute for Health Research, Pamplona, Spain
| | - Pere Santamaria
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Julia McFarlane Diabetes Research Centre, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Alfredo Mayor
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| | - Alberto L García-Basteiro
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
- International Health Department, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Luis Izquierdo
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
15
|
Dobaño C, Vidal M, Santano R, Jiménez A, Chi J, Barrios D, Ruiz-Olalla G, Rodrigo Melero N, Carolis C, Parras D, Serra P, Martínez de Aguirre P, Carmona-Torre F, Reina G, Santamaria P, Mayor A, García-Basteiro AL, Izquierdo L, Aguilar R, Moncunill G. Highly Sensitive and Specific Multiplex Antibody Assays To Quantify Immunoglobulins M, A, and G against SARS-CoV-2 Antigens. J Clin Microbiol 2021. [PMID: 33127841 DOI: 10.1101/2020.06.11.147363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Reliable serological tests are required to determine the prevalence of antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and to characterize immunity to the disease in order to address key knowledge gaps in the coronavirus disease 2019 (COVID-19) pandemic. Quantitative suspension array technology (qSAT) assays based on the xMAP Luminex platform overcome the limitations of rapid diagnostic tests and enzyme-linked immunosorbent assays (ELISAs) with their higher precision, dynamic range, throughput, miniaturization, cost-efficiency, and multiplexing capacity. We developed three qSAT assays for IgM, IgA, and IgG against a panel of eight SARS-CoV-2 antigens, including spike protein (S), nucleocapsid protein (N), and membrane protein (M) constructs. The assays were optimized to minimize the processing time and maximize the signal-to-noise ratio. We evaluated their performances using 128 prepandemic plasma samples (negative controls) and 104 plasma samples from individuals with SARS-CoV-2 diagnosis (positive controls), of whom 5 were asymptomatic, 51 had mild symptoms, and 48 were hospitalized. Preexisting IgG antibodies recognizing N, M, and S proteins were detected in negative controls, which is suggestive of cross-reactivity to common-cold coronaviruses. The best-performing antibody/antigen signatures had specificities of 100% and sensitivities of 95.78% at ≥14 days and 95.65% at ≥21 days since the onset of symptoms, with areas under the curve (AUCs) of 0.977 and 0.999, respectively. Combining multiple markers as assessed by qSAT assays has the highest efficiency, breadth, and versatility to accurately detect low-level antibody responses for obtaining reliable data on the prevalence of exposure to novel pathogens in a population. Our assays will allow gaining insights into antibody correlates of immunity and their kinetics, required for vaccine development to combat the COVID-19 pandemic.
Collapse
Affiliation(s)
- Carlota Dobaño
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Rebeca Santano
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Jordi Chi
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Diana Barrios
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Gemma Ruiz-Olalla
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Natalia Rodrigo Melero
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Carlo Carolis
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Daniel Parras
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Pau Serra
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | - Francisco Carmona-Torre
- Infectious Diseases Division, Clínica Universidad de Navarra, Pamplona, Spain
- Clinical Microbiology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gabriel Reina
- Clínica Universidad de Navarra, Navarra Institute for Health Research, Pamplona, Spain
| | - Pere Santamaria
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Julia McFarlane Diabetes Research Centre, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Alfredo Mayor
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| | - Alberto L García-Basteiro
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
- International Health Department, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Luis Izquierdo
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
16
|
Chauhan R, Gupta N, Tiwari AK, Raina V, Nandi SP. Development of a Novel Multiplex Bead-based Assay for Measuring Autoantibodies on Flow Cytometric Platform. Immunol Invest 2020; 51:588-601. [PMID: 33287608 DOI: 10.1080/08820139.2020.1854782] [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: 10/22/2022]
Abstract
Background: Autoantibodies (AAbs) are important biomarkers for the diagnosis of Autoimmune Diseases (ADs). The detection of AAbs performed by current methods (indirect immunofluorescence test (IIFT)/Immunoblot (dot/line)/enzyme-linked immunosorbent assay ELISA) which have limitations in terms of performing multiple assays to arrive at laboratory diagnosis. We validated a novel multiplex bead-based assay (NMBA) that could quantify five common antibodies, simultaneously, on a flow-cytometry platform.Methods: A total of five recombinant antigens (SS-A Ro60, CENP B, RNP 70, Scl 70 and Histones) were covalently coupled onto beads and tested using known positive sera (positive for AAbs) and analyzed using flow cytometer.Results: The sensitivity, specificity, Positive Predictive Value (PPV) and Negative Predictive Value (NPV) were obtained for each antigen, analyzed by both assays (NMBA and IIFT). It showed comparable or higher values for the NMBA. The Spearman's rank correlation coefficient (Rho) were ≥ 0.97, (P < .05), indicating that multiplexing of the five autoantigens did not alter the results obtained when antigens were tested individually. The mean intra-assay precision measured by coefficient of variation (CV) was7.56 ± 1.6% and the mean inter-assay CV was 10.03 ± 1.34%. The time taken from sample receipt to reporting of results was 90 minutes in NMBA as compared to 150 minutes of IIFTConclusion: The NMBA could quantitatively measure antibodies against five autoantigens, simultaneously in patient's sera. The assay is faster, objective, reproducible, requires low sample volume, and stable. Moreover, the flow cytometer in diagnostic laboratory settings for hematological and transplant immunology tests, can also be used for testing AAbs.
Collapse
Affiliation(s)
- Rajni Chauhan
- Department of Transfusion Medicine, Medanta-The Medicity, Molecular and Transplant Immunology Laboratory, Gurgaon, India.,Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Nikita Gupta
- Department is Molecular Genetics, Chimera Transplant Research Foundation, New Delhi, India
| | - Aseem Kumar Tiwari
- Department of Transfusion Medicine, Medanta-The Medicity, Molecular and Transplant Immunology Laboratory, Gurgaon, India
| | - Vimarsh Raina
- Department is Molecular Genetics, Chimera Transplant Research Foundation, New Delhi, India
| | - Shoma Paul Nandi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| |
Collapse
|
17
|
Contribution of Multiplex Immunoassays to Rheumatoid Arthritis Management: From Biomarker Discovery to Personalized Medicine. J Pers Med 2020; 10:jpm10040202. [PMID: 33142977 PMCID: PMC7712300 DOI: 10.3390/jpm10040202] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 01/18/2023] Open
Abstract
Rheumatoid arthritis (RA) is a multifactorial, inflammatory and progressive autoimmune disease that affects approximately 1% of the population worldwide. RA primarily involves the joints and causes local inflammation and cartilage destruction. Immediate and effective therapies are crucial to control inflammation and prevent deterioration, functional disability and unfavourable progression in RA patients. Thus, early diagnosis is critical to prevent joint damage and physical disability, increasing the chance of achieving remission. A large number of biomarkers have been investigated in RA, although only a few have made it through the discovery and validation phases and reached the clinic. The single biomarker approach mostly used in clinical laboratories is not sufficiently accurate due to its low sensitivity and specificity. Multiplex immunoassays could provide a more complete picture of the disease and the pathways involved. In this review, we discuss the latest proposed protein biomarkers and the advantages of using protein panels for the clinical management of RA. Simultaneous analysis of multiple proteins could yield biomarker signatures of RA subtypes to enable patients to benefit from personalized medicine.
Collapse
|
18
|
Jia A, James E, Lu HY, Sharma M, Modi BP, Biggs CM, Hildebrand KJ, Chomyn A, Erdle S, Kular H, Turvey SE. Clinical IRAK4 deficiency caused by homozygosity for the novel IRAK4 (c.1049delG, p.Gly350Glufs*15) variant. Cold Spring Harb Mol Case Stud 2020; 6:mcs.a005298. [PMID: 32532880 PMCID: PMC7304365 DOI: 10.1101/mcs.a005298] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/15/2020] [Indexed: 11/25/2022] Open
Abstract
The innate immune system allows for rapid recognition of pathogens. Toll-like receptor (TLR) signaling is a key aspect of the innate immune response, and interleukin-1 receptor-associated kinase 4 (IRAK4) plays a vital role in the TLR signaling cascade. Each TLR recognizes a distinct set of pathogen-associated molecular patterns (PAMPs) that encompass conserved microbial components such as lipopolysaccharides and flagellin. Upon binding of PAMPs and TLR activation, TLR intracellular domains initiate the oligomerization of the myeloid differentiation primary response 88 (MyD88), IRAK1, IRAK2, and IRAK4 signaling platform known as the Myddosome complex while also triggering the Toll/IL-1R domain-containing adaptor-inducing IFN-β (TRIF)-dependent pathway. The Myddosome complex initiates signal transduction pathways enabling the activation of NF-κB and mitogen-activated protein kinase (MAPK) transcription factors and the subsequent production of inflammatory cytokines. Human IRAK4 deficiency is an autosomal recessive inborn error of immunity that classically presents with blunted or delayed inflammatory response to infection and susceptibility to a narrow spectrum of pyogenic bacteria, particularly Streptococcus pneumoniae, Staphylococcus aureus, and Pseudomonas aeruginosa. We describe a case of IRAK4 deficiency in an 11-mo-old boy with concurrent S. pneumoniae bacteremia and S. aureus cervical lymphadenitis with a blunted inflammatory response to invasive infection. Although initial clinical immune profiling was unremarkable, a high degree of suspicion for an innate immune defect prompted genetic sequencing. Genetic testing revealed a novel variant in the IRAK4 gene (c.1049delG, p.(Gly350Glufs*15)) predicted to be likely pathogenic. Functional testing showed a loss of IRAK4 protein expression and abolished TLR signaling, confirming the pathogenicity of this novel IRAK4 variant.
Collapse
Affiliation(s)
- Alicia Jia
- Division of Allergy and Clinical Immunology, Department of Pediatrics, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada.,Department of Microbiology and Immunology, Vancouver, British Columbia V5Z 4H4, Canada
| | - Elliot James
- Division of Allergy and Clinical Immunology, Department of Pediatrics, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Henry Y Lu
- Division of Allergy and Clinical Immunology, Department of Pediatrics, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada.,Experimental Medicine Program, Faculty of Medicine, Vancouver, British Columbia V5Z 4H4, Canada
| | - Mehul Sharma
- Division of Allergy and Clinical Immunology, Department of Pediatrics, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada.,Experimental Medicine Program, Faculty of Medicine, Vancouver, British Columbia V5Z 4H4, Canada
| | - Bhavi P Modi
- Department of Medical Genetics, BC Children's Hospital Research Institute, The University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Catherine M Biggs
- Division of Allergy and Clinical Immunology, Department of Pediatrics, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Kyla J Hildebrand
- Division of Allergy and Clinical Immunology, Department of Pediatrics, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Alanna Chomyn
- Division of Allergy and Clinical Immunology, Department of Pediatrics, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Stephanie Erdle
- Division of Allergy and Clinical Immunology, Department of Pediatrics, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Hasandeep Kular
- Division of Allergy and Clinical Immunology, Department of Pediatrics, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Stuart E Turvey
- Division of Allergy and Clinical Immunology, Department of Pediatrics, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada.,Department of Microbiology and Immunology, Vancouver, British Columbia V5Z 4H4, Canada
| |
Collapse
|
19
|
Lau ES, Paniagua SM, Guseh JS, Bhambhani V, Zanni MV, Courchesne P, Lyass A, Larson MG, Levy D, Ho JE. Sex Differences in Circulating Biomarkers of Cardiovascular Disease. J Am Coll Cardiol 2020; 74:1543-1553. [PMID: 31537263 DOI: 10.1016/j.jacc.2019.06.077] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 06/13/2019] [Accepted: 06/24/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Differences in proteomic profiles between men and women may provide insights into the biological pathways that contribute to known sex differences in cardiovascular disease (CVD). OBJECTIVES This study sought to investigate sex differences in circulating biomarkers representative of biological pathways implicated in the development of CVD among Framingham Heart Study participants. METHODS The authors measured 71 circulating CVD protein biomarkers in 7,184 participants (54% women, mean age 49 years). Multivariable models were used to evaluate the associations of sex, menopause, and hormone status with biomarkers. Cox models were used to examine whether sex modified the association of biomarkers with incident CVD. RESULTS Of 71 biomarkers examined, 61 (86%) differed significantly between men and women, of which 37 were higher in women (including adipokines and inflammatory markers such as leptin and C-reactive protein), and 24 were higher in men (including fibrosis and platelet markers such as MMP-8 (matrix metalloproteinase-8) and TIMP-1 (tissue inhibitor of metalloproteinases 1); false discovery rate q < 0.05 for all). Sex differences in biomarker profiles were most pronounced between pre-menopausal women versus men, with attenuated sex differences among post-menopausal women not taking hormone replacement therapy. Sex modified the association of specific biomarkers with incident CVD, including CD14 and apolipoprotein B (pinteraction <0.05 for all). CONCLUSIONS In a predominantly Caucasian population, the authors identified widespread sex differences in circulating biomarkers that reflect distinct pathways implicated in CVD, including inflammation, adiposity, fibrosis, and platelet homeostasis. Menopause and hormone status accounted for some, but not all, of the observed sex differences. Further investigation into factors underlying sex-based differences may provide mechanistic insight into CVD development.
Collapse
Affiliation(s)
- Emily S Lau
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Samantha M Paniagua
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts
| | - James Sawalla Guseh
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Vijeta Bhambhani
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts
| | - Markella V Zanni
- Neuroendocrinology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Paul Courchesne
- Framingham Heart Study, Framingham, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Asya Lyass
- Framingham Heart Study, Framingham, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Martin G Larson
- Framingham Heart Study, Framingham, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Daniel Levy
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Jennifer E Ho
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
20
|
Amodio D, Santilli V, Zangari P, Cotugno N, Manno EC, Rocca S, Rossi P, Cancrini C, Finocchi A, Chassiakos A, Petrovas C, Palma P. How to dissect the plasticity of antigen-specific immune response: a tissue perspective. Clin Exp Immunol 2020; 199:119-130. [PMID: 31626717 PMCID: PMC6954674 DOI: 10.1111/cei.13386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2019] [Indexed: 12/01/2022] Open
Abstract
Generation of antigen-specific humoral responses following vaccination or infection requires the maturation and function of highly specialized immune cells in secondary lymphoid organs (SLO), such as lymph nodes or tonsils. Factors that orchestrate the dynamics of these cells are still poorly understood. Currently, experimental approaches that enable a detailed description of the function of the immune system in SLO have been mainly developed and optimized in animal models. Conversely, methodological approaches in humans are mainly based on the use of blood-associated material because of the challenging access to tissues. Indeed, only few studies in humans were able to provide a discrete description of the complex network of cytokines, chemokines and lymphocytes acting in tissues after antigenic challenge. Furthermore, even fewer data are currently available on the interaction occurring within the complex micro-architecture of the SLO. This information is crucial in order to design particular vaccination strategies, especially for patients affected by chronic and immune compromising medical conditions who are under-vaccinated or who respond poorly to immunizations. Analysis of immune cells in different human tissues by high-throughput technologies, able to obtain data ranging from gene signature to protein expression and cell phenotypes, is needed to dissect the peculiarity of each immune cell in a definite human tissue. The main aim of this review is to provide an in-depth description of the current available methodologies, proven evidence and future perspectives in the analysis of immune mechanisms following immunization or infections in SLO.
Collapse
Affiliation(s)
- D. Amodio
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
- Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
| | - V. Santilli
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - P. Zangari
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - N. Cotugno
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - E. C. Manno
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - S. Rocca
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - P. Rossi
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
- Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
| | - C. Cancrini
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
- Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
| | - A. Finocchi
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
- Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
| | - A. Chassiakos
- Vaccine Research CenterNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMDUSA
| | - C. Petrovas
- Vaccine Research CenterNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMDUSA
| | - P. Palma
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| |
Collapse
|
21
|
Gómez-Banoy N, Guseh JS, Li G, Rubio-Navarro A, Chen T, Poirier B, Putzel G, Rosselot C, Pabón MA, Camporez JP, Bhambhani V, Hwang SJ, Yao C, Perry RJ, Mukherjee S, Larson MG, Levy D, Dow LE, Shulman GI, Dephoure N, Garcia-Ocana A, Hao M, Spiegelman BM, Ho JE, Lo JC. Adipsin preserves beta cells in diabetic mice and associates with protection from type 2 diabetes in humans. Nat Med 2019; 25:1739-1747. [PMID: 31700183 PMCID: PMC7256970 DOI: 10.1038/s41591-019-0610-4] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 09/12/2019] [Indexed: 01/08/2023]
Abstract
Type 2 diabetes is characterized by insulin resistance and a gradual loss of pancreatic beta cell mass and function1,2. Currently, there are no therapies proven to prevent beta cell loss and some, namely insulin secretagogues, have been linked to accelerated beta cell failure, thereby limiting their use in type 2 diabetes3,4. The adipokine adipsin/complement factor D controls the alternative complement pathway and generation of complement component C3a, which acts to augment beta cell insulin secretion5. In contrast to other insulin secretagogues, we show that chronic replenishment of adipsin in diabetic db/db mice ameliorates hyperglycemia and increases insulin levels while preserving beta cells by blocking dedifferentiation and death. Mechanistically, we find that adipsin/C3a decreases the phosphatase Dusp26; forced expression of Dusp26 in beta cells decreases expression of core beta cell identity genes and sensitizes to cell death. In contrast, pharmacological inhibition of DUSP26 improves hyperglycemia in diabetic mice and protects human islet cells from cell death. Pertaining to human health, we show that higher concentrations of circulating adipsin are associated with a significantly lower risk of developing future diabetes among middle-aged adults after adjusting for body mass index (BMI). Collectively, these data suggest that adipsin/C3a and DUSP26-directed therapies may represent a novel approach to achieve beta cell health to treat and prevent type 2 diabetes.
Collapse
Affiliation(s)
- Nicolás Gómez-Banoy
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - J Sawalla Guseh
- Corrigan Minehan Heart Center, Cardiovascular Research Center and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ge Li
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Alfonso Rubio-Navarro
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Tong Chen
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - BreAnne Poirier
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Gregory Putzel
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Carolina Rosselot
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria A Pabón
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - João Paulo Camporez
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Vijeta Bhambhani
- Corrigan Minehan Heart Center, Cardiovascular Research Center and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shih-Jen Hwang
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Chen Yao
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Rachel J Perry
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | | | - Martin G Larson
- Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Daniel Levy
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Lukas E Dow
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Noah Dephoure
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Adolfo Garcia-Ocana
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mingming Hao
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Bruce M Spiegelman
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Jennifer E Ho
- Corrigan Minehan Heart Center, Cardiovascular Research Center and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - James C Lo
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
22
|
The genesis and evolution of bead-based multiplexing. Methods 2019; 158:2-11. [DOI: 10.1016/j.ymeth.2019.01.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/10/2018] [Accepted: 01/14/2019] [Indexed: 12/22/2022] Open
|
23
|
Luo J, Zhang M, Yan B, Li F, Guan S, Chang K, Jiang W, Xu H, Yuan T, Chen M, Deng S. Diagnostic performance of plasma cytokine biosignature combination and MCP-1 as individual biomarkers for differentiating stages Mycobacterium tuberculosis infection. J Infect 2019; 78:281-291. [DOI: 10.1016/j.jinf.2018.10.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022]
|
24
|
Peng M, Qiang L, Xu Y, Li C, Li T, Wang J. IL
‐35 ameliorates collagen‐induced arthritis by promoting
TNF
‐α‐induced apoptosis of synovial fibroblasts and stimulating M2 macrophages polarization. FEBS J 2019; 286:1972-1985. [PMID: 30834683 DOI: 10.1111/febs.14801] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/03/2018] [Accepted: 03/01/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Mingzheng Peng
- Shanghai Key Laboratory of Orthopaedic Implant Department of Orthopaedic Surgery Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine China
| | - Lei Qiang
- Southwest Jiaotong University College of Medicine Chengdu China
| | - Yan Xu
- Southwest Jiaotong University College of Medicine Chengdu China
| | - Cuidi Li
- Med‐X Research Institute School of Biomedical Engineering Shanghai Jiao Tong University China
| | - Tao Li
- Shanghai Key Laboratory of Orthopaedic Implant Department of Orthopaedic Surgery Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic Implant Department of Orthopaedic Surgery Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine China
| |
Collapse
|
25
|
Yamaguchi M, Okamura S, Yamaji T, Iwasaki M, Tsugane S, Shetty V, Koizumi T. Plasma cytokine levels and the presence of colorectal cancer. PLoS One 2019; 14:e0213602. [PMID: 30883594 PMCID: PMC6422333 DOI: 10.1371/journal.pone.0213602] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/25/2019] [Indexed: 02/06/2023] Open
Abstract
Background/Aims Cancer-related activation of cytokine networks are central aspects of tumor development. The goal of the study was to examine the possibility of plasma cytokines for the screening of colorectal cancer (CRC). Methods We carried out a multicenter, hospital-based case-control study in 66 adult Japanese patients with CRC and 87 healthy adult Japanese. A multiplex bead array immunoassay was used to examine 27 different plasma cytokines. Their association with the presence of CRC was evaluated by logistic regression analysis after adjusting for potential confounding factors. Results Thirteen plasma cytokines were notably associated with the presence of CRC (p< 0.05). Receiver operating characteristic analysis revealed that the combinatorial assessment of some of these plasma cytokines showed “good” capability for discriminating between CRC patients and control subjects (area under the curve (AUC): 0.819 for the combination of IL-9, Eotaxin, G-CSF, and TNF-α; 0.832 for the combination of IL-4, IL-8, Eotaxin, IP-10, and TNF-α). Individual cytokine assessments presented lower AUCs (0.657–0.755) than the combinatorial cytokine assessments. Conclusions The levels of several plasma cytokines varied significantly between CRC patients and control subjects, suggesting the possibility of differentially expressed plasma cytokines as potential biomarkers for detecting the presence of CRC. Our results should be validated in other populations.
Collapse
Affiliation(s)
- Masaki Yamaguchi
- Shinshu University, Graduate School of Science & Technology, Department of Mechanical Engineering & Robotics, Ueda, Nagano, Japan
- * E-mail:
| | - Shin Okamura
- Shinshu University, Graduate School of Science & Technology, Department of Mechanical Engineering & Robotics, Ueda, Nagano, Japan
| | - Taiki Yamaji
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Motoki Iwasaki
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Shoichiro Tsugane
- Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Vivek Shetty
- Section of Oral & Maxillofacial Surgery, UCLA Health Sciences Center, Los Angeles, CA, United States of America
| | - Tomonobu Koizumi
- Shinshu University School of Medicine, Department of Comprehensive Cancer Therapy, Matsumoto, Nagano, Japan
| |
Collapse
|
26
|
Reslova N, Huvarova V, Hrdy J, Kasny M, Kralik P. A novel perspective on MOL-PCR optimization and MAGPIX analysis of in-house multiplex foodborne pathogens detection assay. Sci Rep 2019; 9:2719. [PMID: 30804418 PMCID: PMC6389906 DOI: 10.1038/s41598-019-40035-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/06/2019] [Indexed: 12/28/2022] Open
Abstract
Multiplex oligonucleotide ligation-PCR (MOL-PCR) is a rapid method for simultaneous detection of multiple molecular markers within a single reaction. MOL-PCR is increasingly employed in microbial detection assays, where its ability to facilitate identification and further characterization via simple analysis is of great benefit and significantly simplifies routine diagnostics. When adapted to microsphere suspension arrays on a MAGPIX reader, MOL-PCR has the potential to outperform standard nucleic acid-based diagnostic assays. This study represents the guideline towards in-house MOL-PCR assay optimization using the example of foodborne pathogens (bacteria and parasites) with an emphasis on the appropriate choice of crucial parameters. The optimized protocol focused on specific sequence detection utilizes the fluorescent reporter BODIPY-TMRX and self-coupled magnetic microspheres and allows for a smooth and brisk workflow which should serve as a guide for the development of MOL-PCR assays intended for pathogen detection.
Collapse
Affiliation(s)
- Nikol Reslova
- Veterinary Research Institute, Department of Food and Feed Safety, Hudcova 296/70, 621 00, Brno, Czech Republic. .,Faculty of Science, Department of Botany and Zoology, Masaryk University, Kotlářská 2, 611 37, Brno, Czech Republic.
| | - Veronika Huvarova
- Veterinary Research Institute, Department of Food and Feed Safety, Hudcova 296/70, 621 00, Brno, Czech Republic.,Faculty of Science, Department of Experimental Biology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Jakub Hrdy
- Veterinary Research Institute, Department of Food and Feed Safety, Hudcova 296/70, 621 00, Brno, Czech Republic.,Faculty of Science, Department of Experimental Biology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Martin Kasny
- Faculty of Science, Department of Botany and Zoology, Masaryk University, Kotlářská 2, 611 37, Brno, Czech Republic
| | - Petr Kralik
- Veterinary Research Institute, Department of Food and Feed Safety, Hudcova 296/70, 621 00, Brno, Czech Republic
| |
Collapse
|
27
|
van den Berg R, Jongbloed EM, de Schepper EIT, Bierma-Zeinstra SMA, Koes BW, Luijsterburg PAJ. The association between pro-inflammatory biomarkers and nonspecific low back pain: a systematic review. Spine J 2018; 18:2140-2151. [PMID: 29960111 DOI: 10.1016/j.spinee.2018.06.349] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 06/20/2018] [Accepted: 06/20/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT About 85% of the patients with low back pain seeking medical care have nonspecific low back pain (NsLBP), implying that no definitive cause can be identified. Nonspecific low back pain is defined as low back pain and disability which cannot be linked to an underlying pathology, such as cancer, spinal osteomyelitis, fracture, spinal stenosis, cauda equine, ankylosing spondylitis, and visceral-referred pain. Many pain conditions are linked with elevated serum levels of pro-inflammatory biomarkers. Outcomes of interest are NsLBP and the level of pro-inflammatory biomarkers. PURPOSE To unravel the etiology and get better insight in the prognosis of NsLBP, the aim of this study was to assess the association between pro-inflammatory biomarkers and the presence and severity of NsLBP. STUDY DESIGN A systematic literature search was made in Embase, Medline, Cinahl, Webof-science, and Google scholar up to January 19th 2017. METHODS Included were cross-sectional and cohort studies reporting on patients aged over 18 years with NsLBP, in which one or more pro-inflammatory biomarkers were measured in blood plasma. The methodological quality of the included studies was assessed using the Newcastle Ottawa Scale. A best-evidence synthesis was used to summarize the results from the individual studies, meaning that the included studies were ranked according to the consistency of the findings and according to their methodological quality score using the Newcastle Ottawa Scale. RESULTS Included were 10 studies which assessed four different pro-inflammatory biomarkers. For the association between the presence of NsLBP and C-reactive protein (CRP), interleukin 6 (IL-6) and tumor necrosis factor (TNF)-α limited, conflicting and moderate evidence, respectively, was found. For the association between the severity of NsLBP and CRP and IL-6, moderate evidence was found. For the association between the severity of NsLBP and TNF-α and RANTES Regulated on Activation, Normal T Cell Expressed and Secreted conflicting and limited evidence, respectively, was found. CONCLUSIONS This study found moderate evidence for (i) a positive association between the pro-inflammatory biomarkers CRP and IL-6 and the severity of NsLBP, and (ii) a positive association between TNF-α and the presence of NsLBP. Conflicting and limited evidence was found for the association between TNF-α and Regulated on Activation, Normal T Cell Expressed and Secreted and severity of NsLBP, respectively.
Collapse
Affiliation(s)
- R van den Berg
- Department of General Practice, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| | - E M Jongbloed
- Department of General Practice, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - E I T de Schepper
- Department of General Practice, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - S M A Bierma-Zeinstra
- Department of General Practice, Erasmus MC, University Medical Center, Rotterdam, The Netherlands; Department of Orthopedics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - B W Koes
- Department of General Practice, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - P A J Luijsterburg
- Department of General Practice, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
28
|
Çetin A, Şen A, Çetin I, Çimen B, Cimen L, Savas G, Öztürk A, Koker MY. Comparison of ELISA and flow cytometry for measurement of interleukin-1 beta, interleukin-6 and tumor necrosis factor-α. TURKISH JOURNAL OF BIOCHEMISTRY 2018. [DOI: 10.1515/tjb-2017-0164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractBackgroundAlthough majority of the previous studies have shown a good correlation between enzyme linked immuno sorbent assay (ELISA) and flow cytometry in terms of cytokines, two laboratory methods usually were compared with the regression analysis and correlation in the literature. This study aimed at comparing the ELISA and flow cytometry assay for measuring cytokines by using two different statistical methods, regression analysis and Bland-Altman plot.Materials and methodsFifty patients, diagnosed with hypercholesterolemia and expecting high level serum cytokines, and 30 healthy volunteers, expecting normal level serum cytokines, were enrolled in the study. The interleukin-1 beta (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) were measured using ELISA, and compared with obtained levels using flow cytometric method.ResultsAlthough regression analysis showed that the two methods are compatible with measurements of IL-1β, IL-6 and TNF-α, they tended to show dissimilarity with measurements of IL-1β and TNF-α based on Bland-Altman graphs.ConclusionAccording to Bland-Altman plot, our results providing evidence of ELISA and flow cytometry assays were compatible with each other for IL-1β and IL-6 measurements compared to TNF-α measurement. However, our study has a small number of participants, hence this study need to be confirmed by investigations involving more participants.
Collapse
|
29
|
Yao C, Chen G, Song C, Keefe J, Mendelson M, Huan T, Sun BB, Laser A, Maranville JC, Wu H, Ho JE, Courchesne P, Lyass A, Larson MG, Gieger C, Graumann J, Johnson AD, Danesh J, Runz H, Hwang SJ, Liu C, Butterworth AS, Suhre K, Levy D. Genome-wide mapping of plasma protein QTLs identifies putatively causal genes and pathways for cardiovascular disease. Nat Commun 2018; 9:3268. [PMID: 30111768 PMCID: PMC6093935 DOI: 10.1038/s41467-018-05512-x] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/09/2018] [Indexed: 01/17/2023] Open
Abstract
Identifying genetic variants associated with circulating protein concentrations (protein quantitative trait loci; pQTLs) and integrating them with variants from genome-wide association studies (GWAS) may illuminate the proteome's causal role in disease and bridge a knowledge gap regarding SNP-disease associations. We provide the results of GWAS of 71 high-value cardiovascular disease proteins in 6861 Framingham Heart Study participants and independent external replication. We report the mapping of over 16,000 pQTL variants and their functional relevance. We provide an integrated plasma protein-QTL database. Thirteen proteins harbor pQTL variants that match coronary disease-risk variants from GWAS or test causal for coronary disease by Mendelian randomization. Eight of these proteins predict new-onset cardiovascular disease events in Framingham participants. We demonstrate that identifying pQTLs, integrating them with GWAS results, employing Mendelian randomization, and prospectively testing protein-trait associations holds potential for elucidating causal genes, proteins, and pathways for cardiovascular disease and may identify targets for its prevention and treatment.
Collapse
Affiliation(s)
- Chen Yao
- Framingham Heart Study, Framingham, 01702, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - George Chen
- Framingham Heart Study, Framingham, 01702, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Ci Song
- Framingham Heart Study, Framingham, 01702, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, MD, USA
- Department of Medical Sciences, Uppsala University, 75105, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, 75105, Uppsala, Sweden
| | - Joshua Keefe
- Framingham Heart Study, Framingham, 01702, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Michael Mendelson
- Framingham Heart Study, Framingham, 01702, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, MD, USA
- Department of Cardiology, Boston Children's Hospital, Boston, 02115, MA, USA
| | - Tianxiao Huan
- Framingham Heart Study, Framingham, 01702, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Benjamin B Sun
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Annika Laser
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | | | - Hongsheng Wu
- Computer Science and Networking, Wentworth Institute of Technology, Boston, 02115, MA, USA
| | - Jennifer E Ho
- Cardiovascular Research Center and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, 02114, MA, USA
| | - Paul Courchesne
- Framingham Heart Study, Framingham, 01702, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Asya Lyass
- Framingham Heart Study, Framingham, 01702, MA, USA
- Department of Mathematics and Statistics, Boston University, Boston, 02115, MA, USA
| | - Martin G Larson
- Framingham Heart Study, Framingham, 01702, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, 02118, MA, USA
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Johannes Graumann
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, W.G. Kerckhoff Institute, Ludwigstr. 43, D-61231, Bad Nauheim, Germany
| | - Andrew D Johnson
- Framingham Heart Study, Framingham, 01702, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - John Danesh
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
- British Heart Foundation Cambridge Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
- Department of Human Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1RQ, UK
| | - Heiko Runz
- MRL, Merck & Co., Inc, Kenilworth, 07033, NJ, USA
| | - Shih-Jen Hwang
- Framingham Heart Study, Framingham, 01702, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Chunyu Liu
- Framingham Heart Study, Framingham, 01702, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Adam S Butterworth
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Genomics, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, PO 24144, Doha, Qatar
| | - Daniel Levy
- Framingham Heart Study, Framingham, 01702, MA, USA.
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, MD, USA.
| |
Collapse
|
30
|
Verma SK, Albrecht AK, Siebecke V, Klöck G, Kolesnikova TA, Springer S. Comparative validation of a microcapsule-based immunoassay for the detection of proteins and nucleic acids. PLoS One 2018; 13:e0201009. [PMID: 30028867 PMCID: PMC6054379 DOI: 10.1371/journal.pone.0201009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 07/06/2018] [Indexed: 01/12/2023] Open
Abstract
To detect and study diseases, research and clinical laboratories must quantify specific biomarkers in the plasma and urine of patients with precision, sensitivity, and cost-effectiveness. Newly developed techniques, such as particle-based immunoassays, must be validated in these terms against standard methods such as enzyme-linked immunosorbent assays (ELISAs). Here, we compare the performance of assays that use hollow polyelectrolyte microcapsules with assays based on solid plastic beads, and with standard microplate immunoassays. The polyelectrolyte microcapsules detect the disease biomarker beta-2 microglobulin with a fifty-fold increase in sensitivity than polystyrene (PS) beads. For sequence-specific nucleic acid detection, the oligonucleotide-coated microcapsules exhibit a two-fold lower increase in sensitivity over PS beads. The microcapsules also detect the presence of a monoclonal antibody in hybridoma supernatant at a fifty-six-fold increase in sensitivity compared to a microplate assay. Overall, polyelectrolyte microcapsule-based assays are more sensitive for the detection of protein and nucleic acid analytes than PS beads and microplate assays, and they are viable alternatives as a platform for the rapid quantitative detection of analytes at very low concentrations.
Collapse
Affiliation(s)
- Sujit Kumar Verma
- Department of Life Sciences and Chemistry, Jacobs University, Bremen, Germany
| | | | | | - Gerd Klöck
- City University of Applied Sciences, Bremen, Germany
| | | | - Sebastian Springer
- Department of Life Sciences and Chemistry, Jacobs University, Bremen, Germany
- * E-mail:
| |
Collapse
|
31
|
Ho JE, Lyass A, Courchesne P, Chen G, Liu C, Yin X, Hwang SJ, Massaro JM, Larson MG, Levy D. Protein Biomarkers of Cardiovascular Disease and Mortality in the Community. J Am Heart Assoc 2018; 7:e008108. [PMID: 30006491 PMCID: PMC6064847 DOI: 10.1161/jaha.117.008108] [Citation(s) in RCA: 198] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 05/28/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND The discovery of novel and highly predictive biomarkers of cardiovascular disease (CVD) has the potential to improve risk-stratification methods and may be informative regarding biological pathways contributing to disease. METHODS AND RESULTS We used a discovery proteomic platform that targeted high-value proteins for CVD to ascertain 85 circulating protein biomarkers in 3523 Framingham Heart Study participants (mean age, 62 years; 53% women). Using multivariable-adjusted Cox models to account for clinical variables, we found 8 biomarkers associated with incident atherosclerotic CVD, 18 with incident heart failure, 38 with all-cause mortality, and 35 with CVD death (false discovery rate, q<0.05 for all; P-value ranges, 9.8×10-34 to 3.6×10-2). Notably, a number of regulators of metabolic and adipocyte homeostasis were associated with cardiovascular events, including insulin-like growth factor 1 (IGF1), insulin-like growth factor binding protein 1 (IGFBP1), insulin-like growth factor binding protein 2 (IGFBP2), leptin, and adipsin. In a multimarker approach that accounted for clinical factors, growth differentiation factor 15 (GDF15) was associated with all outcomes. In addition, N-terminal pro-b-type natriuretic peptide, C-reactive protein, and leptin were associated with incident heart failure, and C-type lectin domain family 3 member B (CLEC3B; tetranectin), N-terminal pro-b-type natriuretic peptide, arabinogalactan protein 1 (AGP1), soluble receptor for advanced glycation end products (sRAGE), peripheral myelin protein 2 (PMP2), uncarboxylated matrix Gla protein (UCMGP), kallikrein B1 (KLKB1), IGFBP2, IGF1, leptin receptor, and cystatin-C were associated with all-cause mortality in a multimarker model. CONCLUSIONS We identified numerous protein biomarkers that predicted cardiovascular outcomes and all-cause mortality, including biomarkers representing regulators of metabolic homeostasis and inflammatory pathways. Further studies are needed to validate our findings and define clinical utility, with the ultimate goal of improving strategies for CVD prevention.
Collapse
Affiliation(s)
- Jennifer E Ho
- Division of Cardiology, Department of Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Asya Lyass
- Department of Mathematics and Statistics, Boston University, Boston, MA
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA
| | - Paul Courchesne
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA
| | - George Chen
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA
| | - Chunyu Liu
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Xiaoyan Yin
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Joseph M Massaro
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA
- Department of Biostatistics, Boston University School of Public Health, Boston
| | - Martin G Larson
- Department of Mathematics and Statistics, Boston University, Boston, MA
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA
- Department of Biostatistics, Boston University School of Public Health, Boston
| | - Daniel Levy
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| |
Collapse
|
32
|
El Khoury P, Roussel R, Fumeron F, Abou-Khalil Y, Velho G, Mohammedi K, Jacob MP, Steg PG, Potier L, Ghaleb Y, Elbitar S, Ragot S, Andreata F, Caligiuri G, Hadjadj S, Boileau C, Marre M, Abifadel M, Varret M, Hansel B. Plasma proprotein-convertase-subtilisin/kexin type 9 (PCSK9) and cardiovascular events in type 2 diabetes. Diabetes Obes Metab 2018; 20:943-953. [PMID: 29205760 DOI: 10.1111/dom.13181] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/13/2017] [Accepted: 11/30/2017] [Indexed: 12/12/2022]
Abstract
AIM To investigate whether plasma concentrations of proprotein-convertase-subtilisin/kexin type 9 (PCSK9) were associated with cardiovascular (CV) events in two cohorts of patients with type 2 diabetes mellitus. METHODS We considered patients from the DIABHYCAR (n = 3137) and the SURDIAGENE (n = 1468) studies. Baseline plasma PCSK9 concentration was measured using an immunofluorescence assay. In post hoc, but preplanned, analyses we assessed the relationship between PCSK9 and the following endpoints: (1) a combined endpoint of major CV events: CV death, non-fatal myocardial infarction (MI), stroke and heart failure-related hospital admission; (2) a composite of all CV events: MI, stroke, heart failure-related hospital admission, coronary/peripheral angioplasty or bypass, CV death; (3) MI; (4) stroke/transient ischaemic attack (TIA); and (5) CV death. RESULTS In the DIABHYCAR study, plasma PCSK9 tertiles were associated with the incidence of MI, all CV events and stroke/TIA (P for trend <.05). In adjusted Cox analysis, plasma PCSK9 was associated, independently of classic risk factors, with the incidence of major CV events (hazard ratio [HR] for 1-unit increase of log[PCSK9] 1.28 [95% confidence interval {CI} 1.06-1.55]), the incidence of MI (HR 1.66 [95% CI 1.05-2.63]), and the incidence of all CV events (HR 1.22 [95% CI 1.04-1.44]), but not with CV death. Plasma PCSK9 was not associated with the incidence of CV disease in the participants of the SURDIAGENE study with high CV risk treated with statins and insulin. CONCLUSIONS We found that PCSK9 was inconsistently associated with CV events in populations with type 2 diabetes. The association may depend on the level of CV risk and the background treatment.
Collapse
Affiliation(s)
- Petra El Khoury
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
| | - Ronan Roussel
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Frederic Fumeron
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Yara Abou-Khalil
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
| | - Gilberto Velho
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Kamel Mohammedi
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Marie-Paule Jacob
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Philippe Gabriel Steg
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- FACT, HUPNVS, Département de Cardiologie, AP-HP, CHU Xavier Bichat, Paris, France
- NHLI, Imperial College, Royal Brompton Hospital, London, UK
| | - Louis Potier
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Youmna Ghaleb
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Sandy Elbitar
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Stephanie Ragot
- Centre Investigation Clinique 1402, University of Poitiers, Poitiers, France
- Centre Investigation Clinique, CHU Poitiers, Poitiers, France
- Centre Investigation Clinique CIC1402, INSERM, Poitiers, France
| | - Francesco Andreata
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Giusepinna Caligiuri
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Samy Hadjadj
- Centre Investigation Clinique 1402, University of Poitiers, Poitiers, France
- Centre Investigation Clinique, CHU Poitiers, Poitiers, France
- Centre Investigation Clinique CIC1402, INSERM, Poitiers, France
- Pole DUNE, CHU Poitiers, Poitiers, France
- U1082, INSERM, Poitiers, France
| | - Catherine Boileau
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Département de génétique, AP-HP, CHU Xavier Bichat, Paris, France
| | - Michel Marre
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Marianne Abifadel
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
| | - Mathilde Varret
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Boris Hansel
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| |
Collapse
|
33
|
Mucksová J, Chalupský K, Plachý J, Kalina J, Rachačová P, Staněk O, Trefil P. Simultaneous detection of chicken cytokines in plasma samples using the Bio-Plex assay. Poult Sci 2018; 97:1127-1133. [DOI: 10.3382/ps/pex411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 12/06/2017] [Indexed: 11/20/2022] Open
|
34
|
Poetz O, Dieze T, Hammer H, Weiß F, Sommersdorf C, Templin MF, Esdar C, Zimmermann A, Stevanovic S, Bedke J, Stenzl A, Joos TO. Peptide-Based Sandwich Immunoassay for the Quantification of the Membrane Transporter Multidrug Resistance Protein 1. Anal Chem 2018; 90:5788-5794. [DOI: 10.1021/acs.analchem.8b00152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Oliver Poetz
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- SIGNATOPE GmbH Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Theresa Dieze
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Helen Hammer
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- SIGNATOPE GmbH Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Frederik Weiß
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- SIGNATOPE GmbH Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Cornelia Sommersdorf
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- SIGNATOPE GmbH Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Markus F. Templin
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Christina Esdar
- Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | | | - Stefan Stevanovic
- Eberhard Karls University, Department of Immunology, 72076 Tübingen, Germany
| | - Jens Bedke
- Eberhard Karls University, Department of Urology, 72076 Tübingen, Germany
| | - Arnulf Stenzl
- Eberhard Karls University, Department of Urology, 72076 Tübingen, Germany
| | - Thomas O. Joos
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- SIGNATOPE GmbH Markwiesenstrasse 55, 72770 Reutlingen, Germany
| |
Collapse
|
35
|
Evans RL, Pottala JV, Nagata S, Egland KA. Longitudinal autoantibody responses against tumor-associated antigens decrease in breast cancer patients according to treatment modality. BMC Cancer 2018; 18:119. [PMID: 29386014 PMCID: PMC5793406 DOI: 10.1186/s12885-018-4022-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/23/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Metastatic breast cancer (BCa) is most often diagnosed months after completion of treatment of the primary tumor when a patient reports physical symptoms. Besides a physical examination, no other alternative recurrence screening method is recommended for routine follow-up care. Detection of autoantibodies against tumor-associated antigens (TAAs) has demonstrated promise for distinguishing healthy women from patients diagnosed with primary BCa. However, it is unknown what changes occur to patient autoantibody levels during and after treatment. METHODS Three serial blood draws were collected from 200 BCa patients: before treatment, 6 and 12 months after surgery. Patients were categorized according to treatment regimen, including surgery, chemotherapy, radiation, trastuzumab and hormonal therapies. The longitudinal samples were assayed for autoantibody responses against 32 conformation-carrying TAAs using a Luminex multiplex bead assay. RESULTS The treatment modality groups that had the greatest decrease in autoantibody response levels were radiation + hormonal therapy; radiation + chemotherapy; and radiation + hormonal therapy + chemotherapy. For these three treatment groups, autoantibody responses against 9 TAAs (A1AT, ANGPTL4, CAPC, CST2, DKK1, GFRA1, GRN, LGALS3 and LRP10) were significantly reduced at 12 months after surgery compared to before treatment. One TAA, GRP78, had a significantly increased autoantibody response after 12 months. CONCLUSIONS Single treatment regimens alone did not significantly alter autoantibodies levels against the studied TAAs. Radiation treatment was the common denominator of the three most affected groups for significant changes in autoantibody response levels.
Collapse
Affiliation(s)
- Rick L Evans
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD, USA
| | - James V Pottala
- Sanford School of Medicine, University of South Dakota, 2301 East 60th Street North, Sioux Falls, SD, 57104, USA
| | - Satoshi Nagata
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki-City, Osaka, 5670085, Japan
| | - Kristi A Egland
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD, USA. .,Sanford School of Medicine, University of South Dakota, 2301 East 60th Street North, Sioux Falls, SD, 57104, USA.
| |
Collapse
|
36
|
Dysinger M, Marusov G, Fraser S. Quantitative analysis of four protein biomarkers: An automated microfluidic cartridge-based method and its comparison to colorimetric ELISA. J Immunol Methods 2017; 451:1-10. [DOI: 10.1016/j.jim.2017.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/14/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022]
|
37
|
Rho J, Jang W, Hwang I, Lee D, Lee CH, Chung TD. Multiplex immunoassays using virus-tethered gold microspheres by DC impedance-based flow cytometry. Biosens Bioelectron 2017; 102:121-128. [PMID: 29128714 DOI: 10.1016/j.bios.2017.11.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/31/2017] [Accepted: 11/06/2017] [Indexed: 11/17/2022]
Abstract
Bead-based multiplex immunoassays for common use require enhanced sensitivity and effective prevention of non-specific adsorption, as well as miniaturization of the detection device. In this work, we have implemented virus-tethered gold microspheres for multiplex immunoassay applications, employing a DC impedance-based flow cytometer as a detection element. The advantages of virus-tethered gold microspheres, including excellent prevention of non-specific adsorption, are extended to signal enhancement arising from the large quantity of antibody loading on each virion, and to flexible movement of filamentous virus. Individual virus-tethered beads generate their own DC impedance and fluorescence signals, which are simultaneously detected by a chip-based microfluidic flow cytometer. This system successfully realized multiplex immunoassays involving four biomarkers: cardiac troponin I (cTnI), prostate specific antigen (PSA), creatine kinase MB (CK-MB), and myoglobin in undiluted human sera, elevating sensitivity by up to 5.7-fold compared to the beads without virus. Constructive integration between filamentous virus-tethered Au-layered microspheres and use of a microfluidic cytometer suggests a promising strategy for competitive multiplex immunoassay development based on suspension arrays.
Collapse
Affiliation(s)
- Jihun Rho
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Woohyuk Jang
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Inseong Hwang
- InSol Co., Ltd., Yangjae-daero 85-gil, Gangdong-gu, Seoul 05408, Republic of Korea
| | - Dahye Lee
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Chang Heon Lee
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Taek Dong Chung
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea; Program in Nano Science and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Suwon-Si, Gyeonggi-do 16229, Republic of Korea; Advanced Institutes of Convergence Technology, Suwon-Si, Gyeonggi-do 16229, Republic of Korea.
| |
Collapse
|
38
|
Kamińska A, Sprynskyy M, Winkler K, Szymborski T. Ultrasensitive SERS immunoassay based on diatom biosilica for detection of interleukins in blood plasma. Anal Bioanal Chem 2017; 409:6337-6347. [PMID: 28852782 PMCID: PMC5641273 DOI: 10.1007/s00216-017-0566-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/07/2017] [Accepted: 08/02/2017] [Indexed: 01/29/2023]
Abstract
An ultrasensitive surface-enhanced Raman scattering (SERS) immunoassay based on diatom biosilica with integrated gold nanoparticles (AuNPs) for the detection of interleukin 8 (IL-8) in blood plasma has been developed. The SERS sensing originates from unique features of the diatom frustules, which are capable of enhancing the localized surface-plasmon resonance of metal nanostructures. The SERS immune tags ware fabricated by functionalizing 70-nm Au nanoparticles with DTNB (i.e., 5,5′-dithiobis(2-nitrobenzoic acid)), which acted as a Raman reporter molecule, as well as the specific antibodies. These DTNB-labeled immune-AuNPs can form a sandwich structure with IL-8 antigens (infection marker) and the antibodies immobilized on the biosilica material. Our method showed an improved IL-8 detection limit in comparison to standard ELISA methods. The current detection limit for IL-8 using a conventional ELISA test is about 15.6 pg mL−1. The lower detection limit for IL-8 in blood plasma was estimated to be 6.2 pg mL−1. To the best of our knowledge, this is the first report on the recognition of IL-8 in human samples using a SERS-based method. This method clearly possesses high sensitivity to clinically relevant interleukin concentrations in body fluids. The average relative standard deviation of this method is less than 8%, which is sufficient for analytical analysis and comparable to those of classical ELISA methods. This SERS immunoassay also exhibits high biological specificity for the detection of IL-8 antigens. The established SERS immunoassay offers a valuable platform for the ultrasensitive and highly specific detection of immune biomarkers in a clinical setting for medical diagnostics. The SERS-based immunoassay based on naturally generated photonic biosilica for the detection of interleukin 8 (IL-8) in human plasma samples ![]()
Collapse
Affiliation(s)
- Agnieszka Kamińska
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224, Warsaw, Poland.
| | - Myroslav Sprynskyy
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University, 7 Gagarina Str, 87-100, Toruń, Poland
| | - Katarzyna Winkler
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224, Warsaw, Poland
| | - Tomasz Szymborski
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224, Warsaw, Poland
| |
Collapse
|
39
|
Rodrigues V, Baudier JB, Chantal I. Development of a bead-based multiplexed assay for simultaneous quantification of five bovine cytokines by flow cytometry. Cytometry A 2017; 91:901-907. [PMID: 28700121 DOI: 10.1002/cyto.a.23170] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/14/2017] [Accepted: 06/26/2017] [Indexed: 01/01/2023]
Abstract
Quantifying cytokines is extremely important in studies of host-pathogen interactions. Multiplex assays are commercially available but only for human and mouse cytokines. Here a method for the simultaneous quantification of five important bovine cytokines IFNγ, IL-4, IL-10, IL-12, and TNFα in cell culture supernatants, using flow cytometry was reported. Functional beads from BD Biosciences expressing specific APC intensity were used. Commercially available antibodies against bovine cytokines were covalently coupled to beads as capture antibodies. Fixed recombinant cytokines were revealed with a second monoclonal antibody coupled with biotin, then revealed with streptavidin-PE. This complex was analyzed using a standard flow cytometer. Experiments were performed to check no cross reactions had occurred. The limits of detection ranged between 0.08 and 0.4 ng/ml depending on the cytokine, and the linearity between the lower and higher limits was remarkable (R2 > 99.8%). Finally, native cytokines from cell culture supernatants were tested. Results were compared using the standard ELISA test and showed that concentrations of native cytokine in cell culture supernatants were comparable with the two methods, with a wider dynamic range using beads and flow cytometry than with ELISA assays. Bovine IFNγ, IL-4, IL-10, IL-12, and TNFα in culture supernatants can be now simultaneously detected in a single assay, using a standard flow cytometer for both basic and high-throughput analyses. © 2017 International Society for Advancement of Cytometry.
Collapse
|
40
|
Amelioration of adjuvant-induced arthritis in CCDC134-overexpressing transgenic mice. Biochem Biophys Res Commun 2017; 490:111-116. [PMID: 28571739 DOI: 10.1016/j.bbrc.2017.05.166] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/28/2017] [Indexed: 01/23/2023]
Abstract
CCDC134 might be an immune cytokine and plays important and complex roles in the process in vivo. It was proved to illustrate its potent antitumor effects by augmenting CD8+ T-cell-mediated immunity, but its role in the development of rheumatoid arthritis (RA) remains unclear. In this study, we demonstrated that development of adjuvant-induced arthritis and pro-inflammatory responses were more ameliorated in CCDC134-overexpressing transgenic mice than those in WT mice. The underlying mechanism of CCDC134-induced effects involved inhibition of T helper (Th) 1 and Th17 cell differentiation. These findings indicate that overexpression of CCDC134 exerts potent anti-inflammatory effects through selective modulation of pathogenic Th1 and Th17 cells, and might provide insights into the role of CCDC134 as a unique therapeutic agent for the treatment of rheumatoid arthritis.
Collapse
|
41
|
Ellairaja S, Krithiga N, Ponmariappan S, Vasantha VS. Novel Pyrimidine Tagged Silver Nanoparticle Based Fluorescent Immunoassay for the Detection of Pseudomonas aeruginosa. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:1802-1812. [PMID: 28161944 DOI: 10.1021/acs.jafc.6b04790] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A simple pyrimidine-based fluorescent probe (R)-4-(anthracen-9-yl)-6- (naphthalen-1-yl)-1,6-dihydropyrimidine-2-amine (ANDPA) was synthesized through the greener one pot reaction and characterized by IR, NMR, and ESI-Mass. Glucose stabilized silver nanoparticles (Glu-AgNPs) were also synthesized and characterized using UV, IR, XRD, SEM, and TEM. When ANDPA was tagged with Glu-AgNPs, the fluorescent intensity of ANDPA decreased drastically. When the monoclonal antibody (Ab) [immunoglobulin G (IgG)] of Pseudomonas aeruginosa (PA) was attached with ANDPA/Glu-AgNPs, the original intensity of the probe was recovered with minimal enhancement at 446 nm. On further attachment of PA with ANDPA/Glu-AgNPs/PA, the fluorescence intensity of the probe was enhanced obviously at 446 nm with red shift. This phenomenon was further supported by SEM and TEM. The linear range of detection is from 8 to 10-1 CFU/mL, and LOD is 1.5 CFU/mL. The immunosensor was successfully demonstrated to detect Pseudomonas aeruginosa in water, soil, and food products like milk, sugar cane, and orange juices.
Collapse
Affiliation(s)
- Sundaram Ellairaja
- Department of Natural Products Chemistry, School of Chemistry, Madurai Kamaraj University , Madurai-625 021, Tamilnadu, India
| | - Narayanaswamy Krithiga
- Department of Plant Biotechnology, School of Biotechnology, Madurai Kamaraj University , Madurai-625 021, Tamilnadu, India
| | - Sarkaraisamy Ponmariappan
- Biotechnology Division, Defence Research Development & Establishment , Jhansi Road, Gwalior 474002, Madhya Pradesh, India
| | - Vairathevar Sivasamy Vasantha
- Department of Natural Products Chemistry, School of Chemistry, Madurai Kamaraj University , Madurai-625 021, Tamilnadu, India
| |
Collapse
|
42
|
Surenaud M, Manier C, Richert L, Thiébaut R, Levy Y, Hue S, Lacabaratz C. Optimization and evaluation of Luminex performance with supernatants of antigen-stimulated peripheral blood mononuclear cells. BMC Immunol 2016; 17:44. [PMID: 27835944 PMCID: PMC5106791 DOI: 10.1186/s12865-016-0182-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 11/03/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The Luminex bead-based multiplex assay is useful for quantifying immune mediators such as cytokines and chemokines. Cross-comparisons of reagents for this technique from different suppliers have already been performed using serum or plasma but rarely with supernatants collected from antigen-stimulated peripheral blood mononuclear cells (PBMC). Here, we first describe an optimization protocol for cell culture including quantity of cells and culture duration to obtain reproducible cytokine and chemokine quantifications. Then, we compared three different Luminex kit suppliers. RESULTS Intraclass correlation coefficients (ICCs) for a 2-days stimulation protocol were >0.8 for IFNγ and Perforin. The specific concentration was maximal after two or five days of stimulation, depending on the analyte, using 0.5 million PBMC per well, a cell quantity that gave the same level of specific cytokine secretion as 1.0 million. In the second part of the study, Luminex kits from Millipore showed a better working range than Bio-Rad and Ozyme ones. For tuberculin purified protein derivative (PPD)-stimulated samples, the overall mean pooled coefficients of variation (CVs) for all donors and all cytokines was 17.2 % for Bio-Rad, 19.4 % for Millipore and 26.7 % for Ozyme. Although the different kits gave cytokine concentrations that were generally compatible, there were discrepancies for particular cytokines. Finally, evaluation of precision and reproducibility of a 15-plex Millipore kit using a "home-made" internal control showed a mean intra-assay CV <13 % and an inter-assay CV <18 % for each cytokine concentration. CONCLUSIONS A protocol with a single round of stimulation but with two time points gave the best results for assaying different cytokines. Millipore kits appear to be slightly more sensitive than those from Bio-Rad and Ozyme. However, we conclude that the panel of analytes that need to be quantified should be the main determinant of kit selection. Using an internal control we demonstrated that a 15-plex magnetic Milliplex kit displayed good precision and reproducibility. Our findings should help optimize assays for evaluating immune responses during the course of disease or infection, or in response to vaccine or therapy.
Collapse
Affiliation(s)
- Mathieu Surenaud
- INSERM, U955, Equipe 16, Créteil, F-94010, France.,Université Paris Est, Faculté de médecine, Créteil, F-94010, France.,Vaccine Research Institute (VRI), Créteil, F-94010, France
| | - Céline Manier
- INSERM, U955, Equipe 16, Créteil, F-94010, France.,Université Paris Est, Faculté de médecine, Créteil, F-94010, France.,Vaccine Research Institute (VRI), Créteil, F-94010, France
| | - Laura Richert
- Vaccine Research Institute (VRI), Créteil, F-94010, France.,Université Bordeaux, ISPED, Centre INSERM U1219, F-33000, Bordeaux, France.,CHU de Bordeaux, pôle de santé publique, F-33000, Bordeaux, France.,INRIA SISTM, F-33405, Talence, France
| | - Rodolphe Thiébaut
- Vaccine Research Institute (VRI), Créteil, F-94010, France.,Université Bordeaux, ISPED, Centre INSERM U1219, F-33000, Bordeaux, France.,CHU de Bordeaux, pôle de santé publique, F-33000, Bordeaux, France.,INRIA SISTM, F-33405, Talence, France
| | - Yves Levy
- INSERM, U955, Equipe 16, Créteil, F-94010, France.,Université Paris Est, Faculté de médecine, Créteil, F-94010, France.,Vaccine Research Institute (VRI), Créteil, F-94010, France.,AP-HP, Hôpital H. Mondor - A. Chenevier, Service d'Immunologie Clinique et Maladies Infectieuses, F-94010, Créteil, France
| | - Sophie Hue
- INSERM, U955, Equipe 16, Créteil, F-94010, France.,Université Paris Est, Faculté de médecine, Créteil, F-94010, France.,Vaccine Research Institute (VRI), Créteil, F-94010, France.,AP-HP, Hôpital H. Mondor - A. Chenevier, Service d'Immunologie Biologique, F-94010, Créteil, France
| | - Christine Lacabaratz
- INSERM, U955, Equipe 16, Créteil, F-94010, France. .,Université Paris Est, Faculté de médecine, Créteil, F-94010, France. .,Vaccine Research Institute (VRI), Créteil, F-94010, France.
| |
Collapse
|
43
|
Kamburova EG, Wisse BW, Joosten I, Allebes WA, van der Meer A, Hilbrands LB, Baas MC, Spierings E, Hack CE, van Reekum FE, van Zuilen AD, Verhaar M, Bots ML, Drop ACAD, Plaisier L, Seelen MAJ, Sanders JSF, Hepkema BG, Lambeck AJ, Bungener LB, Roozendaal C, Tilanus MGJ, Vanderlocht J, Voorter CE, Wieten L, van Duijnhoven EM, Gelens M, Christiaans MHL, van Ittersum FJ, Nurmohamed A, Lardy NM, Swelsen W, van der Pant KA, van der Weerd NC, Ten Berge IJM, Bemelman FJ, Hoitsma A, van der Boog PJM, de Fijter JW, Betjes MGH, Heidt S, Roelen DL, Claas FH, Otten HG. How can we reduce costs of solid-phase multiplex-bead assays used to determine anti-HLA antibodies? HLA 2016; 88:110-9. [PMID: 27534609 DOI: 10.1111/tan.12860] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 07/24/2016] [Indexed: 11/29/2022]
Abstract
Solid-phase multiplex-bead assays are widely used in transplantation to detect anti-human leukocyte antigen (HLA) antibodies. These assays enable high resolution detection of low levels of HLA antibodies. However, multiplex-bead assays are costly and yield variable measurements that limit the comparison of results between laboratories. In the context of a Dutch national Consortium study we aimed to determine the inter-assay and inter-machine variability of multiplex-bead assays, and we assessed how to reduce the assay reagents costs. Fifteen sera containing a variety of HLA antibodies were used yielding in total 7092 median fluorescence intensities (MFI) values. The inter-assay and inter-machine mean absolute relative differences (MARD) of the screening assay were 12% and 13%, respectively. The single antigen bead (SAB) inter-assay MARD was comparable, but showed a higher lot-to-lot variability. Reduction of screening assay reagents to 50% or 40% of manufacturers' recommendations resulted in MFI values comparable to 100% of the reagents, with an MARD of 12% or 14%, respectively. The MARD of the 50% and 40% SAB assay reagent reductions were 11% and 22%, respectively. From this study, we conclude that the reagents can be reliably reduced at least to 50% of manufacturers' recommendations with virtually no differences in HLA antibody assignments.
Collapse
Affiliation(s)
- E G Kamburova
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - B W Wisse
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - I Joosten
- Laboratory Medicine, Lab. Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W A Allebes
- Laboratory Medicine, Lab. Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A van der Meer
- Laboratory Medicine, Lab. Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - L B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M C Baas
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - E Spierings
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - C E Hack
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F E van Reekum
- Department of Nephrology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - A D van Zuilen
- Department of Nephrology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M Verhaar
- Department of Nephrology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M L Bots
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - A C A D Drop
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - L Plaisier
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M A J Seelen
- Department of Nephrology, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - J S F Sanders
- Department of Nephrology, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - B G Hepkema
- Department of Laboratory Medicine, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - A J Lambeck
- Department of Laboratory Medicine, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - L B Bungener
- Department of Laboratory Medicine, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - C Roozendaal
- Department of Laboratory Medicine, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - M G J Tilanus
- Department of Transplantation Immunology, Tissue Typing Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - J Vanderlocht
- Department of Transplantation Immunology, Tissue Typing Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - C E Voorter
- Department of Transplantation Immunology, Tissue Typing Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - L Wieten
- Department of Transplantation Immunology, Tissue Typing Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - E M van Duijnhoven
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - M Gelens
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - M H L Christiaans
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - F J van Ittersum
- Department of Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - A Nurmohamed
- Department of Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - N M Lardy
- Department of Immunogenetics, Sanquin, Amsterdam, The Netherlands
| | - W Swelsen
- Department of Immunogenetics, Sanquin, Amsterdam, The Netherlands
| | - K A van der Pant
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Centre, Amsterdam, The Netherlands
| | - N C van der Weerd
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Centre, Amsterdam, The Netherlands
| | - I J M Ten Berge
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Centre, Amsterdam, The Netherlands
| | - F J Bemelman
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Centre, Amsterdam, The Netherlands
| | - A Hoitsma
- Dutch Organ Transplant Registry (NOTR), Dutch Transplant Foundation (NTS), Leiden, The Netherlands
| | - P J M van der Boog
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - J W de Fijter
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - M G H Betjes
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.,Department of Nephrology, Erasmus MC, Rotterdam, The Netherlands
| | - S Heidt
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - D L Roelen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - F H Claas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - H G Otten
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
44
|
Singh M, Alabanza A, Gonzalez LE, Wang W, Reeves WB, Hahm JI. Ultratrace level determination and quantitative analysis of kidney injury biomarkers in patient samples attained by zinc oxide nanorods. NANOSCALE 2016; 8:4613-22. [PMID: 26846189 PMCID: PMC4760885 DOI: 10.1039/c5nr08706f] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Determining ultratrace amounts of protein biomarkers in patient samples in a straightforward and quantitative manner is extremely important for early disease diagnosis and treatment. Here, we successfully demonstrate the novel use of zinc oxide nanorods (ZnO NRs) in the ultrasensitive and quantitative detection of two acute kidney injury (AKI)-related protein biomarkers, tumor necrosis factor (TNF)-α and interleukin (IL)-8, directly from patient samples. We first validate the ZnO NRs-based IL-8 results via comparison with those obtained from using a conventional enzyme-linked immunosorbent method in samples from 38 individuals. We further assess the full detection capability of the ZnO NRs-based technique by quantifying TNF-α, whose levels in human urine are often below the detection limits of conventional methods. Using the ZnO NR platforms, we determine the TNF-α concentrations of all 46 patient samples tested, down to the fg per mL level. Subsequently, we screen for TNF-α levels in approximately 50 additional samples collected from different patient groups in order to demonstrate a potential use of the ZnO NRs-based assay in assessing cytokine levels useful for further clinical monitoring. Our research efforts demonstrate that ZnO NRs can be straightforwardly employed in the rapid, ultrasensitive, quantitative, and simultaneous detection of multiple AKI-related biomarkers directly in patient urine samples, providing an unparalleled detection capability beyond those of conventional analysis methods. Additional key advantages of the ZnO NRs-based approach include a fast detection speed, low-volume assay condition, multiplexing ability, and easy automation/integration capability to existing fluorescence instrumentation. Therefore, we anticipate that our ZnO NRs-based detection method will be highly beneficial for overcoming the frequent challenges in early biomarker development and treatment assessment, pertaining to the facile and ultrasensitive quantification of hard-to-trace biomolecules.
Collapse
Affiliation(s)
- Manpreet Singh
- Department of Chemistry, Georgetown University, 37th & O Sts. NW., Washington, DC 20057, USA.
| | - Anginelle Alabanza
- Department of Chemistry, Georgetown University, 37th & O Sts. NW., Washington, DC 20057, USA.
| | - Lorelis E Gonzalez
- Department of Chemistry, Georgetown University, 37th & O Sts. NW., Washington, DC 20057, USA.
| | - Weiwei Wang
- Division of Nephrology, The Penn State College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033, USA
| | - W Brian Reeves
- Division of Nephrology, The Penn State College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033, USA
| | - Jong-in Hahm
- Department of Chemistry, Georgetown University, 37th & O Sts. NW., Washington, DC 20057, USA.
| |
Collapse
|
45
|
Impaired Circulating Angiogenic Cells Mobilization and Metalloproteinase-9 Activity after Dynamic Exercise in Early Metabolic Syndrome. BIOMED RESEARCH INTERNATIONAL 2015; 2015:920356. [PMID: 26557715 PMCID: PMC4628701 DOI: 10.1155/2015/920356] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/22/2015] [Accepted: 05/25/2015] [Indexed: 11/18/2022]
Abstract
Increased levels of adhesion molecules or metalloproteinases (MMPs) may indicate endothelial dysfunction. Exercise mobilizes circulating angiogenic cells (CACs) from bone marrow in healthy subjects, improving vascular function. However, it is unclear whether this mechanism is preserved in the early stages of metabolic syndrome (early MetS). We aimed to evaluate the acute effects of exercise on adhesion molecules, angiogenic factors, MMPs, and CACs in early MetS. Fifteen subjects with early MetS and nine healthy controls underwent an exercise session and a nonexercise session, randomly. Adhesion molecules, angiogenic factors, CACs, and MMPs were evaluated before and after exercise or nonexercise sessions. At baseline, levels of sE-selectin, sICAM-1, and MMP-9 were higher in early MetS than in controls (P ≤ 0.03). After exercise, sE-selectin, sICAM-1, and MMP-9 levels were still higher in early MetS (P < 0.05). Subjects with early MetS presented less CACs (P = 0.02) and higher MMP-9 activity (P ≤ 0.04), while healthy controls presented higher MMP-2 activity after exercise. There was no difference between moments in nonexercise session (P > 0.05). In conclusion, subjects with early MetS already presented impaired endothelial function at rest along with a decrease in CACs and an increase in MMP-9 activity in response to exercise.
Collapse
|
46
|
Karanikola SN, Krücken J, Ramünke S, de Waal T, Höglund J, Charlier J, Weber C, Müller E, Kowalczyk SJ, Kaba J, von Samson-Himmelstjerna G, Demeler J. Development of a multiplex fluorescence immunological assay for the simultaneous detection of antibodies against Cooperia oncophora, Dictyocaulus viviparus and Fasciola hepatica in cattle. Parasit Vectors 2015; 8:335. [PMID: 26084663 PMCID: PMC4492007 DOI: 10.1186/s13071-015-0924-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 06/01/2015] [Indexed: 12/04/2022] Open
Abstract
Background A major constraint for the effective control and management of helminth parasites is the lack of rapid, high-throughput, routine diagnostic tests to assess the health status of individual animals and herds and to identify the parasite species responsible for these helminthoses. The capability of a multiplex platform for the simultaneous detection of three pasture associated parasite species was evaluated and compared to existing ELISAs. Methods The recombinant antigens 14.2 kDa ES protein for Cooperia oncophora, major sperm protein for Dictyocaulus viviparus and Cathepsin L1 for Fasciola hepatica were recombinantly expressed either in Escherichia coli or Pichia pastoris. Antigens were covalently coupled onto magnetic beads. Optimal concentrations for coupling were determined following the examination of serum samples collected from experimentally mono-infected animals, before and after their infection with the target species. Absence of cross-reactivity was further determined with sera from calves mono-infected with Haemonchus contortus, Ostertagia ostertagi and Trichostrongylus colubriformis. Examination of negative serum samples was characterised by low median fluorescence intensity (MFI). Results Establishment of the optimal serum dilution of 1:200 was achieved for all three bead sets. Receiver Operating Characteristic analyses were performed to obtain cut-off MFI values for each parasite separately. Sensitivity and specificity at the chosen cut-off values were close to, or 100 % for all bead sets. Examination of serum samples collected on different days post infection from different animals showed a high reproducibility of the assays. Serum samples were additionally examined with two already established ELISAs, an in-house ELISA using the recombinant MSP as an antigen and a DRG ELISA using Cathepsin L1 for liver fluke. The results between the assays were compared and kappa tests revealed an overall good agreement. Conclusions A versatile bead-based assay using fluorescence detection (xMAP® technology) was developed to simultaneously detect antibodies against C. oncophora, D. viviparus and F. hepatica in cattle serum samples. This platform provides rapid, high-throughput results and is highly sensitive and specific in comparison to existing serological as well as coproscopical diagnostic techniques.
Collapse
Affiliation(s)
- Sofia N Karanikola
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.
| | - Jürgen Krücken
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.
| | - Sabrina Ramünke
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.
| | - Theo de Waal
- UCD School of Agriculture, Food Science and Veterinary Medicine, University College Dublin, Dublin, Ireland.
| | - Johan Höglund
- Department of Biomedical Sciences and Veterinary Public Health, Section for Parasitology (SWEPAR), Swedish University of Agricultural Sciences, Uppsala, Sweden.
| | - Johannes Charlier
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | | | | | - Slawomir J Kowalczyk
- Laboratory of Veterinary Epidemiology and Economics, Faculty of Veterinary Medicine, Warsaw University of Life Science, Warsaw, Poland.
| | - Jaroslaw Kaba
- Laboratory of Veterinary Epidemiology and Economics, Faculty of Veterinary Medicine, Warsaw University of Life Science, Warsaw, Poland.
| | | | - Janina Demeler
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
47
|
Mani A, Ravindran R, Mannepalli S, Vang D, Luciw PA, Hogarth M, Khan IH, Krishnan VV. Data mining strategies to improve multiplex microbead immunoassay tolerance in a mouse model of infectious diseases. PLoS One 2015; 10:e0116262. [PMID: 25614982 PMCID: PMC4304816 DOI: 10.1371/journal.pone.0116262] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 12/04/2014] [Indexed: 11/25/2022] Open
Abstract
Multiplex methodologies, especially those with high-throughput capabilities generate large volumes of data. Accumulation of such data (e.g., genomics, proteomics, metabolomics etc.) is fast becoming more common and thus requires the development and implementation of effective data mining strategies designed for biological and clinical applications. Multiplex microbead immunoassay (MMIA), on xMAP or MagPix platform (Luminex), which is amenable to automation, offers a major advantage over conventional methods such as Western blot or ELISA, for increasing the efficiencies in serodiagnosis of infectious diseases. MMIA allows detection of antibodies and/or antigens efficiently for a wide range of infectious agents simultaneously in host blood samples, in one reaction vessel. In the process, MMIA generates large volumes of data. In this report we demonstrate the application of data mining tools on how the inherent large volume data can improve the assay tolerance (measured in terms of sensitivity and specificity) by analysis of experimental data accumulated over a span of two years. The combination of prior knowledge with machine learning tools provides an efficient approach to improve the diagnostic power of the assay in a continuous basis. Furthermore, this study provides an in-depth knowledge base to study pathological trends of infectious agents in mouse colonies on a multivariate scale. Data mining techniques using serodetection of infections in mice, developed in this study, can be used as a general model for more complex applications in epidemiology and clinical translational research.
Collapse
Affiliation(s)
- Akshay Mani
- Center for Comparative Medicine, University of California Davis, Davis, California, United States of America
| | - Resmi Ravindran
- Center for Comparative Medicine, University of California Davis, Davis, California, United States of America
| | - Soujanya Mannepalli
- Department of Chemistry, California State University, Fresno, California, United States of America
| | - Daniel Vang
- Center for Comparative Medicine, University of California Davis, Davis, California, United States of America
| | - Paul A Luciw
- Center for Comparative Medicine, University of California Davis, Davis, California, United States of America; Department of Pathology and Laboratory Medicine, University of California School of Medicine, Davis, California, United States of America
| | - Michael Hogarth
- Department of Pathology and Laboratory Medicine, University of California School of Medicine, Davis, California, United States of America
| | - Imran H Khan
- Center for Comparative Medicine, University of California Davis, Davis, California, United States of America; Department of Pathology and Laboratory Medicine, University of California School of Medicine, Davis, California, United States of America
| | - Viswanathan V Krishnan
- Center for Comparative Medicine, University of California Davis, Davis, California, United States of America; Department of Chemistry, California State University, Fresno, California, United States of America; Department of Pathology and Laboratory Medicine, University of California School of Medicine, Davis, California, United States of America
| |
Collapse
|
48
|
Stenken JA, Poschenrieder AJ. Bioanalytical chemistry of cytokines--a review. Anal Chim Acta 2015; 853:95-115. [PMID: 25467452 PMCID: PMC4717841 DOI: 10.1016/j.aca.2014.10.009] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/30/2014] [Accepted: 10/08/2014] [Indexed: 02/06/2023]
Abstract
Cytokines are bioactive proteins produced by many different cells of the immune system. Due to their role in different inflammatory disease states and maintaining homeostasis, there is enormous clinical interest in the quantitation of cytokines. The typical standard methods for quantitation of cytokines are immunoassay-based techniques including enzyme-linked immusorbent assays (ELISA) and bead-based immunoassays read by either standard or modified flow cytometers. A review of recent developments in analytical methods for measurements of cytokine proteins is provided. This review briefly covers cytokine biology and the analysis challenges associated with measurement of these biomarker proteins for understanding both health and disease. New techniques applied to immunoassay-based assays are presented along with the uses of aptamers, electrochemistry, mass spectrometry, optical resonator-based methods. Methods used for elucidating the release of cytokines from single cells as well as in vivo collection methods are described.
Collapse
Affiliation(s)
- Julie A Stenken
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA.
| | - Andreas J Poschenrieder
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; Pharmaceutical Radiochemistry, Technische Universität München, Walther-Meißner-Street 3, D-85748 Garching, Germany
| |
Collapse
|
49
|
Grati FR, Vialard F, Gross S. BACs-on-Beads™ (BoBs™) assay for the genetic evaluation of prenatal samples and products of conception. Methods Mol Biol 2015; 1227:259-78. [PMID: 25239751 DOI: 10.1007/978-1-4939-1652-8_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
BACs-on-Beads™ (BoBs™) is a new emerging technology, a modification of comparative genomic hybridization that can be used to detect DNA copy number gains and losses. Here, we describe the application of two different types of BoBs™ assays: (1) Prenatal BoBs (CE-IVD) to detect the most frequent syndromes associated with chromosome microdeletions, as well as the trisomy 13, 18 and 21, and (2) KaryoLite BoBs (RUO) which can detect aneuploidy in all chromosomes by quantifying proximal and terminal regions of each chromosomal arm. The interpretation of the results by BoBsoft™ software is also described. Although BoBs™ may not have the breadth and scope to replace chromosomal microarrays (array comparative genomic hybridization and single nucleotide polymorphism array) in the prenatal setting, particularly when a fetal anomaly has been detected, it is a well suited alternative for FISH or QF-PCR because BoBs™ is comparable, if not superior in terms of cost, turnaround time (TAT) and throughput and accuracy. BoBs™ also has the ability to detect significant fetal mosaicism (≥30% with Prenatal BoBs and ≥50% with KaryoLite BoBs). However, perhaps the greatest strength of this new technology is the fact that unlike FISH or QF-PCR, it has the ability to detect common microdeletion syndromes or additional aneuploidies, both of which may be easily missed despite excellent prenatal sonography. Thus, when BoBs™ is applied in the correct clinical setting and run and analyzed in appropriate laboratories this technique can improve and augment best practices with a personalization of prenatal care.
Collapse
MESH Headings
- Chromosome Deletion
- Chromosomes, Artificial, Bacterial/genetics
- Chromosomes, Artificial, Bacterial/metabolism
- Chromosomes, Human, Pair 13
- Chromosomes, Human, Pair 18
- Chromosomes, Human, Pair 21
- DNA Copy Number Variations
- Female
- Humans
- Male
- Mosaicism
- Precision Medicine
- Pregnancy
- Prenatal Diagnosis/instrumentation
- Prenatal Diagnosis/methods
- Software
- Trisomy/diagnosis
- Trisomy/genetics
Collapse
Affiliation(s)
- Francesca Romana Grati
- Research and Development, Cytogenetics and Molecular Biology, TOMA Advanced Biomedical Assays, S.p.A., Via Francesco Ferrer 25/27, 21052, Busto Arsizio, VA, Italy,
| | | | | |
Collapse
|
50
|
Zhang Y, Zhang W, Zhang Q, Li K, Liu W, Liu Y, Banks CE. Green electrochemical sensing platforms: utilizing hydroxyapatite derived from natural fish scales as a novel electrochemical material for the sensitive detection of kidney injury molecule 1 (KIM-1). Analyst 2014; 139:5362-6. [DOI: 10.1039/c4an00957f] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|