1
|
Kemme S, Canniff JD, Garth KM, Li S, Mysore K, Weinberg A, Feldman AG. Detection of viral RNA and DNA and immune response following administration of live attenuated measles and varicella vaccines in children with chronic liver disease. Am J Transplant 2025; 25:181-188. [PMID: 38901562 DOI: 10.1016/j.ajt.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
Infections preventable by live virus vaccines are surging in the setting of decreased herd immunity. Many children with chronic liver diseases (CLDs) are unimmunized and at increased risk for infection due to guidelines recommending against live vaccines within 4 weeks pretransplant. This prospective study of 21 children with CLD and 13 healthy controls defined the timing of measles virus and varicella-zoster virus (VZV) RNA- and DNA-emia following vaccination and compared immune responses to measles and varicella vaccines in both groups. Measles virus RNA and VZV DNA real-time PCR were measured weekly following vaccination; measles virus RNA was undetectable in all by 14 days postvaccination, but VZV DNA, which can be managed with antivirals, was detected in 1 child in the CLD group at 21 days and 1 control at 28 days postvaccination. Humoral or cell-mediated vaccine response was 100% to measles virus and 94% to VZV in the CLD group postvaccination, whereas it was 100% to both vaccines in controls. Our pilot study suggests that both live vaccines can be safely and effectively administered up to 14 days prior to transplantation in children with CLD. We anticipate this will improve vaccination rates and thus decrease rates of vaccine-preventable infections in vulnerable children with CLD.
Collapse
Affiliation(s)
- Sarah Kemme
- D. Brent Polk Division of Pediatric Gastroenterology, Hepatology, and Nutrition Monroe Carrell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee, USA.
| | - Jennifer D Canniff
- Department of Pediatrics, Medicine, and Pathology, Anschutz Medical Campus, University of Colorado, Aurora, Colorado, USA
| | - Krystle M Garth
- Department of Pediatrics, Medicine, and Pathology, Anschutz Medical Campus, University of Colorado, Aurora, Colorado, USA
| | - Shaobing Li
- Pediatric Infectious Diseases, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Krupa Mysore
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Adriana Weinberg
- Department of Pediatrics, Medicine, and Pathology, Anschutz Medical Campus, University of Colorado, Aurora, Colorado, USA
| | - Amy G Feldman
- Digestive Health Institute, Section of Pediatric Gastroenterology, Hepatology and Nutrition, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
2
|
Abdel-Haq H. Feasibility of Using a Type I IFN-Based Non-Animal Approach to Predict Vaccine Efficacy and Safety Profiles. Vaccines (Basel) 2024; 12:583. [PMID: 38932312 PMCID: PMC11209158 DOI: 10.3390/vaccines12060583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Animal-based tests are used for the control of vaccine quality. However, because highly purified and safe vaccines are now available, alternative approaches that can replace or reduce animal use for the assessment of vaccine outcomes must be established. In vitro tests for vaccine quality control exist and have already been implemented. However, these tests are specifically designed for some next-generation vaccines, and this makes them not readily available for testing other vaccines. Therefore, universal non-animal tests are still needed. Specific signatures of the innate immune response could represent a promising approach to predict the outcome of vaccines by non-animal methods. Type I interferons (IFNs) have multiple immunomodulatory activities, which are exerted through effectors called interferon stimulated genes (ISGs), and are one of the most important immune signatures that might provide potential candidate molecular biomarkers for this purpose. This paper will mainly examine if this idea might be feasible by analyzing all relevant published studies that have provided type I IFN-related biomarkers for evaluating the safety and efficacy profiles of vaccines using an advanced transcriptomic approach as an alternative to the animal methods. Results revealed that such an approach could potentially provide biomarkers predictive of vaccine outcomes after addressing some limitations.
Collapse
Affiliation(s)
- Hanin Abdel-Haq
- Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| |
Collapse
|
3
|
Browne DJ, Miller CM, Doolan DL. Technical pitfalls when collecting, cryopreserving, thawing, and stimulating human T-cells. Front Immunol 2024; 15:1382192. [PMID: 38812513 PMCID: PMC11133553 DOI: 10.3389/fimmu.2024.1382192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
The collection, cryopreservation, thawing, and culture of peripheral blood mononuclear cells (PBMCs) can profoundly influence T cell viability and immunogenicity. Gold-standard PBMC processing protocols have been developed by the Office of HIV/AIDS Network Coordination (HANC); however, these protocols are not universally observed. Herein, we have explored the current literature assessing how technical variation during PBMC processing can influence cellular viability and T cell immunogenicity, noting inconsistent findings between many of these studies. Amid the mounting concerns over scientific replicability, there is growing acknowledgement that improved methodological rigour and transparent reporting is required to facilitate independent reproducibility. This review highlights that in human T cell studies, this entails adopting stringent standardised operating procedures (SOPs) for PBMC processing. We specifically propose the use of HANC's Cross-Network PBMC Processing SOP, when collecting and cryopreserving PBMCs, and the HANC member network International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) PBMC Thawing SOP when thawing PBMCs. These stringent and detailed protocols include comprehensive reporting procedures to document unavoidable technical variations, such as delayed processing times. Additionally, we make further standardisation and reporting recommendations to minimise and document variability during this critical experimental period. This review provides a detailed overview of the challenges inherent to a procedure often considered routine, highlighting the importance of carefully considering each aspect of SOPs for PBMC collection, cryopreservation, thawing, and culture to ensure accurate interpretation and comparison between studies.
Collapse
Affiliation(s)
- Daniel J. Browne
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Catherine M. Miller
- College of Medicine and Dentistry, James Cook University, Cairns, QLD, Australia
| | - Denise L. Doolan
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
4
|
Sumner G, Keller S, Huleatt J, Staack RF, Wagner L, Azadeh M, Bandukwala A, Cao L, Du X, Salinas GF, Garofolo F, Harris S, Hopper S, Irwin C, Ji Q, Joseph J, King L, Kinhikar A, Lu Y, Luo R, Mabrouk O, Malvaux L, Marshall JC, McGuire K, Mikol V, Neely R, Qiu X, Saito Y, Salaun B, Scully I, Smeraglia J, Solstad T, Stoop J, Tang H, Teixeira P, Wang Y, Wright M, Mendez L, Beaver C, Eacret J, Au-Yeung A, Decman V, Dessy F, Eck S, Goihberg P, Alcaide EG, Gonneau C, Grugan K, Hedrick MN, Kar S, Sehra S, Stevens E, Stevens C, Sun Y, McCush F, Williams L, Fischer S, Wu B, Jordan G, Burns C, Cludts I, Coble K, Grimaldi C, Henderson N, Joyce A, Lotz G, Lu Y, Luo L, Neff F, Sperinde G, Stubenrauch KG, Wang Y, Ware M, Xu W. 2022 White Paper on Recent Issues in Bioanalysis: Enzyme Assay Validation, BAV for Primary End Points, Vaccine Functional Assays, Cytometry in Tissue, LBA in Rare Matrices, Complex NAb Assays, Spectral Cytometry, Endogenous Analytes, Extracellular Vesicles Part 2 - Recommendations on Biomarkers/CDx, Flow Cytometry, Ligand-Binding Assays Development & Validation; Emerging Technologies; Critical Reagents Deep Characterization. Bioanalysis 2023; 15:861-903. [PMID: 37584363 DOI: 10.4155/bio-2023-0151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023] Open
Abstract
The 16th Workshop on Recent Issues in Bioanalysis (16th WRIB) took place in Atlanta, GA, USA on September 26-30, 2022. Over 1000 professionals representing pharma/biotech companies, CROs, and multiple regulatory agencies convened to actively discuss the most current topics of interest in bioanalysis. The 16th WRIB included 3 Main Workshops and 7 Specialized Workshops that together spanned 1 week in order to allow exhaustive and thorough coverage of all major issues in bioanalysis, biomarkers, immunogenicity, gene therapy, cell therapy and vaccines. Moreover, in-depth workshops on ICH M10 BMV final guideline (focused on this guideline training, interpretation, adoption and transition); mass spectrometry innovation (focused on novel technologies, novel modalities, and novel challenges); and flow cytometry bioanalysis (rising of the 3rd most common/important technology in bioanalytical labs) were the special features of the 16th edition. As in previous years, WRIB continued to gather a wide diversity of international, industry opinion leaders and regulatory authority experts working on both small and large molecules as well as gene, cell therapies and vaccines to facilitate sharing and discussions focused on improving quality, increasing regulatory compliance, and achieving scientific excellence on bioanalytical issues. This 2022 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2022 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 2) covers the recommendations on LBA, Biomarkers/CDx and Cytometry. Part 1 (Mass Spectrometry and ICH M10) and Part 3 (Gene Therapy, Cell therapy, Vaccines and Biotherapeutics Immunogenicity) are published in volume 15 of Bioanalysis, issues 16 and 14 (2023), respectively.
Collapse
Affiliation(s)
| | | | | | - Roland F Staack
- Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | - Qin Ji
- AbbVie, North Chicago, IL, USA
| | | | | | | | - Yang Lu
- US FDA, Silver Spring, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Priscila Teixeira
- Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | - Yixin Wang
- Bristol-Myers Squibb, Lawrenceville, NJ, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Gregor Jordan
- Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | | | | | | | | | - Neil Henderson
- Integrated Bioanalysis, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Gregor Lotz
- Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | | | | | - Florian Neff
- Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | | | | | | | | | | |
Collapse
|
5
|
de Homdedeu M, Sanchez-Moral L, Violán C, Ràfols N, Ouchi D, Martín B, Peinado MA, Rodríguez-Cortés A, Arch-Sisquella M, Perez-Zsolt D, Muñoz-Basagoiti J, Izquierdo-Useros N, Salvador B, Matllo J, López-Serrano S, Segalés J, Vilaplana C, Torán-Monserrat P, Morros R, Monfà R, Sarrias MR, Cardona PJ. Mycobacterium manresensis induces trained immunity in vitro. iScience 2023; 26:106873. [PMID: 37250788 PMCID: PMC10182650 DOI: 10.1016/j.isci.2023.106873] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 04/24/2023] [Accepted: 05/09/2023] [Indexed: 05/31/2023] Open
Abstract
The COVID-19 pandemic posed a global health crisis, with new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants weakening vaccine-driven protection. Trained immunity could help tackle COVID-19 disease. Our objective was to analyze whether heat-killed Mycobacterium manresensis (hkMm), an environmental mycobacterium, induces trained immunity and confers protection against SARS-CoV-2 infection. To this end, THP-1 cells and primary monocytes were trained with hkMm. The increased secretion of tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, IL-1β, and IL-10, metabolic activity, and changes in epigenetic marks suggested hkMm-induced trained immunity in vitro. Healthcare workers at risk of SARS-CoV-2 infection were enrolled into the MANRECOVID19 clinical trial (NCT04452773) and were administered Nyaditum resae (NR, containing hkMm) or placebo. No significant differences in monocyte inflammatory responses or the incidence of SARS-CoV-2 infection were found between the groups, although NR modified the profile of circulating immune cell populations. Our results show that M. manresensis induces trained immunity in vitro but not in vivo when orally administered as NR daily for 14 days.
Collapse
Affiliation(s)
- Miquel de Homdedeu
- Innate Immunity Group, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- Experimental Tuberculosis Unit, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona (UAB), 08193 Bellaterra, Spain
| | - Lidia Sanchez-Moral
- Innate Immunity Group, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Concepció Violán
- Jordi Gol University Research Institute in Primary Care, 08007 Barcelona, Spain
- North Metropolitan Research Support Unit, Jordi Gol University Research Institute in Primary Care (IDIAP Jordi Gol), Mataró, Spain
- Northern Metropolitan Primary Care Management, Catalan Institute of Health, 08916 Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- Autonomous University of Barcelona (UAB), 08193 Bellaterra, Spain
| | - Neus Ràfols
- Innate Immunity Group, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Dan Ouchi
- Jordi Gol University Research Institute in Primary Care, 08007 Barcelona, Spain
| | - Berta Martín
- Program of Predictive and Personalized Medicine of Cancer, Germans Trias i Pujol Research Institute (PMPPC-IGTP), 08916 Badalona, Spain
| | - Miguel A Peinado
- Program of Predictive and Personalized Medicine of Cancer, Germans Trias i Pujol Research Institute (PMPPC-IGTP), 08916 Badalona, Spain
| | - Alhelí Rodríguez-Cortés
- Department of Pharmacology, Toxicology, and Therapeutics, Veterinary Faculty, Autonomous University of Barcelona, 08193 Bellaterra, Spain
| | - Marta Arch-Sisquella
- Experimental Tuberculosis Unit, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | | | | | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, 08916 Badalona, Spain
- Centre for Biomedical Research on Infectious Diseases (CIBERINFEC), Madrid, Spain
| | - Betlem Salvador
- Jordi Gol University Research Institute in Primary Care, 08007 Barcelona, Spain
| | - Joan Matllo
- Department of Prevention and Risks, Germans Trias i Pujol University Hospital, Northern Metropolitan Territorial Management, Catalan Health Institute, 08916 Badalona, Spain
| | - Sergi López-Serrano
- Joint IRTA-UAB Research Unit in Animal Health, Animal Health Research Center (CReSA), Autonomous University of Barcelona (UAB), 08193 Bellaterra, Spain
- Institute of Agrifood Research and Technology, Animal Health Program, Animal Health Research Center (CReSA), Autonomous University of Barcelona (UAB), 08193 Bellaterra, Spain
- OIE Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Spain
| | - Joaquim Segalés
- Joint IRTA-UAB Research Unit in Animal Health, Animal Health Research Center (CReSA), Autonomous University of Barcelona (UAB), 08193 Bellaterra, Spain
- OIE Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Spain
- Department of Animal Health and Anatomy, Faculty of Veterinary Medicine, Autonomous University of Barcelona (UAB), 08193 Bellaterra, Spain
| | - Cristina Vilaplana
- Experimental Tuberculosis Unit, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona (UAB), 08193 Bellaterra, Spain
- Centre for Biomedical Research on Respiratory Diseases (CIBERES), Madrid, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
- Direcció Clínica Territorial de Malalties Infeccioses i Salut Internacional de Gerència Territorial Metropolitana Nord, Barcelona, Spain
| | - Pere Torán-Monserrat
- Jordi Gol University Research Institute in Primary Care, 08007 Barcelona, Spain
- North Metropolitan Research Support Unit, Jordi Gol University Research Institute in Primary Care (IDIAP Jordi Gol), Mataró, Spain
- Northern Metropolitan Primary Care Management, Catalan Institute of Health, 08916 Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Rosa Morros
- Jordi Gol University Research Institute in Primary Care, 08007 Barcelona, Spain
| | - Ramon Monfà
- Jordi Gol University Research Institute in Primary Care, 08007 Barcelona, Spain
| | - Maria-Rosa Sarrias
- Innate Immunity Group, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- Centre for Biomedical Research on Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
| | - Pere-Joan Cardona
- Experimental Tuberculosis Unit, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona (UAB), 08193 Bellaterra, Spain
- Centre for Biomedical Research on Respiratory Diseases (CIBERES), Madrid, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| |
Collapse
|
6
|
Odak I, Sikora R, Riemann L, Bayir LM, Beck M, Drenker M, Xiao Y, Schneider J, Dammann E, Stadler M, Eder M, Ganser A, Förster R, Koenecke C, Schultze-Florey CR. Spectral flow cytometry cluster analysis of therapeutic donor lymphocyte infusions identifies T cell subsets associated with outcome in patients with AML relapse. Front Immunol 2022; 13:999163. [PMID: 36275657 PMCID: PMC9579313 DOI: 10.3389/fimmu.2022.999163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Identification of immune phenotypes linked to durable graft-versus-leukemia (GVL) response following donor lymphocyte infusions (DLI) is of high clinical relevance. In this prospective observational study of 13 AML relapse patients receiving therapeutic DLI, we longitudinally investigated changes in differentiation stages and exhaustion markers of T cell subsets using cluster analysis of 30-color spectral flow cytometry during 24 months follow-up. DLI cell products and patient samples after DLI were analyzed and correlated to the clinical outcome. Analysis of DLI cell products revealed heterogeneity in the proportions of naïve and antigen experienced T cells. Cell products containing lower levels of effector memory (eff/m) cells and higher amounts of naïve CD4+ and CD8+ T cells were associated with long-term remission. Furthermore, investigation of patient blood samples early after DLI showed that patients relapsing during the study period, had higher levels of CD4+ eff/m T cells and expressed a mosaic of surface molecules implying an exhausted functional state. Of note, this observation preceded the clinical diagnosis of relapse by five months. On the other hand, patients with continuous remission retained lower levels of exhausted CD4+ eff/m T cells more than four months post DLI. Moreover, lower frequencies of exhausted CD8+ eff/m T cells as well as higher amounts of CD4+temra CD45RO+ T cells were present in this group. These results imply the formation of functional long-term memory pool of T cells. Finally, unbiased sample analysis showed that DLI cell products with low levels of eff/m cells both in CD4+ and CD8+ T cell subpopulations associate with a lower relapse incidence. Additionally, competing risk analysis of patient samples taken early after DLI revealed that patients with high amounts of exhausted CD4+ eff/m T cells in their blood exhibited significantly higher rates of relapse. In conclusion, differentially activated T cell clusters, both in the DLI product and in patients post infusion, were associated with AML relapse after DLI. Our study suggests that differences in DLI cell product composition might influence GVL. In-depth monitoring of T cell dynamics post DLI might increase safety and efficacy of this immunotherapy, while further studies are needed to assess the functionality of T cells found in the DLI.
Collapse
Affiliation(s)
- Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- *Correspondence: Christian R. Schultze-Florey, ; Ivan Odak,
| | - Ruth Sikora
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Lennart Riemann
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Lâle M. Bayir
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Maleen Beck
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Melanie Drenker
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Yankai Xiao
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Jessica Schneider
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Elke Dammann
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Michael Stadler
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Matthias Eder
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christian Koenecke
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Christian R. Schultze-Florey
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- *Correspondence: Christian R. Schultze-Florey, ; Ivan Odak,
| |
Collapse
|
7
|
Rules of thumb to obtain, isolate, and preserve porcine peripheral blood mononuclear cells. Vet Immunol Immunopathol 2022; 251:110461. [PMID: 35870231 DOI: 10.1016/j.vetimm.2022.110461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022]
Abstract
One of the most used biospecimens in immunology are peripheral blood mononuclear cells (PBMC). PBMC are particularly useful when evaluating immunity through responses of circulating B- and T-cells, during an infection, or after a vaccination. While several reviews and research papers have been published aiming to point out critical steps when sampling, isolating, and cryopreserving human PBMC -or even analyzing any parameter before sampling that could impair the immune assays' outcomes-, there are almost no publications in swine research dealing with these topics. As it has been demonstrated, several factors, such as stress, circadian rhythmicity, or the anticoagulant used have serious negative impact, not only on the separation performance of PBMC, but also on the ulterior immune assays. The present review aims to discuss studies carried out in humans that could shed some light for swine research. When possible, publications in pigs are also discussed. The main goal of the review is to encourage swine researchers to standardize protocols to obtain, manage and preserve porcine PBMC, as well as to minimize, or at least to consider, the bias that some parameters might induce in their studies before, during and after isolating PBMC.
Collapse
|
8
|
Li Y, Mateu E, Díaz I. Impact of Cryopreservation on Viability, Phenotype, and Functionality of Porcine PBMC. Front Immunol 2021; 12:765667. [PMID: 34912338 PMCID: PMC8666977 DOI: 10.3389/fimmu.2021.765667] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
The use of frozen peripheral blood mononuclear cells (PBMC) is common in immunological studies. The impact of freezing PBMC has been assessed using human and mice cells, but little information is available regarding domestic animals. In the present study, the phenotype and functionality of frozen porcine PBMC were examined. In a preliminary experiment, three freezing media: fetal bovine serum plus 10% dimethyl sulfoxide, PSC cryopreservation kit, and Cryostor CS10, were compared regarding the preservation of cell viability and the response of PBMC to mitogens after thawing. After being stored one month in liquid nitrogen, cell viability was above 89% for all freezing media. The ELISPOT IFN-gamma (IFN-γ) results in response to PHA and of IgG ELISPOT in response to R848+IL-2 were similar to those obtained using fresh PBMC. In the second set of experiments, PBMC were obtained from five pigs vaccinated against Porcine reproductive and respiratory syndrome virus (PRRSV) and then frozen using Cryostor CS10. Recovered cells were phenotyped by flow cytometry using anti-CD3, CD4, CD8, and CD21 antibodies and were used to assess the PRRSV-specific responses in a proliferation experiment, an IFN-γ ELISPOT, and an IgG ELISPOT, and compared to the results obtained with fresh cells. The antigen-specific responses of frozen cells were significantly (p<0.05) impaired in the proliferation assay, particularly for CD4/CD8 double-positive T-cells and for CD21+ cells. Freezing resulted in decreased proliferation when Con A, but not PHA, was used. In ELISPOT, cryopreservation resulted in a decreased frequency of IFN-γ-secreting cells in response to PRRSV (p<0.05) but the response to PHA was not affected. No differences were observed in the IgG ELISPOT after polyclonal activation. Taken together, cryopreservation of porcine PBMC had a significant impact on the magnitude of recall antigen responses and therefore, it may affect the response of effector/memory cells but seems not to have a major impact on naïve T-cells. These results may help to the better use of frozen porcine PBMC, and to the interpretation of the results obtained from them.
Collapse
Affiliation(s)
- Yanli Li
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Enric Mateu
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,Centre de Recerca en Sanitat Animal, Institut de Recerca en Tecnologies Agroalimentàries (IRTA-CReSA), Bellaterra, Spain.,World Organisation for Animal Health (OIE) Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Bellaterra, Spain
| | - Ivan Díaz
- Centre de Recerca en Sanitat Animal, Institut de Recerca en Tecnologies Agroalimentàries (IRTA-CReSA), Bellaterra, Spain.,World Organisation for Animal Health (OIE) Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Bellaterra, Spain
| |
Collapse
|
9
|
Cryopreservation of NK and T Cells Without DMSO for Adoptive Cell-Based Immunotherapy. BioDrugs 2021; 35:529-545. [PMID: 34427899 DOI: 10.1007/s40259-021-00494-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 10/20/2022]
Abstract
Dimethylsufoxide (DMSO) being universally used as a cryoprotectant in clinical adoptive cell-therapy settings to treat hematological malignancies and solid tumors is a growing concern, largely due to its broad toxicities. Its use has been associated with significant clinical side effects-cardiovascular, neurological, gastrointestinal, and allergic-in patients receiving infusions of cell-therapy products. DMSO has also been associated with altered expression of natural killer (NK) and T-cell markers and their in vivo function, not to mention difficulties in scaling up DMSO-based cryoprotectants, which introduce manufacturing challenges for autologous and allogeneic cellular therapies, including chimeric antigen receptor (CAR)-T and CAR-NK cell therapies. Interest in developing alternatives to DMSO has resulted in the evaluation of a variety of sugars, proteins, polymers, amino acids, and other small molecules and osmolytes as well as modalities to efficiently enable cellular uptake of these cryoprotectants. However, the DMSO-free cryopreservation of NK and T cells remains difficult. They represent heterogeneous cell populations that are sensitive to freezing and thawing. As a result, clinical use of cryopreserved cell-therapy products has not moved past the use of DMSO. Here, we present the state of the art in the development and use of cryopreservation options that do not contain DMSO toward clinical solutions to enable the global deployment of safer adoptively transferred cell-based therapies.
Collapse
|
10
|
Gómez-Archila LG, Palomino-Schätzlein M, Zapata-Builes W, Galeano E. Development of an optimized method for processing peripheral blood mononuclear cells for 1H-nuclear magnetic resonance-based metabolomic profiling. PLoS One 2021; 16:e0247668. [PMID: 33630921 PMCID: PMC7906414 DOI: 10.1371/journal.pone.0247668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 02/11/2021] [Indexed: 01/04/2023] Open
Abstract
Human peripheral blood mononuclear cells (PBMCs) are part of the innate and adaptive immune system, and form a critical interface between both systems. Studying the metabolic profile of PBMC could provide valuable information about the response to pathogens, toxins or cancer, the detection of drug toxicity, in drug discovery and cell replacement therapy. The primary purpose of this study was to develop an improved processing method for PBMCs metabolomic profiling with nuclear magnetic resonance (NMR) spectroscopy. To this end, an experimental design was applied to develop an alternative method to process PBMCs at low concentrations. The design included the isolation of PBMCs from the whole blood of four different volunteers, of whom 27 cell samples were processed by two different techniques for quenching and extraction of metabolites: a traditional one using organic solvents and an alternative one employing a high-intensity ultrasound probe, the latter with a variation that includes the use of deproteinizing filters. Finally, all the samples were characterized by 1H-NMR and the metabolomic profiles were compared by the method. As a result, two new methods for PBMCs processing, called Ultrasound Method (UM) and Ultrasound and Ultrafiltration Method (UUM), are described and compared to the Folch Method (FM), which is the standard protocol for extracting metabolites from cell samples. We found that UM and UUM were superior to FM in terms of sensitivity, processing time, spectrum quality, amount of identifiable, quantifiable metabolites and reproducibility.
Collapse
Affiliation(s)
- León Gabriel Gómez-Archila
- Grupo de Investigación en Sustancias Bioactivas, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia (UdeA), Medellín, Colombia
| | | | - Wildeman Zapata-Builes
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia (UdeA), Medellín, Colombia
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medelín, Colombia
| | - Elkin Galeano
- Grupo de Investigación en Sustancias Bioactivas, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia (UdeA), Medellín, Colombia
| |
Collapse
|
11
|
Song J, Lv F, Dai RJ, Deng YL. Rapid identification and structural characterization of polyoxypregnane glycosides in Dregea sinensis by HPLC-MS n and HRMS. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2021; 23:9-19. [PMID: 32009448 DOI: 10.1080/10286020.2020.1715951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 01/08/2020] [Indexed: 06/10/2023]
Abstract
By HPLC-MSn and HRMS analyses, the structures of 52 polyoxypregnane glycosides were rapidly inferred from Dregea sinensis Hemsl on the basis of their sodium-cationized molecules [M + Na]+ and predominant diagnostic ions resulting from the saccharic chain on C3 and the neutral loss of substituent on C11 and C12. Compounds 1 and 7 significantly inhibited LPS-stimulated splenocyte proliferation in vitro.[Formula: see text].
Collapse
Affiliation(s)
- Juan Song
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Fang Lv
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Rong-Ji Dai
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yu-Lin Deng
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
12
|
Kazem N, Sulzgruber P, Thaler B, Baumgartner J, Koller L, Laufer G, Steinlechner B, Hohensinner P, Wojta J, Niessner A. CD8+CD28null T Lymphocytes are Associated with the Development of Atrial Fibrillation after Elective Cardiac Surgery. Thromb Haemost 2020; 120:1182-1187. [DOI: 10.1055/s-0040-1713096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Abstract
Background Postoperative atrial fibrillation (POAF) is assumed as a complex and multifactorial interaction of different pathogenic factors. Data suggests an inflammatory process as the main trigger of this specific type of atrial fibrillation. CD8+ T lymphocytes that lack the surface protein CD28 were found to be crucially involved in chronic inflammatory processes within the cardiovascular system. Of utmost interest, these so-called CD8+CD28null T cells are known to present with autoaggressive behavior and deleterious cytotoxic effects on human tissue.
Methods A total of 129 patients undergoing elective cardiac valve and/or coronary artery bypass graft surgery were enrolled. Fluorescence-activated cell sorting was performed to investigate lymphocyte subsets. Patients were stratified in two subgroups according to patients developing POAF (n = 60) and individuals free of POAF (n = 69).
Results Comparing patients developing POAF to individuals free of POAF, the fraction of CD8+ lymphocytes was significantly higher in individuals developing POAF (30.5% [POAF] vs. 25.7% [no-POAF]; p = 0.021). Interestingly, also the fraction of CD8+CD28null T lymphocytes was significantly higher in the POAF subgroup (66.7% [POAF] vs. 61.6% [non-POAF]; p = 0.043). Multivariate logistic regression proved that the fraction of CD8+CD28null cells is a strong and independent prognosticator for the development of POAF with an adjusted odds ratio per 1 standard deviation of 3.21 (95% confidence interval 1.01–10.18; p = 0.048).
Conclusion We found that cytotoxic CD8+CD28null T lymphocytes proved to be a strong and independent predictor for the development of POAF after elective cardiac surgery. Our results potentially indicate an autoimmune impact of this preexisting, highly cytotoxic T cell subset in the pathogenesis of POAF.
Collapse
Affiliation(s)
- Niema Kazem
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Patrick Sulzgruber
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Barbara Thaler
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Johanna Baumgartner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Lorenz Koller
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Günther Laufer
- Division of Cardiac Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Barbara Steinlechner
- Department of Anesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria
| | - Philipp Hohensinner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Johann Wojta
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Alexander Niessner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Martikainen MV, Roponen M. Cryopreservation affected the levels of immune responses of PBMCs and antigen-presenting cells. Toxicol In Vitro 2020; 67:104918. [PMID: 32565220 DOI: 10.1016/j.tiv.2020.104918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 10/24/2022]
Abstract
The effect of cryopreservation on antigen-presenting cells (APCs) is understudied. It is important to understand the effects of cryopreservation on these cells as they play a major role in immune responses, and they could be utilized in different clinical applications. In this study, we compared fresh and cryopreserved PBMCs in regards of their general immune responsiveness and, furthermore, the effect of cryopreservation on the circulating APCs among PBMCs. We stimulated fresh and cryopreserved PBMCs (N = 6) with LPS or Poly(I:C).Cytokine production of PBMCs and expression of functional markers CD80 and ILT4 on major types of APCs, dendritic cells (DCs) and monocytes, were analysed. We also analysed whether cryopreservation affects different subtypes of DCs (plasmacytoid and myeloid DCs) differently. Cryopreserved PBMCs produced less cytokines than fresh cells in response to stimulation, but the response profiles were comparable. Cryopreservation had also an effect on the relative proportions of APCs. Stimuli-induced responses were somewhat parallel but weaker than those observed in fresh cells. This study suggests that the use of cryopreserved cells is more suitable in studies that assess general responses to stimuli instead of measuring exact levels of reactions. Thus, the interpretation and comparison of the results of different studies should not be done without considering the differences in cryopreservation techniques and their effects on PBMCs and, more specifically, on APCs.
Collapse
Affiliation(s)
- Maria-Viola Martikainen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Marjut Roponen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
14
|
Chen H, Schürch CM, Noble K, Kim K, Krutzik PO, O'Donnell E, Vander Tuig J, Nolan GP, McIlwain DR. Functional comparison of PBMCs isolated by Cell Preparation Tubes (CPT) vs. Lymphoprep Tubes. BMC Immunol 2020; 21:15. [PMID: 32228458 PMCID: PMC7106580 DOI: 10.1186/s12865-020-00345-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/12/2020] [Indexed: 11/10/2022] Open
Abstract
Background Cryopreserved human peripheral blood mononuclear cells (PBMCs) are a commonly used sample type for a variety of immunological assays. Many factors can affect the quality of PBMCs, and careful consideration and validation of an appropriate PBMC isolation and cryopreservation method is important for well-designed clinical studies. A major point of divergence in PBMC isolation protocols is the collection of blood, either directly into vacutainers pre-filled with density gradient medium or the use of conical tubes containing a porous barrier to separate the density gradient medium from blood. To address potential differences in sample outcome, we isolated, cryopreserved, and compared PBMCs using parallel protocols differing only in the use of one of two common tube types for isolation. Methods Whole blood was processed in parallel using both Cell Preparation Tubes™ (CPT, BD Biosciences) and Lymphoprep™ Tubes (Axis-Shield) and assessed for yield and viability prior to cryopreservation. After thawing, samples were further examined by flow cytometry for cell yield, cell viability, frequency of 10 cell subsets, and capacity for stimulation-dependent CD4+ and CD8+ T cell intracellular cytokine production. Results No significant differences in cell recovery, viability, frequency of immune cell subsets, or T cell functionality between PBMC samples isolated using CPT or Lymphoprep tubes were identified. Conclusion CPT and Lymphoprep tubes are effective and comparable methods for PBMC isolation for immunological studies.
Collapse
Affiliation(s)
- Han Chen
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian M Schürch
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | | | - Garry P Nolan
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - David R McIlwain
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA. .,WCCT Global Inc., Cypress, CA, USA.
| |
Collapse
|
15
|
Diks AM, Bonroy C, Teodosio C, Groenland RJ, de Mooij B, de Maertelaere E, Neirynck J, Philippé J, Orfao A, van Dongen JJM, Berkowska MA. Impact of blood storage and sample handling on quality of high dimensional flow cytometric data in multicenter clinical research. J Immunol Methods 2019; 475:112616. [PMID: 31181213 DOI: 10.1016/j.jim.2019.06.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/21/2019] [Accepted: 06/04/2019] [Indexed: 01/20/2023]
Abstract
Obtaining reliable and reproducible high quality data in multicenter clinical research settings requires design of optimal standard operating procedures. While the need for standardization in sample processing and data analysis is well-recognized, the impact of sample handling in the pre-analytical phase remains underestimated. We evaluated the impact of sample storage time (≈transport time) and temperature, type of anticoagulant, and limited blood volume on reproducibility of flow cytometric studies. EDTA and Na-Heparin samples processed with the EuroFlow bulk lysis protocol, stained and stored at 4 °C showed fairly stable expression of cell surface markers and distribution of the major leukocyte populations for up to 72 h. Additional sample fixation (1% PFA, Fix & Perm) did not have any beneficial effects. Blood samples stored for <24 h at room temperature before processing and staining seemed suitable for reliable immunophenotyping, although losses in absolute cell numbers were observed. The major losses were observed in myeloid cells and monocytes, while lymphocytes seemed less affected. Expression of cell surface markers and population distribution were more stable in Na-Heparin blood than in EDTA blood. However, storage of Na-Heparin samples was associated with faster decrease in leukocyte counts over time. Whole blood fixation strategies (Cyto-Chex, TransFix) improved long-term population distribution, but were detrimental for expression of cellular markers. The main conclusions from this study on healthy donor blood samples were successfully confirmed in EDTA clinical (patient) blood samples with different time delays until processing. Finally, we recognized the need for adjustments in bulk lysis in case of insufficient blood volumes. Despite clear overall conclusions, individual markers and cell populations had different preferred conditions. Therefore, specific guidelines for sample handling should always be adjusted to the clinical application and the main target leukocyte population.
Collapse
Affiliation(s)
- A M Diks
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - C Bonroy
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium; Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - C Teodosio
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - R J Groenland
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - B de Mooij
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - E de Maertelaere
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - J Neirynck
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - J Philippé
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium; Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - A Orfao
- Cancer Research Centre (IBMCC, USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - J J M van Dongen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands.
| | - M A Berkowska
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| |
Collapse
|
16
|
Biospecimen Science of Blood for Peripheral Blood Mononuclear Cell (PBMC) Functional Applications. CURRENT PATHOBIOLOGY REPORTS 2019. [DOI: 10.1007/s40139-019-00192-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Sasaki E, Momose H, Hiradate Y, Ishii KJ, Mizukami T, Hamaguchi I. In vitro marker gene expression analyses in human peripheral blood mononuclear cells: A tool to assess safety of influenza vaccines in humans. J Immunotoxicol 2018. [PMID: 29521144 DOI: 10.1080/1547691x.2018.1447052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Vaccines are inoculated in healthy individuals from children to the elderly, and thus high levels of safety and consistency of vaccine quality in each lot must meet the required specifications by using preclinical and lot release testing. Because vaccines are inoculated into humans, recapitulation of biological reactions in humans should be considered for test methods. We have developed a new method to evaluate the safety of influenza vaccines using biomarker gene expression in mouse and rat models. Some biomarker genes are already known to be expressed in human lymphocytes, macrophages and dendritic cells; therefore, we considered some of these genes might be common biomarkers for human and mice to evaluate influenza vaccine safety. In this study, we used human peripheral blood mononuclear cells (PBMC) as a primary assessment tool to confirm the usefulness of potential marker genes in humans. Analysis of marker gene expression in PBMC revealed biomarker gene expressions were dose-relatedly increased in toxic reference influenza vaccine (RE)-stimulated PBMC. Although some marker genes showed increased expression in hemagglutinin split vaccine-stimulated PBMC, their expression levels were lower than that of RE in PBMC from two different donors. Many marker gene expressions correlated with chemokine production. Marker genes such as IRF7 were associated with other Type 1 interferon (IFN)-associated signals and were highly expressed in the CD304+ plasmacytoid dendritic cell (pDC) population. These results suggest PBMC and their marker genes may be useful for vaccine safety evaluation in humans.
Collapse
Affiliation(s)
- Eita Sasaki
- a Department of Safety Research on Blood and Biological Products , National Institute of Infectious Diseases , Tokyo , Japan
| | - Haruka Momose
- a Department of Safety Research on Blood and Biological Products , National Institute of Infectious Diseases , Tokyo , Japan
| | - Yuki Hiradate
- a Department of Safety Research on Blood and Biological Products , National Institute of Infectious Diseases , Tokyo , Japan
| | - Ken J Ishii
- b Laboratory of Adjuvant Innovation , National Institutes of Biomedical Innovation, Health and Nutrition , Osaka , Japan.,c Laboratory of Vaccine Science , WPI Immunology Frontier Research Center, Osaka University , Osaka , Japan
| | - Takuo Mizukami
- a Department of Safety Research on Blood and Biological Products , National Institute of Infectious Diseases , Tokyo , Japan
| | - Isao Hamaguchi
- a Department of Safety Research on Blood and Biological Products , National Institute of Infectious Diseases , Tokyo , Japan
| |
Collapse
|
18
|
Tompa A, Nilsson-Bowers A, Faresjö M. Subsets of CD4+, CD8+, and CD25hi Lymphocytes Are in General Not Influenced by Isolation and Long-Term Cryopreservation. THE JOURNAL OF IMMUNOLOGY 2018; 201:1799-1809. [DOI: 10.4049/jimmunol.1701409] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 07/12/2018] [Indexed: 12/21/2022]
|
19
|
Ding Y, Zhou L, Xia Y, Wang W, Wang Y, Li L, Qi Z, Zhong L, Sun J, Tang W, Liang F, Xiao H, Qin T, Luo Y, Zhao X, Shu Z, Ru Y, Dai R, Wang H, Wang Y, Zhang Y, Zhang S, Gao C, Du H, Zhang X, Chen Z, Wang X, Song H, Yang J, Zhao X. Reference values for peripheral blood lymphocyte subsets of healthy children in China. J Allergy Clin Immunol 2018; 142:970-973.e8. [PMID: 29746882 DOI: 10.1016/j.jaci.2018.04.022] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 04/23/2018] [Accepted: 04/30/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Yuan Ding
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lina Zhou
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Xia
- Department of Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Wei Wang
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ying Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Li Li
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhongxiang Qi
- Department of Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Linqing Zhong
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jinqiao Sun
- Department of Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Wenjing Tang
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Fangfang Liang
- Department of Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Haijuan Xiao
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tao Qin
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Luo
- Department of Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Xuezhen Zhao
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhou Shu
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Ru
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Rongxin Dai
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Wang
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yanping Wang
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yongjie Zhang
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Suqian Zhang
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Cong Gao
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hongqiang Du
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xuan Zhang
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhaolong Chen
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaochuan Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China.
| | - Hongmei Song
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Jun Yang
- Department of Immunology, Shenzhen Children's Hospital, Shenzhen, China.
| | - Xiaodong Zhao
- Ministry of Education, Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
20
|
Ten Brinke A, Marek-Trzonkowska N, Mansilla MJ, Turksma AW, Piekarska K, Iwaszkiewicz-Grześ D, Passerini L, Locafaro G, Puñet-Ortiz J, van Ham SM, Hernandez-Fuentes MP, Martínez-Cáceres EM, Gregori S. Monitoring T-Cell Responses in Translational Studies: Optimization of Dye-Based Proliferation Assay for Evaluation of Antigen-Specific Responses. Front Immunol 2017; 8:1870. [PMID: 29312346 PMCID: PMC5742609 DOI: 10.3389/fimmu.2017.01870] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/08/2017] [Indexed: 01/05/2023] Open
Abstract
Adoptive therapy with regulatory T cells or tolerance-inducing antigen (Ag)-presenting cells is innovative and promising therapeutic approach to control undesired and harmful activation of the immune system, as observed in autoimmune diseases, solid organ and bone marrow transplantation. One of the critical issues to elucidate the mechanisms responsible for success or failure of these therapies and define the specificity of the therapy is the evaluation of the Ag-specific T-cell responses. Several efforts have been made to develop suitable and reproducible assays. Here, we focus on dye-based proliferation assays. We highlight with practical examples the fundamental issues to take into consideration for implementation of an effective and sensitive dye-based proliferation assay to monitor Ag-specific responses in patients. The most critical points were used to design a road map to set up and analyze the optimal assay to assess Ag-specific T-cell responses in patients undergoing different treatments. This is the first step to optimize monitoring of tolerance induction, allowing comparison of outcomes of different clinical studies. The road map can also be applied to other therapeutic interventions, not limited to tolerance induction therapies, in which Ag-specific T-cell responses are relevant such as vaccination approaches and cancer immunotherapy.
Collapse
Affiliation(s)
- Anja Ten Brinke
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Natalia Marek-Trzonkowska
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Maria J. Mansilla
- Immunology Division, Department of Cellular Biology, Germans Trias i Pujol University Hospital and Research Institute, Physiology, and Immunology, Universitat Autònoma Barcelona, Barcelona, Spain
| | - Annelies W. Turksma
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Karolina Piekarska
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Laura Passerini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Grazia Locafaro
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Joan Puñet-Ortiz
- Immunology Division, Department of Cellular Biology, Germans Trias i Pujol University Hospital and Research Institute, Physiology, and Immunology, Universitat Autònoma Barcelona, Barcelona, Spain
| | - S. Marieke van Ham
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | | | - Eva M. Martínez-Cáceres
- Immunology Division, Department of Cellular Biology, Germans Trias i Pujol University Hospital and Research Institute, Physiology, and Immunology, Universitat Autònoma Barcelona, Barcelona, Spain
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
21
|
Blood lymphocyte subsets identify optimal responders to IFN-beta in MS. J Neurol 2017; 265:24-31. [PMID: 29027004 DOI: 10.1007/s00415-017-8625-6] [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: 07/11/2017] [Revised: 08/30/2017] [Accepted: 09/20/2017] [Indexed: 12/20/2022]
Abstract
Response to interferon-beta (IFN-beta) treatment is heterogeneous in multiple sclerosis (MS). We aimed to search for biomarkers predicting no evidence of disease activity (NEDA) status upon IFN-beta treatment in MS. 119 patients with relapsing-remitting MS (RRMS) initiating IFN-beta treatment were included in the study, and followed prospectively for 2 years. Neutralizing antibodies (NAb) were explored in serum samples obtained after 6 and 12 months of IFN-beta treatment. Soluble cytokines and blood lymphocytes were studied in basal samples by ELISA and flow cytometry, respectively. 9% of patients developed NAb. These antibodies were more frequent in patients receiving IFN-beta 1b than in those treated subcutaneous (p = 0.008) or intramuscular (p < 0.0001) IFN-beta 1a. No patient showing NAb remained NEDA during follow-up. Basal immunological variables are also associated with patient response. Percentages below 3% of CD19 + CD5 + cells (AUC 0.74, CI 0.63-0.84; OR 10.68, CI 3.55-32.15, p < 0.0001; Likelihood ratio 4.28) or above 2.6% of CD8 + perforin + T cells (AUC 0.79, CI 0.63-0.96; OR 6.11, CI 2.0-18.6, p = 0.0009; Likelihood ratio 5.47) increased the probability of achieving NEDA status during treatment. Basal blood immune cell subsets contribute to identify MS patients with a high probability of showing an optimal response to IFN-beta.
Collapse
|
22
|
Selective Expression of CCR10 and CXCR3 by Circulating Human Herpes Simplex Virus-Specific CD8 T Cells. J Virol 2017; 91:JVI.00810-17. [PMID: 28701399 DOI: 10.1128/jvi.00810-17] [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: 05/15/2017] [Accepted: 07/03/2017] [Indexed: 01/13/2023] Open
Abstract
Herpes simplex virus (HSV) infection is restricted to epithelial cells and neurons and is controlled by CD8 T cells. These cells both traffic to epithelial sites of recurrent lytic infection and to ganglia and persist at the dermal-epidermal junction for up to 12 weeks after lesion resolution. We previously showed that cutaneous lymphocyte-associated antigen (CLA), a functional E-selectin ligand (ESL), is selectively expressed on circulating HSV-2-specific CD8 T cells. CLA/ESL mediates adhesion of T cells to inflamed vascular endothelium. Later stages in T-cell homing involve chemokines (Ch) and lymphocyte chemokine receptors (ChR) for vascular wall arrest and diapedesis. Several candidate ChR have been implicated in skin homing. We measured cell surface ChR on HSV-specific human peripheral blood CD8 T cells and extended our studies to HSV-1. We observed preferential cell surface expression of CCR10 and CXCR3 by HSV-specific CD8 T cells compared to CD8 T cells specific for control viruses, Epstein-Barr virus (EBV) and cytomegalovirus (CMV), and compared to bulk memory CD8 T cells. CXCR3 ligand mRNA levels were selectively increased in skin biopsy specimens from persons with recurrent HSV-2, while the mRNA levels of the CCR10 ligand CCL27 were equivalent in lesion and control skin. Our data are consistent with a model in which CCL27 drives baseline recruitment of HSV-specific CD8 T cells expressing CCR10, while interferon-responsive CXCR3 ligands recruit additional cells in response to virus-driven inflammation.IMPORTANCE HSV-2 causes very localized recurrent infections in the skin and genital mucosa. Virus-specific CD8 T cells home to the site of recurrent infection and participate in viral clearance. The exit of T cells from the blood involves the use of chemokine receptors on the T-cell surface and chemokines that are present in infected tissue. In this study, circulating HSV-2-specific CD8 T cells were identified using specific fluorescent tetramer reagents, and their expression of several candidate skin-homing-associated chemokine receptors was measured using flow cytometry. We found that two chemokine receptors, CXCR3 and CCR10, are upregulated on HSV-specific CD8 T cells in blood. The chemokines corresponding to these receptors are also expressed in infected tissues. Vaccine strategies to prime CD8 T cells to home to HSV lesions should elicit these chemokine receptors if possible to increase the homing of vaccine-primed cells to sites of infection.
Collapse
|
23
|
Consuegra I, Rodríguez-Aierbe C, Santiuste I, Bosch A, Martínez-Marín R, Fortuto MA, Díaz T, Martí S, Muñoz-Fernández MÁ. Isolation Methods of Peripheral Blood Mononuclear Cells in Spanish Biobanks: An Overview. Biopreserv Biobank 2017; 15:305-309. [DOI: 10.1089/bio.2016.0105] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Irene Consuegra
- Spanish HIV HGM BioBank, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | | | - Inés Santiuste
- Instituto de Investigación Marqués de Valdecilla, Biobanco Valdecilla, Santander, Spain
| | - Anna Bosch
- Biobanco Hospital Clínic–IDIBAPS, Institut d'Investigacions Biomèdiques August Pi i Sunyer, IDIBAPS, Barcelona, Spain
| | | | - M Antonia Fortuto
- Clínica Universidad de Navarra, Biobanco de la Universidad de Navarra, Pamplona, Spain
| | - Tatiana Díaz
- Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | - Salvador Martí
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER BIOBANK, Valencia, Spain
| | - M Ángeles Muñoz-Fernández
- Spanish HIV HGM BioBank, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
24
|
Brief Report: L-Selectin (CD62L) Is Downregulated on CD4+ and CD8+ T Lymphocytes of HIV-1-Infected Individuals Naive for ART. J Acquir Immune Defic Syndr 2017; 72:492-7. [PMID: 27003497 DOI: 10.1097/qai.0000000000000999] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The expression of L-selectin (CD62L) in HIV-1 infection has not been extensively investigated. Here, we measured CD62L expression on T-cell subsets of HIV-1-infected individuals naive for antiretroviral therapy (ART-naive) or receiving therapy (ART), and seronegative control subjects (HIV-neg). We found reduced frequencies of CD62L(+) cells among CD4(+) and CD8(+) T cells from ART-naive as compared with ART and HIV-neg groups, particularly within naive and central memory subsets. CD62L expression on T cells inversely correlated with viral load and rapidly increased after ART initiation. Plasma sCD62L levels did not correlate with CD62L expression, being higher in all HIV-1-infected individuals as compared with HIV-neg subjects. Finally, CD62L downregulation was found associated with the expression of the CD38 activation marker in CD8(+) T cells, but not in CD4(+) T cells. We suggest that CD62L downregulation due to unconstrained HIV-1 replication may have important consequences for T-cell circulation and function and for disease progression.
Collapse
|
25
|
Ben-Ami E, Barysauskas CM, Solomon S, Tahlil K, Malley R, Hohos M, Polson K, Loucks M, Severgnini M, Patel T, Cunningham A, Rodig SJ, Hodi FS, Morgan JA, Merriam P, Wagner AJ, Shapiro GI, George S. Immunotherapy with single agent nivolumab for advanced leiomyosarcoma of the uterus: Results of a phase 2 study. Cancer 2017; 123:3285-3290. [PMID: 28440953 DOI: 10.1002/cncr.30738] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/10/2017] [Accepted: 03/22/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND Immunotherapy has changed the therapeutic landscape in oncology. Advanced uterine leiomyosarcoma (ULMS) remains an incurable disease in most cases, and despite new drug approvals, improvements in overall survival have been modest at best. The goal of this study was to evaluate programmed-death 1 (PD-1) inhibition with nivolumab in this patient population. METHODS This single-center phase 2 trial completed enrollment between May and October 2015. Patients received 3 mg/kg of intravenous nivolumab on day 1 of each 2-week cycle until disease progression or unacceptable toxicity. The primary endpoint was objective response rate. We assessed PD-1, PD-ligand 1 (PD-L1), and PD-L2 expression in archival tumor samples and variations in immune-phenotyping of circulating immune cells during treatment. RESULTS Twelve patients were enrolled in the first stage of the 2-stage design. A median of 5 (range, 2-6) 2-week cycles of nivolumab were administered. Of the 12 patients, none responded to treatment. The overall median progression-free survival was 1.8 months (95% confidence interval, 0.8-unknown). The study did not open the second stage due to lack of benefit as defined by the statistical plan. Archival samples were available for 83% of patients. PD-1 (>3% of cells), PD-L1, and PD-L2 (>5% and >10% of tumor cells, respectively) expression were observed in 20%, 20%, and 90% of samples, respectively. No significant differences were observed between pre- and posttreatment cell phenotypes. CONCLUSION Single-agent nivolumab did not demonstrate a benefit in this cohort of previously treated advanced ULMS patients. Further biomarker-driven approaches and studies evaluating combined immune checkpoint-modulators should be considered. Cancer 2017;123:3285-90. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Eytan Ben-Ami
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Constance M Barysauskas
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sarah Solomon
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kadija Tahlil
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Rita Malley
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Melissa Hohos
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kathleen Polson
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Margaret Loucks
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mariano Severgnini
- Center for Immuno-oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tara Patel
- Center for Immuno-oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amy Cunningham
- Center for Immuno-oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Scott J Rodig
- Center for Immuno-oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - F Stephen Hodi
- Center for Immuno-oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jeffrey A Morgan
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Priscilla Merriam
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Andrew J Wagner
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Suzanne George
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
26
|
Calleros-Basilio L, Cortés MA, García-Jerez A, Luengo-Rodríguez A, Orozco-Agudo A, Valdivielso JM, Rodríguez-Puyol D, Rodríguez-Puyol M. Quality Assurance of Samples and Processes in the Spanish Renal Research Network (REDinREN) Biobank. Biopreserv Biobank 2016; 14:499-510. [PMID: 27541936 DOI: 10.1089/bio.2015.0095] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Biobanks are useful platforms to build bridges between basic, translational, and clinical research and clinical care. They are repositories of high-quality human biological samples ideal for evaluating their histological characteristics and also their genome, transcriptome, and proteome. The Spanish Renal Research Network Biobank contains more than 76,500 well-preserved frozen samples of a wide variety of kidney diseases, collected from 5450 patients seen by over 70 nephrology services throughout the Spanish territory. OBJECTIVE To determine and to report the results of the quality control of samples and processes conducted in our biobank, implemented in accordance with the requirements of the ISO 9001:2008 international standard. STUDY DESIGN Two types of quality controls were performed: (1) systematic, that is, measurement of viable peripheral blood mononuclear cells (PBMCs) obtained and purity of nucleic acids and (2) ad-hoc, that is, viability of thawed PBMC, DNA extraction process reproducibility, and the integrity and functionality of nucleic acids, implemented on a routine basis. METHODS AND RESULTS PBMC isolation by Ficoll yielded reproducible results and its cryopreserved viability was >90%. Acceptable A260/A280 ratios were obtained for the vast majority of the DNA (n = 2328) and RNA (n = 78) samples analyzed. DNA integrity was demonstrated by agarose gels and by β-globulin gene polymerase chain reaction (PCR) amplification of 1327 and 989 bp fragments. DNA of acceptable quality had at least three bands of β-globulin amplified obtained (n = 26/30). RNA integrity number (RIN) determinations obtained RIN numbers ≥7 (n = 87/96). The amplifiability of nucleic acids was confirmed by qPCR and RT-qPCR of β-actin and GAPDH genes. Long storage or delayed processing time did not affect the quality of the samples analyzed. The processes of DNA extraction also yielded reproducible results. CONCLUSIONS These results clearly indicate that our PBMC, DNA, and RNA stored samples meet the required quality standards to be used for biomedical research, ensuring their long-term preservation.
Collapse
Affiliation(s)
- Laura Calleros-Basilio
- 1 Physiology Unit, Department of Systems Biology, Medicine School, Alcala University , Madrid, Spain .,2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain
| | - María Alicia Cortés
- 3 CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Medicine School, Universidad Nacional del Nordeste , Corrientes, Argentina
| | - Andrea García-Jerez
- 1 Physiology Unit, Department of Systems Biology, Medicine School, Alcala University , Madrid, Spain .,2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain
| | - Alicia Luengo-Rodríguez
- 1 Physiology Unit, Department of Systems Biology, Medicine School, Alcala University , Madrid, Spain .,2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain
| | - Ana Orozco-Agudo
- 1 Physiology Unit, Department of Systems Biology, Medicine School, Alcala University , Madrid, Spain .,2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain
| | - José Manuel Valdivielso
- 2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain .,4 Department of Experimental Nephrology, Institut de Recerca Biomédica de Lleida, Universitat de Lleida , Lleida, Spain
| | - Diego Rodríguez-Puyol
- 2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain .,5 Nephrology Section and Research Unit, Hospital Universitario Príncipe de Asturias , Alcalá de Henares, Madrid, Spain
| | - Manuel Rodríguez-Puyol
- 1 Physiology Unit, Department of Systems Biology, Medicine School, Alcala University , Madrid, Spain .,2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain
| |
Collapse
|
27
|
Virtual Global Transplant Laboratory Standard Operating Procedures for Blood Collection, PBMC Isolation, and Storage. Transplant Direct 2016; 2:e101. [PMID: 27795993 PMCID: PMC5068195 DOI: 10.1097/txd.0000000000000613] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 05/27/2016] [Indexed: 11/26/2022] Open
Abstract
Research on human immune responses frequently involves the use of peripheral blood mononuclear cells (PBMC) immediately, or at significantly delayed timepoints, after collection. This requires PBMC isolation from whole blood and cryopreservation for some applications. It is important to standardize protocols for blood collection, PBMC isolation, cryopreservation, and thawing that maximize survival and functionality of PBMC at the time of analysis. This resource includes detailed protocols describing blood collection tubes, isolation of PBMC using a density gradient, cryopreservation of PBMC, and thawing of cells as well as preparation for functional assays. For each protocol, we include important considerations, such as timing, storage temperatures, and freezing rate. In addition, we provide alternatives so that researchers can make informed decisions in determining the optimal protocol for their application.
Collapse
|
28
|
Kääriö H, Nieminen JK, Karvonen AM, Huttunen K, Schröder PC, Vaarala O, von Mutius E, Pfefferle PI, Schaub B, Pekkanen J, Hirvonen MR, Roponen M. Circulating Dendritic Cells, Farm Exposure and Asthma at Early Age. Scand J Immunol 2016; 83:18-25. [PMID: 26368653 DOI: 10.1111/sji.12389] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 09/09/2015] [Indexed: 12/23/2022]
Abstract
Farm environment has been shown to protect from childhood asthma. Underlying immunological mechanisms are not clear yet, including the role of dendritic cells (DCs). The aim was to explore whether asthma and farm exposures are associated with the proportions and functional properties of DCs from 4.5-year-old children in a subgroup of the Finnish PASTURE birth cohort study. Myeloid DCs (mDCs), plasmacytoid DCs (pDCs) and CD86 expression on mDCs ex vivo (n = 100) identified from peripheral blood mononuclear cells (PBMCs) were analysed using flow cytometry. MDCs and production of interleukin (IL)-6 and tumour necrosis factor alpha (TNF-α) by mDCs were analysed after 5 h in vitro stimulation with lipopolysaccharide (LPS) (n = 88). Prenatal and current farm exposures (farming, stables, hay barn and farm milk) were assessed from questionnaires. Asthma at age 6 years was defined as a doctor's diagnosis and symptoms; atopic sensitization was defined by antigen-specific IgE measurements. Asthma was positively associated with CD86 expression on mDCs ex vivo [adjusted odds ratio (aOR) 4.83, 95% confidence interval (CI) 1.51-15.4] and inversely with IL-6 production in mDCs after in vitro stimulation with LPS (aOR 0.19, 95% CI 0.04-0.82). In vitro stimulation with LPS resulted in lower percentage of mDCs in the farm PBMC cultures as compared to non-farm PBMC cultures. Our results suggest an association between childhood asthma and functional properties of DCs. Farm exposure may have immunomodulatory effects by decreasing mDC proportions.
Collapse
Affiliation(s)
- H Kääriö
- Department of Environmental Science, University of Eastern Finland, Kuopio, Finland
| | - J K Nieminen
- Immune Response Unit, Department of Vaccination and Immune Protection, National Institute for Health and Welfare, Helsinki, Finland
| | - A M Karvonen
- Department of Health Protection, National Institute for Health and Welfare, Kuopio, Finland
| | - K Huttunen
- Department of Environmental Science, University of Eastern Finland, Kuopio, Finland
| | - P C Schröder
- Department of Pulmonary and Allergy, LMU Munich, University Children's Hospital, Munich, Germany
| | - O Vaarala
- Institute of Clinical Medicine, University of Helsinki, Helsinki, Finland
| | - E von Mutius
- Department of Pulmonary and Allergy, LMU Munich, University Children's Hospital, Munich, Germany
| | - P I Pfefferle
- Institute of Laboratory Medicine, Pathobiochemistry and Molecular Diagnostics, Philipps University, Marburg, Germany
| | - B Schaub
- Department of Pulmonary and Allergy, LMU Munich, University Children's Hospital, Munich, Germany
| | - J Pekkanen
- Department of Health Protection, National Institute for Health and Welfare, Kuopio, Finland.,Department of Public Health, University of Helsinki, Helsinki, Finland
| | - M-R Hirvonen
- Department of Environmental Science, University of Eastern Finland, Kuopio, Finland.,Department of Health Protection, National Institute for Health and Welfare, Kuopio, Finland
| | - M Roponen
- Department of Environmental Science, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
29
|
Comparative phenotypical analysis of B cells in fresh and cryopreserved mononuclear cells from blood and tissue of rhesus macaques. J Immunol Methods 2016; 433:59-68. [PMID: 26970138 DOI: 10.1016/j.jim.2016.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 12/13/2022]
Abstract
Cryopreservation of peripheral blood mononuclear cells (PBMCs) is common for large clinical trials for which phenotypical characterization of lymphocytes is retrospectively performed in specialized core laboratories. It is therefore essential to assess the comparability between fresh and frozen samples. No side-by-side comparison of B and plasma cells of rhesus macaques (RM), which serve as useful models for several human diseases has been conducted until now. Hence, we performed an extensive comparative analysis between fresh and thawed mononuclear cells (MNCs) from blood and various tissues of healthy RM to analyze for the possible effects of cryopreservation on phenotype and functionality. Our data demonstrate that -80°C cryopreservation induces profound changes compared to fresh ex vivo-derived material. Percentages of B cells were stable in PBMCs, but were increased in all organs analyzed. The expression of CD27, a marker for differentiation between naïve and memory B cells, was massively reduced in PBMCs and MNC from organs with the most severe changes observed in cells from bone marrow (BM). Additionally, similar low percentages of CD27(+) memory B cells were detected in PBMCs and BM samples stored in liquid nitrogen. Therefore, cryopreservation is not suitable for the phenotypical and functional characterization of B cells. Further optimization of cryoconservation protocols monitoring the surface expression of CD27, which was identified as a marker for the quality of cryopreserved material of RM, will be essential.
Collapse
|
30
|
Wang L, Hückelhoven A, Hong J, Jin N, Mani J, Chen BA, Schmitt M, Schmitt A. Standardization of cryopreserved peripheral blood mononuclear cells through a resting process for clinical immunomonitoring-Development of an algorithm. Cytometry A 2016; 89:246-58. [DOI: 10.1002/cyto.a.22813] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 10/18/2015] [Accepted: 12/11/2015] [Indexed: 01/05/2023]
Affiliation(s)
- Lei Wang
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Angela Hückelhoven
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Jian Hong
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Nan Jin
- Department of Hematology; Zhongda Hospital, Southeast University; Nanjing China
| | - Jiju Mani
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Bao-an Chen
- Department of Hematology; Zhongda Hospital, Southeast University; Nanjing China
| | - Michael Schmitt
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Anita Schmitt
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| |
Collapse
|
31
|
Eschbaumer M, Stenfeldt C, Pacheco JM, Rekant SI, Arzt J. Effect of storage conditions on subpopulations of peripheral blood T lymphocytes isolated from naïve cattle and cattle infected with foot-and-mouth disease virus. Vet Clin Pathol 2016; 45:110-5. [PMID: 26802284 DOI: 10.1111/vcp.12327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Immunophenotyping of blood lymphocytes by flow cytometry is important in infectious disease research. In animal experiments and other longitudinal studies, the processing, prompt staining, and analysis of fresh samples is a logistical challenge and daily assay variation can confound data interpretation. OBJECTIVE This study examined the feasibility of cryopreservation and deferred analysis of bovine peripheral blood T lymphocytes from normal or infected animals. METHODS Peripheral blood mononuclear cells were collected from 4 naïve Holstein steers and 4 steers infected with foot-and-mouth-disease virus serotype Asia1. Identical aliquots were labeled and analyzed immediately, labeled for deferred analysis, or stored at -70°C or over liquid nitrogen for up to 3 weeks before labeling. RESULTS Freezing of unlabeled cells induced statistically significant changes in phenotypic recognition. In infected animals, the γδ T-cell population increased by 28% and CD8(+) αβT cells by 32%, while total CD3(+) cells decreased by 16%, and CD4(+) αβT cells decreased by 12%. Subsequent storage of frozen cells for the duration of the study, however, had no significant effect. There was less than 20% relative change in subpopulation sizes, and storage at -70°C or over liquid nitrogen was equivalent. CONCLUSIONS Depending on the objectives and practical limitations of a study, deferred labeling of peripheral blood lymphocytes can be a viable option. Although frozen storage of lymphocytes can introduce some artifactual distortion of relative cell populations, frozen cells can be maintained in storage until all samples in a longitudinal study can be analyzed in batch under standardized conditions and without introducing further bias.
Collapse
Affiliation(s)
- Michael Eschbaumer
- Plum Island Animal Disease Center (PIADC), Foreign Animal Disease Research Unit (FADRU), Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Greenport, NY, USA.,Oak Ridge Institute for Science and Education, PIADC Research Participation Program, Oak Ridge, TN, USA
| | - Carolina Stenfeldt
- Plum Island Animal Disease Center (PIADC), Foreign Animal Disease Research Unit (FADRU), Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Greenport, NY, USA.,Oak Ridge Institute for Science and Education, PIADC Research Participation Program, Oak Ridge, TN, USA
| | - Juan M Pacheco
- Plum Island Animal Disease Center (PIADC), Foreign Animal Disease Research Unit (FADRU), Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Greenport, NY, USA
| | - Steven I Rekant
- Plum Island Animal Disease Center (PIADC), Foreign Animal Disease Research Unit (FADRU), Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Greenport, NY, USA.,Oak Ridge Institute for Science and Education, PIADC Research Participation Program, Oak Ridge, TN, USA
| | - Jonathan Arzt
- Plum Island Animal Disease Center (PIADC), Foreign Animal Disease Research Unit (FADRU), Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Greenport, NY, USA
| |
Collapse
|
32
|
Grützner E, Stirner R, Arenz L, Athanasoulia AP, Schrödl K, Berking C, Bogner JR, Draenert R. Kinetics of human myeloid-derived suppressor cells after blood draw. J Transl Med 2016; 14:2. [PMID: 26733325 PMCID: PMC4702395 DOI: 10.1186/s12967-015-0755-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/14/2015] [Indexed: 12/18/2022] Open
Abstract
Background Human myeloid-derived suppressor cells (MDSC) have been described as a group of immature myeloid cells which exert immunosuppressive action by inhibiting function of T lymphocytes. While there is a huge scientific interest to study these cells in multiple human diseases, the methodological approach varies substantially between published studies. This is problematic as human MDSC seem to be a sensible cell type concerning not only cryopreservation but also time point after blood draw. To date data on delayed blood processing influencing cell numbers and phenotype is missing. We therefore evaluated the kinetics of granulocytic MDSC (gMDSC) and monocytic MDSC (mMDSC) frequencies after blood draw in order to determine the best time point for analysis of this recently defined cell type. Methods In this study, we isolated peripheral blood mononuclear cells (PBMC) of patients with HIV infection or solid tumors directly after blood draw. We then analyzed the frequencies of gMDSC and mMDSC 2, 4 and 6 h after blood draw and after an overnight rest by FACS analysis using the standard phenotypic markers. In addition, part of the cells was frozen directly after PBMC preparation and was measured after thawing. Results gMDSC levels showed no significant difference using fresh PBMC over time with a limitation for the overnight sample. However they were massively diminished after freezing (p = 0.0001 for all subjects). In contrast, frequencies of fresh mMDSC varied over time with no difference between time point 2 and 4 h but a significantly reduction after 6 h and overnight rest (p = 0.0005 and p = 0.005 respectively). Freezing of PBMC decreased the yield of mMDSC reaching statistical significance (p = 0.04). For both MDSC subgroups, FACS analysis became more difficult over time due to less sharp divisions between populations. Conclusions According to our data human MDSC need to be studied on fresh PBMC. gMDSC can be studied with delay, mMDSC however should be studied no later than 4 h after blood draw. These results are crucial as an increasing number of clinical trials aim at analyzing MDSC nowadays and the logistics of blood processing implies delayed sample processing in some cases. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0755-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eva Grützner
- Sektion Klinische Infektiologie, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Pettenkoferstr. 8a, 80336, Munich, Germany.
| | - Renate Stirner
- Sektion Klinische Infektiologie, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Pettenkoferstr. 8a, 80336, Munich, Germany.
| | - Lukas Arenz
- Sektion Klinische Infektiologie, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Pettenkoferstr. 8a, 80336, Munich, Germany.
| | - Anastasia P Athanasoulia
- Sektion Klinische Infektiologie, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Pettenkoferstr. 8a, 80336, Munich, Germany.
| | - Kathrin Schrödl
- Medizinische Klinik und Poliklinik V, Klinikum der Universität München, Ziemssenstr. 1, 80336, Munich, Germany.
| | - Carola Berking
- Klinik und Poliklinik für Dermatologie und Allergologie, Frauenlobstraße 9-11, 80337, Munich, Germany.
| | - Johannes R Bogner
- Sektion Klinische Infektiologie, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Pettenkoferstr. 8a, 80336, Munich, Germany.
| | - Rika Draenert
- Sektion Klinische Infektiologie, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Pettenkoferstr. 8a, 80336, Munich, Germany.
| |
Collapse
|
33
|
Tooley JE, Vudattu N, Choi J, Cotsapas C, Devine L, Raddassi K, Ehlers MR, McNamara JG, Harris KM, Kanaparthi S, Phippard D, Herold KC. Changes in T-cell subsets identify responders to FcR-nonbinding anti-CD3 mAb (teplizumab) in patients with type 1 diabetes. Eur J Immunol 2015; 46:230-41. [PMID: 26518356 DOI: 10.1002/eji.201545708] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 08/30/2015] [Accepted: 10/16/2015] [Indexed: 12/15/2022]
Abstract
The mechanisms whereby immune therapies affect progression of type 1 diabetes (T1D) are not well understood. Teplizumab, an FcR nonbinding anti-CD3 mAb, has shown efficacy in multiple randomized clinical trials. We previously reported an increase in the frequency of circulating CD8(+) central memory (CD8CM) T cells in clinical responders, but the generalizability of this finding and the molecular effects of teplizumab on these T cells have not been evaluated. We analyzed data from two randomized clinical studies of teplizumab in patients with new- and recent-onset T1D. At the conclusion of therapy, clinical responders showed a significant reduction in circulating CD4(+) effector memory T cells. Afterward, there was an increase in the frequency and absolute number of CD8CM T cells. In vitro, teplizumab expanded CD8CM T cells by proliferation and conversion of non-CM T cells. Nanostring analysis of gene expression of CD8CM T cells from responders and nonresponders versus placebo-treated control subjects identified decreases in expression of genes associated with immune activation and increases in expression of genes associated with T-cell differentiation and regulation. We conclude that CD8CM T cells with decreased activation and regulatory gene expression are associated with clinical responses to teplizumab in patients with T1D.
Collapse
Affiliation(s)
- James E Tooley
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Nalini Vudattu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jinmyung Choi
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Chris Cotsapas
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Lesley Devine
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Khadir Raddassi
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | | | - James G McNamara
- National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | | | | | | | - Kevan C Herold
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
34
|
Finney C, Serghides L. An In Vitro Model for Measuring Immune Responses to Malaria in the Context of HIV Co-infection. J Vis Exp 2015. [PMID: 26485041 DOI: 10.3791/52969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Malaria and HIV co-infection is a growing health priority. However, most research on malaria or HIV currently focuses on each infection individually. Although understanding the disease dynamics for each of these pathogens independently is vital, it is also important that the interactions between these pathogens are investigated and understood. We have developed a versatile in vitro model of HIV-malaria co-infection to study host immune responses to malaria in the context of HIV infection. Our model allows the study of secreted factors in cellular supernatants, cell surface and intracellular protein markers, as well as RNA expression levels. The experimental design and methods used limit variability and promote data reliability and reproducibility. All pathogens used in this model are natural human pathogens (Plasmodium falciparum and HIV-1), and all infected cells are naturally infected and used fresh. We use human erythrocytes parasitized with P. falciparum and maintained in continuous in vitro culture. We obtain freshly isolated peripheral blood mononuclear cells from chronically HIV-infected volunteers. Every condition used has an appropriate control (P. falciparum parasitized vs. normal erythrocytes), and every HIV-infected donor has an HIV uninfected control, from which cells are harvested on the same day. This model provides a realistic environment to study the interactions between malaria parasites and human immune cells in the context of HIV infection.
Collapse
Affiliation(s)
| | - Lena Serghides
- Toronto General Research Institute, University Health Network;
| |
Collapse
|
35
|
Martikainen MV, Kääriö H, Karvonen A, Schröder PC, Renz H, Kaulek V, Dalphin JC, von Mutius E, Schaub B, Pekkanen J, Hirvonen MR, Roponen M. Farm exposures are associated with lower percentage of circulating myeloid dendritic cell subtype 2 at age 6. Allergy 2015; 70:1278-87. [PMID: 26119336 DOI: 10.1111/all.12682] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND Early life farm exposures have been shown to decrease the risk of allergic diseases. Dendritic cells (DCs) may mediate asthma-protective effect of farm exposures as they play an important role in the development of immunity and tolerance. Our aim was to investigate whether the numbers and phenotypes of circulating DCs at age 6 are associated with farming, asthma, and atopy in a selected sample of French and Finnish children from the PASTURE study. METHODS We studied 82 farm and 86 nonfarm children with and without asthma. Using flow cytometry, BDCA1+ CD11c+ myeloid DC1s (mDC1), BDCA3+(high) mDC2s and BDCA2+ plasmacytoid DCs (pDCs) were identified and expressions of CD86, immunoglobulin-like transcript 3 (ILT3) and ILT4 were analyzed. Questionnaires were used to assess prenatal and lifetime patterns of farm exposures and to define asthma. Atopic sensitization was defined by specific IgE measurements. RESULTS The percentage of mDC2 cells was lower in farm children (0.033 ± 0.001) than in nonfarm children (0.042 ± 0.001; P = 0.008). Similar associations were found between mDC2 percentage and prenatal (P = 0.02) and lifetime exposure to farm milk (P = 0.03) and stables (P = 0.003), but these associations were not independent from farming. Asthma was positively associated with ILT4 + mDCs (P = 0.04) and negatively with CD86 + pDCs (P = 0.048) but only in nonfarm children. CONCLUSIONS Inverse association between farm exposure and mDC2 percentage suggest that this DC subset may play a role in farm-related immunoregulation.
Collapse
Affiliation(s)
- M.-V. Martikainen
- Department of Environmental Science; University of Eastern Finland; Kuopio Finland
| | - H. Kääriö
- Department of Environmental Science; University of Eastern Finland; Kuopio Finland
| | - A. Karvonen
- Department of Health Protection; National Institute for Health and Welfare; Kuopio Finland
| | - P. C. Schröder
- Department of Allergy and Pulmonary; University Children's Hospital; Dr. von Hauner Children's Hospital; LMU Munich; Munich Germany
- Member of the German Center for Lung Research (DZL); Munich; Germany
| | - H. Renz
- Institute of Laboratory Medicine, Pathobiochemistry and Molecular Diagnostics; Philipps University; Marburg Germany
| | - V. Kaulek
- Department of Respiratory Disease; UMR/CNRS 6249 Chrono-Environment; University Hospital of Besançon; Besançon France
| | - J.-C. Dalphin
- Department of Respiratory Disease; UMR/CNRS 6249 Chrono-Environment; University Hospital of Besançon; Besançon France
| | - E. von Mutius
- Department of Allergy and Pulmonary; University Children's Hospital; Dr. von Hauner Children's Hospital; LMU Munich; Munich Germany
- Member of the German Center for Lung Research (DZL); Munich; Germany
| | - B. Schaub
- Department of Allergy and Pulmonary; University Children's Hospital; Dr. von Hauner Children's Hospital; LMU Munich; Munich Germany
- Member of the German Center for Lung Research (DZL); Munich; Germany
| | - J. Pekkanen
- Department of Public Health; University of Helsinki; Helsinki Finland
| | - M.-R. Hirvonen
- Department of Environmental Science; University of Eastern Finland; Kuopio Finland
- Department of Health Protection; National Institute for Health and Welfare; Kuopio Finland
| | - M. Roponen
- Department of Environmental Science; University of Eastern Finland; Kuopio Finland
| |
Collapse
|
36
|
Corkum CP, Ings DP, Burgess C, Karwowska S, Kroll W, Michalak TI. Immune cell subsets and their gene expression profiles from human PBMC isolated by Vacutainer Cell Preparation Tube (CPT™) and standard density gradient. BMC Immunol 2015; 16:48. [PMID: 26307036 PMCID: PMC4549105 DOI: 10.1186/s12865-015-0113-0] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 08/17/2015] [Indexed: 01/25/2023] Open
Abstract
Background High quality genetic material is an essential pre-requisite when analyzing gene expression using microarray technology. Peripheral blood mononuclear cells (PBMC) are frequently used for genomic analyses, but several factors can affect the integrity of nucleic acids prior to their extraction, including the methods of PBMC collection and isolation. Due to the lack of the relevant data published, we compared the Ficoll-Paque density gradient centrifugation and BD Vacutainer cell preparation tube (CPT) protocols to determine if either method offered a distinct advantage in preparation of PBMC-derived immune cell subsets for their use in gene expression analysis. We evaluated the yield and purity of immune cell subpopulations isolated from PBMC derived by both methods, the quantity and quality of extracted nucleic acids, and compared gene expression in PBMC and individual immune cell types from Ficoll and CPT isolation protocols using Affymetrix microarrays. Results The mean yield and viability of fresh PBMC acquired by the CPT method (1.16 × 106 cells/ml, 93.3 %) were compatible to those obtained with Ficoll (1.34 × 106 cells/ml, 97.2 %). No differences in the mean purity, recovery, and viability of CD19+ (B cells), CD8+ (cytotoxic T cells), CD4+ (helper T cell) and CD14+ (monocytes) positively selected from CPT- or Ficoll-isolated PBMC were found. Similar quantities of high quality RNA and DNA were extracted from PBMC and immune cells obtained by both methods. Finally, the PBMC isolation methods tested did not impact subsequent recovery and purity of individual immune cell subsets and, importantly, their gene expression profiles. Conclusions Our findings demonstrate that the CPT and Ficoll PBMC isolation protocols do not differ in their ability to purify high quality immune cell subpopulations. Since there was no difference in the gene expression profiles between immune cells obtained by these two methods, the Ficoll isolation can be substituted by the CPT protocol without conceding phenotypic changes of immune cells and compromising the gene expression studies. Given that the CPT protocol is less elaborate, minimizes cells’ handling and processing time, this method offers a significant operating advantage, especially in large-scale clinical studies aiming at dissecting gene expression in PBMC and PBMC-derived immune cell subpopulations. Electronic supplementary material The online version of this article (doi:10.1186/s12865-015-0113-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher P Corkum
- Molecular Virology and Hepatology Research Group, Division of BioMedical Sciences, Faculty of Medicine, Health Sciences Centre, Memorial University, St. John's, NL, A1B3V6, Canada.
| | - Danielle P Ings
- Molecular Virology and Hepatology Research Group, Division of BioMedical Sciences, Faculty of Medicine, Health Sciences Centre, Memorial University, St. John's, NL, A1B3V6, Canada.
| | | | - Sylwia Karwowska
- Novartis Oncology Companion Diagnostics, Cambridge, MA, 02139, USA.
| | - Werner Kroll
- Novartis Oncology Companion Diagnostics, Cambridge, MA, 02139, USA. .,Present address: Quidel Corporation, San Diego, CA, 92130, USA.
| | - Tomasz I Michalak
- Molecular Virology and Hepatology Research Group, Division of BioMedical Sciences, Faculty of Medicine, Health Sciences Centre, Memorial University, St. John's, NL, A1B3V6, Canada.
| |
Collapse
|
37
|
Litvinov IS. The reasons for the nonmonotonic influence of extracellular calcium ion concentrations on the capacity of CD4+ T cells in human peripheral blood to polyclonal activation. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2015. [DOI: 10.1134/s106816201504010x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Hamot G, Ammerlaan W, Mathay C, Kofanova O, Betsou F. Method Validation for Automated Isolation of Viable Peripheral Blood Mononuclear Cells. Biopreserv Biobank 2015; 13:152-63. [DOI: 10.1089/bio.2014.0054] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Gael Hamot
- Integrated BioBank of Luxemburg (IBBL), Luxembourg
| | | | - Conny Mathay
- Integrated BioBank of Luxemburg (IBBL), Luxembourg
| | | | - Fay Betsou
- Integrated BioBank of Luxemburg (IBBL), Luxembourg
| |
Collapse
|
39
|
Parnell GP, Booth DR. Whole blood transcriptomic analysis to identify clinical biomarkers of drug response. Methods Mol Biol 2015; 1175:35-43. [PMID: 25150865 DOI: 10.1007/978-1-4939-0956-8_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Since most immunological and hematological conditions might be expected to alter whole blood gene expression, its examination can lead to insights into disease processes, and biomarkers to assess molecular phenotypes, disease states, progression and response to therapy. In this chapter we describe collection and storage of RNA from whole blood, techniques to measure gene expression, and analytical approaches to identify the dysregulated gene expression using pathway and clustering analysis, gene set enrichment, heat map approaches, and cell subset deconvolution.
Collapse
Affiliation(s)
- Grant P Parnell
- Westmead Millennium Institute, University of Sydney, Hawkesbury Road, Sydney, NSW, 2145, Australia
| | | |
Collapse
|
40
|
Posevitz-Fejfár A, Posevitz V, Gross CC, Bhatia U, Kurth F, Schütte V, Bar-Or A, Meuth SG, Wiendl H. Effects of blood transportation on human peripheral mononuclear cell yield, phenotype and function: implications for immune cell biobanking. PLoS One 2014; 9:e115920. [PMID: 25541968 PMCID: PMC4277394 DOI: 10.1371/journal.pone.0115920] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 11/30/2014] [Indexed: 01/25/2023] Open
Abstract
Human biospecimen collection, processing and preservation are rapidly emerging subjects providing essential support to clinical as well as basic researchers. Unlike collection of other biospecimens (e.g. DNA and serum), biobanking of viable immune cells, such as peripheral blood mononuclear cells (PBMC) and/or isolated immune cell subsets is still in its infancy. While certain aspects of processing and freezing conditions have been studied in the past years, little is known about the effect of blood transportation on immune cell survival, phenotype and specific functions. However, especially for multicentric and cooperative projects it is vital to precisely know those effects. In this study we investigated the effect of blood shipping and pre-processing delay on immune cell phenotype and function both on cellular and subcellular levels. Peripheral blood was collected from healthy volunteers (n = 9): at a distal location (shipped overnight) and in the central laboratory (processed immediately). PBMC were processed in the central laboratory and analyzed post-cryopreservation. We analyzed yield, major immune subset distribution, proliferative capacity of T cells, cytokine pattern and T-cell receptor signal transduction. Results show that overnight transportation of blood samples does not globally compromise T- cell subsets as they largely retain their phenotype and proliferative capacity. However, NK and B cell frequencies, the production of certain PBMC-derived cytokines and IL-6 mediated cytokine signaling pathway are altered due to transportation. Various control experiments have been carried out to compare issues related to shipping versus pre-processing delay on site. Our results suggest the implementation of appropriate controls when using multicenter logistics for blood transportation aiming at subsequent isolation of viable immune cells, e.g. in multicenter clinical trials or studies analyzing immune cells/subsets. One important conclusion might be that despite changes due to overnight shipment, highly standardized central processing (and analysis) could be superior to multicentric de-central processing with more difficult standardization.
Collapse
Affiliation(s)
- Anita Posevitz-Fejfár
- University Hospital Muenster, Department of Neurology, Albert-Schweitzer-Campus 1, Muenster, Germany
- * E-mail: (HW); (APF)
| | - Vilmos Posevitz
- University Hospital Muenster, Department of Neurology, Albert-Schweitzer-Campus 1, Muenster, Germany
| | - Catharina C. Gross
- University Hospital Muenster, Department of Neurology, Albert-Schweitzer-Campus 1, Muenster, Germany
| | - Urvashi Bhatia
- University Hospital Muenster, Department of Neurology, Albert-Schweitzer-Campus 1, Muenster, Germany
| | - Frank Kurth
- University Hospital Muenster, Department of Neurology, Albert-Schweitzer-Campus 1, Muenster, Germany
| | - Verena Schütte
- University Hospital Muenster, Department of Neurology, Albert-Schweitzer-Campus 1, Muenster, Germany
| | - Amit Bar-Or
- Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Sven G. Meuth
- University Hospital Muenster, Department of Neurology, Albert-Schweitzer-Campus 1, Muenster, Germany
| | - Heinz Wiendl
- University Hospital Muenster, Department of Neurology, Albert-Schweitzer-Campus 1, Muenster, Germany
- * E-mail: (HW); (APF)
| |
Collapse
|
41
|
Association of chronic hepatitis C infection with T-cell phenotypes in HIV-negative and HIV-positive women. J Acquir Immune Defic Syndr 2014; 67:295-303. [PMID: 25314250 DOI: 10.1097/qai.0000000000000310] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Hepatitis C virus (HCV) viremia is thought to have broad systemic effects on the cellular immune system that go beyond its impact on just those T cells that are HCV specific. However, previous studies of chronic HCV and circulating T-cell subsets (activation and differentiation phenotypes) in HIV negatives used general population controls, rather than a risk-appropriate comparison group. Studies in HIV positives did not address overall immune status (total CD4⁺ count). METHODS We used fresh blood from HIV-positive and at-risk HIV-negative women, with and without chronic HCV, to measure percentages of activated CD4⁺ and CD8⁺ T cells, Tregs, and T-cell differentiation phenotypes (naive, central memory, effector memory (EM), and terminally differentiated effector). This included 158 HIV negatives and 464 HIV positives, of whom 18 and 63, respectively, were HCV viremic. RESULTS In multivariate models of HIV negatives, HCV viremia was associated with 25% fewer naive CD4⁺ (P = 0.03), 33% more EM CD4⁺ (P = 0.0002), and 37% fewer central memory CD8⁺ (P = 0.02) T cells. Among HIV positives, we observed only 1 of these 3 relationships: higher percentage of EM CD4⁺ among HCV viremic women. Furthermore, the association with EM CD4⁺ among HIV positives was limited to individuals with diminished immune status (total CD4⁺ count ≤500 cells/μL), as were associations of HCV viremia with higher percentages of activated CD4⁺ and Tregs. Among HIV positives with high CD4⁺ count, no significant associations were observed. CONCLUSIONS These data suggest that HCV viremia in HIV negatives is associated with accelerated T-cell differentiation, but among HIV positives, the impact of HCV viremia is less straightforward and varies by total CD4v count.
Collapse
|
42
|
Lohman-Payne B, Sandifer T, OhAinle M, Crudder C, Lynch J, Omenda MM, Maroa J, Fowke K, John-Stewart GC, Farquhar C. In-utero infection with HIV-1 associated with suppressed lymphoproliferative responses at birth. Clin Exp Immunol 2014; 178:86-93. [PMID: 24853045 DOI: 10.1111/cei.12386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2014] [Indexed: 11/28/2022] Open
Abstract
In-utero exposure to HIV-1 may affect the immune system of the developing child and may induce HIV-1-specific immune responses, even in the absence of HIV-1 infection. We evaluated lymphoproliferative capacity at birth among 40 HIV-1-uninfected infants born to HIV-1-infected mothers and 10 infants who had acquired HIV-1 in utero. Cord blood mononuclear cells were assayed using [(3) H]-thymidine incorporation for proliferation in response to HIV-1 p55-gag and the control stimuli phytohaemagglutinin (PHA), Staphylococcus enterotoxin B (SEB) and allogeneic cells. In response to HIV-1 p55-gag, eight (20%) HIV-1-exposed, uninfected (EU) infants had a stimulation index (SI) ≥ 2 and three (30%) in-utero HIV-1 infected infants had SI ≥2. The frequency and magnitude of responses to HIV-1 p55-gag were low overall, and did not differ statistically between groups. However, proliferative responses to control stimuli were significantly higher in EU infants than in infants infected in utero, with a median SI in response to PHA of 123 [interquartile range (IQR) 77-231] versus 18 (IQR 4-86) between EU and infected infants, respectively (P < 0·001). Among infected infants, gestational maturity was associated with the strength of HIV-1 p55-gag response (P < 0·001); neither maternal nor infant HIV-1 viral load was associated. In summary, EU and HIV-1-infected infants mounted HIV-1-specific lymphoproliferative responses at similar rates (20-30%), and although global immune function was preserved among EU infants, neonatal immune responses were significantly compromised by HIV-1 infection. Such early lymphoproliferative compromise may, in part, explain rapid progression to AIDS and death among HIV-1-infected infants.
Collapse
Affiliation(s)
- B Lohman-Payne
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya; Department of Medicine, University of Washington, Seattle, Washington, USA; Department of Global Health, University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Strickler HD, Martinson J, Desai S, Xie X, Burk RD, Anastos K, Massad LS, Minkoff H, Xue X, D'Souza G, Levine AM, Colie C, Watts DH, Palefsky JM, Landay A. The relation of plasmacytoid dendritic cells (pDCs) and regulatory T-cells (Tregs) with HPV persistence in HIV-infected and HIV-uninfected women. Viral Immunol 2014; 27:20-5. [PMID: 24494969 DOI: 10.1089/vim.2013.0097] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Other than CD4+ count, the immunologic factors that underlie the relationship of HIV/AIDS with persistent oncogenic HPV (oncHPV) and cervical cancer are not well understood. Plasmacytoid dendritic cells (pDCs) and regulatory T-cells (Tregs) are of particular interest. pDCs have both effector and antigen presenting activity and, in HIV-positive patients, low pDC levels are associated with opportunistic infections. Tregs downregulate immune responses, and are present at high levels in HIV-positives. The current pilot study shows for the first time that low pDC and high Treg levels may be significantly associated with oncHPV persistence in both HIV-positive and HIV-negative women. Larger studies are now warranted.
Collapse
Affiliation(s)
- Howard D Strickler
- 1 Department of Epidemiology and Population Health, Albert Einstein College of Medicine , Bronx, New York
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Circulating B-lymphocytes as potential biomarkers of tuberculosis infection activity. PLoS One 2014; 9:e106796. [PMID: 25192196 PMCID: PMC4156407 DOI: 10.1371/journal.pone.0106796] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 08/02/2014] [Indexed: 12/25/2022] Open
Abstract
Accurate biomarkers of Mycobacterium tuberculosis infection activity would significantly improve early diagnosis, treatment and management of M. tuberculosis infection. We hypothesised that circulating B-lymphocytes may be useful biomarkers of tuberculosis (TB) infection status in highly TB-endemic settings. Ex-vivo and in-vitro mycobacteria-specific B-cell ELISPOT assays were used to examine the plasmablast (PB) and memory B-cell (MBC) responses in the peripheral blood of adult, healthy, community controls (n = 151) and of active TB patients (n = 48) living in Uganda. Frequencies of mycobacteria-specific PBs were markedly higher in active TB patients compared to healthy controls, and, conversely, MBCs were markedly higher in the healthy controls compared to active TB patients. In addition, the community controls with evidence of latent TB infection had higher peripheral blood PB and MBC responses than those without evidence of TB infection. These data demonstrate that peripheral blood B-cell responses are differentially modulated during latent and active M. tuberculosis infection, and suggest that the PB to MBC ratio may be a useful biomarker of TB infection activity.
Collapse
|
45
|
The Center for HIV/AIDS Vaccine Immunology (CHAVI) multi-site quality assurance program for cryopreserved human peripheral blood mononuclear cells. J Immunol Methods 2014; 409:21-30. [PMID: 24910414 DOI: 10.1016/j.jim.2014.05.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 05/23/2014] [Accepted: 05/28/2014] [Indexed: 01/17/2023]
Abstract
The Center for HIV/AIDS Vaccine Immunology (CHAVI) consortium was established to determine the host and virus factors associated with HIV transmission, infection and containment of virus replication, with the goal of advancing the development of an HIV protective vaccine. Studies to meet this goal required the use of cryopreserved Peripheral Blood Mononuclear Cell (PBMC) specimens, and therefore it was imperative that a quality assurance (QA) oversight program be developed to monitor PBMC samples obtained from study participants at multiple international sites. Nine site-affiliated laboratories in Africa and the USA collected and processed PBMCs, and cryopreserved PBMC were shipped to CHAVI repositories in Africa and the USA for long-term storage. A three-stage program was designed, based on Good Clinical Laboratory Practices (GCLP), to monitor PBMC integrity at each step of this process. The first stage evaluated the integrity of fresh PBMCs for initial viability, overall yield, and processing time at the site-affiliated laboratories (Stage 1); for the second stage, the repositories determined post-thaw viability and cell recovery of cryopreserved PBMC, received from the site-affiliated laboratories (Stage 2); the third stage assessed the long-term specimen storage at each repository (Stage 3). Overall, the CHAVI PBMC QA oversight program results highlight the relative importance of each of these stages to the ultimate goal of preserving specimen integrity from peripheral blood collection to long-term repository storage.
Collapse
|
46
|
Kofanova OA, Davis K, Glazer B, De Souza Y, Kessler J, Betsou F. Viable mononuclear cell stability study for implementation in a proficiency testing program: impact of shipment conditions. Biopreserv Biobank 2014; 12:206-16. [PMID: 24955735 PMCID: PMC4955601 DOI: 10.1089/bio.2013.0090] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The impact of shipping temperatures and preservation media used during transport of either peripheral blood mononuclear cells (PBMCs) or Jurkat cells was assessed, in view of implementing of a proficiency testing scheme on mononuclear cell viability. Samples were analyzed before and after shipment at different temperatures (ambient temperature, dry ice, and liquid nitrogen) and in different preservation media (serum with cryoprotectant, commercial cryopreservation solution, and room temperature transport medium). Sample quality was assessed by viability assays (Trypan Blue dye exclusion, flow cytometry, Cell Analysis System cell counting (CASY)), and by ELISpot functional assay. The liquid nitrogen storage and shipment were found to be the most stable conditions to preserve cell viability and functionality. However, we show that alternative high quality shipment conditions for viable cells are dry ice shipment and commercial cryopreservation solution. These were also cost-efficient shipment conditions, satisfying the requirements of a proficiency testing scheme for viable mononuclear cells. Room temperature transport medium dramatically and adversely affected the integrity of mononuclear cells.
Collapse
Affiliation(s)
- Olga A. Kofanova
- Integrated BioBank Of Luxembourg (IBBL), 6 rue Nicolas Ernest Barblé, L-1210, Luxembourg
| | - Kristine Davis
- PPD Vaccines and Biologics Laboratory, Wayne, Pennsylvania
| | | | | | - Joseph Kessler
- PPD Vaccines and Biologics Laboratory, Wayne, Pennsylvania
| | - Fotini Betsou
- Integrated BioBank Of Luxembourg (IBBL), 6 rue Nicolas Ernest Barblé, L-1210, Luxembourg
| |
Collapse
|
47
|
Sambor A, Garcia A, Berrong M, Pickeral J, Brown S, Rountree W, Sanchez A, Pollara J, Frahm N, Keinonen S, Kijak GH, Roederer M, Levine G, D'Souza MP, Jaimes M, Koup R, Denny T, Cox J, Ferrari G. Establishment and maintenance of a PBMC repository for functional cellular studies in support of clinical vaccine trials. J Immunol Methods 2014; 409:107-16. [PMID: 24787274 DOI: 10.1016/j.jim.2014.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/28/2014] [Accepted: 04/08/2014] [Indexed: 11/19/2022]
Abstract
A large repository of cryopreserved peripheral blood mononuclear cells (PBMCs) samples was created to provide laboratories testing the specimens from human immunodeficiency virus-1 (HIV-1) vaccine clinical trials the material for assay development, optimization, and validation. One hundred thirty-one PBMC samples were collected using leukapheresis procedure between 2007 and 2013 by the Comprehensive T cell Vaccine Immune Monitoring Consortium core repository. The donors included 83 human immunodeficiency virus-1 (HIV-1) seronegative and 32 HIV-1 seropositive subjects. The samples were extensively characterized for the ability of T cell subsets to respond to recall viral antigens including cytomegalovirus, Epstein-Barr virus, influenza virus, and HIV-1 using Interferon-gamma (IFN-γ) enzyme linked immunospot (ELISpot) and IFN-γ/interleukin 2 (IL-2) intracellular cytokine staining (ICS) assays. A subset of samples was evaluated over time to determine the integrity of the cryopreserved samples in relation to recovery, viability, and functionality. The principal results of our study demonstrate that viable and functional cells were consistently recovered from the cryopreserved samples. Therefore, we determined that this repository of large size cryopreserved cellular samples constitutes a unique resource for laboratories that are involved in optimization and validation of assays to evaluate T, B, and NK cellular functions in the context of clinical trials.
Collapse
Affiliation(s)
- Anna Sambor
- Foundation for National Institutes of Health, Bethesda, MD, USA
| | - Ambrosia Garcia
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA
| | | | | | - Sara Brown
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA
| | - Wes Rountree
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA
| | - Ana Sanchez
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA
| | | | - Nicole Frahm
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sarah Keinonen
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA
| | - Gustavo H Kijak
- Viral Genetics Section, US Military HIV Research Program, Henry M Jackson Foundation for the Advancement of Military Medicine, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | | | - Gail Levine
- Foundation for National Institutes of Health, Bethesda, MD, USA
| | | | | | - Richard Koup
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Thomas Denny
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA
| | - Josephine Cox
- International AIDS Vaccine Initiative, New York, NY, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
48
|
Shete A, Jayawant P, Thakar M, Kurle S, Singh DP, Paranjape RS. Differential modulation of phenotypic composition of HIV-infected and -uninfected PBMCs during cryopreservation. J Immunoassay Immunochem 2014; 34:333-45. [PMID: 23859784 DOI: 10.1080/15321819.2012.741087] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This article was designed to determine variations in phenotypic composition of fresh and frozen PBMCs for assessing utility of cryopreserved PBMCs for phenotypic assays. Relative percentages of effector memory cells increased significantly as against percentages of naïve cells which showed significant decrease after cryopreservation in HIV-uninfected samples. These differences were not significant in HIV-infected individuals. There was no significant difference in the expression of activation markers in fresh and frozen PBMCs except the HLA DR expression on CD8 cells in HIV-infected individuals, which was significantly decreased in frozen PBMCs. Thus, cryopreservation resulted in differential effect on phenotypic composition of PBMCs in HIV-infected and -uninfected individuals.
Collapse
Affiliation(s)
- Ashwini Shete
- National AIDS Research Institute, Bhosari, Pune, India.
| | | | | | | | | | | |
Collapse
|
49
|
Germann A, Oh YJ, Schmidt T, Schön U, Zimmermann H, von Briesen H. Temperature fluctuations during deep temperature cryopreservation reduce PBMC recovery, viability and T-cell function. Cryobiology 2013; 67:193-200. [PMID: 23850825 DOI: 10.1016/j.cryobiol.2013.06.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 06/24/2013] [Accepted: 06/26/2013] [Indexed: 10/26/2022]
Abstract
The ability to analyze cryopreserved peripheral blood mononuclear cell (PBMC) from biobanks for antigen-specific immunity is necessary to evaluate response to immune-based therapies. To ensure comparable assay results, collaborative research in multicenter trials needs reliable and reproducible cryopreservation that maintains cell viability and functionality. A standardized cryopreservation procedure is comprised of not only sample collection, preparation and freezing but also low temperature storage in liquid nitrogen without any temperature fluctuations, to avoid cell damage. Therefore, we have developed a storage approach to minimize suboptimal storage conditions in order to maximize cell viability, recovery and T-cell functionality. We compared the influence of repeated temperature fluctuations on cell health from sample storage, sample sorting and removal in comparison to sample storage without temperature rises. We found that cyclical temperature shifts during low temperature storage reduce cell viability, recovery and immune response against specific-antigens. We showed that samples handled under a protective hood system, to avoid or minimize such repeated temperature rises, have comparable cell viability and cell recovery rates to samples stored without any temperature fluctuations. Also T-cell functionality could be considerably increased with the use of the protective hood system compared to sample handling without such a protection system. This data suggests that the impact of temperature fluctuation on cell integrity should be carefully considered in future clinical vaccine trials and consideration should be given to optimal sample storage conditions.
Collapse
Affiliation(s)
- Anja Germann
- (a)Fraunhofer Institute for Biomedical Engineering, Ensheimerstr. 48, 66386 St. Ingbert, Germany
| | | | | | | | | | | |
Collapse
|
50
|
Ogunjimi B, Hens N, Malfait R, Van Tendeloo V, Smits E. Creating a robust framework for the analysis of cryopreserved samples in quantitative immunological experiments. J Immunol Methods 2013; 392:63-7. [DOI: 10.1016/j.jim.2013.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 03/07/2013] [Accepted: 03/07/2013] [Indexed: 01/31/2023]
|