1
|
Wang X, Ma S, Twardowski P, Lau C, Chan YS, Wong K, Xiao S, Wang J, Wu X, Frankel P, Wilson TG, Synold TW, Presant C, Dorff T, Yu J, Sadava D, Chen S. Reduction of myeloid-derived suppressor cells in prostate cancer murine models and patients following white button mushroom treatment. Clin Transl Med 2024; 14:e70048. [PMID: 39390760 PMCID: PMC11467013 DOI: 10.1002/ctm2.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND In a previously reported Phase I trial, we observed therapy-associated declines in circulating myeloid-derived suppressor cells (MDSCs) with the administration of white button mushroom (WBM) tablets in prostate cancer (PCa) patients. These observations led us to hypothesise that WBM could mitigate PCa progression by suppressing MDSCs. METHODS We performed bidirectional translational research to examine the immunomodulatory effects of WBM consumption in both syngeneic murine PCa models and patients with PCa participating in an ongoing randomised Phase II trial (NCT04519879). RESULTS In murine models, WBM treatment significantly suppressed tumour growth with a reduction in both the number and function of MDSCs, which in turn promoted antitumour immune responses mediated by T cells and natural killer (NK) cells. In patients, after consumption of WBM tablets for 3 months, we observed a decline in circulating polymorphonuclear MDSCs (PMN-MDSCs), along with an increase in cytotoxic CD8+ T and NK cells. Furthermore, single immune cell profiling of peripheral blood from WBM-treated patients showed suppressed STAT3/IRF1 and TGFβ signalling in circulating PMN-MDSCs. Subclusters of PMN-MDSCs presented transcriptional profiles associated with responsiveness to fungi, neutrophil chemotaxis, leukocyte aggregation, and regulation of inflammatory response. Finally, in mouse models of PCa, we found that WBM consumption enhanced the anticancer activity of anti-PD-1 antibodies, indicating that WBM may be used as an adjuvant therapy with immune checkpoint inhibitors. CONCLUSION Our results from PCa murine models and patients provide mechanistic insights into the immunomodulatory effects of WBM and provide a scientific foundation for WBM as a nutraceutical intervention to delay or prevent PCa progression. HIGHLIGHTS White button mushroom (WBM) treatment resulted in a reduction in pro-tumoural MDSCs, notably polymorphonuclear MDSCs (PMN-MDSCs), along with activation of anti-tumoural T and NK cells. Human single immune cell gene expression profiling shed light on the molecular alterations induced by WBM, specifically on PMN-MDSCs. A proof-of-concept study combining WBM with PD-1 blockade in murine models revealed an additive effect on tumour regression and survival outcomes, highlighting the clinical relevance of WBM in cancer management.
Collapse
Affiliation(s)
- Xiaoqiang Wang
- Department of Cancer Biology & Molecular MedicineBeckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Shoubao Ma
- Department of Hematology and Hematopoietic Cell TransplantationCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Przemyslaw Twardowski
- Department of Urology and Urologic OncologyProvidence Saint John's Cancer InstituteSanta MonicaCaliforniaUSA
| | - Clayton Lau
- Department of SurgeryCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Yin S. Chan
- Department of Cancer Biology & Molecular MedicineBeckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Kelly Wong
- Department of Cancer Biology & Molecular MedicineBeckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Sai Xiao
- Department of Hematology and Hematopoietic Cell TransplantationCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Jinhui Wang
- Integrative Genomics CoreBeckman Research Institute, City of HopeMonroviaCaliforniaUSA
| | - Xiwei Wu
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Paul Frankel
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Timothy G. Wilson
- Department of Urology and Urologic OncologyProvidence Saint John's Cancer InstituteSanta MonicaCaliforniaUSA
| | - Timothy W Synold
- Department of Medical Oncology & Therapeutics ResearchCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Cary Presant
- Department of Medical Oncology & Therapeutics ResearchCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Tanya Dorff
- Department of Medical Oncology & Therapeutics ResearchCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell TransplantationCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - David Sadava
- Department of Cancer Biology & Molecular MedicineBeckman Research Institute, City of HopeDuarteCaliforniaUSA
| | - Shiuan Chen
- Department of Cancer Biology & Molecular MedicineBeckman Research Institute, City of HopeDuarteCaliforniaUSA
| |
Collapse
|
2
|
Lee WH, Graham CE, Wiggin HR, Nolan HK, Graham KJ, Korell F, Leick MB, Barselau AL, Emmanuel-Alejandro E, Trailor MA, Gildea JM, Preffer F, Frigault MJ, Maus MV, Gallagher KME. Optimization of a flow cytometry test for routine monitoring of B cell maturation antigen targeted CAR in peripheral blood. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2024; 106:162-170. [PMID: 38418432 DOI: 10.1002/cyto.b.22165] [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: 10/17/2023] [Revised: 01/11/2024] [Accepted: 02/06/2024] [Indexed: 03/01/2024]
Abstract
Chimeric antigen receptor (CAR) modified T cell therapies targeting BCMA have displayed impressive activity in the treatment of multiple myeloma. There are currently two FDA licensed products, ciltacabtagene autoleucel and idecabtagene vicleucel, for treating relapsed and refractory disease. Although correlative analyses performed by product manufacturers have been reported in clinical trials, there are limited options for reliable BCMA CAR T detection assays for physicians and researchers looking to explore it as a biomarker for clinical outcome. Given the known association of CAR T cell expansion kinetics with toxicity and response, being able to quantify BCMA CAR T cells routinely and accurately in the blood of patients can serve as a valuable asset. Here, we optimized an accurate and sensitive flow cytometry test using a PE-conjugated soluble BCMA protein, with a lower limit of quantitation of 0.19% of CD3+ T cells, suitable for use as a routine assay for monitoring the frequency of BCMA CAR T cells in the blood of patients receiving either ciltacabtagene autoleucel or idecabtagene vicleucel.
Collapse
Affiliation(s)
- Won-Ho Lee
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Charlotte E Graham
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Hadley R Wiggin
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hannah K Nolan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kiana J Graham
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Felix Korell
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark B Leick
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexis L Barselau
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Estelle Emmanuel-Alejandro
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael A Trailor
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Juliane M Gildea
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Frederic Preffer
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Matthew J Frigault
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Kathleen M E Gallagher
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Hubens WHG, Maié T, Schnitker M, Bocova L, Puri D, Wessiepe M, Kramer J, Rink L, Koschmieder S, Costa IG, Wagner W. Targeted DNA Methylation Analysis Facilitates Leukocyte Counts in Dried Blood Samples. Clin Chem 2023; 69:1283-1294. [PMID: 37708296 DOI: 10.1093/clinchem/hvad143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/10/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND Cell-type specific DNA methylation (DNAm) can be employed to determine the numbers of leukocyte subsets in blood. In contrast to conventional methods for leukocyte counts, which are based on cellular morphology or surface marker protein expression, the cellular deconvolution based on DNAm levels is applicable for frozen or dried blood. Here, we further enhanced targeted DNAm assays for leukocyte counts in clinical application. METHODS DNAm profiles of 40 different studies were compiled to identify CG dinucleotides (CpGs) with cell-type specific DNAm using a computational framework, CimpleG. DNAm levels at these CpGs were then measured with digital droplet PCR in venous blood from 160 healthy donors and 150 patients with various hematological disorders. Deconvolution was further validated with venous blood (n = 75) and capillary blood (n = 31) that was dried on Whatman paper or on Mitra microsampling devices. RESULTS In venous blood, automated cell counting or flow cytometry correlated well with epigenetic estimates of relative leukocyte counts for granulocytes (r = 0.95), lymphocytes (r = 0.97), monocytes (r = 0.82), CD4 T cells (r = 0.84), CD8 T cells (r = 0.94), B cells (r = 0.96), and NK cells (r = 0.72). Similar correlations and precisions were achieved for dried blood samples. Spike-in with a reference plasmid enabled accurate epigenetic estimation of absolute leukocyte counts from dried blood samples, correlating with conventional venous (r = 0.86) and capillary (r = 0.80) blood measurements. CONCLUSIONS The advanced selection of cell-type specific CpGs and utilization of digital droplet PCR analysis provided accurate epigenetic blood counts. Analysis of dried blood facilitates self-sampling with a finger prick, thereby enabling easier accessibility to testing.
Collapse
Affiliation(s)
- Wouter H G Hubens
- Institute for Stem Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Tiago Maié
- Institute for Computational Genomics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Matthis Schnitker
- Institute for Stem Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Ledio Bocova
- Institute for Stem Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Deepika Puri
- Institute for Stem Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Martina Wessiepe
- Institute for Transfusion Medicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Jan Kramer
- Division of Nephrology and Transplantation Unit, Department of Internal Medicine I, University of Lübeck, Lübeck, Germany
- LADR Laboratory Group Dr. Kramer & Colleagues, Geesthacht, Germany
| | - Lothar Rink
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Wolfgang Wagner
- Institute for Stem Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| |
Collapse
|
4
|
Pacheco GA, Andrade CA, Gálvez NM, Vázquez Y, Rodríguez-Guilarte L, Abarca K, González PA, Bueno SM, Kalergis AM. Characterization of the humoral and cellular immunity induced by a recombinant BCG vaccine for the respiratory syncytial virus in healthy adults. Front Immunol 2023; 14:1215893. [PMID: 37533867 PMCID: PMC10390696 DOI: 10.3389/fimmu.2023.1215893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/27/2023] [Indexed: 08/04/2023] Open
Abstract
Introduction The human respiratory syncytial virus (hRSV) is responsible for most respiratory tract infections in infants. Even though currently there are no approved hRSV vaccines for newborns or infants, several candidates are being developed. rBCG-N-hRSV is a vaccine candidate previously shown to be safe in a phase I clinical trial in adults (clinicaltrials.gov identifier #NCT03213405). Here, secondary immunogenicity analyses were performed on these samples. Methods PBMCs isolated from immunized volunteers were stimulated with hRSV or mycobacterial antigens to evaluate cytokines and cytotoxic T cell-derived molecules and the expansion of memory T cell subsets. Complement C1q binding and IgG subclass composition of serum antibodies were assessed. Results Compared to levels detected prior to vaccination, perforin-, granzyme B-, and IFN-γ-producing PBMCs responding to stimulus increased after immunization, along with their effector memory response. N-hRSV- and mycobacterial-specific antibodies from rBCG-N-hRSV-immunized subjects bound C1q. Conclusion Immunization with rBCG-N-hRSV induces cellular and humoral immune responses, supporting that rBCG-N-hRSV is immunogenic and safe in healthy individuals. Clinical trial registration https://classic.clinicaltrials.gov/ct2/show/, identifier NCT03213405.
Collapse
Affiliation(s)
- Gaspar A. Pacheco
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A. Andrade
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M.S. Gálvez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yaneisi Vázquez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Linmar Rodríguez-Guilarte
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katia Abarca
- Millennium Institute on Immunology and Immunotherapy, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Enfermedades Infecciosas e Inmunología Pediá trica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
5
|
Rogers NM, Zammit N, Nguyen-Ngo D, Souilmi Y, Minhas N, Meijles DN, Self E, Walters SN, Warren J, Cultrone D, El-Rashid M, Li J, Chtanova T, O'Connell PJ, Grey ST. The impact of the cytoplasmic ubiquitin ligase TNFAIP3 gene variation on transcription factor NF-κB activation in acute kidney injury. Kidney Int 2023; 103:1105-1119. [PMID: 37097268 DOI: 10.1016/j.kint.2023.02.030] [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: 08/23/2021] [Revised: 02/08/2023] [Accepted: 02/23/2023] [Indexed: 04/26/2023]
Abstract
Nuclear factor κB (NF-κB) activation is a deleterious molecular mechanism that drives acute kidney injury (AKI) and manifests in transplanted kidneys as delayed graft function. The TNFAIP3 gene encodes A20, a cytoplasmic ubiquitin ligase and a master negative regulator of the NF- κB signaling pathway. Common population-specific TNFAIP3 coding variants that reduce A20's enzyme function and increase NF- κB activation have been linked to heightened protective immunity and autoimmune disease, but have not been investigated in AKI. Here, we functionally identified a series of unique human TNFAIP3 coding variants linked to the autoimmune genome-wide association studies single nucleotide polymorphisms of F127C; namely F127C;R22Q, F127C;G281E, F127C;W448C and F127C;N449K that reduce A20's anti-inflammatory function in an NF- κB reporter assay. To investigate the impact of TNFAIP3 hypomorphic coding variants in AKI we tested a mouse Tnfaip3 hypomorph in a model of ischemia reperfusion injury (IRI). The mouse Tnfaip3 coding variant I325N increases NF- κB activation without overt inflammatory disease, providing an immune boost as I325N mice exhibit enhanced innate immunity to a bacterial challenge. Surprisingly, despite exhibiting increased intra-kidney NF- κB activation with inflammation in IRI, the kidney of I325N mice was protected. The I325N variant influenced the outcome of IRI by changing the dynamic expression of multiple cytoprotective mechanisms, particularly by increasing NF- κB-dependent anti-apoptotic factors BCL-2, BCL-XL, c-FLIP and A20, altering the active redox state of the kidney with a reduction of superoxide levels and the enzyme super oxide dismutase-1, and enhancing cellular protective mechanisms including increased Foxp3+ T cells. Thus, TNFAIP3 gene variants represent a kidney and population-specific molecular factor that can dictate the course of IRI.
Collapse
Affiliation(s)
- Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; Renal and Transplant Medicine Unit, Westmead Hospital, Westmead, New South Wales, Australia; Westmead Clinical School, University of Sydney, New South Wales, Australia
| | - Nathan Zammit
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Danny Nguyen-Ngo
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Yassine Souilmi
- Australian Centre for Ancient DNA, School of Biological Sciences, University of Adelaide, South Australia, Australia; Environment Institute, Faculty of Sciences, University of Adelaide, South Australia, Australia
| | - Nikita Minhas
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Eleanor Self
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Stacey N Walters
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Joanna Warren
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Daniele Cultrone
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Maryam El-Rashid
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Jennifer Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Tatyana Chtanova
- Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Innate and Tumour Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Philip J O'Connell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; Renal and Transplant Medicine Unit, Westmead Hospital, Westmead, New South Wales, Australia; Westmead Clinical School, University of Sydney, New South Wales, Australia
| | - Shane T Grey
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
6
|
Serra V, Orrù V, Lai S, Lobina M, Steri M, Cucca F, Fiorillo E. Comparison of Whole Blood Cryopreservation Methods for Extensive Flow Cytometry Immunophenotyping. Cells 2022; 11:cells11091527. [PMID: 35563832 PMCID: PMC9103885 DOI: 10.3390/cells11091527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/24/2022] Open
Abstract
Fresh blood immunophenotyping by flow cytometry, based on the reliable simultaneous detection of several markers in a cell, is the method of choice to study the circulating human immune system. Especially in large and multicenter studies, high sample quality is difficult to achieve, and adequate collection and storage of samples with fine-tuned whole blood cryopreservation is mandatory. Here, we compared the quality of immunophenotypic data obtained from fresh blood with those obtained after five cryopreservation methods by quantifying the levels of 41 immune cell populations. They comprised B and T lymphocyte subsets and their maturation stages, as well as monocytes and granulocytes. Three methods used fixative solutions and two other methods used dimethyl sulfoxide solutions to preserve cell viability. The fixative methods prevented detection of markers critical for identification of B and T cell subsets, including CD27, CXCR3, and CCR6. The other two methods permitted reliable discrimination of most immune-cell populations in thawed samples, though some cell frequencies varied compared to the corresponding fresh sample. Of those two methods, the one preserving blood in media containing dimethyl sulfoxide produced results that were most similar to those with fresh samples.
Collapse
Affiliation(s)
- Valentina Serra
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Monserrato, 09042 Cagliari, Italy; (V.O.); (S.L.); (M.L.); (M.S.); (F.C.); (E.F.)
- Correspondence:
| | - Valeria Orrù
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Monserrato, 09042 Cagliari, Italy; (V.O.); (S.L.); (M.L.); (M.S.); (F.C.); (E.F.)
| | - Sandra Lai
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Monserrato, 09042 Cagliari, Italy; (V.O.); (S.L.); (M.L.); (M.S.); (F.C.); (E.F.)
| | - Monia Lobina
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Monserrato, 09042 Cagliari, Italy; (V.O.); (S.L.); (M.L.); (M.S.); (F.C.); (E.F.)
| | - Maristella Steri
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Monserrato, 09042 Cagliari, Italy; (V.O.); (S.L.); (M.L.); (M.S.); (F.C.); (E.F.)
| | - Francesco Cucca
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Monserrato, 09042 Cagliari, Italy; (V.O.); (S.L.); (M.L.); (M.S.); (F.C.); (E.F.)
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Edoardo Fiorillo
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Monserrato, 09042 Cagliari, Italy; (V.O.); (S.L.); (M.L.); (M.S.); (F.C.); (E.F.)
| |
Collapse
|
7
|
Yang JHM, Ward-Hartstonge KA, Perry DJ, Blanchfield JL, Posgai AL, Wiedeman AE, Diggins K, Rahman A, Tree TIM, Brusko TM, Levings MK, James EA, Kent SC, Speake C, Homann D, Long SA. Guidelines for standardizing T-cell cytometry assays to link biomarkers, mechanisms, and disease outcomes in type 1 diabetes. Eur J Immunol 2022; 52:372-388. [PMID: 35025103 PMCID: PMC9006584 DOI: 10.1002/eji.202049067] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 11/10/2021] [Accepted: 12/22/2021] [Indexed: 11/11/2022]
Abstract
Cytometric immunophenotyping is a powerful tool to discover and implement T-cell biomarkers of type 1 diabetes (T1D) progression and response to clinical therapy. Although many discovery-based T-cell biomarkers have been described, to date, no such markers have been widely adopted in standard practice. The heterogeneous nature of T1D and lack of standardized assays and experimental design across studies is a major barrier to the broader adoption of T-cell immunophenotyping assays. There is an unmet need to harmonize the design of immunophenotyping assays, including those that measure antigen-agnostic cell populations, such that data collected from different clinical trial sites and T1D cohorts are comparable, yet account for cohort-specific features and different drug mechanisms of action. In these Guidelines, we aim to provide expert advice on how to unify aspects of study design and practice. We provide recommendations for defining cohorts, method implementation, as well as tools for data analysis and reporting by highlighting and building on selected successes. Harmonization of cytometry-based T-cell assays will allow researchers to better integrate findings across trials, ultimately enabling the identification and validation of biomarkers of disease progression and treatment response in T1D.
Collapse
Affiliation(s)
- Jennie H. M. Yang
- Department of Immunobiology, Faculty of Life Sciences & Medicine, King’s College, London, UK
- National Institute of Health Research Biomedical Research Centre at Guy’s and St. Thomas’ National Health Service Foundation Trust, King’s College London, London, UK
| | - Kirsten A. Ward-Hartstonge
- Department of Surgery, University of British Columbia, Vancouver, California, USA
- BC Children’s Hospital Research Institute, Vancouver, California, USA
| | - Daniel J. Perry
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - J. Lori Blanchfield
- Center for Translational Research, Benaroya Research Institute, Seattle, Washington, USA
| | - Amanda L. Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Alice E. Wiedeman
- Center for Translational Research, Benaroya Research Institute, Seattle, Washington, USA
| | - Kirsten Diggins
- Center for Translational Research, Benaroya Research Institute, Seattle, Washington, USA
| | - Adeeb Rahman
- Human Immune Monitoring Center, Hess Center for Science and Medicine, Icahn School of Medicine, New York, New York, USA
| | - Timothy I. M. Tree
- Department of Immunobiology, Faculty of Life Sciences & Medicine, King’s College, London, UK
- National Institute of Health Research Biomedical Research Centre at Guy’s and St. Thomas’ National Health Service Foundation Trust, King’s College London, London, UK
| | - Todd M. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Megan K. Levings
- Department of Surgery, University of British Columbia, Vancouver, California, USA
- BC Children’s Hospital Research Institute, Vancouver, California, USA
- School of Biomedical Engineering, University of British Columbia, California, USA
| | - Eddie A. James
- Center for Translational Research, Benaroya Research Institute, Seattle, Washington, USA
| | - Sally C. Kent
- Diabetes Center of Excellence, University of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute, Seattle, Washington, USA
| | - Dirk Homann
- Precision Immunology Institute, Icahn School of Medicine, New York, New York, USA
- Diabetes, Obesity, & Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - S. Alice Long
- Center for Translational Research, Benaroya Research Institute, Seattle, Washington, USA
| | | |
Collapse
|
8
|
Shaikh A, Olkhanud PB, Gangaplara A, Kone A, Patel S, Gucek M, Fitzhugh CD. Thrombospondin-1, Platelet Factor 4, and Galectin-1 are Associated with Engraftment in Patients with Sickle Cell Disease Who Underwent Haploidentical HSCT. Transplant Cell Ther 2022; 28:249.e1-249.e13. [PMID: 35131485 PMCID: PMC9176382 DOI: 10.1016/j.jtct.2022.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 11/17/2022]
Abstract
Sickle cell disease (SCD) is an inherited red blood cell disorder that leads to significant morbidity and early mortality. The most widely available curative approach remains allogeneic hematopoietic stem cell transplantation (HSCT). HLA-haploidentical (haplo) HSCT expands the donor pool considerably and is a practical alternative for these patients, but traditionally with an increased risk of allograft rejection. Biomarkers in patient plasma could potentially help predict HSCT outcome and allow treatment at an early stage to reverse or prevent graft rejection. Reliable, noninvasive methods to predict engraftment or rejection early after HSCT are needed. We sought to detect variations in the plasma proteomes of patients who engrafted compared with those who rejected their grafts. We used a mass spectrometry-based proteomics approach to identify candidate biomarkers associated with engraftment and rejection by comparing plasma samples obtained from 9 engrafted patients and 10 patients who experienced graft rejection. A total of 1378 proteins were identified, 45 of which were differentially expressed in the engrafted group compared with the rejected group. Based on bioinformatics analysis results, information from the literature, and immunoassay availability, 7 proteins-thrombospondin-1 (Tsp-1), platelet factor 4 (Pf-4), talin-1, moesin, cell division control protein 42 homolog (CDC42), galectin-1 (Gal-1), and CD9-were selected for further analysis. We compared these protein concentrations among 35 plasma samples (engrafted, n = 9; rejected, n = 10; healthy volunteers, n = 8; nontransplanted SCD, n = 8). ELISA analysis confirmed the significant up-regulation of Tsp-1, Pf-4, and Gal-1 in plasma samples from engrafted patients compared with rejected patients, healthy African American volunteers, and the nontransplanted SCD group (P < .01). By receiver operating characteristic analysis, these 3 proteins distinguished engrafted patients from the other groups (area under the curve, >0.8; P < .05). We then evaluated the concentration of these 3 proteins in samples collected pre-HSCT and at days +30, +60, +100, and +180 post-HSCT. The results demonstrate that Tsp-1 and Pf-4 stratified engrafted patients as early as day 60 post-HSCT (P < .01), and that Gal-1 was significantly higher in engrafted patients as early as day 30 post-HSCT (P < .01). We also divided the rejected group into those who experienced primary (n = 5) and secondary graft rejection (n = 5) and found that engrafted patients had significantly higher Tsp-1 levels compared with patients who developed primary graft rejection at days +60 and +100 (P < .05), as well as higher Pf-4 levels compared with patients who developed primary graft rejection at post-transplantation (PT) day 100. Furthermore, Tsp-1 levels were significantly higher at PT days 60 and 100 and Pf-4 levels were higher at PT day 100 in engrafted patients compared with those who experienced secondary graft rejection. Increased concentrations of plasma Gal-1, Tsp-1, and Pf-4 could reflect increased T regulatory cells, IL-10, and TGF-β, which are essential players in the initiation of immunologic tolerance. These biomarkers may provide opportunities for preemptive intervention to minimize the incidence of graft rejection.
Collapse
Affiliation(s)
- Ahmad Shaikh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland; Department of Biology, The Catholic University of America, Washington, DC; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Purevdorj B Olkhanud
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Arunakumar Gangaplara
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Abdoul Kone
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Sajni Patel
- Proteomics Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Marjan Gucek
- Proteomics Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Courtney D Fitzhugh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
9
|
|
10
|
Vazquez J, Sheerar D, Stanic AK, Patankar MS. Multiparameter Flow Cytometry for Detailed Characterization of Peritoneal Immune Cells from Patients with Ovarian Cancer. Methods Mol Biol 2022; 2424:43-58. [PMID: 34918286 DOI: 10.1007/978-1-0716-1956-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Multiparameter flow cytometry is a convenient and efficient method for thorough phenotyping of cells, and especially immune cells from various tissues. We have successfully used multiparameter flow cytometry to characterize immune cells from patients with ovarian cancer and leveraged dimensionality reduction and machine learning for optimized visualization and analysis. Herein, we describe our optimized and established protocols for the labeling of cells with fluorophore-conjugated antibody panels, followed by details on data acquisition. Finally, we describe methods for analysis of the flow cytometry data using both FlowJo as well as R package, Cytofkit, for multidimensional data visualization.
Collapse
Affiliation(s)
- Jessica Vazquez
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| | - Dagna Sheerar
- University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Aleksandar K Stanic
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Manish S Patankar
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
11
|
Cuervo Florez M, Bruner J, Zarrinpar A. Progress and challenges in diagnosis and treatment of rejection following liver transplantation. Curr Opin Organ Transplant 2021; 26:669-674. [PMID: 34581291 DOI: 10.1097/mot.0000000000000924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Liver biopsy remains the most widely utilized method for diagnosis of allograft rejection following liver transplantation. However, associated risks and limitations present an opportunity for emerging noninvasive diagnostic techniques to improve upon the current standard of care. This review evaluates progress toward development of new noninvasive methods for the monitoring and diagnosing of allograft rejection. RECENT FINDINGS Recent studies investigate the potential of a variety of analytes. Quantification of dd-cfDNA and of DSA show potential to indicate status of allograft rejection and aid in immunosuppression modulation. Moreover, mRNA microarray profiling of differentially expressed genes, as well as characterization of cytokine responses and immunophenotypic shifts following liver transplantation, may predict and recognize rejection events. SUMMARY Noninvasive methods are not yet ready to replace liver biopsy as the standard of care for diagnosis of allograft rejection, though several assays and biomarkers have shown promising preliminary results. As noninvasive techniques become validated in clinical settings, their integration with current diagnostic methods is likely to foster increased sensitivity, specificity, and reliability of diagnosis.
Collapse
Affiliation(s)
- Mateo Cuervo Florez
- Department of Surgery, College of Medicine, University of Florida, Florida, USA
| | | | | |
Collapse
|
12
|
Baumgaertner P, Sankar M, Herrera F, Benedetti F, Barras D, Thierry AC, Dangaj D, Kandalaft LE, Coukos G, Xenarios I, Guex N, Harari A. Unsupervised Analysis of Flow Cytometry Data in a Clinical Setting Captures Cell Diversity and Allows Population Discovery. Front Immunol 2021; 12:633910. [PMID: 33995353 PMCID: PMC8119773 DOI: 10.3389/fimmu.2021.633910] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/12/2021] [Indexed: 11/13/2022] Open
Abstract
Data obtained with cytometry are increasingly complex and their interrogation impacts the type and quality of knowledge gained. Conventional supervised analyses are limited to pre-defined cell populations and do not exploit the full potential of data. Here, in the context of a clinical trial of cancer patients treated with radiotherapy, we performed longitudinal flow cytometry analyses to identify multiple distinct cell populations in circulating whole blood. We cross-compared the results from state-of-the-art recommended supervised analyses with results from MegaClust, a high-performance data-driven clustering algorithm allowing fast and robust identification of cell-type populations. Ten distinct cell populations were accurately identified by supervised analyses, including main T, B, dendritic cell (DC), natural killer (NK) and monocytes subsets. While all ten subsets were also identified with MegaClust, additional cell populations were revealed (e.g. CD4+HLA-DR+ and NKT-like subsets), and DC profiling was enriched by the assignment of additional subset-specific markers. Comparison between transcriptomic profiles of purified DC populations and publicly available datasets confirmed the accuracy of the unsupervised clustering algorithm and demonstrated its potential to identify rare and scarcely described cell subsets. Our observations show that data-driven analyses of cytometry data significantly enrich the amount and quality of knowledge gained, representing an important step in refining the characterization of immune responses.
Collapse
Affiliation(s)
- Petra Baumgaertner
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Martial Sankar
- Vital-IT, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Fernanda Herrera
- Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Fabrizio Benedetti
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - David Barras
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Anne-Christine Thierry
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Denarda Dangaj
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Lana E Kandalaft
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Ioannis Xenarios
- Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Nicolas Guex
- Vital-IT, Swiss Institute of Bioinformatics, Lausanne, Switzerland.,Bioinformatics Competence Center (BICC), University of Lausanne, Lausanne, Switzerland
| | - Alexandre Harari
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
13
|
Marfil-Garza BA, Hefler J, Bermudez De Leon M, Pawlick R, Dadheech N, Shapiro AMJ. Progress in Translational Regulatory T Cell Therapies for Type 1 Diabetes and Islet Transplantation. Endocr Rev 2021; 42:198-218. [PMID: 33247733 DOI: 10.1210/endrev/bnaa028] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Regulatory T cells (Tregs) have become highly relevant in the pathophysiology and treatment of autoimmune diseases, such as type 1 diabetes (T1D). As these cells are known to be defective in T1D, recent efforts have explored ex vivo and in vivo Treg expansion and enhancement as a means for restoring self-tolerance in this disease. Given their capacity to also modulate alloimmune responses, studies using Treg-based therapies have recently been undertaken in transplantation. Islet transplantation provides a unique opportunity to study the critical immunological crossroads between auto- and alloimmunity. This procedure has advanced greatly in recent years, and reports of complete abrogation of severe hypoglycemia and long-term insulin independence have become increasingly reported. It is clear that cellular transplantation has the potential to be a true cure in T1D, provided the remaining barriers of cell supply and abrogated need for immune suppression can be overcome. However, the role that Tregs play in islet transplantation remains to be defined. Herein, we synthesize the progress and current state of Treg-based therapies in T1D and islet transplantation. We provide an extensive, but concise, background to understand the physiology and function of these cells and discuss the clinical evidence supporting potency and potential Treg-based therapies in the context of T1D and islet transplantation. Finally, we discuss some areas of opportunity and potential research avenues to guide effective future clinical application. This review provides a basic framework of knowledge for clinicians and researchers involved in the care of patients with T1D and islet transplantation.
Collapse
Affiliation(s)
| | - Joshua Hefler
- Department of Surgery, University of Alberta, Edmonton, Canada
| | - Mario Bermudez De Leon
- Department of Molecular Biology, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey, Nuevo Leon, Mexico
| | - Rena Pawlick
- Department of Surgery, University of Alberta, Edmonton, Canada
| | | | - A M James Shapiro
- Department of Surgery, University of Alberta, Edmonton, Canada.,Clinical Islet Transplant Program, University of Alberta, Edmonton, Canada
| |
Collapse
|
14
|
An Overview of Flow Cytometry: Its Principles and Applications in Allergic Disease Research. Methods Mol Biol 2021; 2223:169-182. [PMID: 33226595 DOI: 10.1007/978-1-0716-1001-5_13] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Flow cytometry is a popular technique used for both clinical and research purposes. It involves laser-based technology to characterize cells based on size, shape, and complexity. Additionally, flow cytometers are equipped with the ability to take fluorescence measurements at multiple wavelengths. This capability makes the flow cytometer a practical resource in the utilization of fluorescently conjugated antibodies, fluorescent proteins, DNA binding dyes, viability dyes, and ion indicator dyes. As the technology advances, the number of parameters a flow cytometer can measure has increased tremendously, and now some has the capacity to analyze 30-50 or more parameters on a single cell. Here, we describe the basic principles involved in the mechanics and procedures of flow cytometry along with an insight into applications of flow cytometry techniques for biomedical and allergic disease research.
Collapse
|
15
|
El-Rashid M, Nguyen-Ngo D, Minhas N, Meijles DN, Li J, Ghimire K, Julovi S, Rogers NM. Repurposing of metformin and colchicine reveals differential modulation of acute and chronic kidney injury. Sci Rep 2020; 10:21968. [PMID: 33319836 PMCID: PMC7738483 DOI: 10.1038/s41598-020-78936-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury (AKI) is a major health problem affecting millions of patients globally. There is no effective treatment for AKI and new therapies are urgently needed. Novel drug development, testing and progression to clinical trials is overwhelmingly expensive. Drug repurposing is a more cost-effective measure. We identified 2 commonly used drugs (colchicine and metformin) that alter inflammatory cell function and signalling pathways characteristic of AKI, and tested them in models of acute and chronic kidney injury to assess therapeutic benefit. We assessed the renoprotective effects of colchicine or metformin in C57BL/6 mice challenged with renal ischemia reperfusion injury (IRI), treated before or after injury. All animals underwent analysis of renal function and biomolecular phenotyping at 24 h, 48 h and 4 weeks after injury. Murine renal tubular epithelial cells were studied in response to in vitro mimics of IRI. Pre-emptive treatment with colchicine or metformin protected against AKI, with lower serum creatinine, improved histological changes and decreased TUNEL staining. Pro-inflammatory cytokine profile and multiple markers of oxidative stress were not substantially different between groups. Metformin augmented expression of multiple autophagic proteins which was reversed by the addition of hydroxychloroquine. Colchicine led to an increase in inflammatory cells within the renal parenchyma. Chronic exposure after acute injury to either therapeutic agent in the context of reduced renal mass did not mitigate the development of fibrosis, with colchicine significantly worsening an ischemic phenotype. These data indicate that colchicine and metformin affect acute and chronic kidney injury differently. This has significant implications for potential drug repurposing, as baseline renal disease must be considered when selecting medication.
Collapse
Affiliation(s)
- Maryam El-Rashid
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Danny Nguyen-Ngo
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Nikita Minhas
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Jennifer Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Kedar Ghimire
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Sohel Julovi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia. .,Westmead Clinical Medical School, University of Sydney, Camperdown, NSW, Australia. .,Renal Division, Westmead Hospital, Sydney, NSW, Australia. .,Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
16
|
Petsiou A, Paschou SA, Vartholomatos G, Chatzigianni K, Kolaitis N, Giotaki E, Bondinas GP, Moustakas AK, Karamoutsios A, Zervou E, Tigas S, Tsatsoulis A, Papadopoulos GK. A modified flow cytometry method for objective estimation of human CD4 + regulatory T cells (CD4 + Tregs) in peripheral blood, via CD4/CD25/CD45RO/FoxP3 labeling. CYTOMETRY PART B-CLINICAL CYTOMETRY 2019; 98:259-269. [PMID: 31571372 DOI: 10.1002/cyto.b.21841] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/26/2019] [Accepted: 08/04/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Several methods exist for flow-cytometric estimation of human peripheral blood CD4+ T regulatory cells (CD4+ Tregs). METHODS We report our experience with the estimation of human CD4+ Tregs via three different characterizations using flow cytometry (CD25high FoxP3+ , CD25high CD127low/- FoxP3+ , and CD4+ CD25high/int CD45ROFoxP3+ ) in normal subjects. We have used these methods on the control populations from two studies (32 and 36 subjects, respectively), the latter two methods retrospectively on the subjects of the first study. The six CD4+ T cell fractions obtained by the third method were differentially colored to ascertain the distribution of these cell fractions in the CD25/FoxP3, CD45RO/FoxP3, and CD25/CD127 dot plots from CD4/CD25/CD45RO/FoxP3 and CD4/CD25/CD45RO/CD127 panels. RESULTS Each approach gives significantly different estimates of Tregs (expressed as percentage of CD4+ T cells), with the second almost invariably yielding higher percentages than the other two. Only the third approach can distinguish among effector and naïve Tregs and FoxP3+ non-Tregs. Analysis of CD25/CD127 dot plots reveals that Treg delineation via the widely used definition of CD4+ CD25high CD127low/- cells unavoidably yields a mixture of nearly all effector and most of naïve Tregs, as well as FoxP3+ non-Tregs plus other cells. Delineation of effector/naïve Tregs and FoxP3+ non-Tregs is possible via CD45RO/CD25 dot plots but not by CD45RO/FoxP3 counterparts (as done previously) because of overlapping FoxP3 intensities among Tregs and non-Tregs. CONCLUSION Our comparison shows that CD4/CD25/CD45RO/FoxP3 panels are an objective means of estimating effector and naïve Tregs via colored dot plots, aiding thus in Treg delineation in health and detecting aberrations in disease.
Collapse
Affiliation(s)
- Asimina Petsiou
- Unit of Molecular Biology, Laboratory of Hematology, University Hospital of Ioannina, Ioannina, Greece
| | - Stavroula A Paschou
- Department of Endocrinology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Georgios Vartholomatos
- Unit of Molecular Biology, Laboratory of Hematology, University Hospital of Ioannina, Ioannina, Greece
| | - Katerina Chatzigianni
- Unit of Molecular Biology, Laboratory of Hematology, University Hospital of Ioannina, Ioannina, Greece
| | - Nikolaos Kolaitis
- Laboratory of Hematology, University Hospital of Ioannina, Ioannina, Greece
| | - Eleni Giotaki
- Department of Nursing, Technological Educational Institute of Epirus, Ioannina, Greece
| | - George P Bondinas
- Laboratory of Biophysics, Biochemistry, Bioprocessing and Bioproducts, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece
| | - Antonis K Moustakas
- Department of Food Science and Technology, Technological Educational Institute of Ionian Islands, Argostoli, Greece
| | - Achilleas Karamoutsios
- Laboratory of Animal Health-Food Hygiene and Quality, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece
| | - Eleftheria Zervou
- Department of Bloodbank, University Hospital of Ioannina, Ioannina, Greece
| | - Stelios Tigas
- Department of Endocrinology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Agathocles Tsatsoulis
- Department of Endocrinology, University of Ioannina School of Medicine, Ioannina, Greece
| | - George K Papadopoulos
- Laboratory of Biophysics, Biochemistry, Bioprocessing and Bioproducts, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece
| |
Collapse
|