1
|
Thitilertdecha P, Poungpairoj P, Tantithavorn V, Ammaranond P, Onlamoon N. Determination of cell expansion and surface molecule expression on anti-CD3/28 expanded CD4 + T cells. Scand J Immunol 2019; 90:e12808. [PMID: 31322752 DOI: 10.1111/sji.12808] [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: 10/31/2018] [Revised: 07/03/2019] [Accepted: 07/15/2019] [Indexed: 11/30/2022]
Abstract
CD4+ T cell immunotherapy has potential for treatment in HIV-infected patients. A large number of expanded CD4+ T cells and confirmation of functional-related phenotypes are required for ensuring the successful outcomes of treatment. Freshly isolated CD4+ T cells from healthy donors were activated with anti-CD3/28-coated magnetic beads at different bead-to-cell ratios and cultured in the absence and presence of IL-2 supplementation for 3 weeks. Fold expansion, cell viability, growth kinetic and lymphocyte subset identities were determined. Data demonstrated that a 1:1 bead-to-cell ratio rendered the highest expansion of 1044-fold with 88% viability and 99.5% purity followed by the 2:1 and 0.5:1 ratios. No significant difference in proliferation and phenotypes was found between non-IL-2 and IL-2 supplementation groups. Several specific surface molecule expressions of the expanded cells including chemokine receptors, adhesion molecules, co-stimulatory molecules, activation molecules, maturation markers, cytokine receptors and other molecules were altered when compared to the unexpanded cells. This optimized expansion protocol using the 1:1 bead-to-cell ratio of anti-CD3/28-coated magnetic beads and culture condition without IL-2 supplementation provided the satisfactory yield with good reproducibility. Specific surface molecule expressions of the expanded cells presented potential roles in proliferation, differentiation, homeostasis, apoptosis and organ homing.
Collapse
Affiliation(s)
- Premrutai Thitilertdecha
- Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkoknoi, Bangkok, Thailand.,Biomedical Research Incubator Unit, Research Group and Research Network Division, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkoknoi, Bangkok, Thailand
| | - Poonsin Poungpairoj
- Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkoknoi, Bangkok, Thailand.,Biomedical Research Incubator Unit, Research Group and Research Network Division, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkoknoi, Bangkok, Thailand
| | - Varangkana Tantithavorn
- Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkoknoi, Bangkok, Thailand.,Biomedical Research Incubator Unit, Research Group and Research Network Division, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkoknoi, Bangkok, Thailand
| | - Palanee Ammaranond
- Department of Transfusion Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| | - Nattawat Onlamoon
- Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkoknoi, Bangkok, Thailand.,Biomedical Research Incubator Unit, Research Group and Research Network Division, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkoknoi, Bangkok, Thailand
| |
Collapse
|
2
|
Thitilertdecha P, Suwannachod P, Poungpairoj P, Tantithavorn V, Khowawisetsut L, Ammaranond P, Onlamoon N. A closed-culture system using a GMP-grade culture bag and anti-CD3/28 coated bead stimulation for CD4 + T cell expansion from healthy and HIV-infected donors. J Immunol Methods 2018; 460:17-25. [PMID: 29894747 DOI: 10.1016/j.jim.2018.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 11/29/2022]
Abstract
CD4 immunotherapy is potentially useful in immune reconstitution of CD4+ T cells for HIV-infected patients. Transfusion of anti-CD3/28 expanded CD4+ T cells is also proved to be safe and effective in both SIV-infected macaques and HIV-infected patients. However, there is no such standardized and practical protocol available for cell production in order to use in clinics. This study thus aimed to develop a closed-culture system for in vitro CD4+ T lymphocyte expansion by using a commercially available GMP-grade culture bag and anti-CD3/28 activation. Freshly isolated CD4+ T cells by immunorosette formation from healthy donors and cryopreserved CD4+ T cells from HIV-infected patients with CD4 count over 500 cells/μL were stimulated with anti-CD3/28 coated beads. The activated cells were then expanded in conventional culture flasks and GMP-grade culture bags for three weeks. Fold expansion, cell viability, growth kinetic and phenotypic characters were observed. Results revealed that purified CD4+ T cells from healthy individuals cultured in flasks showed better expansion than those cultured in bags (797-fold and 331-fold, respectively), whereas, their cell viability, growth kinetic and expanded CD4+ T cell purity were almost similar. A large-scale production was also conducted and supported consistency of cell proliferation in the closed-culture system. Frozen CD4+ T lymphocytes from the patients were able to remain their growth function and well expanded with a good yield of 415-fold, 85% viability and 96% purity of CD4+ T cells at the end of a 3-week culture in bags. This developed closed-culture system using culture bags and anti-CD3/28 coated beads, therefore, can achieve a large number of expanded CD4+ T lymphocytes with good reproducibility, suggesting a promising protocol required for adoptive immunotherapy.
Collapse
Affiliation(s)
- Premrutai Thitilertdecha
- Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Biomedical Research Incubator Unit, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pornpichaya Suwannachod
- Graduate program in Immunology, Department of Immunulogy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Poonsin Poungpairoj
- Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Biomedical Research Incubator Unit, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Varangkana Tantithavorn
- Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Biomedical Research Incubator Unit, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ladawan Khowawisetsut
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Palanee Ammaranond
- Department of Transfusion Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Nattawat Onlamoon
- Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Biomedical Research Incubator Unit, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
3
|
Extensive Proliferation of Cd4+ Lymphocyte by Both Phytohaemagglutinin A and Anti-Cd2/Cd3/ Cd28 Macsibeads. ACTA MEDICA BULGARICA 2018. [DOI: 10.2478/amb-2018-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Background: Lymphocytes proliferate considerably following appropriate stimulation in vitro. Autologous T cells are obtained from whole blood or tissue sites in relatively limited amounts. We need a method to expand these cells efficiently, study their functions and manipulate them to create appropriate cells for transferring to the patient with infection and cancer. Objectives: The aim of this study is to determine proliferation ability of two different stimulators on CD4+ lymphocytes. Methods: Lymphocytes were isolated from blood samples of healthy donors after removing adherent cells (monocytes).The efficacy of MACSiBead™ coated with anti-CD2, anti-CD3, anti-CD28 (anti-CD2/CD3/CD28) was compared with Phytohaemagglutinin A (PHA) on CD4+ lymphocytes proliferation using carboxyfluorescein diacetate succinimidyl ester (CFSE) in cell culture media. The percentage of proliferating cells was analyzed using flow cytometry. Results: Both stimulators induced extensive proliferation of CD4+ lymphocytes but proliferation ability of PHA was higher compared to stimulation by anti-CD2/CD3/CD28 MACSiBead™. The proliferation rate of cells stimulated by PHA was 93.8% ± 3.37% whereas it was 85.2% ± 4.7% in cells stimulated by anti-CD2/CD3/CD28 MACSiBead™. Conclusions: Our results show that MACSiBead™ along with PHA can be used to obtain a large number of expanded CD4+ lymphocytes.
Collapse
|
4
|
Hou H, Zhou Y, Yu J, Mao L, Bosco MJ, Wang J, Lu Y, Mao L, Wu X, Wang F, Sun Z. Establishment of the Reference Intervals of Lymphocyte Function in Healthy Adults Based on IFN-γ Secretion Assay upon Phorbol-12-Myristate-13-Acetate/Ionomycin Stimulation. Front Immunol 2018; 9:172. [PMID: 29467761 PMCID: PMC5808316 DOI: 10.3389/fimmu.2018.00172] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/19/2018] [Indexed: 11/13/2022] Open
Abstract
The function of lymphocytes is the key to reflect the immune status of hosts. Evaluation of lymphocyte function is a useful tool to monitor the effect of immunosuppressive treatment and predict the prognosis of immune-mediated diseases (e.g., cancer, autoimmune diseases, and infectious diseases). As the lymphocytes have various activities, such as activation, cytotoxicity, and cytokine secretion, it is a challenge to evaluate the function of lymphocytes in clinical practice and the reference intervals (RIs) of lymphocyte function are rarely reported. The present study showed that the secretion of IFN-γ was well correlated with the activation, chemotaxis, and cytotoxicity of CD4+, CD8+ T cells, and NK cells, which suggests that IFN-γ production can be used as a symbol of lymphocyte function. We therefore created a simple method to detect the function of CD4+, CD8+ T cells, and NK cells simultaneously according to IFN-γ secretion by using whole blood instead of peripheral blood mononuclear cells. We further established the RIs of lymphocyte function (CD4+ T cells: 15.31-34.98%; CD8+ T cells: 26.11-66.59%; NK cells: 39.43-70.79%) in healthy adults. This method showed good reproducibility for the evaluation of lymphocyte function. The established RIs were suitable for use in other centers based on the validation data. We also validated the RIs in individuals with different immune status, and the results showed that kidney transplant recipients and infants (0-1 year) had a decreased lymphocyte function, whereas T cells in systemic lupus erythematosus patients exhibited an opposite trend. Overall, we have successfully established the RIs of lymphocyte function in healthy adults in a simple way, which might be of important clinical value in the diagnosis, monitoring, and prognosis of immune-related diseases.
Collapse
Affiliation(s)
- Hongyan Hou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Zhou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lie Mao
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Munyemana Jean Bosco
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanfang Lu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liyan Mao
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohui Wu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyong Sun
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Khamaikawin W, Saoin S, Nangola S, Chupradit K, Sakkhachornphop S, Hadpech S, Onlamoon N, Ansari AA, Byrareddy SN, Boulanger P, Hong SS, Torbett BE, Tayapiwatana C. Combined Antiviral Therapy Using Designed Molecular Scaffolds Targeting Two Distinct Viral Functions, HIV-1 Genome Integration and Capsid Assembly. MOLECULAR THERAPY-NUCLEIC ACIDS 2015; 4:e249. [PMID: 26305555 PMCID: PMC4560793 DOI: 10.1038/mtna.2015.22] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 07/14/2015] [Indexed: 01/06/2023]
Abstract
Designed molecular scaffolds have been proposed as alternative therapeutic agents against HIV-1. The ankyrin repeat protein (Ank(GAG)1D4) and the zinc finger protein (2LTRZFP) have recently been characterized as intracellular antivirals, but these molecules, used individually, do not completely block HIV-1 replication and propagation. The capsid-binder Ank(GAG)1D4, which inhibits HIV-1 assembly, does not prevent the genome integration of newly incoming viruses. 2LTRZFP, designed to target the 2-LTR-circle junction of HIV-1 cDNA and block HIV-1 integration, would have no antiviral effect on HIV-1-infected cells. However, simultaneous expression of these two molecules should combine the advantage of preventive and curative treatments. To test this hypothesis, the genes encoding the N-myristoylated Myr(+)Ank(GAG)1D4 protein and the 2LTRZFP were introduced into human T-cells, using a third-generation lentiviral vector. SupT1 cells stably expressing 2LTRZFP alone or with Myr(+)Ank(GAG)1D4 showed a complete resistance to HIV-1 in viral challenge. Administration of the Myr(+)Ank(GAG)1D4 vector to HIV-1-preinfected SupT1 cells resulted in a significant antiviral effect. Resistance to viral infection was also observed in primary human CD4+ T-cells stably expressing Myr(+)Ank(GAG)1D4, and challenged with HIV-1, SIVmac, or SHIV. Our data suggest that our two anti-HIV-1 molecular scaffold prototypes are promising antiviral agents for anti-HIV-1 gene therapy.
Collapse
Affiliation(s)
- Wannisa Khamaikawin
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Somphot Saoin
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Sawitree Nangola
- Division of Clinical Immunology and Transfusion Sciences, School of Allied Health Sciences, University of Phayao, Phayao, Thailand
| | - Koollawat Chupradit
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | | | - Sudarat Hadpech
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Nattawat Onlamoon
- Division of Instruments for Research, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Aftab A Ansari
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Siddappa N Byrareddy
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Pierre Boulanger
- University Lyon 1 & INRA UMR-754, Retrovirus and Comparative Pathology, Lyon, France
| | - Saw-See Hong
- University Lyon 1 & INRA UMR-754, Retrovirus and Comparative Pathology, Lyon, France
| | - Bruce E Torbett
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Chatchai Tayapiwatana
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
6
|
Onlamoon N, Petphong V, Sukapirom K, Wang S, Ammaranond P, Pattanapanyasat K. Production of anti-CD3/28 expanded CD4⁺ T lymphocytes from HIV-infected patients with different degrees of disease progression. Immunotherapy 2015; 7:765-75. [PMID: 26250407 DOI: 10.2217/imt.15.41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS CD4+ T lymphocytes from HIV-infected patients with different degrees of disease progression based on CD4 count were expanded in vitro using anti-CD3/28-coated beads. MATERIALS & METHODS Purified CD4+ T lymphocytes from healthy subjects and patients were expanded for 3 weeks. Moreover, the improvement of cell expansion by IL-2 supplementation was also determined. RESULTS Expanded CD4+ T lymphocytes from patients had lower fold expansion when compared with healthy subjects. Furthermore, patients with high CD4 counts had higher fold expansion than patients with low CD4 count, and IL-2 supplementation further increased cell expansion. CONCLUSIONS Anti-CD3/28 activation failed to potently induce expansion of CD4+ T lymphocytes from patients. However, the cell expansion could be improved by IL-2 supplementation.
Collapse
Affiliation(s)
- Nattawat Onlamoon
- Department of Research & Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.,Center for Emerging & Neglected Infectious Diseases, Mahidol University, Nakhon Pathom, Thailand
| | - Vajee Petphong
- Department of Research & Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.,Master of Science Program in Immunology, Department of Immunology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kasama Sukapirom
- Department of Research & Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Siyu Wang
- Department of Research & Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.,Master of Science Program in Immunology, Department of Immunology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Palanee Ammaranond
- Department of Transfusion Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Kovit Pattanapanyasat
- Department of Research & Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.,Center for Emerging & Neglected Infectious Diseases, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
7
|
Hong JJ, Villinger F, Courtney CL. PAS-positive extracellular deposits within germinal centers of hyperplastic follicles during SIV infection in a rhesus macaque. J Med Primatol 2014; 43:374-7. [PMID: 24628065 DOI: 10.1111/jmp.12109] [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] [Accepted: 02/20/2014] [Indexed: 01/26/2023]
Abstract
BACKGROUND Lymphoid tissue remodeling is characteristic of chronic simian immunodeficiency virus infection. METHODS A rhesus macaque infected with SIVmac239 was necropsied and its lymphoid tissues subjected to histopathology characterization. RESULTS Germinal centers in spleen and lymph nodes contained PAS-positive, non-amyloid extracellular deposits, decreased T follicular helper cells, and normal density of Ki67(+) B cells. CONCLUSIONS A possible mechanism for PAS-positive deposits includes exaggerated involution of SIV-induced follicular hyperplasia secondary to virus-associated immune reaction.
Collapse
Affiliation(s)
- Jung Joo Hong
- Division of Pathology, Yerkes National Primate Research Center, Atlanta, GA, USA
| | | | | |
Collapse
|
8
|
Chono H, Saito N, Tsuda H, Shibata H, Ageyama N, Terao K, Yasutomi Y, Mineno J, Kato I. In vivo safety and persistence of endoribonuclease gene-transduced CD4+ T cells in cynomolgus macaques for HIV-1 gene therapy model. PLoS One 2011; 6:e23585. [PMID: 21858176 PMCID: PMC3157387 DOI: 10.1371/journal.pone.0023585] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 07/20/2011] [Indexed: 02/03/2023] Open
Abstract
Background MazF is an endoribonuclease encoded by Escherichia coli that specifically cleaves the ACA sequence of mRNA. In our previous report, conditional expression of MazF in the HIV-1 LTR rendered CD4+ T lymphocytes resistant to HIV-1 replication. In this study, we examined the in vivo safety and persistence of MazF-transduced cynomolgus macaque CD4+ T cells infused into autologous monkeys. Methodology/Principal Findings The in vivo persistence of the gene-modified CD4+ T cells in the peripheral blood was monitored for more than half a year using quantitative real-time PCR and flow cytometry, followed by experimental autopsy in order to examine the safety and distribution pattern of the infused cells in several organs. Although the levels of the MazF-transduced CD4+ T cells gradually decreased in the peripheral blood, they were clearly detected throughout the experimental period. Moreover, the infused cells were detected in the distal lymphoid tissues, such as several lymph nodes and the spleen. Histopathological analyses of tissues revealed that there were no lesions related to the infused gene modified cells. Antibodies against MazF were not detected. These data suggest the safety and the low immunogenicity of MazF-transduced CD4+ T cells. Finally, gene modified cells harvested from the monkey more than half a year post-infusion suppressed the replication of SHIV 89.6P. Conclusions/Significance The long-term persistence, safety and continuous HIV replication resistance of the mazF gene-modified CD4+ T cells in the non-human primate model suggests that autologous transplantation of mazF gene-modified cells is an attractive strategy for HIV gene therapy.
Collapse
Affiliation(s)
- Hideto Chono
- Center for Cell and Gene Therapy, Takara Bio Inc, Otsu, Shiga, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Wilen CB, Wang J, Tilton JC, Miller JC, Kim KA, Rebar EJ, Sherrill-Mix SA, Patro SC, Secreto AJ, Jordan APO, Lee G, Kahn J, Aye PP, Bunnell BA, Lackner AA, Hoxie JA, Danet-Desnoyers GA, Bushman FD, Riley JL, Gregory PD, June CH, Holmes MC, Doms RW. Engineering HIV-resistant human CD4+ T cells with CXCR4-specific zinc-finger nucleases. PLoS Pathog 2011; 7:e1002020. [PMID: 21533216 PMCID: PMC3077364 DOI: 10.1371/journal.ppat.1002020] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 02/18/2011] [Indexed: 11/18/2022] Open
Abstract
HIV-1 entry requires the cell surface expression of CD4 and either the CCR5 or CXCR4 coreceptors on host cells. Individuals homozygous for the ccr5Δ32 polymorphism do not express CCR5 and are protected from infection by CCR5-tropic (R5) virus strains. As an approach to inactivating CCR5, we introduced CCR5-specific zinc-finger nucleases into human CD4+ T cells prior to adoptive transfer, but the need to protect cells from virus strains that use CXCR4 (X4) in place of or in addition to CCR5 (R5X4) remains. Here we describe engineering a pair of zinc finger nucleases that, when introduced into human T cells, efficiently disrupt cxcr4 by cleavage and error-prone non-homologous DNA end-joining. The resulting cells proliferated normally and were resistant to infection by X4-tropic HIV-1 strains. CXCR4 could also be inactivated in ccr5Δ32 CD4+ T cells, and we show that such cells were resistant to all strains of HIV-1 tested. Loss of CXCR4 also provided protection from X4 HIV-1 in a humanized mouse model, though this protection was lost over time due to the emergence of R5-tropic viral mutants. These data suggest that CXCR4-specific ZFNs may prove useful in establishing resistance to CXCR4-tropic HIV for autologous transplant in HIV-infected individuals.
Collapse
Affiliation(s)
- Craig B. Wilen
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jianbin Wang
- Sangamo BioSciences, Richmond, California, United States of America
| | - John C. Tilton
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | | | - Kenneth A. Kim
- Sangamo BioSciences, Richmond, California, United States of America
| | - Edward J. Rebar
- Sangamo BioSciences, Richmond, California, United States of America
| | - Scott A. Sherrill-Mix
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Sean C. Patro
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Anthony J. Secreto
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Andrea P. O. Jordan
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Gary Lee
- Sangamo BioSciences, Richmond, California, United States of America
| | - Joshua Kahn
- Sangamo BioSciences, Richmond, California, United States of America
| | - Pyone P. Aye
- Divisions of Regenerative Medicine and Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
| | - Bruce A. Bunnell
- Divisions of Regenerative Medicine and Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
| | - Andrew A. Lackner
- Divisions of Regenerative Medicine and Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
| | - James A. Hoxie
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Gwenn A. Danet-Desnoyers
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Frederic D. Bushman
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - James L. Riley
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | | | - Carl H. June
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | | | - Robert W. Doms
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
10
|
Abstract
Among the microorganisms that cause diseases of medical or veterinary importance, the only group that is entirely dependent on the host, and hence not easily amenable to therapy via pharmaceuticals, is the viruses. Since viruses are obligate intracellular pathogens, and therefore depend a great deal on cellular processes, direct therapy of viral infections is difficult. Thus, modifying or targeting nonspecific or specific immune responses is an important aspect of intervention of ongoing viral infections. However, as a result of the unavailability of effective vaccines and the extended duration of manifestation, chronic viral infections are the most suitable for immunotherapies. We present an overview of various immunological strategies that have been applied for treating viral infections after exposure to the infectious agent.
Collapse
Affiliation(s)
- Nagendra R Hegde
- Bharat Biotech Foundation, Genome Valley, Turkapally, Shameerpet Mandal, Hyderabad 500078, India.
| | | | | | | |
Collapse
|
11
|
Shedlock DJ, Talbott KT, Morrow MP, Ferraro B, Hokey DA, Muthumani K, Weiner DB. Ki-67 staining for determination of rhesus macaque T cell proliferative responses ex vivo. Cytometry A 2010; 77:275-84. [PMID: 20104580 DOI: 10.1002/cyto.a.20857] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The capacity for robust proliferation upon re-infection is a hallmark of adaptive immunity and the basis of vaccination. A widely used animal model for the study of human disease is the rhesus macaque (RM), where capacity for proliferation can be assessed ex vivo using carboxyfluorescein succinimidyl ester (CFSE)-based dilution assays. However, we show over the course of the standard ex vivo proliferation assay that CFSE-labeling at commonly used dye concentrations induces significant cell death, but that this phenomenon is dose-dependent. Here, we describe an alternative semiquantitative method for estimating T cell proliferative responses that avoids the putative biases associated with chemical modification. RM peripheral blood mononuclear cells were stimulated ex vivo with cognate peptides for 5 days, immunostained for intracellular Ki-67, and then analyzed by flow cytometry. We describe a gating strategy using Ki-67 and side light scatter, also a marker of blastogenesis, which correlates strongly with data from CFSE dilution. We show that this method is a valid tool for measuring RM antigen-specific cellular proliferation ex vivo and can be used as an alternative to CFSE dilution assays.
Collapse
Affiliation(s)
- Devon J Shedlock
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Muñoz NM, Trobridge GD, Kiem HP. Ex vivo expansion and lentiviral transduction of Macaca nemestrina CD4+ T cells. J Med Primatol 2009; 38:438-43. [PMID: 19793180 DOI: 10.1111/j.1600-0684.2009.00383.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Macaca nemestrina is a nonhuman primate used as a model in preclinical studies of hematopoietic stem cell transplantation and adoptive transfer of T cells. Adoptive T cell transfer studies typically require ex vivo expansion of substantial numbers of T cells prior to their reinfusion into the subject. METHODS Pigtailed macaque peripheral blood CD4(+) cells were expanded using CD3 and CD28 antibody-coated beads. These cells were transformed using Herpesvirus saimiri and were also transduced with HIV-1 based lentiviral vectors. RESULTS We report an efficient method for the ex vivo expansion of CD4(+) T cells from Macaca nemestrina peripheral blood. With this protocol, primary CD4(+) T cells can be expanded between 300- to 6000-fold during 24-day period and can be efficiently transduced with lentiviral vectors. Furthermore, these T cells can be transformed by Herpesvirus saimiri and maintained in culture for several months. The transformed T cell lines can be productively infected with the simian immunodeficiency virus (SIV) strain SIV(mac239). CONCLUSIONS We have established methods for the expansion and transformation of primary M. nemestrina CD4(+) T cells and demonstrated the utility of these methods for several applications.
Collapse
Affiliation(s)
- Nina M Muñoz
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | |
Collapse
|
13
|
Greene JM, Burwitz BJ, Blasky AJ, Mattila TL, Hong JJ, Rakasz EG, Wiseman RW, Hasenkrug KJ, Skinner PJ, O'Connor SL, O'Connor DH. Allogeneic lymphocytes persist and traffic in feral MHC-matched mauritian cynomolgus macaques. PLoS One 2008; 3:e2384. [PMID: 18545705 PMCID: PMC2408966 DOI: 10.1371/journal.pone.0002384] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Accepted: 05/05/2008] [Indexed: 12/27/2022] Open
Abstract
Background Thus far, live attenuated SIV has been the most successful method for vaccinating macaques against pathogenic SIV challenge; however, it is not clear what mechanisms are responsible for this protection. Adoptive transfer studies in mice have been integral to understanding live attenuated vaccine protection in models like Friend virus. Previous adoptive transfers in primates have failed as transferred cells are typically cleared within hours after transfer. Methodology/ Principal Findings Here we describe adoptive transfer studies in Mauritian origin cynomolgus macaques (MCM), a non-human primate model with limited MHC diversity. Cells transferred between unrelated MHC-matched macaques persist for at least fourteen days but are rejected within 36 hours in MHC-mismatched macaques. Cells trafficked from the blood to peripheral lymphoid tissues within 12 hours of transfer. Conclusions/Significance MHC-matched MCM provide the first viable primate model for adoptive transfer studies. Because macaques infected with SIV are the best model for HIV/AIDS pathogenesis, we can now directly study the correlates of protective immune responses to AIDS viruses. For example, plasma viral loads following pathogenic SIV challenge are reduced by several orders of magnitude in macaques previously immunized with attenuated SIV. Adoptive transfer of lymphocyte subpopulations from vaccinated donors into SIV-naïve animals may define the immune mechanisms responsible for protection and guide future vaccine development.
Collapse
Affiliation(s)
- Justin M. Greene
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, United States of America
| | - Benjamin J. Burwitz
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, United States of America
| | - Alex J. Blasky
- Wisconsin National Primate Research Center, University of Wisconsin—Madison, Madison, Wisconsin, United States of America
| | - Teresa L. Mattila
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Jung Joo Hong
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin—Madison, Madison, Wisconsin, United States of America
| | - Roger W. Wiseman
- Wisconsin National Primate Research Center, University of Wisconsin—Madison, Madison, Wisconsin, United States of America
| | - Kim J. Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Shelby L. O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, United States of America
| | - David H. O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, University of Wisconsin—Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|