1
|
Yang J, Zhang N, Luo T, Yang M, Shen W, Tan Z, Xia Y, Zhang L, Zhou X, Lei Q, Guo A. TCellSI: A novel method for T cell state assessment and its applications in immune environment prediction. IMETA 2024; 3:e231. [PMID: 39429885 PMCID: PMC11487559 DOI: 10.1002/imt2.231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 10/22/2024]
Abstract
T cell is an indispensable component of the immune system and its multifaceted functions are shaped by the distinct T cell types and their various states. Although multiple computational models exist for predicting the abundance of diverse T cell types, tools for assessing their states to characterize their degree of resting, activation, and suppression are lacking. To address this gap, a robust and nuanced scoring tool called T cell state identifier (TCellSI) leveraging Mann-Whitney U statistics is established. The TCellSI methodology enables the evaluation of eight distinct T cell states-Quiescence, Regulating, Proliferation, Helper, Cytotoxicity, Progenitor exhaustion, Terminal exhaustion, and Senescence-from transcriptome data, providing T cell state scores (TCSS) for samples through specific marker gene sets and a compiled reference spectrum. Validated against sizeable pseudo-bulk and actual bulk RNA-seq data across a range of T cell types, TCellSI not only accurately characterizes T cell states but also surpasses existing well-discovered signatures in reflecting the nature of T cells. Significantly, the tool demonstrates predictive value in the immune environment, correlating T cell states with patient prognosis and responses to immunotherapy. For better utilization, the TCellSI is readily accessible through user-friendly R package and web server (https://guolab.wchscu.cn/TCellSI/). By offering insights into personalized cancer therapies, TCellSI has the potential to improve treatment outcomes and efficacy.
Collapse
Affiliation(s)
- Jing‐Min Yang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
- Department of Thoracic SurgeryWest China Biomedical Big Data Center, West China Hospital, Sichuan UniversityChengduChina
| | - Nan Zhang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
- Department of Thoracic SurgeryWest China Biomedical Big Data Center, West China Hospital, Sichuan UniversityChengduChina
| | - Tao Luo
- BGI Education CenterUniversity of Chinese Academy of SciencesShenzhenChina
| | - Mei Yang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Wen‐Kang Shen
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Zhen‐Lin Tan
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Yun Xia
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Libin Zhang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, School of Biomedical InformaticsThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Qian Lei
- Department of Thoracic SurgeryWest China Biomedical Big Data Center, West China Hospital, Sichuan UniversityChengduChina
| | - An‐Yuan Guo
- Department of Thoracic SurgeryWest China Biomedical Big Data Center, West China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
2
|
Chen S, Nguyen TD, Lee KZ, Liu D. Ex vivo T cell differentiation in adoptive immunotherapy manufacturing: Critical process parameters and analytical technologies. Biotechnol Adv 2024; 77:108434. [PMID: 39168355 DOI: 10.1016/j.biotechadv.2024.108434] [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: 11/09/2023] [Revised: 08/01/2024] [Accepted: 08/17/2024] [Indexed: 08/23/2024]
Abstract
Adoptive immunotherapy shows great promise as a treatment for cancer and other diseases. Recent evidence suggests that the therapeutic efficacy of these cell-based therapies can be enhanced by the enrichment of less-differentiated T cell subpopulations in the therapeutic product, giving rise to a need for advanced manufacturing technologies capable of enriching these subpopulations through regulation of T cell differentiation. Studies have shown that modifying certain critical process control parameters, such as cytokines, metabolites, amino acids, and culture environment, can effectively manipulate T cell differentiation in ex vivo cultures. Advanced process analytical technologies (PATs) are crucial for monitoring these parameters and the assessment of T cell differentiation during culture. In this review, we examine such critical process parameters and PATs, with an emphasis on their impact on enriching less-differentiated T cell population. We also discuss the limitations of current technologies and advocate for further efforts from the community to establish more stringent critical process parameters (CPPs) and develop more at-line/online PATs that are specific to T cell differentiation. These advancements will be essential to enable the manufacturing of more efficacious adoptive immunotherapy products.
Collapse
Affiliation(s)
- Sixun Chen
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, 20 Biopolis Way, 138668, Singapore
| | - Tan Dai Nguyen
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, 20 Biopolis Way, 138668, Singapore
| | - Kang-Zheng Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, 20 Biopolis Way, 138668, Singapore
| | - Dan Liu
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, 20 Biopolis Way, 138668, Singapore.
| |
Collapse
|
3
|
Ioannou M, Simon MS, Borkent J, Wijkhuijs A, Berghmans R, Haarman BC, Drexhage HA. Higher T central and lower effector memory cells in bipolar disorder: A differentiation abnormality? Brain Behav Immun Health 2024; 38:100764. [PMID: 38600952 PMCID: PMC11004065 DOI: 10.1016/j.bbih.2024.100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/04/2024] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
The aim of this study was to elucidate the nature of T cell abnormalities in bipolar disorder (BD). With the use of multicolor flow cytometry, we first quantified the composition of the different memory and pro-inflammatory immune subpopulations in samples of 58 patients with BD and compared them to 113 healthy controls. Second, to assess if cytomegalovirus infection was related to the resulted immune subpopulation compositions in the two groups, we measured cytomegalovirus-specific antibodies in serum. Thirdly, we assessed differences between the two groups in the serum levels of the immune cell differentiation factor interleukin-7. Compared to healthy controls, patients showed significantly higher T helper-17, T regulatory and T central memory cells (CD4+ and CD8+). Besides, patients showed significantly lower CD4+ T effector memory and CD4+ T effector memory re-expressing RA cells. Cytomegalovirus infection was not related to the observed abnormalities, with the exception of T helper-17 cells. This immune subpopulation was significantly higher only in patients seropositive to cytomegalovirus infection. Finally, interleukin-7 levels were significantly lower in BD compared to healthy controls. In conclusion, the aberrant levels of T memory cell populations in BD may suggest a T cell differentiation abnormality. The role of interleukin-7 in this putative abnormality should be further investigated.
Collapse
Affiliation(s)
- Magdalini Ioannou
- Department of Psychiatry, University of Groningen and University Medical Centre Groningen, Groningen, the Netherlands
| | - Maria S. Simon
- Department of Psychiatry and Psychotherapy, Ludwig Maximilians University, Munich, Germany
| | - Jenny Borkent
- Department of Psychiatry, University of Groningen and University Medical Centre Groningen, Groningen, the Netherlands
| | - Annemarie Wijkhuijs
- Department of Immunology, Erasmus Universiteit Rotterdam and University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Raf Berghmans
- Advanced Practical Diagnostics BV, Turnhout, Belgium
| | - Bartholomeus C.M. Haarman
- Department of Psychiatry, University of Groningen and University Medical Centre Groningen, Groningen, the Netherlands
| | - Hemmo A. Drexhage
- Department of Immunology, Erasmus Universiteit Rotterdam and University Medical Centre Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
4
|
Rakebrandt N, Yassini N, Kolz A, Schorer M, Lambert K, Goljat E, Estrada Brull A, Rauld C, Balazs Z, Krauthammer M, Carballido JM, Peters A, Joller N. Innate acting memory Th1 cells modulate heterologous diseases. Proc Natl Acad Sci U S A 2024; 121:e2312837121. [PMID: 38838013 PMCID: PMC11181110 DOI: 10.1073/pnas.2312837121] [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: 07/26/2023] [Accepted: 05/08/2024] [Indexed: 06/07/2024] Open
Abstract
Through immune memory, infections have a lasting effect on the host. While memory cells enable accelerated and enhanced responses upon rechallenge with the same pathogen, their impact on susceptibility to unrelated diseases is unclear. We identify a subset of memory T helper 1 (Th1) cells termed innate acting memory T (TIA) cells that originate from a viral infection and produce IFN-γ with innate kinetics upon heterologous challenge in vivo. Activation of memory TIA cells is induced in response to IL-12 in combination with IL-18 or IL-33 but is TCR independent. Rapid IFN-γ production by memory TIA cells is protective in subsequent heterologous challenge with the bacterial pathogen Legionella pneumophila. In contrast, antigen-independent reactivation of CD4+ memory TIA cells accelerates disease onset in an autoimmune model of multiple sclerosis. Our findings demonstrate that memory Th1 cells can acquire additional TCR-independent functionality to mount rapid, innate-like responses that modulate susceptibility to heterologous challenges.
Collapse
Affiliation(s)
- Nikolas Rakebrandt
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Nima Yassini
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Anna Kolz
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
| | - Michelle Schorer
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Katharina Lambert
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Eva Goljat
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Anna Estrada Brull
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Celine Rauld
- Novartis Biomedical Research, 4002Basel, Switzerland
| | - Zsolt Balazs
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Michael Krauthammer
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | | | - Anneli Peters
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
| | - Nicole Joller
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| |
Collapse
|
5
|
Shi J, Wu W, Chen D, Liao Z, Sheng T, Wang Y, Yao Y, Wu Q, Liu F, Zhou R, Zhu C, Shen X, Mao Z, Ding Y, Wang W, Dotti G, Sun J, Liang X, Fang W, Zhao P, Li H, Gu Z. Lyophilized lymph nodes for improved delivery of chimeric antigen receptor T cells. NATURE MATERIALS 2024; 23:844-853. [PMID: 38448658 DOI: 10.1038/s41563-024-01825-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/31/2024] [Indexed: 03/08/2024]
Abstract
Lymph nodes are crucial organs of the adaptive immune system, orchestrating T cell priming, activation and tolerance. T cell activity and function are highly regulated by lymph nodes, which have a unique structure harbouring distinct cells that work together to detect and respond to pathogen-derived antigens. Here we show that implanted patient-derived freeze-dried lymph nodes loaded with chimeric antigen receptor T cells improve delivery to solid tumours and inhibit tumour recurrence after surgery. Chimeric antigen receptor T cells can be effectively loaded into lyophilized lymph nodes, whose unaltered meshwork and cytokine and chemokine contents promote chimeric antigen receptor T cell viability and activation. In mouse models of cell-line-derived human cervical cancer and patient-derived pancreatic cancer, delivery of chimeric antigen receptor T cells targeting mesothelin via the freeze-dried lymph nodes is more effective in preventing tumour recurrence when compared to hydrogels containing T-cell-supporting cytokines. This tissue-mediated cell delivery strategy holds promise for controlled release of various cells and therapeutics with long-term activity and augmented function.
Collapse
Affiliation(s)
- Jiaqi Shi
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Wei Wu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ziyan Liao
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Tao Sheng
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yanfang Wang
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yuejun Yao
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qing Wu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Feng Liu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Ruyi Zhou
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chaojie Zhu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinyuan Shen
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gianpietro Dotti
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Jie Sun
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Xiao Liang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Hongjun Li
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Jinhua Institute, Zhejiang University, Jinhua, China.
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Jinhua Institute, Zhejiang University, Jinhua, China.
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Gordon KS, Perez CR, Garmilla A, Lam MSY, Aw JJ, Datta A, Lauffenburger DA, Pavesi A, Birnbaum ME. Pooled screening for CAR function identifies novel IL13Rα2-targeted CARs for treatment of glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.586240. [PMID: 38766252 PMCID: PMC11100612 DOI: 10.1101/2024.04.04.586240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Chimeric antigen receptor therapies have demonstrated potent efficacy in treating B cell malignancies, but have yet to meaningfully translate to solid tumors. Here, we utilize our pooled screening platform, CARPOOL, to expedite the discovery of CARs with anti-tumor functions necessary for solid tumor efficacy. We performed selections in primary human T cells expressing a library of 1.3×10 6 3 rd generation CARs targeting IL13Rα2, a cancer testis antigen commonly expressed in glioblastoma. Selections were performed for cytotoxicity, proliferation, memory formation, and persistence upon repeated antigen challenge. Each enriched CAR robustly produced the phenotype for which it was selected, and one enriched CAR triggered potent cytotoxicity and long-term proliferation upon in vitro tumor rechallenge. It also showed significantly improved persistence and comparable antigen-specific tumor control in a microphysiological human in vitro model and a xenograft model of human glioblastoma. Taken together, this work demonstrates the utility of extending CARPOOL to diseases beyond hematological malignancies and represents the largest exploration of signaling combinations in human primary cells to date.
Collapse
|
7
|
Yin R, Melton S, Huseby ES, Kardar M, Chakraborty AK. How persistent infection overcomes peripheral tolerance mechanisms to cause T cell-mediated autoimmune disease. Proc Natl Acad Sci U S A 2024; 121:e2318599121. [PMID: 38446856 PMCID: PMC10945823 DOI: 10.1073/pnas.2318599121] [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: 10/24/2023] [Accepted: 02/06/2024] [Indexed: 03/08/2024] Open
Abstract
T cells help orchestrate immune responses to pathogens, and their aberrant regulation can trigger autoimmunity. Recent studies highlight that a threshold number of T cells (a quorum) must be activated in a tissue to mount a functional immune response. These collective effects allow the T cell repertoire to respond to pathogens while suppressing autoimmunity due to circulating autoreactive T cells. Our computational studies show that increasing numbers of pathogenic peptides targeted by T cells during persistent or severe viral infections increase the probability of activating T cells that are weakly reactive to self-antigens (molecular mimicry). These T cells are easily re-activated by the self-antigens and contribute to exceeding the quorum threshold required to mount autoimmune responses. Rare peptides that activate many T cells are sampled more readily during severe/persistent infections than in acute infections, which amplifies these effects. Experiments in mice to test predictions from these mechanistic insights are suggested.
Collapse
Affiliation(s)
- Rose Yin
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Samuel Melton
- Physics of Living Systems, Department of Physics, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Eric S. Huseby
- Basic Pathology, Department of Pathology, University of Massachusetts Medical School, Worcester, MA01655
| | - Mehran Kardar
- Physics of Living Systems, Department of Physics, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Arup K. Chakraborty
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Physics of Living Systems, Department of Physics, Massachusetts Institute of Technology, Cambridge, MA02139
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA02139
| |
Collapse
|
8
|
Amoriello R, Memo C, Ballerini L, Ballerini C. The brain cytokine orchestra in multiple sclerosis: from neuroinflammation to synaptopathology. Mol Brain 2024; 17:4. [PMID: 38263055 PMCID: PMC10807071 DOI: 10.1186/s13041-024-01077-7] [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: 11/21/2023] [Accepted: 01/18/2024] [Indexed: 01/25/2024] Open
Abstract
The central nervous system (CNS) is finely protected by the blood-brain barrier (BBB). Immune soluble factors such as cytokines (CKs) are normally produced in the CNS, contributing to physiological immunosurveillance and homeostatic synaptic scaling. CKs are peptide, pleiotropic molecules involved in a broad range of cellular functions, with a pivotal role in resolving the inflammation and promoting tissue healing. However, pro-inflammatory CKs can exert a detrimental effect in pathological conditions, spreading the damage. In the inflamed CNS, CKs recruit immune cells, stimulate the local production of other inflammatory mediators, and promote synaptic dysfunction. Our understanding of neuroinflammation in humans owes much to the study of multiple sclerosis (MS), the most common autoimmune and demyelinating disease, in which autoreactive T cells migrate from the periphery to the CNS after the encounter with a still unknown antigen. CNS-infiltrating T cells produce pro-inflammatory CKs that aggravate local demyelination and neurodegeneration. This review aims to recapitulate the state of the art about CKs role in the healthy and inflamed CNS, with focus on recent advances bridging the study of adaptive immune system and neurophysiology.
Collapse
Affiliation(s)
- Roberta Amoriello
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy.
| | - Christian Memo
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy
| | - Laura Ballerini
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy
| | - Clara Ballerini
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.
| |
Collapse
|
9
|
Rüterbusch MJ, Hondowicz BD, Takehara KK, Pruner KB, Griffith TS, Pepper M. Allergen exposure functionally alters influenza-specific CD4+ Th1 memory cells in the lung. J Exp Med 2023; 220:e20230112. [PMID: 37698553 PMCID: PMC10497397 DOI: 10.1084/jem.20230112] [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: 01/17/2023] [Revised: 07/11/2023] [Accepted: 08/22/2023] [Indexed: 09/13/2023] Open
Abstract
CD4+ lung-resident memory T cells (TRM) generated in response to influenza infection confer effective protection against subsequent viral exposures. Whether these cells can be altered by environmental antigens and cytokines released during heterologous, antigen-independent immune responses is currently unclear. We therefore investigated how influenza-specific CD4+ Th1 TRM in the lung are impacted by a subsequent Th2-inducing respiratory house dust mite (HDM) exposure. Although naïve influenza-specific CD4+ T cells in the lymph nodes do not respond to HDM, influenza-specific CD4+ TRM in the lungs do respond to a subsequent allergen exposure by decreasing expression of the transcription factor T-bet. This functional alteration is associated with decreased IFN-γ production upon restimulation and improved disease outcomes following heterosubtypic influenza challenge. Further investigation revealed that ST2 signaling in CD4+ T cells during allergic challenge is necessary to induce these changes in lung-resident influenza-specific CD4+ TRM. Thus, heterologous antigen exposure or ST2-signaling can drive persistent changes in CD4+ Th1 TRM populations and impact protection upon reinfection.
Collapse
Affiliation(s)
- Mikel J. Rüterbusch
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Brian D. Hondowicz
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Kennidy K. Takehara
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Kurt B. Pruner
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN, USA
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Marion Pepper
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
10
|
Yang LR, Li L, Meng MY, Li TT, Zhao YY, Yang SL, Gao H, Tang WW, Yang Y, Yang LL, Wang WJ, Liao LW, Hou ZL. IL-7 promotes CD19-directed CAR-T cells proliferation through miRNA-98-5p by targeting CDKN1A. Int Immunopharmacol 2023; 124:110974. [PMID: 37757633 DOI: 10.1016/j.intimp.2023.110974] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
CAR-T targeting CD19 have achieved significant effects in the treatment of B-line leukemia and lymphoma. However, the treated patients frequently relapsed and could not achieve complete remission. Therefore, improving the proliferation and cytotoxicity of CAR-T cells, reducing exhaustion and enhancing infiltration capacity are still issues to be solved. The IL-7 has been shown to enhance the memory characteristics of CAR-T cells, but the specific mechanism has yet to be elaborated. miRNAs play an important role in T cell activity. However, whether miRNA is involved in the activation of CAR-T cells by IL-7 has not yet been reported. Our previous study had established the 3rd generation CAR-T cells. The present study further found that IL-7 significantly increased the proliferation of anti-CD19 CAR-T cells, the ratio of CD4 + CAR + cells and the S phase of cell cycle. In vivo study NAMALWA xenograft model showed that IL-7-stimulated CAR-T cells possessed stronger tumoricidal efficiency. Further we validated that IL-7 induced CAR-T cells had low expression of CDKN1A and high expression of miRNA-98-5p. Additionally, CDKN1A was associated with miRNA-98-5p. Our results, for the first time, suggested IL-7 could conspicuously enhance the proliferation of CAR-T cells through miRNA-98-5p targeting CDKN1A expression, which should be applied to CAR-T production.
Collapse
Affiliation(s)
- Li-Rong Yang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Department of Oncology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Lin Li
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Yunnan Cell Biology and Clinical Translation Research Center, China
| | - Ming-Yao Meng
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Yunnan Cell Biology and Clinical Translation Research Center, China
| | - Tian-Tian Li
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Kunming Medical University, Kunming, Yunnan Province, China
| | - Yi-Yi Zhao
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Yunnan Cell Biology and Clinical Translation Research Center, China
| | - Song-Lin Yang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Kunming Medical University, Kunming, Yunnan Province, China
| | - Hui Gao
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Yunnan Cell Biology and Clinical Translation Research Center, China
| | - Wei-Wei Tang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Yunnan Cell Biology and Clinical Translation Research Center, China
| | - Yang Yang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Kunming Medical University, Kunming, Yunnan Province, China
| | - Li-Li Yang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Kunming Medical University, Kunming, Yunnan Province, China
| | - Wen-Ju Wang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Yunnan Cell Biology and Clinical Translation Research Center, China
| | - Li-Wei Liao
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Yunnan Cell Biology and Clinical Translation Research Center, China.
| | - Zong-Liu Hou
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, China; Key Laboratory of Tumor Immunological Prevention and Treatment, Yunnan Province, China; Yunnan Cell Biology and Clinical Translation Research Center, China.
| |
Collapse
|
11
|
Chen K, Shi Y, Luo W, Zhang T, Bao K, Huang C. SMIM20: a new biological signal associated with the prognosis of glioblastoma. Transl Cancer Res 2023; 12:2754-2763. [PMID: 37969370 PMCID: PMC10643959 DOI: 10.21037/tcr-23-796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/13/2023] [Indexed: 11/17/2023]
Abstract
Background Glioblastoma multiforme (GBM) is the most prevalent fatal central nervous system tumor. Notably, the survival rates after surgical intervention and active radiotherapy are not optimistic. Therefore, identifying new GBM-related biomarkers is a top priority in current research. Methods Transcriptome and clinical information of patients with GBM were obtained from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. According to the SMIM20 expression levels, the samples were divided into high- and low-expression groups and used for differential expression gene (DEG) analysis. Functional enrichment analyses, including Gene Ontology (GO), gene set enrichment analysis, and immune cell infiltration, were performed on SMIM20-related DEGs. Subsequently, univariate and multivariate Cox regression analyses were performed to screen the risk factors associated with the poor prognosis of SMIM20, and the clinical significance of SMIM20 in GBM was explored by constructing a prognostic nomogram. Results In total, 156 DEGs were screened, of which 131 were upregulated and 25 were downregulated. Kaplan-Meier analysis revealed that the total survival time of the SMIM20 high expression group was significantly lower than that of the SMIM20 low-expression group. Finally, the nomogram map had good predictive value for evaluating GBM prognosis of patients. Conclusions High expression of SMIM20 is associated with poor outcomes in GBM. The DEGs and pathways identified in this study reveal potential molecular mechanisms underlying the occurrence and progression of GBM. Our study identifies potential new biomarkers and therapeutic targets for the treatment of GBM.
Collapse
Affiliation(s)
- Kai Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yu Shi
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenzhang Luo
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Tianyu Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Kunyang Bao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Changren Huang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Neurosurgical Clinical Research Center of Sichuan Province, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Functions, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Martyshkina YS, Tereshchenko VP, Bogdanova DA, Rybtsov SA. Reliable Hallmarks and Biomarkers of Senescent Lymphocytes. Int J Mol Sci 2023; 24:15653. [PMID: 37958640 PMCID: PMC10647376 DOI: 10.3390/ijms242115653] [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: 10/06/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
The phenomenon of accumulation of senescent adaptive immunity cells in the elderly is attracting attention due to the increasing risk of global epidemics and aging of the global population. Elderly people are predisposed to various infectious and age-related diseases and are at higher risk of vaccination failure. The accumulation of senescent cells increases age-related background inflammation, "Inflammaging", causing lymphocyte exhaustion and cardiovascular, neurodegenerative, autoimmune and cancer diseases. Here, we present a comprehensive contemporary review of the mechanisms and phenotype of senescence in the adaptive immune system. Although modern research has not yet identified specific markers of aging lymphocytes, several sets of markers facilitate the separation of the aging population based on normal memory and exhausted cells for further genetic and functional analysis. The reasons for the higher predisposition of CD8+ T-lymphocytes to senescence compared to the CD4+ population are also discussed. We point out approaches for senescent-lymphocyte-targeting markers using small molecules (senolytics), antibodies and immunization against senescent cells. The suppression of immune senescence is the most relevant area of research aimed at developing anti-aging and anti-cancer therapy for prolonging the lifespan of the global population.
Collapse
Affiliation(s)
- Yuliya S. Martyshkina
- Division of Immunobiology and Biomedicine, Center for Genetics and Life Sciences, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia; (Y.S.M.)
| | - Valeriy P. Tereshchenko
- Resource Center for Cell Technology and Immunology, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia
| | - Daria A. Bogdanova
- Division of Immunobiology and Biomedicine, Center for Genetics and Life Sciences, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia; (Y.S.M.)
| | - Stanislav A. Rybtsov
- Resource Center for Cell Technology and Immunology, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia
| |
Collapse
|
13
|
Doratt BM, Sureshchandra S, True H, Rincon M, Marshall NE, Messaoudi I. Mild/asymptomatic COVID-19 in unvaccinated pregnant mothers impairs neonatal immune responses. JCI Insight 2023; 8:e172658. [PMID: 37698937 PMCID: PMC10629812 DOI: 10.1172/jci.insight.172658] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023] Open
Abstract
Maternal SARS-CoV-2 infection triggers placental inflammation and alters cord blood immune cell composition. However, most studies focus on outcomes of severe maternal infection. Therefore, we analyzed cord blood and chorionic villi from newborns of unvaccinated mothers who experienced mild/asymptomatic SARS-CoV-2 infection during pregnancy. We investigated immune cell rewiring using flow cytometry, single-cell RNA sequencing, and functional readouts using ex vivo stimulation with TLR agonists and pathogens. Maternal infection was associated with increased frequency of memory T and B cells and nonclassical monocytes in cord blood. Ex vivo T and B cell responses to stimulation were attenuated, suggesting a tolerogenic state. Maladaptive responses were also observed in cord blood monocytes, where antiviral responses were dampened but responses to bacterial TLRs were increased. Maternal infection was also associated with expansion and activation of placental Hofbauer cells, secreting elevated levels of myeloid cell-recruiting chemokines. Moreover, we reported increased activation of maternally derived monocytes/macrophages in the fetal placenta that were transcriptionally primed for antiviral responses. Our data indicate that even in the absence of vertical transmission or symptoms in the neonate, mild/asymptomatic maternal COVID-19 altered the transcriptional and functional state in fetal immune cells in circulation and in the placenta.
Collapse
Affiliation(s)
- Brianna M. Doratt
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Suhas Sureshchandra
- Department of Physiology and Biophysics, School of Medicine, and
- Institute for Immunology, University of California, Irvine, California, USA
| | - Heather True
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Monica Rincon
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Nicole E. Marshall
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
14
|
Khan MT, Mahmud A, Islam MM, Sumaia MSN, Rahim Z, Islam K, Iqbal A. Multi-epitope vaccine against drug-resistant strains of Mycobacterium tuberculosis: a proteome-wide subtraction and immunoinformatics approach. Genomics Inform 2023; 21:e42. [PMID: 37813638 PMCID: PMC10584640 DOI: 10.5808/gi.23021] [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: 03/28/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 10/11/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis, one of the most deadly infections in humans. The emergence of multidrug-resistant and extensively drug-resistant Mtb strains presents a global challenge. Mtb has shown resistance to many frontline antibiotics, including rifampicin, kanamycin, isoniazid, and capreomycin. The only licensed vaccine, Bacille Calmette-Guerin, does not efficiently protect against adult pulmonary tuberculosis. Therefore, it is urgently necessary to develop new vaccines to prevent infections caused by these strains. We used a subtractive proteomics approach on 23 virulent Mtb strains and identified a conserved membrane protein (MmpL4, NP_214964.1) as both a potential drug target and vaccine candidate. MmpL4 is a non-homologous essential protein in the host and is involved in the pathogen-specific pathway. Furthermore, MmpL4 shows no homology with anti-targets and has limited homology to human gut microflora, potentially reducing the likelihood of adverse effects and cross-reactivity if therapeutics specific to this protein are developed. Subsequently, we constructed a highly soluble, safe, antigenic, and stable multi-subunit vaccine from the MmpL4 protein using immunoinformatics. Molecular dynamics simulations revealed the stability of the vaccine-bound Toll-like receptor-4 complex on a nanosecond scale, and immune simulations indicated strong primary and secondary immune responses in the host. Therefore, our study identifies a new target that could expedite the design of effective therapeutics, and the designed vaccine should be validated. Future directions include an extensive molecular interaction analysis, in silico cloning, wet-lab experiments, and evaluation and comparison of the designed candidate as both a DNA vaccine and protein vaccine.
Collapse
Affiliation(s)
- Md Tahsin Khan
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Araf Mahmud
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Md. Muzahidul Islam
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Mst. Sayedatun Nessa Sumaia
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Zeaur Rahim
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, Bangladesh
| | - Kamrul Islam
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Asif Iqbal
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
| |
Collapse
|
15
|
Doratt BM, Sureshchandra S, True H, Rincon M, Marshall N, Messaoudi I. Mild/Asymptomatic Maternal SARS-CoV-2 Infection Leads to Immune Paralysis in Fetal Circulation and Immune Dysregulation in Fetal-Placental Tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540233. [PMID: 37214938 PMCID: PMC10197637 DOI: 10.1101/2023.05.10.540233] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Few studies have addressed the impact of maternal mild/asymptomatic SARS-CoV-2 infection on the developing neonatal immune system. In this study, we analyzed umbilical cord blood and placental chorionic villi from newborns of unvaccinated mothers with mild/asymptomatic SARSCoV-2 infection during pregnancy using flow cytometry, single-cell transcriptomics, and functional assays. Despite the lack of vertical transmission, levels of inflammatory mediators were altered in cord blood. Maternal infection was also associated with increased memory T, B cells, and non-classical monocytes as well as increased activation. However, ex vivo responses to stimulation were attenuated. Finally, within the placental villi, we report an expansion of fetal Hofbauer cells and infiltrating maternal macrophages and rewiring towards a heightened inflammatory state. In contrast to cord blood monocytes, placental myeloid cells were primed for heightened antiviral responses. Taken together, this study highlights dysregulated fetal immune cell responses in response to mild maternal SARS-CoV-2 infection during pregnancy.
Collapse
Affiliation(s)
- Brianna M. Doratt
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington KY 40536
| | - Suhas Sureshchandra
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine CA 92697
- Institute for Immunology, University of California, Irvine CA 92697
| | - Heather True
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington KY 40536
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington KY 40536
| | - Monica Rincon
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland OR 97239
| | - Nicole Marshall
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland OR 97239
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington KY 40536
| |
Collapse
|
16
|
Kao CY, Mills JA, Burke CJ, Morse B, Marques BF. Role of Cytokines and Growth Factors in the Manufacturing of iPSC-Derived Allogeneic Cell Therapy Products. BIOLOGY 2023; 12:biology12050677. [PMID: 37237491 DOI: 10.3390/biology12050677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/23/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023]
Abstract
Cytokines and other growth factors are essential for cell expansion, health, function, and immune stimulation. Stem cells have the additional reliance on these factors to direct differentiation to the appropriate terminal cell type. Successful manufacturing of allogeneic cell therapies from induced pluripotent stem cells (iPSCs) requires close attention to the selection and control of cytokines and factors used throughout the manufacturing process, as well as after administration to the patient. This paper employs iPSC-derived natural killer cell/T cell therapeutics to illustrate the use of cytokines, growth factors, and transcription factors at different stages of the manufacturing process, ranging from the generation of iPSCs to controlling of iPSC differentiation into immune-effector cells through the support of cell therapy after patient administration.
Collapse
Affiliation(s)
- Chen-Yuan Kao
- Process and Product Development, Century Therapeutics, Philadelphia, PA 19104, USA
| | - Jason A Mills
- Process and Product Development, Century Therapeutics, Philadelphia, PA 19104, USA
| | - Carl J Burke
- Process and Product Development, Century Therapeutics, Philadelphia, PA 19104, USA
| | - Barry Morse
- Research and Development, Century Therapeutics, Philadelphia, PA 19104, USA
| | - Bruno F Marques
- Process and Product Development, Century Therapeutics, Philadelphia, PA 19104, USA
| |
Collapse
|
17
|
Immunomodulatory action of Lactococcuslactis. J Biosci Bioeng 2023; 135:1-9. [PMID: 36428209 DOI: 10.1016/j.jbiosc.2022.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/07/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2022]
Abstract
Fermented foods are gaining popularity due to health-promoting properties with high levels of nutrients, phytochemicals, bioactive compounds, and probiotic microorganisms. Due to its unique fermentation process, Lactococcus lactis plays a key role in the food business, notably in the manufacturing of dairy products. The superior biological activities of L. lactis in these functional foods include anti-inflammatory and immunomodulatory capabilities. L. lactis boosted growth performance, controlled amino acid profiles, intestinal immunology, and microbiota. Besides that, the administration of L. lactis increased the rate of infection clearance. Innate and acquired immune responses would be upregulated in both local and systemic compartments, resulting in these consequences. L. lactis is often employed in the food sector and is currently being exploited as a delivery vehicle for biological research. These bacteria are being eyed as potential candidates for biotechnological applications. With this in mind, we reviewed the immunomodulatory effects of different L. lactis strains.
Collapse
|
18
|
Soluble factors from TLR4- or TCR-activated cells contribute to stability of the resting phenotype and increase the expression of CXCR4 of human memory CD4 T cells. Immunol Res 2022; 71:388-403. [PMID: 36539634 DOI: 10.1007/s12026-022-09345-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022]
Abstract
It has been proposed that cytokines can induce activation of resting T cells in an antigen-independent manner. However, experimental conditions have included the use of fetal serum and nanogram concentrations of added cytokines. To evaluate the effect of cytokines and chemokines generated by activated immune cells on the phenotypic profile of human memory CD4 T cells, the cells were cultured in FBS-free conditions in the presence of IL-15 and 5% of hAB serum and incubated with conditioned medium (CM) obtained from PBMC activated through the TCR using anti-CD3/CD28/CD2 antibodies (TCR-CM) or through TLR4 using bacterial LPS (TLR4-CM). Cytokines and chemokines present in the CMs were evaluated by ProcartaPlex immunoassay. Cell viability, proliferation, and surface markers were determined by flow cytometry on day 2, 5, and 8 of culture. Cell viability was maintained by TLR4-CM plus IL-15 for 8 days but decreased in the presence of the TCR-CM plus IL-15. In combination with IL-15, the TLR4-CM, but not the TCR-CM, maintained the expression of CD3 and CD4 stable. Both conditions stabilized the expression of CD45RO and CCR5. Thus, the TLR4-CM better supported the viability and stability of the memory phenotype. None of the CMs induced proliferation or expression of activation markers; however, they induced an increased expression of CXCR4. This study indicates that resting memory CD4 T cells are not activated by, but may be sensitive to soluble factors produced by antigen or PAMP-stimulated cells, which may contribute to their homeostasis and favor the CXCR4 expression.
Collapse
|
19
|
Svensson-Arvelund J, Cuadrado-Castano S, Pantsulaia G, Kim K, Aleynick M, Hammerich L, Upadhyay R, Yellin M, Marsh H, Oreper D, Jhunjhunwala S, Moussion C, Merad M, Brown BD, García-Sastre A, Brody JD. Expanding cross-presenting dendritic cells enhances oncolytic virotherapy and is critical for long-term anti-tumor immunity. Nat Commun 2022; 13:7149. [PMID: 36418317 PMCID: PMC9684150 DOI: 10.1038/s41467-022-34791-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
Immunotherapies directly enhancing anti-tumor CD8+ T cell responses have yielded measurable but limited success, highlighting the need for alternatives. Anti-tumor T cell responses critically depend on antigen presenting dendritic cells (DC), and enhancing mobilization, antigen loading and activation of these cells represent an attractive possibility to potentiate T cell based therapies. Here we show that expansion of DCs by Flt3L administration impacts in situ vaccination with oncolytic Newcastle Disease Virus (NDV). Mechanistically, NDV activates DCs and sensitizes them to dying tumor cells through upregulation of dead-cell receptors and synergizes with Flt3L to promote anti-tumor CD8+ T cell cross-priming. In vivo, Flt3L-NDV in situ vaccination induces parallel amplification of virus- and tumor-specific T cells, including CD8+ T cells reactive to newly-described neoepitopes, promoting long-term tumor control. Cross-presenting conventional Type 1 DCs are indispensable for the anti-tumor, but not anti-viral, T cell response, and type I IFN-dependent CD4+ Th1 effector cells contribute to optimal anti-tumor immunity. These data demonstrate that mobilizing DCs to increase tumor antigen cross-presentation improves oncolytic virotherapy and that neoepitope-specific T cells can be induced without individualized, ex vivo manufactured vaccines.
Collapse
Affiliation(s)
- Judit Svensson-Arvelund
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, 582 25, Sweden.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Sara Cuadrado-Castano
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gvantsa Pantsulaia
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kristy Kim
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mark Aleynick
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Linda Hammerich
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Hepatology and Gastroenterology, Campus Virchow- Klinikum, Charité Universitätsmedizin Berlin, Berlin, 13353, Germany
| | - Ranjan Upadhyay
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Henry Marsh
- Celldex Therapeutics, Inc, Needham, MA, 02494, USA
| | | | | | | | - Miriam Merad
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Brian D Brown
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Adolfo García-Sastre
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Joshua D Brody
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
20
|
Matheson LS, Petkau G, Sáenz-Narciso B, D'Angeli V, McHugh J, Newman R, Munford H, West J, Chakraborty K, Roberts J, Łukasiak S, Díaz-Muñoz MD, Bell SE, Dimeloe S, Turner M. Multiomics analysis couples mRNA turnover and translational control of glutamine metabolism to the differentiation of the activated CD4 + T cell. Sci Rep 2022; 12:19657. [PMID: 36385275 PMCID: PMC9669047 DOI: 10.1038/s41598-022-24132-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
The ZFP36 family of RNA-binding proteins acts post-transcriptionally to repress translation and promote RNA decay. Studies of genes and pathways regulated by the ZFP36 family in CD4+ T cells have focussed largely on cytokines, but their impact on metabolic reprogramming and differentiation is unclear. Using CD4+ T cells lacking Zfp36 and Zfp36l1, we combined the quantification of mRNA transcription, stability, abundance and translation with crosslinking immunoprecipitation and metabolic profiling to determine how they regulate T cell metabolism and differentiation. Our results suggest that ZFP36 and ZFP36L1 act directly to limit the expression of genes driving anabolic processes by two distinct routes: by targeting transcription factors and by targeting transcripts encoding rate-limiting enzymes. These enzymes span numerous metabolic pathways including glycolysis, one-carbon metabolism and glutaminolysis. Direct binding and repression of transcripts encoding glutamine transporter SLC38A2 correlated with increased cellular glutamine content in ZFP36/ZFP36L1-deficient T cells. Increased conversion of glutamine to α-ketoglutarate in these cells was consistent with direct binding of ZFP36/ZFP36L1 to Gls (encoding glutaminase) and Glud1 (encoding glutamate dehydrogenase). We propose that ZFP36 and ZFP36L1 as well as glutamine and α-ketoglutarate are limiting factors for the acquisition of the cytotoxic CD4+ T cell fate. Our data implicate ZFP36 and ZFP36L1 in limiting glutamine anaplerosis and differentiation of activated CD4+ T cells, likely mediated by direct binding to transcripts of critical genes that drive these processes.
Collapse
Affiliation(s)
- Louise S Matheson
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| | - Georg Petkau
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Beatriz Sáenz-Narciso
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Vanessa D'Angeli
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.,Present Address: IONTAS, The Works, Unity Campus, Cambridge, CB22 3EF, UK
| | - Jessica McHugh
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.,Present Address: Nature Reviews Rheumatology, The Campus, 4 Crinan Street, London, N1 9XW, UK
| | - Rebecca Newman
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.,Present Address: Immunology Research Unit, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, Herts, UK
| | - Haydn Munford
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, IBR, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - James West
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Krishnendu Chakraborty
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.,Present Address: Bioanalysis, Immunogenicity and Biomarkers (BIB), IVIVT, GSK, Stevenage, SG1 2NY, UK
| | - Jennie Roberts
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Sebastian Łukasiak
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.,Present Address: Discovery Biology, Discovery Science, R&D, AstraZeneca, Cambridge, UK
| | - Manuel D Díaz-Muñoz
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.,Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Inserm UMR1291, CNRS UMR5051, University Paul Sabatier, CHU Purpan, BP3028, 31024, Toulouse, France
| | - Sarah E Bell
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Sarah Dimeloe
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, IBR, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Martin Turner
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
21
|
Zhang P, Zhu H. Cytokines in Thyroid-Associated Ophthalmopathy. J Immunol Res 2022; 2022:2528046. [PMID: 36419958 PMCID: PMC9678454 DOI: 10.1155/2022/2528046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 09/07/2023] Open
Abstract
Thyroid-associated ophthalmopathy (TAO), also known as thyroid eye disease (TED) or Graves' orbitopathy (GO), is a complex autoimmune condition causing visual impairment, disfigurement, and harm to patients' physical and mental health. The pathogenesis of TAO has not been fully elucidated, and the mainstream view is that coantigens shared by the thyroid and orbit trigger remodeling of extraocular muscles and orbital connective tissues through an inflammatory response. In recent years, cytokines and the immune responses they mediate have been crucial in disease progression, and currently, common evidence has shown that drugs targeting cytokines, such as tocilizumab, infliximab, and adalimumab, may be novel targets for therapy. In this review, we summarize the research development of different cytokines in TAO pathogenesis in the hope of discovering new therapeutic targets.
Collapse
Affiliation(s)
- Pengbo Zhang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Huang Zhu
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
22
|
Ge T, Phung A, Jhala G, Trivedi P, Principe N, De George DJ, Pappas EG, Litwak S, Sanz‐Villanueva L, Catterall T, Fynch S, Boon L, Kay TW, Chee J, Krishnamurthy B, Thomas HE. Diabetes induced by checkpoint inhibition in nonobese diabetic mice can be prevented or reversed by a JAK1/JAK2 inhibitor. Clin Transl Immunology 2022; 11:e1425. [PMID: 36325490 PMCID: PMC9618467 DOI: 10.1002/cti2.1425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES Immune checkpoint inhibitors have achieved clinical success in cancer treatment, but this treatment causes immune-related adverse events, including type 1 diabetes (T1D). Our aim was to test whether a JAK1/JAK2 inhibitor, effective at treating spontaneous autoimmune diabetes in nonobese diabetic (NOD) mice, can prevent diabetes secondary to PD-L1 blockade. METHODS Anti-PD-L1 antibody was injected into NOD mice to induce diabetes, and JAK1/JAK2 inhibitor LN3103801 was administered by oral gavage to prevent diabetes. Flow cytometry was used to study T cells and beta cells. Mesothelioma cells were inoculated into BALB/c mice to induce a transplantable tumour model. RESULTS Anti-PD-L1-induced diabetes was associated with increased immune cell infiltration in the islets and upregulated MHC class I on islet cells. Anti-PD-L1 administration significantly increased islet T cell proliferation and islet-specific CD8+ T cell numbers in peripheral lymphoid organs. JAK1/JAK2 inhibitor treatment blocked IFNγ-mediated MHC class I upregulation on beta cells and T cell proliferation mediated by cytokines that use the common γ chain receptor. As a result, anti-PD-L1-induced diabetes was prevented by JAK1/JAK2 inhibitor administered before or after checkpoint inhibitor therapy. Diabetes was also reversed when the JAK1/JAK2 inhibitor was administered after the onset of anti-PD-L1-induced hyperglycaemia. Furthermore, JAK1/JAK2 inhibitor intervention after checkpoint inhibitors did not reverse or abrogate the antitumour effects in a transplantable tumour model. CONCLUSION A JAK1/JAK2 inhibitor can prevent and reverse anti-PD-L1-induced diabetes by blocking IFNγ and γc cytokine activities. Our study provides preclinical validation of JAK1/JAK2 inhibitor use in checkpoint inhibitor-induced diabetes.
Collapse
Affiliation(s)
- Tingting Ge
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia,The University of MelbourneParkvilleVICAustralia
| | - Amber‐Lee Phung
- National Centre for Asbestos Related Diseases, Institute for Respiratory HealthThe University of Western AustraliaCrawleyWAAustralia
| | - Gaurang Jhala
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia
| | - Prerak Trivedi
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia
| | - Nicola Principe
- National Centre for Asbestos Related Diseases, Institute for Respiratory HealthThe University of Western AustraliaCrawleyWAAustralia
| | - David J De George
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia,The University of MelbourneParkvilleVICAustralia
| | - Evan G Pappas
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia
| | - Sara Litwak
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia
| | - Laura Sanz‐Villanueva
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia,The University of MelbourneParkvilleVICAustralia
| | - Tara Catterall
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia
| | - Stacey Fynch
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia
| | | | - Thomas W Kay
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia,The University of MelbourneParkvilleVICAustralia
| | - Jonathan Chee
- National Centre for Asbestos Related Diseases, Institute for Respiratory HealthThe University of Western AustraliaCrawleyWAAustralia
| | - Balasubramanian Krishnamurthy
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia,The University of MelbourneParkvilleVICAustralia
| | - Helen E Thomas
- Immunology and Diabetes UnitSt Vincent's InstituteFitzroyVICAustralia,The University of MelbourneParkvilleVICAustralia
| |
Collapse
|
23
|
Terahara K, Sato T, Adachi Y, Tonouchi K, Onodera T, Moriyama S, Sun L, Takano T, Nishiyama A, Kawana-Tachikawa A, Matano T, Matsumura T, Shinkai M, Isogawa M, Takahashi Y. SARS-CoV-2-specific CD4 + T cell longevity correlates with Th17-like phenotype. iScience 2022; 25:104959. [PMID: 35992306 PMCID: PMC9384329 DOI: 10.1016/j.isci.2022.104959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/01/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Determinants of memory T cell longevity following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection remain unknown. In addition, phenotypes associated with memory T cell longevity, antibody titers, and disease severity are incompletely understood. Here, we longitudinally analyzed SARS-CoV-2-specific T cell and antibody responses of a unique cohort with similar numbers of mild, moderate, and severe coronavirus disease 2019 cases. The half-lives of CD4+ and CD8+ T cells were longer than those of antibody titers and showed no clear correlation with disease severity. When CD4+ T cells were divided into Th1-, Th2-, Th17-, and Tfh-like subsets, the Th17-like subset showed a longer half-life than other subsets, indicating that Th17-like cells are most closely correlated with T cell longevity. In contrast, Th2- and Tfh-like T cells were more closely correlated with antibody titers than other subsets. These results suggest that distinct CD4+ T cell subsets are associated with longevity and antibody responses. Th17-like CD4+ T cells showed a longer half-life than other CD4+ T cell subsets Anti-RBD-IgG titers were associated with Th2- and Tfh-like CD4 T cells CD45RA+CD8+ T cells were correlated with disease severity during the early phase
Collapse
Affiliation(s)
- Kazutaka Terahara
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Takashi Sato
- Tokyo Shinagawa Hospital; Tokyo, 140-8522, Japan
| | - Yu Adachi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Keisuke Tonouchi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.,Department of Life Science and Medical Bioscience, Waseda University, Tokyo, 162-8480, Japan
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Saya Moriyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Lin Sun
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Tomohiro Takano
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Ayae Nishiyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Ai Kawana-Tachikawa
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Takayuki Matsumura
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | | | - Masanori Isogawa
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|
24
|
Saidakova EV. Lymphopenia and Mechanisms of T-Cell Regeneration. CELL AND TISSUE BIOLOGY 2022; 16:302-311. [PMID: 35967247 PMCID: PMC9358362 DOI: 10.1134/s1990519x2204006x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 11/24/2022]
Abstract
Chronic lymphopenia, in particular, T-lymphocyte deficiency, increases the risk of death from cancer, cardiovascular and respiratory diseases and serves as a risk factor for a severe course and poor outcome of infectious diseases such as COVID-19. The regeneration of T-lymphocytes is a complex multilevel process, many questions of which still remain unanswered. The present review considers two main pathways of increasing the T-cell number in lymphopenia: production in the thymus and homeostatic proliferation in the periphery. Literature data on the signals that regulate each pathway are summarized. Their contribution to the quantitative and qualitative restoration of the immune cell pool is analyzed. The features of CD4+ and CD8+ T-lymphocytes’ regeneration are considered.
Collapse
Affiliation(s)
- E. V. Saidakova
- Institute of Ecology and Genetics of Microorganisms, Ural Branch, Russian Academy of Sciences—Branch of Perm Federal Research Center, Ural Branch, Russian Academy of Sciences, 614081 Perm, Russia
| |
Collapse
|
25
|
Ansari A, Sachan S, Jit BP, Sharma A, Coshic P, Sette A, Weiskopf D, Gupta N. An efficient immunoassay for the B cell help function of SARS-CoV-2-specific memory CD4 + T cells. CELL REPORTS METHODS 2022; 2:100224. [PMID: 35571764 PMCID: PMC9085463 DOI: 10.1016/j.crmeth.2022.100224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/27/2021] [Accepted: 04/28/2022] [Indexed: 04/30/2023]
Abstract
The B cell "help" function of CD4+ T cells is an important mechanism of adaptive immunity. Here, we describe improved antigen-specific T-B cocultures for quantitative measurement of T cell-dependent B cell responses, with as few as ∼90 T cells. Utilizing M. tuberculosis (Mtb), we show that early priming and activation of CD4+ T cells is important for productive interaction between T and B cells and that similar effects are achieved by supplementing cocultures with monocytes. We find that monocytes promote survivability of B cells via BAFF and stem cell growth factor (SCGF)/C-type lectin domain family 11 member A (CLEC11A), but this alone does not fully recapitulate the effects of monocyte supplementation. Importantly, we demonstrate improved activation and immunological output of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific memory CD4+ T-B cell cocultures with the inclusion of monocytes. This method may therefore provide a more sensitive assay to evaluate the B cell help quality of memory CD4+ T cells, for example, after vaccination or natural infection.
Collapse
Affiliation(s)
- Asgar Ansari
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Shilpa Sachan
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Bimal Prasad Jit
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Poonam Coshic
- Department of Transfusion Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Nimesh Gupta
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| |
Collapse
|
26
|
Vdovenko D, Balbi C, Di Silvestre D, Passignani G, Puspitasari YM, Zarak-Crnkovic M, Mauri P, Camici GG, Lüscher TF, Eriksson U, Vassalli G. Microvesicles released from activated CD4 + T cells alter microvascular endothelial cell function. Eur J Clin Invest 2022; 52:e13769. [PMID: 35316536 PMCID: PMC9287044 DOI: 10.1111/eci.13769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/18/2021] [Accepted: 01/02/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Microvesicles are vesicles shed by plasma membranes following cell activation and apoptosis. The role of lymphocyte-derived microvesicles in endothelial function remains poorly understood. METHODS CD4+ T cells isolated from peripheral blood of healthy human donors were stimulated using anti-CD3/anti-CD28-coated beads. Proteomic profiling of microvesicles was performed using linear discriminant analysis (LDA) from activated T cells (MV.Act) and nonactivated T cells (MV.NAct). In addition, data processing analysis was performed using MaxQUANT workflow. Differentially expressed proteins found in MV.Act or MV.NAct samples with identification frequency = 100%, which were selected by both LDA (p < .01) and MaxQUANT (p < .01) workflows, were defined as "high-confidence" differentially expressed proteins. Functional effects of MV.Act on human primary microvascular endothelial cells were analysed. RESULTS T cells released large amounts of microvesicles upon stimulation. Proteomic profiling of microvesicles using LDA identified 2279 proteins (n = 2110 and n = 851 proteins in MV.Act and MV.NAct, respectively). Protein-protein interaction network models reconstructed from both differentially expressed proteins (n = 594; LDA p ≤ .01) and "high-confidence" differentially expressed proteins (n = 98; p ≤ .01) revealed that MV.Act were enriched with proteins related to immune responses, protein translation, cytoskeleton organisation and TNFα-induced apoptosis. For instance, MV.Act were highly enriched with IFN-γ, a key proinflammatory pathway related to effector CD4+ T cells. Endothelial cell incubation with MV.Act induced superoxide generation, apoptosis, endothelial wound healing impairment and endothelial monolayer barrier disruption. CONCLUSIONS T cell receptor-mediated activation of CD4+ T cells stimulates the release of microvesicles enriched with proteins involved in immune responses, inflammation and apoptosis. T cell-derived microvesicles alter microvascular endothelial function and barrier permeability, potentially promoting tissue inflammation.
Collapse
Affiliation(s)
- Daria Vdovenko
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Carolina Balbi
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Laboratory of Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino-EOC, Lugano, Switzerland.,Laboratories for Translational Research-EOC, Bellinzona, Switzerland
| | | | | | | | | | | | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Royal Brompton & Harefield Hospital, Imperial College, London, UK
| | - Urs Eriksson
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Department of Medicine, GZO - Zurich Regional Health Center, Wetzikon, Switzerland
| | - Giuseppe Vassalli
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Laboratory of Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino-EOC, Lugano, Switzerland.,Laboratories for Translational Research-EOC, Bellinzona, Switzerland.,Department of Biomedicine, Università della Svizzera Italiana (USI), Lugano, Switzerland
| |
Collapse
|
27
|
An In-Vitro Study of the Expansion and Transcriptomics of CD4+ and CD8+ Naïve and Memory T Cells Stimulated by IL-2, IL-7 and IL-15. Cells 2022; 11:cells11101701. [PMID: 35626739 PMCID: PMC9139303 DOI: 10.3390/cells11101701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/15/2022] [Accepted: 05/18/2022] [Indexed: 02/01/2023] Open
Abstract
The growth of T cells ex vivo for the purpose of T cell therapies is a rate-limiting step in the overall process for cancer patients to achieve remission. Growing T cells is a fiscally-, time-, and resource-intensive process. Cytokines have been shown to accelerate the growth of T cells, specifically IL-2, IL-7, and IL-15. Here a design of experiments was conducted to optimize the growth rate of different naïve and memory T cell subsets using combinations of cytokines. Mathematical models were developed to study the impact of IL-2, IL-7, and IL-15 on the growth of T cells. The results show that CD4+ and CD8+ naïve T cells grew effectively using moderate IL-2 and IL-7 in combination, and IL-7, respectively. CD4+ and CD8+ memory cells favored moderate IL-2 and IL-15 in combination and moderate IL-7 and IL-15 in combination, respectively. A statistically significant interaction was observed between IL-2 and IL-7 in the growth data of CD4+ naïve T cells, while the interaction between IL-7 and IL-15 was found for CD8+ naïve T cells. The important genes and related signaling pathways and metabolic reactions were identified from the RNA sequencing data for each of the four subsets stimulated by each of the three cytokines. This systematic investigation lays the groundwork for studying other T cell subsets.
Collapse
|
28
|
Heterogeneity of Latency Establishment in the Different Human CD4
+
T Cell Subsets Stimulated with IL-15. J Virol 2022; 96:e0037922. [PMID: 35499323 PMCID: PMC9131862 DOI: 10.1128/jvi.00379-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
HIV integrates into the host genome, creating a viral reservoir of latently infected cells that persists despite effective antiretroviral treatment. CD4-positive (CD4+) T cells are the main contributors to the HIV reservoir. CD4+ T cells are a heterogeneous population, and the mechanisms of latency establishment in the different subsets, as well as their contribution to the reservoir, are still unclear. In this study, we analyzed HIV latency establishment in different CD4+ T cell subsets stimulated with interleukin 15 (IL-15), a cytokine that increases both susceptibility to infection and reactivation from latency. Using a dual-reporter virus that allows discrimination between latent and productive infection at the single-cell level, we found that IL-15-treated primary human CD4+ T naive and CD4+ T stem cell memory (TSCM) cells are less susceptible to HIV infection than CD4+ central memory (TCM), effector memory (TEM), and transitional memory (TTM) cells but are also more likely to harbor transcriptionally silent provirus. The propensity of these subsets to harbor latent provirus compared to the more differentiated memory subsets was independent of differential expression of pTEFb components. Microscopy analysis of NF-κB suggested that CD4+ T naive cells express smaller amounts of nuclear NF-κB than the other subsets, partially explaining the inefficient long terminal repeat (LTR)-driven transcription. On the other hand, CD4+ TSCM cells display similar levels of nuclear NF-κB to CD4+ TCM, CD4+ TEM, and CD4+ TTM cells, indicating the availability of transcription initiation and elongation factors is not solely responsible for the inefficient HIV gene expression in the CD4+ TSCM subset. IMPORTANCE The formation of a latent reservoir is the main barrier to HIV cure. Here, we investigated how HIV latency is established in different CD4+ T cell subsets in the presence of IL-15, a cytokine that has been shown to efficiently induce latency reversal. We observed that, even in the presence of IL-15, the less differentiated subsets display lower levels of productive HIV infection than the more differentiated subsets. These differences were not related to different expression of pTEFb, and modest differences in NF-κB were observed for CD4+ T naive cells only, implying the involvement of other mechanisms. Understanding the molecular basis of latency establishment in different CD4+ T cell subsets might be important for tailoring specific strategies to reactivate HIV transcription in all the CD4+ T subsets that compose the latent reservoir.
Collapse
|
29
|
Kwiecień I, Rutkowska E, Sokołowski R, Bednarek J, Raniszewska A, Jahnz-Różyk K, Rzepecki P, Domagała-Kulawik J. Effector Memory T Cells and CD45RO+ Regulatory T Cells in Metastatic vs. Non-Metastatic Lymph Nodes in Lung Cancer Patients. Front Immunol 2022; 13:864497. [PMID: 35585972 PMCID: PMC9108231 DOI: 10.3389/fimmu.2022.864497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/07/2022] [Indexed: 11/26/2022] Open
Abstract
Lymphocytes play a leading role in regulation of the immune system in lung cancer patients. The recognition of T cells profile may help in prediction of effectiveness of anticancer immunotherapy. The aim of the study was to determine the dominant subpopulation of CD4+ and CD8+ lymphocytes in metastatic and non-metastatic lymph nodes (LNs) of lung cancer patients. LNs aspirates were obtained during EBUS/TBNA procedure and cells were analyzed by flow cytometry. We showed a higher percentage of CD4+ and CD8+ effector memory T cells in the metastatic than in the non-metastatic LNs (28.6 vs. 15.3% and 28.6 vs. 14.0%, p< 0.05). The proportion of CD45RO+ T regulatory cells (CD45RO+ Tregs) was higher in the metastatic LNs than in the non-metastatic ones (65.6 vs. 31%, p< 0.05). We reported the significant differences in T cell subsets depending on the lung cancer metastatic process. We observed that the effector memory T cells were predominant subpopulations in metastatic LNs. Lymphocyte profile in LNs is easy to evaluate by flow cytometry of EBUS/TBNA samples and may reflect the immune status in lung cancer.
Collapse
Affiliation(s)
- Iwona Kwiecień
- Department of Internal Medicine and Hematology, Laboratory of Flow Cytometry, Military Institute of Medicine, Warsaw, Poland
- *Correspondence: Iwona Kwiecień, ;
| | - Elżbieta Rutkowska
- Department of Internal Medicine and Hematology, Laboratory of Flow Cytometry, Military Institute of Medicine, Warsaw, Poland
| | - Rafał Sokołowski
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine, Warsaw, Poland
| | - Joanna Bednarek
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine, Warsaw, Poland
| | - Agata Raniszewska
- Department of Internal Medicine and Hematology, Laboratory of Flow Cytometry, Military Institute of Medicine, Warsaw, Poland
| | - Karina Jahnz-Różyk
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine, Warsaw, Poland
| | - Piotr Rzepecki
- Department of Internal Medicine and Hematology, Military Institute of Medicine, Warsaw, Poland
| | - Joanna Domagała-Kulawik
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
30
|
Heikkilä N, Hetemäki I, Sormunen S, Isoniemi H, Kekäläinen E, Saramäki J, Arstila TP. Peripheral differentiation patterns of human T cells. Eur J Immunol 2022; 52:882-894. [PMID: 35307831 PMCID: PMC9313577 DOI: 10.1002/eji.202149465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/12/2022] [Accepted: 03/15/2022] [Indexed: 11/08/2022]
Abstract
Long-term T-cell memory is dependent on the maintenance of memory T cells in the lymphoid tissues, and at the surface interfaces that provide entry routes for pathogens. However, much of the current information on human T-cell memory is based on analyzing circulating T cells. Here, we have studied the distribution and age-related changes of memory T-cell subsets in samples from blood, mesenteric LNs, spleen, and ileum, obtained from donors ranging in age from 5 days to 67 years of age. Our data show that the main reservoir of polyclonal naive cells is found in the LNs, and the resting memory subsets capable of self-renewal are also prominent there. In contrast, nondividing but functionally active memory subsets dominate the spleen, and especially the ileum. In general, the replacement of naive cells with memory subsets continues throughout our period of observation, with no apparent plateau. In conclusion, the analysis of lymphoid and nonlymphoid tissues reveals a dynamic pattern of changes distinct to each tissue, and with substantial differences between CD4+ and CD8+ compartments.
Collapse
Affiliation(s)
- Nelli Heikkilä
- Translational Immunology Research Program (TRIMM), Research Programs Unit (RPU), Department of Bacteriology and Immunology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Iivo Hetemäki
- Translational Immunology Research Program (TRIMM), Research Programs Unit (RPU), Department of Bacteriology and Immunology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Silja Sormunen
- Department of Computer Science, Aalto University, Espoo, Finland
| | - Helena Isoniemi
- Division of Transplantation and Liver Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Eliisa Kekäläinen
- Translational Immunology Research Program (TRIMM), Research Programs Unit (RPU), Department of Bacteriology and Immunology, Medicum, Faculty of Medicine, HUSLAB Clinical Microbiology, University of Helsinki, and Helsinki University Central Hospital, Helsinki, Finland
| | - Jari Saramäki
- Department of Computer Science, Aalto University, Espoo, Finland
| | - T Petteri Arstila
- Translational Immunology Research Program (TRIMM), Research Programs Unit (RPU), Department of Bacteriology and Immunology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
31
|
Rajamanickam A, Pavan Kumar N, Pandiaraj AN, Selvaraj N, Munisankar S, Renji RM, Venkataramani V, Murhekar M, Thangaraj JWV, Muthusamy SK, Chethrapilly Purushothaman GK, Bhatnagar T, Ponnaiah M, Ramasamy S, Velusamy S, Babu S. Characterization of memory T cell subsets and common γ-chain cytokines in convalescent COVID-19 individuals. J Leukoc Biol 2022; 112:201-212. [PMID: 35258122 PMCID: PMC9088480 DOI: 10.1002/jlb.5cova0721-392rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/30/2021] [Accepted: 01/21/2022] [Indexed: 01/08/2023] Open
Abstract
T cells are thought to be an important correlates of protection against SARS‐CoV2 infection. However, the composition of T cell subsets in convalescent individuals of SARS‐CoV2 infection has not been well studied. The authors determined the lymphocyte absolute counts, the frequency of memory T cell subsets, and the plasma levels of common γ−chain in 7 groups of COVID‐19 individuals, based on days since RT‐PCR confirmation of SARS‐CoV‐2 infection. The data show that both absolute counts and frequencies of lymphocytes as well as, the frequencies of CD4+ central and effector memory cells increased, and the frequencies of CD4+ naïve T cells, transitional memory, stem cell memory T cells, and regulatory cells decreased from Days 15–30 to Days 61–90 and plateaued thereafter. In addition, the frequencies of CD8+ central memory, effector, and terminal effector memory T cells increased, and the frequencies of CD8+ naïve cells, transitional memory, and stem cell memory T cells decreased from Days 15–30 to Days 61–90 and plateaued thereafter. The plasma levels of IL‐2, IL‐7, IL‐15, and IL‐21—common γc cytokines started decreasing from Days 15–30 till Days 151–180. Severe COVID‐19 patients exhibit decreased levels of lymphocyte counts and frequencies, higher frequencies of naïve cells, regulatory T cells, lower frequencies of central memory, effector memory, and stem cell memory, and elevated plasma levels of IL‐2, IL‐7, IL‐15, and IL‐21. Finally, there was a significant correlation between memory T cell subsets and common γc cytokines. Thus, the study provides evidence of alterations in lymphocyte counts, memory T cell subset frequencies, and common γ−chain cytokines in convalescent COVID‐19 individuals.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | | | - Arul Nancy Pandiaraj
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Nandhini Selvaraj
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Saravanan Munisankar
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Rachel Mariam Renji
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Vijayalakshmi Venkataramani
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Manoj Murhekar
- National Institute of Epidemiology (ICMR), Near Ambattur, Ayapakkam, Chennai, India
| | | | | | | | - Tarun Bhatnagar
- National Institute of Epidemiology (ICMR), Near Ambattur, Ayapakkam, Chennai, India
| | - Manickam Ponnaiah
- National Institute of Epidemiology (ICMR), Near Ambattur, Ayapakkam, Chennai, India
| | | | | | - Subash Babu
- ICMR-NIRT-International Center for Excellence in Research, Chennai, India
| |
Collapse
|
32
|
Wonderlich ER, Reece MD, Kulpa DA. Ex Vivo Differentiation of Resting CD4+ T Lymphocytes Enhances Detection of Replication Competent HIV-1 in Viral Outgrowth Assays. Methods Mol Biol 2022; 2407:315-331. [PMID: 34985673 DOI: 10.1007/978-1-0716-1871-4_21] [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
Quantifying the number of cells harboring inducible and replication competent HIV-1 provirus is critical to evaluating HIV-1 cure interventions, but precise quantification of the latent reservoir has proven to be technically challenging. Existing protocols to quantify the frequency of replication-competent HIV-1 in resting CD4+ T cells from long-term ART treated individuals have helped to investigate the dynamics of reservoir stability, however these approaches have significant barriers to the induction of HIV-1 expression required to effectively evaluate the intact reservoir. Differentiation of CD4+ T cells to an effector memory phenotype is a successful strategy for promoting latency reversal in vitro, and significantly enhances the performance and sensitivity of viral outgrowth assays.
Collapse
Affiliation(s)
| | - Monica D Reece
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, and Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Deanna A Kulpa
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, and Yerkes National Primate Research Center, Atlanta, GA, USA.
| |
Collapse
|
33
|
Zhang G, Zheng G, Jiang F, Wu T, Wu L. Granzyme B and perforin produced by SEC2 mutant-activated human CD4 + T cells and CD8 + T cells induce apoptosis of K562 leukemic cells by the mitochondrial apoptotic pathway. Int J Biol Macromol 2021; 190:284-290. [PMID: 34492245 DOI: 10.1016/j.ijbiomac.2021.08.225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
Staphylococcal enterotoxin C2 (SEC2), a classical representative of superantigens, activates T cells that produce massive cytokines. This characteristic makes SEC2 a promising candidate drug for cancer immunotherapy. Previous study showed that ST-4, a SEC2 mutant, enhanced recognition of mouse T-cell receptor Vβ regions, and activated the increased number of T cells that produced more cytokines. However, the underlying molecular mechanism for stimulation of human peripheral blood mononuclear cells (PBMCs) and antitumor effect on human tumor cells remains unknown. Herein, we showed that ST-4 significantly activated TCR Vβ 12, 13A, 14, 15, 17, and 20 CD4+ and CD8+ T cells, which produced substantial amounts of granzyme B and perforin. These cytokines exhibited antitumor effect on K562 cells by promoting apoptosis and inducing S-phase cell cycle arrest. Conversely, the granzyme B inhibitor or perforin inhibitor significantly weakened antitumor effect of ST-4, accompanied by a decrease of cleaved proapoptotic BAX and cytochrome c, and an increase of antiapoptotic BCL2. Taken together, these data suggest that granzyme B and perforin produced by ST-4-activated CD4+ T cells and CD8+ T cells play a pivotal role in inducing K562 cell apoptosis by the mitochondrial apoptotic pathway, and support ST-4 as a potential candidate for cancer immunotherapy.
Collapse
Affiliation(s)
- Guojun Zhang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area District, 110122 Shenyang, Liaoning, People's Republic of China
| | - Guoliang Zheng
- Department of Gastric Surgery, Cancer Hospital of China Medical University (Liaoning Cancer Hospital and Institute), No. 44, Xiaoheyan Road, Shenhe District, 110042 Shenyang, Liaoning, People's Republic of China
| | - Fengli Jiang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area District, 110122 Shenyang, Liaoning, People's Republic of China
| | - Tianyi Wu
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area District, 110122 Shenyang, Liaoning, People's Republic of China
| | - Lizhao Wu
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area District, 110122 Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
34
|
Park J, Daniels J, Wartewig T, Ringbloom KG, Martinez-Escala ME, Choi S, Thomas JJ, Doukas PG, Yang J, Snowden C, Law C, Lee Y, Lee K, Zhang Y, Conran C, Tegtmeyer K, Mo SH, Pease DR, Jothishankar B, Kwok PY, Abdulla FR, Pro B, Louissaint A, Boggon TJ, Sosman J, Guitart J, Rao D, Ruland J, Choi J. Integrated genomic analyses of cutaneous T-cell lymphomas reveal the molecular bases for disease heterogeneity. Blood 2021; 138:1225-1236. [PMID: 34115827 PMCID: PMC8499046 DOI: 10.1182/blood.2020009655] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 05/20/2021] [Indexed: 11/20/2022] Open
Abstract
Cutaneous T-cell lymphomas (CTCLs) are a clinically heterogeneous collection of lymphomas of the skin-homing T cell. To identify molecular drivers of disease phenotypes, we assembled representative samples of CTCLs from patients with diverse disease subtypes and stages. Via DNA/RNA-sequencing, immunophenotyping, and ex vivo functional assays, we identified the landscape of putative driver genes, elucidated genetic relationships between CTCLs across disease stages, and inferred molecular subtypes in patients with stage-matched leukemic disease. Collectively, our analysis identified 86 putative driver genes, including 19 genes not previously implicated in this disease. Two mutations have never been described in any cancer. Functionally, multiple mutations augment T-cell receptor-dependent proliferation, highlighting the importance of this pathway in lymphomagenesis. To identify putative genetic causes of disease heterogeneity, we examined the distribution of driver genes across clinical cohorts. There are broad similarities across disease stages. Many driver genes are shared by mycosis fungoides (MF) and Sezary syndrome (SS). However, there are significantly more structural variants in leukemic disease, leading to highly recurrent deletions of putative tumor suppressors that are uncommon in early-stage skin-centered MF. For example, TP53 is deleted in 7% and 87% of MF and SS, respectively. In both human and mouse samples, PD1 mutations drive aggressive behavior. PD1 wild-type lymphomas show features of T-cell exhaustion. PD1 deletions are sufficient to reverse the exhaustion phenotype, promote a FOXM1-driven transcriptional signature, and predict significantly worse survival. Collectively, our findings clarify CTCL genetics and provide novel insights into pathways that drive diverse disease phenotypes.
Collapse
Affiliation(s)
- Joonhee Park
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Jay Daniels
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Tim Wartewig
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Munich, Germany
| | - Kimberly G Ringbloom
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | | | - Jane J Thomas
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Jingyi Yang
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Caroline Snowden
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Calvin Law
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Yujin Lee
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Katie Lee
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Yancong Zhang
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Kyle Tegtmeyer
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Samuel H Mo
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Balaji Jothishankar
- Department of Medicine, Section of Dermatology, University of Chicago Pritzker School of Medicine, Chicago, IL
| | - Pui-Yan Kwok
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Farah R Abdulla
- Division of Dermatology, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Barbara Pro
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Abner Louissaint
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Titus J Boggon
- Department of Pharmacology and
- Department of Molecular Biology and Biophysics, Yale University School of Medicine, New Haven, CT
| | - Jeffrey Sosman
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | | | - Deepak Rao
- Division of Rheumatology, Inflammation, Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany; and
- German Center for Infection Research (DZIF), Munich, Germany
| | - Jaehyuk Choi
- Department of Dermatology, and
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| |
Collapse
|
35
|
Cimen Bozkus C, Blazquez AB, Enokida T, Bhardwaj N. A T-cell-based immunogenicity protocol for evaluating human antigen-specific responses. STAR Protoc 2021; 2:100758. [PMID: 34458873 PMCID: PMC8377590 DOI: 10.1016/j.xpro.2021.100758] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Determining the antigen specificities of the endogenous T-cell repertoire is important for screening naturally occurring or therapy-induced T-cell immunity and may help identify novel targets for T-cell-based therapies. Here, we describe a rapid, sensitive, and high-throughput protocol for expanding antigen-specific T cells from human peripheral blood mononuclear cells in vitro following peptide stimulation and detecting antigen-specific effector cytokine formation by flow cytometry. Our approach can be applied to examining specific T-cell subsets from various tissues. For complete details on the use and execution of this protocol, please refer to Roudko et al. (2020) and Cimen Bozkus et al. (2019).
Collapse
Affiliation(s)
- Cansu Cimen Bozkus
- Department of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10127, USA
- Corresponding author
| | - Ana Belen Blazquez
- Department of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10127, USA
- Global Biological Specimen and Imaging Management (GBS&IM), Bristol Myers Squibb, Princeton, NJ 08540, USA
| | - Tomohiro Enokida
- Department of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10127, USA
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo, Chiba, Japan
| | - Nina Bhardwaj
- Department of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10127, USA
- Corresponding author
| |
Collapse
|
36
|
Singh DK, Tousif S, Bhaskar A, Devi A, Negi K, Moitra B, Ranganathan A, Dwivedi VP, Das G. Luteolin as a potential host-directed immunotherapy adjunct to isoniazid treatment of tuberculosis. PLoS Pathog 2021; 17:e1009805. [PMID: 34415976 PMCID: PMC8409628 DOI: 10.1371/journal.ppat.1009805] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 09/01/2021] [Accepted: 07/16/2021] [Indexed: 12/31/2022] Open
Abstract
Tuberculosis (TB) remains a major health problem throughout the world with one third of the population latently infected and ~1.74 million deaths annually. Current therapy consists of multiple antibiotics and a lengthy treatment regimen, which is associated with risk for the generation of drug-resistant Mycobacterium tuberculosis variants. Therefore, alternate host directed strategies that can shorten treatment length and enhance anti-TB immunity during the treatment phase are urgently needed. Here, we show that Luteolin, a plant-derived hepatoprotective immunomodulator, when administered along with isoniazid as potential host directed therapy promotes anti-TB immunity, reduces the length of TB treatment and prevents disease relapse. Luteolin also enhances long-term anti-TB immunity by promoting central memory T cell responses. Furthermore, we found that Luteolin enhances the activities of natural killer and natural killer T cells, both of which exhibit antitubercular attributes. Therefore, the addition of Luteolin to conventional antibiotic therapy may provide a means to avoid the development of drug-resistance and to improve disease outcome. The current TB therapy is lengthy, expensive, and may induce severe hepatotoxicity in patients, often leading to premature withdrawal from therapy, which is associated with the risk of generating drug-resistant strains. We considered inclusion of a hepatoprotective immunomodulator, Luteolin, as a potential host directed adjunct to available therapy as a means to improve its efficacy by enhancing host protective immunity. Luteolin-Isoniazid combination therapy induces improved central memory T cell responses. The boosted immune responses permitted reduction of treatment duration, improved treatment outcome and efficiently prevented disease relapse. Luteolin treatment rendered the host resistant against reinfection and reactivation of the disease, which is a major challenge following conventional TB treatment. We conclude that Luteolin is an effective adjunctive immunomodulator for designing anti-TB immunotherapeutics that can provide superior host protection.
Collapse
Affiliation(s)
- Dhiraj Kumar Singh
- Special Centre for Molecular Medicine (SCMM), Jawaharlal Nehru University, New Delhi, India
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Sultan Tousif
- Special Centre for Molecular Medicine (SCMM), Jawaharlal Nehru University, New Delhi, India
| | - Ashima Bhaskar
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Annu Devi
- Special Centre for Molecular Medicine (SCMM), Jawaharlal Nehru University, New Delhi, India
| | - Kriti Negi
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Barnani Moitra
- Special Centre for Molecular Medicine (SCMM), Jawaharlal Nehru University, New Delhi, India
| | - Anand Ranganathan
- Special Centre for Molecular Medicine (SCMM), Jawaharlal Nehru University, New Delhi, India
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Ved Prakash Dwivedi
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Gobardhan Das
- Special Centre for Molecular Medicine (SCMM), Jawaharlal Nehru University, New Delhi, India
- * E-mail:
| |
Collapse
|
37
|
Basar R, Uprety N, Ensley E, Daher M, Klein K, Martinez F, Aung F, Shanley M, Hu B, Gokdemir E, Nunez Cortes AK, Mendt M, Reyes Silva F, Acharya S, Laskowski T, Muniz-Feliciano L, Banerjee PP, Li Y, Li S, Melo Garcia L, Lin P, Shaim H, Yates SG, Marin D, Kaur I, Rao S, Mak D, Lin A, Miao Q, Dou J, Chen K, Champlin RE, Shpall EJ, Rezvani K. Generation of glucocorticoid-resistant SARS-CoV-2 T cells for adoptive cell therapy. Cell Rep 2021; 36:109432. [PMID: 34270918 PMCID: PMC8260499 DOI: 10.1016/j.celrep.2021.109432] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 04/15/2021] [Accepted: 06/30/2021] [Indexed: 12/15/2022] Open
Abstract
Adoptive cell therapy with virus-specific T cells has been used successfully to treat life-threatening viral infections, supporting application of this approach to coronavirus disease 2019 (COVID-19). We expand severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) T cells from the peripheral blood of COVID-19-recovered donors and non-exposed controls using different culture conditions. We observe that the choice of cytokines modulates the expansion, phenotype, and hierarchy of antigenic recognition by SARS-CoV-2 T cells. Culture with interleukin (IL)-2/4/7, but not under other cytokine-driven conditions, results in more than 1,000-fold expansion in SARS-CoV-2 T cells with a retained phenotype, function, and hierarchy of antigenic recognition compared with baseline (pre-expansion) samples. Expanded cytotoxic T lymphocytes (CTLs) are directed against structural SARS-CoV-2 proteins, including the receptor-binding domain of Spike. SARS-CoV-2 T cells cannot be expanded efficiently from the peripheral blood of non-exposed controls. Because corticosteroids are used for management of severe COVID-19, we propose an efficient strategy to inactivate the glucocorticoid receptor gene (NR3C1) in SARS-CoV-2 CTLs using CRISPR-Cas9 gene editing.
Collapse
Affiliation(s)
- Rafet Basar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nadima Uprety
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emily Ensley
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kimberly Klein
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fernando Martinez
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fleur Aung
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mayra Shanley
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bingqian Hu
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elif Gokdemir
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana Karen Nunez Cortes
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mayela Mendt
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francia Reyes Silva
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sunil Acharya
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tamara Laskowski
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luis Muniz-Feliciano
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pinaki P Banerjee
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ye Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sufang Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luciana Melo Garcia
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Lin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hila Shaim
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, USA
| | - Sean G Yates
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, USA
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Indreshpal Kaur
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sheetal Rao
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Duncan Mak
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Angelique Lin
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Miao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jinzhuang Dou
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
38
|
Thysen AH, Waage J, Larsen JM, Rasmussen MA, Stokholm J, Chawes B, Fink NR, Pedersen TM, Wolsk H, Thorsteinsdottir S, Litman T, Renz H, Bønnelykke K, Bisgaard H, Brix S. Distinct immune phenotypes in infants developing asthma during childhood. Sci Transl Med 2021; 12:12/529/eaaw0258. [PMID: 32024797 DOI: 10.1126/scitranslmed.aaw0258] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 08/22/2019] [Accepted: 01/10/2020] [Indexed: 12/16/2022]
Abstract
Early exposure to environmental triggers may elicit trajectories to chronic inflammatory disease through deregulated immune responses. To address relations between early immune competence and development of childhood asthma, we performed functional immune profiling of 186 parameters in blood of 541 18-month-old infants and examined links between their response phenotype and development of transient or persistent disease at 6 years of age. An abnormal neutrophil-linked antiviral response was associated with increased risk of transient asthma. Children who exhibited persistent asthma at year 6 showed enhanced interleukin-5 (IL-5) and IL-13 production in stimulated T cells at 18 months of age, which was associated with early life bacterial colonization of the airways. These findings highlight the early appearance of distinct immune characteristics in infants developing different asthma endotypes during childhood.
Collapse
Affiliation(s)
- Anna Hammerich Thysen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, DK-2200 Copenhagen, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Johannes Waage
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Jeppe Madura Larsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Morten Arendt Rasmussen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, DK-2200 Copenhagen, Denmark.,Department of Food Science, Faculty of Science, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Jakob Stokholm
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Bo Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Nadia Rahman Fink
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Tine Marie Pedersen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Helene Wolsk
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Sunna Thorsteinsdottir
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Thomas Litman
- Explorative Biology, LEO Pharma, DK-2750 Ballerup, Denmark
| | - Harald Renz
- Institute of Laboratory Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps Universität Marburg, German Center for Lung Research (DZL), 35043 Marburg, Germany
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
39
|
Matthews NC, Burton CS, Alfred A. Low-density neutrophils in chronic graft versus host disease (cGVHD) are primarily immature CD10 - and enhance T cell activation. Clin Exp Immunol 2021; 205:257-273. [PMID: 33932293 DOI: 10.1111/cei.13612] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/15/2021] [Accepted: 04/23/2021] [Indexed: 12/17/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is a frequent complication of allogeneic haematopoietic stem cell transplantation. Low density neutrophils (LDNs) in autoimmunity, which shares disease features with cGVHD, are proinflammatory, whereas those in cancer and sepsis suppress T cell immunity. Mature LDNs can be distinguished from immature LDNs on the basis of expression of CD10 and suppressive neutrophils can be identified using lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) expression. The functionality of LDNs in cGVHD has not been specifically investigated. Here, we have determined the relative contribution of immature and mature neutrophils to LDNs in cGVHD and assessed whether these were suppressive or potentially proinflammatory. Peripheral blood LDNs and normal density neutrophils (NDNs) from 30 cGVHD patients and NDNs from 10 healthy controls (HCs) were immunophenotyped by flow cytometry. The ability of LDNs and NDNs to influence T cell proliferation and cytokine production in co-cultures was quantified. To further characterize LDNs, their propensity to undergo constitutive apoptosis and differentiate ex vivo was assessed. LDNs were elevated in cGVHD versus HCs, heterogeneous in phenotype, with a predominance of immature CD10- cells in most patients, but some mature CD10+ LOX-1+ LDNs were also detected. LDNs enhanced autologous T cell proliferation, interleukin (IL)-6 and interferon (IFN)-γ production. LDN, but not NDN, CD10 expression was inversely correlated with LOX-1, which correlated with IL-6 production. LDNs resisted apoptosis and differentiated into antigen-presenting/neutrophil-hybrid-like cells, which co-expressed major histocompatibility complex (MHC) class II HLA-DR and immuno-inhibitory programmed cell death ligand 1 (PD-L1), but did not suppress T cell proliferation. These data suggest LDNs in cGVHD are predominantly immature, proinflammatory and may have pathogenic potential.
Collapse
Affiliation(s)
- Nick C Matthews
- Department of Haematology, The Rotherham NHS Foundation Trust, Rotherham, United Kingdom
| | - Charlotte S Burton
- Department of Haematology, The Rotherham NHS Foundation Trust, Rotherham, United Kingdom
| | - Arun Alfred
- Department of Haematology, The Rotherham NHS Foundation Trust, Rotherham, United Kingdom
| |
Collapse
|
40
|
Devi-Marulkar P, Moraes-Cabe C, Campagne P, Corre B, Meghraoui-Kheddar A, Bondet V, Llibre A, Duffy D, Maillart E, Papeix C, Pellegrini S, Michel F. Altered Immune Phenotypes and HLA-DQB1 Gene Variation in Multiple Sclerosis Patients Failing Interferon β Treatment. Front Immunol 2021; 12:628375. [PMID: 34113337 PMCID: PMC8185344 DOI: 10.3389/fimmu.2021.628375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/23/2021] [Indexed: 11/25/2022] Open
Abstract
Background Interferon beta (IFNβ) has been prescribed as a first-line disease-modifying therapy for relapsing-remitting multiple sclerosis (RRMS) for nearly three decades. However, there is still a lack of treatment response markers that correlate with the clinical outcome of patients. Aim To determine a combination of cellular and molecular blood signatures associated with the efficacy of IFNβ treatment using an integrated approach. Methods The immune status of 40 RRMS patients, 15 of whom were untreated and 25 that received IFNβ1a treatment (15 responders, 10 non-responders), was investigated by phenotyping regulatory CD4+ T cells and naïve/memory T cell subsets, by measurement of circulating IFNα/β proteins with digital ELISA (Simoa) and analysis of ~600 immune related genes including 159 interferon-stimulated genes (ISGs) with the Nanostring technology. The potential impact of HLA class II gene variation in treatment responsiveness was investigated by genotyping HLA-DRB1, -DRB3,4,5, -DQA1, and -DQB1, using as a control population the Milieu Interieur cohort of 1,000 French healthy donors. Results Clinical responders and non-responders displayed similar plasma levels of IFNβ and similar ISG profiles. However, non-responders mainly differed from other subject groups with reduced circulating naïve regulatory T cells, enhanced terminally differentiated effector memory CD4+ TEMRA cells, and altered expression of at least six genes with immunoregulatory function. Moreover, non-responders were enriched for HLA-DQB1 genotypes encoding DQ8 and DQ2 serotypes. Interestingly, these two serotypes are associated with type 1 diabetes and celiac disease. Overall, the immune signatures of non-responders suggest an active disease that is resistant to therapeutic IFNβ, and in which CD4+ T cells, likely restricted by DQ8 and/or DQ2, exert enhanced autoreactive and bystander inflammatory activities.
Collapse
Affiliation(s)
- Priyanka Devi-Marulkar
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Carolina Moraes-Cabe
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Pascal Campagne
- Center of Bioinformatics, Biostatistics and Integrative Biology, Institut Pasteur, Paris, France
| | - Béatrice Corre
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Aida Meghraoui-Kheddar
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Vincent Bondet
- Translational Immunology Laboratory, Department of Immunology, Institut Pasteur, Paris, France
| | - Alba Llibre
- Translational Immunology Laboratory, Department of Immunology, Institut Pasteur, Paris, France
| | - Darragh Duffy
- Translational Immunology Laboratory, Department of Immunology, Institut Pasteur, Paris, France
| | | | - Caroline Papeix
- Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France
| | - Sandra Pellegrini
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Frédérique Michel
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| |
Collapse
|
41
|
Zhao Y, Cai C, Samir J, Palgen JL, Keoshkerian E, Li H, Bull RA, Luciani F, An H, Lloyd AR. Human CD8 T-stem cell memory subsets phenotypic and functional characterization are defined by expression of CD122 or CXCR3. Eur J Immunol 2021; 51:1732-1747. [PMID: 33844287 DOI: 10.1002/eji.202049057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/07/2021] [Accepted: 04/07/2021] [Indexed: 11/12/2022]
Abstract
Long-lived T-memory stem cells (TSCM ) are key to both naturally occurring and vaccine-conferred protection against infection. These cells are characterized by the CD45RA+ CCR7+ CD95+ phenotype. Significant heterogeneity within the TSCM population is recognized, but distinguishing surface markers and functional characterization of potential subsets are lacking. Human CD8 TSCM subsets were identified in healthy subjects who had been previously exposed to CMV or Influenza (Flu) virus in flow cytometry by expression of CD122 or CXCR3, and then characterized in proliferation, multipotency, self-renewal, and intracellular cytokine production (TNF-α, IL-2, IFN-γ), together with transcriptomic profiles. The TSCM CD122hi -expressing subset (versus CD122lo ) demonstrated greater proliferation, greater multipotency, and enhanced polyfunctionality with higher frequencies of triple positive (TNF-α, IL-2, IFN-γ) cytokine-producing cells upon exposure to recall antigen. The TSCM CXCR3lo subpopulation also had increased proliferation and polyfunctional cytokine production. Transcriptomic analysis further showed that the TSCM CD122hi population had increased expression of activation and homing molecules, such as Ccr6, Cxcr6, Il12rb, and Il18rap, and downregulated cell proliferation inhibitors, S100A8 and S100A9. These data reveal that the TSCM CD122hi phenotype is associated with increased proliferation, enhanced multipotency and polyfunctionality with an activated memory-cell like transcriptional profile, and hence, may be favored for induction by immunization and for adoptive immunotherapy.
Collapse
Affiliation(s)
- Yanran Zhao
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Curtis Cai
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Jerome Samir
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Jean-Louis Palgen
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Elizabeth Keoshkerian
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Hui Li
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Rowena A Bull
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Fabio Luciani
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia.,School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Hongyan An
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Andrew R Lloyd
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| |
Collapse
|
42
|
Hirahara K, Kokubo K, Aoki A, Kiuchi M, Nakayama T. The Role of CD4 + Resident Memory T Cells in Local Immunity in the Mucosal Tissue - Protection Versus Pathology. Front Immunol 2021; 12:616309. [PMID: 33968018 PMCID: PMC8097179 DOI: 10.3389/fimmu.2021.616309] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/25/2021] [Indexed: 01/12/2023] Open
Abstract
Memory T cells are crucial for both local and systemic protection against pathogens over a long period of time. Three major subsets of memory T cells; effector memory T (TEM) cells, central memory T (TCM) cells, and tissue-resident memory T (TRM) cells have been identified. The most recently identified subset, TRM cells, is characterized by the expression of the C-type lectin CD69 and/or the integrin CD103. TRM cells persist locally at sites of mucosal tissue, such as the lung, where they provide frontline defense against various pathogens. Importantly, however, TRM cells are also involved in shaping the pathology of inflammatory diseases. A number of pioneering studies revealed important roles of CD8+ TRM cells, particularly those in the local control of viral infection. However, the protective function and pathogenic role of CD4+ TRM cells that reside within the mucosal tissue remain largely unknown. In this review, we discuss the ambivalent feature of CD4+ TRM cells in the protective and pathological immune responses. We also review the transcriptional and epigenetic characteristics of CD4+ TRM cells in the lung that have been elucidated by recent technical approaches. A better understanding of the function of CD4+ TRM cells is crucial for the development of both effective vaccination against pathogens and new therapeutic strategies for intractable inflammatory diseases, such as inflammatory bowel diseases and chronic allergic diseases.
Collapse
Affiliation(s)
- Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,AMED-PRIME, Japan Agency for Medical Research and Development, Chiba, Japan
| | - Kota Kokubo
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ami Aoki
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masahiro Kiuchi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Chiba, Japan
| |
Collapse
|
43
|
Kellogg C, Equils O. The role of the thymus in COVID-19 disease severity: implications for antibody treatment and immunization. Hum Vaccin Immunother 2021; 17:638-643. [PMID: 33064620 PMCID: PMC7993178 DOI: 10.1080/21645515.2020.1818519] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/27/2020] [Indexed: 12/20/2022] Open
Abstract
The thymus is a largely neglected organ but plays a significant role in the regulation of adaptive immune responses. The effect of aging on the thymus and immune senescence is well established, and the resulting inflammaging is found to be implicated in the development of many chronic diseases including atherosclerosis, hypertension and type 2 diabetes. Both aging and diseases of inflammaging are associated with severe COVID-19 disease, and a dysfunctional thymus may be a predisposing factor. In addition, insults on the thymus during childhood may lead to abnormal thymic function and may explain severe COVID-19 disease among younger individuals; therefore, measurement of thymic function may assist COVID-19 care. Those with poor thymic function may be treated prophylactically with convalescent serum or recombinant antibodies, and they may respond better to high-dose or adjuvanted COVID-19 vaccines. Treatments inducing thymic regeneration may improve patients' overall health and may be incorporated in COVID-19 management.
Collapse
Affiliation(s)
- Caitlyn Kellogg
- University of California, San Diego School of Medicine, San Diego, CA, USA
- Public Health Education , MiOra Foundation, Los Angeles, CA, USA
| | - Ozlem Equils
- Public Health Education , MiOra Foundation, Los Angeles, CA, USA
| |
Collapse
|
44
|
Ji N, Mukherjee N, Reyes RM, Gelfond J, Javors M, Meeks JJ, McConkey DJ, Shu ZJ, Ramamurthy C, Dennett R, Curiel TJ, Svatek RS. Rapamycin enhances BCG-specific γδ T cells during intravesical BCG therapy for non-muscle invasive bladder cancer: a randomized, double-blind study. J Immunother Cancer 2021; 9:jitc-2020-001941. [PMID: 33653802 PMCID: PMC7929866 DOI: 10.1136/jitc-2020-001941] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Although intravesical BCG is the standard treatment of high-grade non-muscle invasive bladder cancer (NMIBC), response rates remain unsatisfactory. In preclinical models, rapamycin enhances BCG vaccine efficacy against tuberculosis and the killing capacity of γδ T cells, which are critical for BCG's antitumor effects. Here, we monitored immunity, safety, and tolerability of rapamycin combined with BCG in patients with NMIBC. METHODS A randomized double-blind trial of oral rapamycin (0.5 or 2.0 mg daily) versus placebo for 1 month was conducted in patients with NMIBC concurrently receiving 3 weekly BCG instillations (NCT02753309). The primary outcome was induction of BCG-specific γδ T cells, measured as a percentage change from baseline. Post-BCG urinary cytokines and immune cells were examined as surrogates for local immune response in the bladder. Secondary outcomes measured were adverse events (AEs) and tolerability using validated patient-reported questionnaires. RESULTS Thirty-one patients were randomized (11 placebo, 8 rapamycin 2.0 mg, and 12 rapamycin 0.5 mg). AEs were similar across groups and most were grade 1-2. One (12.5%) patient randomized to 2.0 mg rapamycin was taken off treatment due to stomatitis. No significant differences in urinary symptoms, bowel function, or bother were observed between groups. The median (IQR) percentage change in BCG-specific γδ T cells from baseline per group was as follows: -26% (-51% to 24%) for placebo, 9.6% (-59% to 117%) for rapamycin 0.5 mg (versus placebo, p=0.18), and 78.8% (-31% to 115%) for rapamycin 2.0 mg (versus placebo, p=0.03). BCG-induced cytokines showed a progressive increase in IL-8 (p=0.02) and TNF-α (p=0.04) over time for patients on rapamycin 2.0 mg, whereas patients receiving placebo had no significant change in urinary cytokines. Compared with placebo, patients receiving 2.0 mg rapamycin had increased urinary γδ T cells at the first week of BCG (p=0.02). CONCLUSIONS Four weeks of 0.5 and 2.0 mg oral rapamycin daily is safe and tolerable in combination with BCG for patients with NMIBC. Rapamycin enhances BCG-specific γδ T cell immunity and boosts urinary cytokines during BCG treatment. Further study is needed to determine long-term rapamycin safety, tolerability and effects on BCG efficacy.
Collapse
Affiliation(s)
- Niannian Ji
- Experimental Developmental Therapeutics (EDT) Program, Mays Cancer Center at UT Health MD Anderson, San Antonio, Texas, USA.,Department of Urology, UT Health San Antonio, San Antonio, Texas, USA
| | - Neelam Mukherjee
- Experimental Developmental Therapeutics (EDT) Program, Mays Cancer Center at UT Health MD Anderson, San Antonio, Texas, USA.,Department of Urology, UT Health San Antonio, San Antonio, Texas, USA
| | - Ryan M Reyes
- Experimental Developmental Therapeutics (EDT) Program, Mays Cancer Center at UT Health MD Anderson, San Antonio, Texas, USA.,Division of Hematology/Medical Oncology, UT Health San Antonio, San Antonio, Texas, USA
| | - Jonathan Gelfond
- Department of Epidemiology and Biostatistics, UT Health San Antonio, San Antonio, Texas, USA
| | - Martin Javors
- Department of Psychiatry, UT Health San Antonio, San Antonio, Texas, USA
| | - Joshua J Meeks
- Departments of Urology, and Biochemistry and Molecular Genetics, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - David J McConkey
- Greenberg Bladder Cancer Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zhen-Ju Shu
- Experimental Developmental Therapeutics (EDT) Program, Mays Cancer Center at UT Health MD Anderson, San Antonio, Texas, USA.,Department of Urology, UT Health San Antonio, San Antonio, Texas, USA
| | - Chethan Ramamurthy
- Experimental Developmental Therapeutics (EDT) Program, Mays Cancer Center at UT Health MD Anderson, San Antonio, Texas, USA.,Division of Hematology/Medical Oncology, UT Health San Antonio, San Antonio, Texas, USA
| | - Ryan Dennett
- Experimental Developmental Therapeutics (EDT) Program, Mays Cancer Center at UT Health MD Anderson, San Antonio, Texas, USA.,Department of Urology, UT Health San Antonio, San Antonio, Texas, USA
| | - Tyler J Curiel
- Experimental Developmental Therapeutics (EDT) Program, Mays Cancer Center at UT Health MD Anderson, San Antonio, Texas, USA.,Division of Hematology/Medical Oncology, UT Health San Antonio, San Antonio, Texas, USA
| | - Robert S Svatek
- Experimental Developmental Therapeutics (EDT) Program, Mays Cancer Center at UT Health MD Anderson, San Antonio, Texas, USA .,Department of Urology, UT Health San Antonio, San Antonio, Texas, USA
| |
Collapse
|
45
|
Topham DJ, DeDiego ML, Nogales A, Sangster MY, Sant A. Immunity to Influenza Infection in Humans. Cold Spring Harb Perspect Med 2021; 11:a038729. [PMID: 31871226 PMCID: PMC7919402 DOI: 10.1101/cshperspect.a038729] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review discusses the human immune responses to influenza infection with some insights from studies using animal models, such as experimental infection of mice. Recent technological advances in the study of human immune responses have greatly added to our knowledge of the infection and immune responses, and therefore much of the focus is on recent studies that have moved the field forward. We consider the complexity of the adaptive response generated by many sequential encounters through infection and vaccination.
Collapse
Affiliation(s)
- David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Marta L DeDiego
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Cientificas, 28049 Madrid, Spain
| | - Aitor Nogales
- Instituto Nacional de Investigación y Tecnologia Agraria y Ailmentaria, 28040 Madrid, Spain
| | - Mark Y Sangster
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Andrea Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| |
Collapse
|
46
|
Fatma F, Tripathi DK, Srivastava M, Srivastava KK, Arora A. Immunological characterization of chimeras of high specificity antigens from Mycobacterium tuberculosis H37Rv. Tuberculosis (Edinb) 2021; 127:102054. [PMID: 33550109 DOI: 10.1016/j.tube.2021.102054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 12/20/2020] [Accepted: 01/21/2021] [Indexed: 10/22/2022]
Abstract
Tuberculosis remains a serious global health problem. BCG is the only prophylactic TB vaccine and it shows variable protective efficacy. Chimeric protein subunit vaccines hold great potential as stand-alone vaccines or heterologous BCG prime boosters. We have designed a protein chimera, PP31, by combining Mtb ESAT-6 family antigen Rv1198 and MoCo biosynthesis family antigen Rv3111. Further, PP31 was extended by addition of latency antigen Rv1813c to yield PP43. Immunization of BALB/c mice with PP31 or PP43 with FIA adjuvant elicited strong humoral immune response. Restimulation of splenocytes of the immunized mice lead to significant proliferation of lymphocytes, secretion of cytokines IFN-γ, TNF, IL-2 of the Th1 class, IL-17A of the Th17 class, and IL-6. PP31 and PP43 also induced intracellular cytokine expression (IFN-γ, TNF, and IL-2) from both CD4+-CD44high and CD8+-CD44high T-cells. Antigen-specific IFN-γ+/IL-2+ double positive CD4+ T-cells were significantly higher in case of PP43 than PP31-immunized mice and control group. PP43 showed protection equivalent to heat-inactivated BCG in response to challenge of the immunized mice with Mtb H37Ra. Based on its immunogenicity and protective efficacy, PP43 appears to be a potential candidate for further development as a subunit vaccine against TB.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Bacterial/blood
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/immunology
- Bacterial Proteins/administration & dosage
- Bacterial Proteins/immunology
- Cell Proliferation/drug effects
- Cells, Cultured
- Cytokines/metabolism
- Epitopes
- Female
- Humans
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunization
- Immunogenicity, Vaccine
- Lymphocyte Activation/drug effects
- Lymphocyte Subsets/drug effects
- Lymphocyte Subsets/immunology
- Lymphocyte Subsets/metabolism
- Mice, Inbred BALB C
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/immunology
- Tuberculosis/blood
- Tuberculosis/immunology
- Tuberculosis/microbiology
- Tuberculosis/prevention & control
- Tuberculosis Vaccines/administration & dosage
- Tuberculosis Vaccines/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
- Mice
Collapse
Affiliation(s)
- Farheen Fatma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Dinesh K Tripathi
- Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Mrigank Srivastava
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Kishore K Srivastava
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
| | - Ashish Arora
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
47
|
Chauhan P, Nair A, Patidar A, Dandapat J, Sarkar A, Saha B. A primer on cytokines. Cytokine 2021; 145:155458. [PMID: 33581983 DOI: 10.1016/j.cyto.2021.155458] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/19/2022]
Abstract
Cytokines are pleiotropic polypeptides that control the development of and responses mediated by immune cells. Cytokine classification predominantly relies on [1] the target receptor(s), [2] the primary structural features of the extracellular domains of their receptors, and [3] their receptor composition. Functionally, cytokines are either pro-inflammatory or anti-inflammatory, hematopoietic colony-stimulating factors, developmental and would healing maintaining immune homeostasis. When the balance in C can form complex networks amongst themselves that may affect the homeostasis and diseases. Cytokines can affect resistance and susceptibility for many diseases and their availability in the host cytokine production and interaction is disturbed, immunopathogenesis sets in. Therefore, cytokine-targeting bispecific, and chimeric antibodies form a significant mode of immnuo-therapeutics Although the field has grown deep and wide, many areas of cytokine biology remain unknown. Here, we have reviewed these cytokines along with the organization, signaling, and functions through respective cytokine-receptor-families. Being part of the special issue on the Role of Cytokines in Leishmaniasis, this review is intended to be used as an organized primer on cytokines and not a resource for detailed discussion- for which a two-volume Handbook of cytokines is available- on each of the cytokines. Priming the readers on cytokines, we next brief the role of cytokines in Leishmaniasis. In the brief, we do not provide an account of each of the involved cytokines known to date, instead, we offer a temporal relationship between the cytokines and the progress of the infection towards the alternate outcomes- healing or non-healing- of the infection.
Collapse
Affiliation(s)
- Prashant Chauhan
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Arathi Nair
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Ashok Patidar
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Jagneshwar Dandapat
- P.G. Department of Biotechnology, Utkal University, Bhubaneswar 751004, India
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar 751024, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India; Trident Academy of Creative Technology, Bhubaneswar 751024, India; Department of Allied Health Sciences, BLDE (Deemed University), Vijayapura 562135, India.
| |
Collapse
|
48
|
Kathamuthu GR, Moideen K, Sridhar R, Baskaran D, Babu S. Altered plasma levels of βC and γC chain cytokines and post-treatment modulation in tuberculous lymphadenitis. Cytokine 2020; 138:155405. [PMID: 33341600 DOI: 10.1016/j.cyto.2020.155405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Alterations in β common (βC) and γ common (γC) chain cytokines have been described in pulmonary tuberculosis. However, their role in tuberculous lymphadenitis (TBL) disease has not been assessed. METHODS Thus, in the present study, we have examined the systemic levels of βC and γC chain cytokines in TBL, latent tuberculosis (LTB) and healthy control (HC) individuals. We have examined the discriminatory potential of both family of cytokines using ROC analysis. Finally, we measured the pre and post-treatment responses of these cytokines after anti-tuberculosis treatment. RESULTS TBL individuals exhibit significantly increased (IL-3) and diminished systemic levels of (IL-5, GM-CSF) βC cytokines compared to LTB and HC individuals. TBL individuals also exhibit significantly diminished (IL-2, IL-7) and elevated (IL-4, IL-9) levels of γC cytokines compared to LTB and/or HC. ROC analysis shows a clear discriminatory capacity of both βC (IL-5) and γC (IL-2) chain cytokines to distinguish TBL from LTB and HCs. The systemic levels of βC chain cytokines were not significantly altered, but in contrast γC (IL-2 and IL-7) cytokines were significantly modulated after treatment. Finally, no significant correlation was observed for βC and γC chain cytokines with their respective lymphocyte count of TBL individuals. CONCLUSIONS Hence, we conclude that altered plasma levels of βC and γC cytokines are the characteristics of immune alteration in TBL disease and certain cytokines were modulated after treatment.
Collapse
Affiliation(s)
- Gokul Raj Kathamuthu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India; National Institute for Research in Tuberculosis (NIRT), Chennai, India.
| | - Kadar Moideen
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India
| | | | - Dhanaraj Baskaran
- National Institute for Research in Tuberculosis (NIRT), Chennai, India
| | - Subash Babu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India; Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
49
|
Minamihata K, Hamada Y, Kagawa G, Ramadhan W, Higuchi A, Moriyama K, Wakabayashi R, Goto M, Kamiya N. Dual-Functionalizable Streptavidin-SpyCatcher-Fused Protein-Polymer Hydrogels as Scaffolds for Cell Culture. ACS APPLIED BIO MATERIALS 2020; 3:7734-7742. [PMID: 35019513 DOI: 10.1021/acsabm.0c00940] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Hydrogels possessing the ability to control cell functions have great potential as artificial substrates for cell culture. Herein, we report dual-functionalizable protein-polymer hybrid hydrogels prepared by thiol oxidation catalyzed by horseradish peroxidase and a phenolic molecule. A chimera protein of streptavidin (SA) and the SpyCatcher protein, with a cysteine residue at its N-terminus, (C-SA-SC) was constructed and co-cross-linked with thiol-functionalized four-arm polyethylene glycol (PEG-SH) to obtain hydrogels possessing two orthogonal conjugation moieties. Hydrogel formation using C-SA-SC conjugated with biotinylated or SpyTagged functional molecules (premodification strategy) resulted in the formation of hydrogels with a uniform distribution of the functional molecules. Postmodification of the functional molecules of the C-SA-SC hydrogel with biotin or SpyTag could alter the three-dimensional (3D) spatial distribution of the functional molecules within the hydrogels depending on the mode of conjugation (SA/biotin or SpyCatcher/SpyTag), the size of the functional molecules, and the length of time of the modification. NIH-3T3 cells cultured on a C-SA-SC hydrogel, dual-functionalized with a biotinylated-Arg-Gly-Asp-Ser (RGDS) peptide and a basic fibroblast growth factor (bFGF) with SpyTag, showed cell adhesion to the PEG-SH-based hydrogels and cell morphological changes in response to the immobilized RGDS peptide and the bFGF. Moreover, the cells showed higher proliferation on the dual-functionalized C-SA-SC hydrogel than the cells cultured on hydrogels without either the RGDS peptide or the bFGF, demonstrating the benefits of dual-functionalizable hydrogels. The C-SA-SC hydrogel presented in this study is capable of being orthogonally functionalized by two different functional molecules with different 3D distributions of each molecule within the hydrogel and thus has the potential for use as a cell culturing scaffold for creating artificial cellular microstructures.
Collapse
Affiliation(s)
- Kosuke Minamihata
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yusei Hamada
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Genki Kagawa
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Wahyu Ramadhan
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Ayato Higuchi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Kousuke Moriyama
- Department of Chemical and Biological Engineering, National Institute of Technology, Sasebo College, Okishin-cho, Sasebo, Nagasaki 857-1193, Japan
| | - Rie Wakabayashi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masahiro Goto
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Noriho Kamiya
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
50
|
Kalfaoglu B, Almeida-Santos J, Tye CA, Satou Y, Ono M. T-cell dysregulation in COVID-19. Biochem Biophys Res Commun 2020; 538:204-210. [PMID: 33220925 PMCID: PMC7648511 DOI: 10.1016/j.bbrc.2020.10.079] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
T-cells play key roles in immunity to COVID-19 as well as the development of severe disease. T-cell immunity to COVID-19 is mediated through differentiated CD4+ T-cells and cytotoxic CD8+ T-cells, although their differentiation is often atypical and ambiguous in COVID-19 and single cell dynamics of key genes need to be characterized. Notably, T-cells are dysregulated in severe COVID-19 patients, although their molecular features are still yet to be fully revealed. Importantly, it is not clear which T-cell activities are beneficial and protective and which ones can contribute to the development of severe COVID-19. In this article, we examine the latest evidence and discuss the key features of T-cell responses in COVID-19, showing how T-cells are dysregulated in severe COVID-19 patients. Particularly, we highlight the impairment of FOXP3 induction in CD4+ T-cells and how the impaired FOXP3 expression can lead to the differentiation of abnormally activated (hyperactivated) T-cells and the dysregulated T-cell responses in severe patients. Furthermore, we characterise the feature of hyperactivated T-cells, showing their potential contribution to T-cell dysregulation and immune-mediated tissue destruction (immunopathology) in COVID-19.
Collapse
Affiliation(s)
| | - José Almeida-Santos
- Department of Life Sciences, Imperial College London, UK; Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | - Yorifumi Satou
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Japan
| | - Masahiro Ono
- Department of Life Sciences, Imperial College London, UK; International Research Center for Medical Sciences, Kumamoto University, Japan.
| |
Collapse
|