1
|
Zhu E, Yu J, Li YR, Ma F, Wang YC, Liu Y, Li M, Kim YJ, Zhu Y, Hahn Z, Zhou Y, Brown J, Zhang Y, Pelegrini M, Hsiai T, Yang L, Huang Y. Biomimetic cell stimulation with a graphene oxide antigen-presenting platform for developing T cell-based therapies. NATURE NANOTECHNOLOGY 2024:10.1038/s41565-024-01781-4. [PMID: 39313679 DOI: 10.1038/s41565-024-01781-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/08/2024] [Indexed: 09/25/2024]
Abstract
Chimeric antigen receptor (CAR)-engineered T cells represent a front-line therapy for cancers. However, the current CAR T cell manufacturing protocols do not adequately reproduce immunological synapse formation. Here, in response to this limitation, we have developed a flexible graphene oxide antigen-presenting platform (GO-APP) that anchors antibodies onto graphene oxide. By decorating anti-CD3 (αCD3) and anti-CD28 (αCD28) on graphene oxide (GO-APP3/28), we achieved remarkable T cell proliferation. In vitro interactions between GO-APP3/28 and T cells closely mimic the in vivo immunological synapses between antigen-presenting cells and T cells. This immunological synapse mimicry shows a high capacity for stimulating T cell proliferation while preserving their multifunctionality and high potency. Meanwhile, it enhances CAR gene-engineering efficiency, yielding a more than fivefold increase in CAR T cell production compared with the standard protocol. Notably, GO-APP3/28 stimulated appropriate autocrine interleukin-2 (IL-2) in T cells and overcame the in vitro reliance on external IL-2 supplementation, offering an opportunity to culture T cell-based products independent of IL-2 supplementation.
Collapse
Affiliation(s)
- Enbo Zhu
- Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jiaji Yu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yan-Ruide Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Feiyang Ma
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yu-Chen Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yang Liu
- Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Miao Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yu Jeong Kim
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zoe Hahn
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yang Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - James Brown
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yuchong Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matteo Pelegrini
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tzung Hsiai
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Yu Huang
- Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Biemond M, Vremec D, Gray DHD, Hodgkin PD, Heinzel S. Programmed death receptor 1 (PD-1) ligand Fc fusion proteins reduce T-cell proliferation in vitro independently of PD-1. Immunol Cell Biol 2024; 102:117-130. [PMID: 38069638 PMCID: PMC10952853 DOI: 10.1111/imcb.12714] [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: 05/23/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 02/02/2024]
Abstract
Programmed death receptor 1 (PD-1) is an inhibitory receptor on T cells shown to restrain T-cell proliferation. PD-1 immune checkpoint blockade has emerged as a highly promising approach in cancer treatment. Much of our understanding of the function of PD-1 is derived from in vitro T-cell activation assays. Here we set out to further investigate how T cells integrate inhibitory signals such as PD-1 in vitro using the PD-1 agonist, PD-1 ligand 1 (PD-L1) fusion protein (PD-L1.Fc), coimmobilized alongside anti-CD3 agonist monoclonal antibody (mAb) on plates to deliver PD-1 signals to wild-type and PD-1-/- CD8+ T cells. Surprisingly, we found that the PD-L1.Fc fusion protein inhibited T-cell proliferation independently of PD-1. This PD-L1.Fc inhibition was observed in the presence and absence of CD28 and interleukin-2 signaling. Binding of PD-L1.Fc was restricted to PD-1-expressing T cells and thus inhibition was not mediated by the interaction of PD-L1.Fc with CD80 or other yet unknown binding partners. Furthermore, a similar PD-1-independent reduction of T-cell proliferation was observed with plate-bound PD-L2.Fc. Hence, our results suggest that the coimmobilization of PD-1 ligand fusion proteins with anti-CD3 mAb leads to a reduction of T-cell engagement with plate-bound anti-CD3 mAb. This study demonstrates a nonspecific mechanism of T-cell inhibition when PD-L1.Fc or PD-L2.Fc fusion proteins are delivered in a plate-bound coimmobilization assay and highlights the importance of careful optimization of assay systems and reagents when interpreting their influence on T-cell proliferation.
Collapse
Affiliation(s)
- Melissa Biemond
- Immunology DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVICAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleVICAustralia
- Present address:
Department of ImmunologyLeiden University Medical CenterLeidenThe Netherlands
| | - David Vremec
- Immunology DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVICAustralia
| | - Daniel HD Gray
- Immunology DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVICAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleVICAustralia
| | - Philip D Hodgkin
- Immunology DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVICAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleVICAustralia
| | - Susanne Heinzel
- Immunology DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVICAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleVICAustralia
| |
Collapse
|
3
|
Heinzel S, Cheon H, Belz GT, Hodgkin PD. Survival and division fate programs are preserved but retuned during the naïve to memory CD8 + T-cell transition. Immunol Cell Biol 2024; 102:46-57. [PMID: 37840018 PMCID: PMC10952575 DOI: 10.1111/imcb.12699] [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: 08/11/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/17/2023]
Abstract
Memory T cells are generated from naïve precursors undergoing proliferation during the initial immune response. Both naïve and memory T cells are maintained in a resting, quiescent state and respond to activation with a controlled proliferative burst and differentiation into effector cells. This similarity in the maintenance and response dynamics points to the preservation of key cellular fate programs; however, whether memory T cells have acquired intrinsic changes in these programs that may contribute to the enhanced immune protection in a recall response is not fully understood. Here we used a quantitative model-based analysis of proliferation and survival kinetics of in vitro-stimulated murine naïve and memory CD8+ T cells in response to homeostatic and activating signals to establish intrinsic similarities or differences within these cell types. We show that resting memory T cells display heightened sensitivity to homeostatic cytokines, responding to interleukin (IL)-2 in addition to IL-7 and IL-15. The proliferative response to αCD3 was equal in size and kinetics, demonstrating that memory T cells undergo the same controlled division burst and automated return to quiescence as naïve T cells. However, perhaps surprisingly, we observed reduced expansion of αCD3-stimulated memory T cells in response to activating signals αCD28 and IL-2 compared with naïve T cells. Overall, we demonstrate that although sensitivities to cytokine and costimulatory signals have shifted, fate programs regulating the scale of the division burst are conserved in memory T cells.
Collapse
Affiliation(s)
- Susanne Heinzel
- Immunology DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVICAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleVICAustralia
| | - HoChan Cheon
- Immunology DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVICAustralia
| | - Gabrielle T Belz
- Immunology DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVICAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleVICAustralia
- Frazer InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Philip D Hodgkin
- Immunology DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVICAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleVICAustralia
| |
Collapse
|
4
|
Farchione AJ, Cheon H, Hodgkin PD, Bryant VL. Quantifying Human Naïve B Cell Proliferation Kinetics and Differentiation in Controlled In Vitro Cell Culture. Methods Mol Biol 2024; 2826:167-187. [PMID: 39017893 DOI: 10.1007/978-1-0716-3950-4_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Division tracking dyes like Cell Trace Violet (CTV) enable the quantification of cell proliferation, division, and survival kinetics of human naïve B cell responses in vitro. Human naïve B cells exhibit distinct responses to different stimuli, with CpG and anti-Ig inducing a T cell-independent (TI) response, while CD40L and IL-21 promote a T cell-dependent (TD) response that induces isotype switching and differentiation into antibody-secreting cells (ASCs). Both stimulation methods yield valuable insights into the intrinsic programming of B cell health within individuals, making them useful for clinical investigations. For instance, quantitative analysis from these B cell populations could reveal biologically meaningful measurements such as the average number of division rounds and the time to cells' fate. Here, we describe a novel in vitro culture setup for CTV-labelled human naïve B cells and a method for obtaining precise time-based data on proliferation, division-linked isotype switching, and differentiation.
Collapse
Affiliation(s)
- Anthony J Farchione
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - HoChan Cheon
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Philip D Hodgkin
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Vanessa L Bryant
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
- Department Clinical Immunology and Allergy, The Royal Melbourne Hospital, Parkville, VIC, Australia.
| |
Collapse
|
5
|
Straube R, Schmidt BJ. A mean-field description for the expansion kinetics of activated T cell populations. Proc Natl Acad Sci U S A 2023; 120:e2305774120. [PMID: 37910551 PMCID: PMC10636355 DOI: 10.1073/pnas.2305774120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 09/22/2023] [Indexed: 11/03/2023] Open
Abstract
When lymphocytes encounter their cognate antigen, they become activated and undergo a limited number of cell divisions during which they differentiate into memory or effector cells or die. While the dynamics of individual cells are often heterogeneous, the expansion kinetics at the population level are highly reproducible, suggesting a mean-field description. To generate a finite division destiny, we consider two scenarios: Cells stop dividing after a certain number of iterations or their death rate increases with each cell division. The dynamics of the combined system can be mapped to a partial differential equation, and for a suitable choice of the activation rate, we obtain simple analytical solutions for the total cell number and the mean number of divisions per cell which can well describe the signal-dependent T cell expansion kinetics from in vitro experiments. Interestingly, only the division cessation mechanism yields an expression for the division destiny that does not contradict experiments. We show that the generation-dependent decrease of the division rate in individual cells leads to a time-dependent decrease at the population level which is consistent with a "time-to-die" control mechanism for the division destiny as suggested previously. We also derive mean-field equations for the total cell number which provide a basis for implementing T cell expansion kinetics into quantitative systems pharmacology models for immuno-oncology and CAR-T cell therapies.
Collapse
Affiliation(s)
- Ronny Straube
- Department of Clinical Pharmacology, Pharmacometrics, & Bioanalysis, Bristol Myers Squibb, Princeton, NJ08540
| | - Brian J. Schmidt
- Department of Clinical Pharmacology, Pharmacometrics, & Bioanalysis, Bristol Myers Squibb, Princeton, NJ08540
| |
Collapse
|
6
|
Guo M, Abd-Rabbo D, Bertol BC, Carew M, Lukhele S, Snell LM, Xu W, Boukhaled GM, Elsaesser H, Halaby MJ, Hirano N, McGaha TL, Brooks DG. Molecular, metabolic, and functional CD4 T cell paralysis in the lymph node impedes tumor control. Cell Rep 2023; 42:113047. [PMID: 37651234 PMCID: PMC10578141 DOI: 10.1016/j.celrep.2023.113047] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 07/14/2023] [Accepted: 08/11/2023] [Indexed: 09/02/2023] Open
Abstract
CD4 T cells are central effectors of anti-cancer immunity and immunotherapy, yet the regulation of CD4 tumor-specific T (TTS) cells is unclear. We demonstrate that CD4 TTS cells are quickly primed and begin to divide following tumor initiation. However, unlike CD8 TTS cells or exhaustion programming, CD4 TTS cell proliferation is rapidly frozen in place by a functional interplay of regulatory T cells and CTLA4. Together these mechanisms paralyze CD4 TTS cell differentiation, redirecting metabolic circuits, and reducing their accumulation in the tumor. The paralyzed state is actively maintained throughout cancer progression and CD4 TTS cells rapidly resume proliferation and functional differentiation when the suppressive constraints are alleviated. Overcoming their paralysis established long-term tumor control, demonstrating the importance of rapidly crippling CD4 TTS cells for tumor progression and their potential restoration as therapeutic targets.
Collapse
Affiliation(s)
- Mengdi Guo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Diala Abd-Rabbo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Bruna C Bertol
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Madeleine Carew
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sabelo Lukhele
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Laura M Snell
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Microbiology and Immunology and Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wenxi Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Giselle M Boukhaled
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Heidi Elsaesser
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Marie Jo Halaby
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Naoto Hirano
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Tracy L McGaha
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - David G Brooks
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
7
|
Burt P, Thurley K. Distribution modeling quantifies collective T H cell decision circuits in chronic inflammation. SCIENCE ADVANCES 2023; 9:eadg7668. [PMID: 37703364 PMCID: PMC10881075 DOI: 10.1126/sciadv.adg7668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 08/11/2023] [Indexed: 09/15/2023]
Abstract
Immune responses are tightly regulated by a diverse set of interacting immune cell populations. Alongside decision-making processes such as differentiation into specific effector cell types, immune cells initiate proliferation at the beginning of an inflammation, forming two layers of complexity. Here, we developed a general mathematical framework for the data-driven analysis of collective immune cell dynamics. We identified qualitative and quantitative properties of generic network motifs, and we specified differentiation dynamics by analysis of kinetic transcriptome data. Furthermore, we derived a specific, data-driven mathematical model for T helper 1 versus T follicular helper cell-fate decision dynamics in acute and chronic lymphocytic choriomeningitis virus infections in mice. The model recapitulates important dynamical properties without model fitting and solely by using measured response-time distributions. Model simulations predict different windows of opportunity for perturbation in acute and chronic infection scenarios, with potential implications for optimization of targeted immunotherapy.
Collapse
Affiliation(s)
- Philipp Burt
- Systems Biology of Inflammation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Institute for Theoretical Biophysics, Humboldt University, Berlin, Germany
| | - Kevin Thurley
- Systems Biology of Inflammation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Biomathematics Division, Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
8
|
De Boer RJ, Yates AJ. Modeling T Cell Fate. Annu Rev Immunol 2023; 41:513-532. [PMID: 37126420 PMCID: PMC11100019 DOI: 10.1146/annurev-immunol-101721-040924] [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: 05/02/2023]
Abstract
Many of the pathways that underlie the diversification of naive T cells into effector and memory subsets, and the maintenance of these populations, remain controversial. In recent years a variety of experimental tools have been developed that allow us to follow the fates of cells and their descendants. In this review we describe how mathematical models provide a natural language for describing the growth, loss, and differentiation of cell populations. By encoding mechanistic descriptions of cell behavior, models can help us interpret these new datasets and reveal the rules underpinning T cell fate decisions, both at steady state and during immune responses.
Collapse
Affiliation(s)
- Rob J De Boer
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Utrecht, The Netherlands;
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA;
| |
Collapse
|
9
|
Guo M, Abd-Rabbo D, Bertol B, Carew M, Lukhele S, Snell LM, Xu W, Boukhaled GM, Elsaesser H, Halaby MJ, Hirano N, McGaha TL, Brooks DG. Molecular, metabolic and functional CD4 T cell paralysis impedes tumor control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.15.536946. [PMID: 37131587 PMCID: PMC10153152 DOI: 10.1101/2023.04.15.536946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
CD4 T cells are important effectors of anti-tumor immunity, yet the regulation of CD4 tumor-specific T (T TS ) cells during cancer development is still unclear. We demonstrate that CD4 T TS cells are initially primed in the tumor draining lymph node and begin to divide following tumor initiation. Distinct from CD8 T TS cells and previously defined exhaustion programs, CD4 T TS cell proliferation is rapidly frozen in place and differentiation stunted by a functional interplay of T regulatory cells and both intrinsic and extrinsic CTLA4 signaling. Together these mechanisms paralyze CD4 T TS cell differentiation, redirecting metabolic and cytokine production circuits, and reducing CD4 T TS cell accumulation in the tumor. Paralysis is actively maintained throughout cancer progression and CD4 T TS cells rapidly resume proliferation and functional differentiation when both suppressive reactions are alleviated. Strikingly, Treg depletion alone reciprocally induced CD4 T TS cells to themselves become tumor-specific Tregs, whereas CTLA4 blockade alone failed to promote T helper differentiation. Overcoming their paralysis established long-term tumor control, demonstrating a novel immune evasion mechanism that specifically cripples CD4 T TS cells to favor tumor progression.
Collapse
|
10
|
Chen M, Li M, Budai MM, Rice AP, Kimata JT, Mohan M, Wang J. Clearance of HIV-1 or SIV reservoirs by promotion of apoptosis and inhibition of autophagy: Targeting intracellular molecules in cure-directed strategies. J Leukoc Biol 2022; 112:1245-1259. [PMID: 35362118 PMCID: PMC9522917 DOI: 10.1002/jlb.4mr0222-606] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/15/2022] [Indexed: 12/24/2022] Open
Abstract
The reservoirs of the HIV display cellular properties resembling long-lived immune memory cells that could be exploited for viral clearance. Our interest in developing a cure for HIV stems from the studies of immunologic memory against infections. We and others have found that long-lived immune memory cells employ prosurvival autophagy and antiapoptotic mechanisms to protect their longevity. Here, we describe the rationale for the development of an approach to clear HIV-1 by selective elimination of host cells harboring replication-competent HIV (SECH). While reactivation of HIV-1 in the host cells with latency reversing agents (LRAs) induces viral gene expression leading to cell death, LRAs also simultaneously up-regulate prosurvival antiapoptotic molecules and autophagy. Mechanistically, transcription factors that promote HIV-1 LTR-directed gene expression, such as NF-κB, AP-1, and Hif-1α, can also enhance the expression of cellular genes essential for cell survival and metabolic regulation, including Bcl-xL, Mcl-1, and autophagy genes. In the SECH approach, we inhibit the prosurvival antiapoptotic molecules and autophagy induced by LRAs, thereby allowing maximum killing of host cells by the induced HIV-1 proteins. SECH treatments cleared HIV-1 infections in humanized mice in vivo and in HIV-1 patient PBMCs ex vivo. SECH also cleared infections by the SIV in rhesus macaque PBMCs ex vivo. Research efforts are underway to improve the efficacy and safety of SECH and to facilitate the development of SECH as a therapeutic approach for treating people with HIV.
Collapse
Affiliation(s)
- Min Chen
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
| | - Min Li
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Research Institute, Houston, Texas, USA
| | - Marietta M. Budai
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Research Institute, Houston, Texas, USA
| | - Andrew P. Rice
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Jason T. Kimata
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Mahesh Mohan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Jin Wang
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Surgery, Weill Cornell Medical College, Cornell University, New York, New York, USA
| |
Collapse
|
11
|
Belluccini G, López-García M, Lythe G, Molina-París C. Counting generations in birth and death processes with competing Erlang and exponential waiting times. Sci Rep 2022; 12:11289. [PMID: 35789162 PMCID: PMC9253354 DOI: 10.1038/s41598-022-14202-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 05/09/2022] [Indexed: 11/09/2022] Open
Abstract
Lymphocyte populations, stimulated in vitro or in vivo, grow as cells divide. Stochastic models are appropriate because some cells undergo multiple rounds of division, some die, and others of the same type in the same conditions do not divide at all. If individual cells behave independently, then each cell can be imagined as sampling from a probability density of times to division and death. The exponential density is the most mathematically and computationally convenient choice. It has the advantage of satisfying the memoryless property, consistent with a Markov process, but it overestimates the probability of short division times. With the aim of preserving the advantages of a Markovian framework while improving the representation of experimentally-observed division times, we consider a multi-stage model of cellular division and death. We use Erlang-distributed (or, more generally, phase-type distributed) times to division, and exponentially distributed times to death. We classify cells into generations, using the rule that the daughters of cells in generation n are in generation [Formula: see text]. In some circumstances, our representation is equivalent to established models of lymphocyte dynamics. We find the growth rate of the cell population by calculating the proportions of cells by stage and generation. The exponent describing the late-time cell population growth, and the criterion for extinction of the population, differs from what would be expected if N steps with rate [Formula: see text] were equivalent to a single step of rate [Formula: see text]. We link with a published experimental dataset, where cell counts were reported after T cells were transferred to lymphopenic mice, using Approximate Bayesian Computation. In the comparison, the death rate is assumed to be proportional to the generation and the Erlang time to division for generation 0 is allowed to differ from that of subsequent generations. The multi-stage representation is preferred to a simple exponential in posterior distributions, and the mean time to first division is estimated to be longer than the mean time to subsequent divisions.
Collapse
Affiliation(s)
| | | | - Grant Lythe
- School of Mathematics, University of Leeds, Leeds, LS2 9JT, UK
| | - Carmen Molina-París
- School of Mathematics, University of Leeds, Leeds, LS2 9JT, UK.
- T-6, Theoretical Biology and Biophysics, Theoretical Division, Los Alamos National Laboratory, Los Alamos, NM, USA.
| |
Collapse
|
12
|
Burt P, Cornelis R, Geißler G, Hahne S, Radbruch A, Chang HD, Thurley K. Data-Driven Mathematical Model of Apoptosis Regulation in Memory Plasma Cells. Cells 2022; 11:cells11091547. [PMID: 35563853 PMCID: PMC9102437 DOI: 10.3390/cells11091547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/13/2022] [Accepted: 04/21/2022] [Indexed: 12/04/2022] Open
Abstract
Memory plasma cells constitutively produce copious amounts of antibodies, imposing a critical risk factor for autoimmune disease. We previously found that plasma cell survival requires secreted factors such as APRIL and direct contact to stromal cells, which act in concert to activate NF-κB- and PI3K-dependent signaling pathways to prevent cell death. However, the regulatory properties of the underlying biochemical network are confounded by the complexity of potential interaction and cross-regulation pathways. Here, based on flow-cytometric quantification of key signaling proteins in the presence or absence of the survival signals APRIL and contact to the stromal cell line ST2, we generated a quantitative model of plasma cell survival. Our model emphasizes the non-redundant nature of the two plasma cell survival signals APRIL and stromal cell contact, and highlights a requirement for differential regulation of individual caspases. The modeling approach allowed us to unify distinct data sets and derive a consistent picture of the intertwined signaling and apoptosis pathways regulating plasma cell survival.
Collapse
Affiliation(s)
- Philipp Burt
- German Rheumatism Research Center, 10117 Berlin, Germany; (P.B.); (R.C.); (G.G.); (S.H.); (A.R.)
- Institute for Theoretical Biology, Humboldt University, 10115 Berlin, Germany
| | - Rebecca Cornelis
- German Rheumatism Research Center, 10117 Berlin, Germany; (P.B.); (R.C.); (G.G.); (S.H.); (A.R.)
| | - Gustav Geißler
- German Rheumatism Research Center, 10117 Berlin, Germany; (P.B.); (R.C.); (G.G.); (S.H.); (A.R.)
- Institute for Theoretical Biology, Humboldt University, 10115 Berlin, Germany
| | - Stefanie Hahne
- German Rheumatism Research Center, 10117 Berlin, Germany; (P.B.); (R.C.); (G.G.); (S.H.); (A.R.)
| | - Andreas Radbruch
- German Rheumatism Research Center, 10117 Berlin, Germany; (P.B.); (R.C.); (G.G.); (S.H.); (A.R.)
| | - Hyun-Dong Chang
- German Rheumatism Research Center, 10117 Berlin, Germany; (P.B.); (R.C.); (G.G.); (S.H.); (A.R.)
- Institute of Biotechnology, Department of Cytometry, Technische Universität, 10623 Berlin, Germany
- Correspondence: (H.-D.C.); (K.T.)
| | - Kevin Thurley
- German Rheumatism Research Center, 10117 Berlin, Germany; (P.B.); (R.C.); (G.G.); (S.H.); (A.R.)
- Institute for Theoretical Biology, Humboldt University, 10115 Berlin, Germany
- Biomathematics Division, Institute of Experimental Oncology, University Hospital Bonn, 53127 Bonn, Germany
- Correspondence: (H.-D.C.); (K.T.)
| |
Collapse
|
13
|
Varadarajan SN, Mathew KA, Chandrasekharan A, Lupitha SS, Lekshmi A, Mini M, Darvin P, Santhoshkumar TR. Real-time visualization and quantitation of cell death and cell cycle progression in 2D and 3D cultures utilizing genetically encoded probes. J Cell Biochem 2022; 123:782-797. [PMID: 35106828 DOI: 10.1002/jcb.30222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 12/23/2021] [Accepted: 01/11/2022] [Indexed: 12/17/2022]
Abstract
Cancer cells grown as 3D-structures are better models for mimicking in vivo conditions than the 2D-culture systems employable in drug discovery applications. Cell cycle and cell death are important determinants for preclinical drug screening and tumor growth studies in laboratory conditions. Though several 3D-models and live-cell compatible approaches are available, a method for simultaneous real-time detection of cell cycle and cell death is required. Here we demonstrate a high-throughput adaptable method using genetically encoded fluorescent probes for the real-time quantitative detection of cell death and cell cycle. The cell-cycle indicator cdt1-Kusabira orange (KO) is stably integrated into cancer cells and further transfected with the Fluorescence Resonance Energy Transfer-based ECFP-DEVD-EYFP caspase activation sensor. The nuclear cdt1-KO expression serves as the readout for cell-cycle, and caspase activation is visualized by ECFP/EYFP ratiometric imaging. The image-based platform allowed imaging of growing spheres for prolonged periods in 3D-culture with excellent single-cell resolution through confocal microscopy. High-throughput screening (HTS) adaptation was achieved by targeting the caspase-sensor at the nucleus, which enabled the quantitation of cell death in 3D-models. The HTS using limited compound libraries, identified two lead compounds that induced caspase-activation both in 2D and 3D-cultures. This is the first report of an approach for noninvasive stain-free quantitative imaging of cell death and cell cycle with potential drug discovery applications.
Collapse
Affiliation(s)
| | - Krupa Ann Mathew
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Aneesh Chandrasekharan
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | | | - Asha Lekshmi
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Minsa Mini
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Pramod Darvin
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - T R Santhoshkumar
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
14
|
Cheon H, Kan A, Prevedello G, Oostindie SC, Dovedi SJ, Hawkins ED, Marchingo JM, Heinzel S, Duffy KR, Hodgkin PD. Cyton2: A Model of Immune Cell Population Dynamics That Includes Familial Instructional Inheritance. FRONTIERS IN BIOINFORMATICS 2021; 1:723337. [PMID: 36303793 PMCID: PMC9581048 DOI: 10.3389/fbinf.2021.723337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Lymphocytes are the central actors in adaptive immune responses. When challenged with antigen, a small number of B and T cells have a cognate receptor capable of recognising and responding to the insult. These cells proliferate, building an exponentially growing, differentiating clone army to fight off the threat, before ceasing to divide and dying over a period of weeks, leaving in their wake memory cells that are primed to rapidly respond to any repeated infection. Due to the non-linearity of lymphocyte population dynamics, mathematical models are needed to interrogate data from experimental studies. Due to lack of evidence to the contrary and appealing to arguments based on Occam's Razor, in these models newly born progeny are typically assumed to behave independently of their predecessors. Recent experimental studies, however, challenge that assumption, making clear that there is substantial inheritance of timed fate changes from each cell by its offspring, calling for a revision to the existing mathematical modelling paradigms used for information extraction. By assessing long-term live-cell imaging of stimulated murine B and T cells in vitro, we distilled the key phenomena of these within-family inheritances and used them to develop a new mathematical model, Cyton2, that encapsulates them. We establish the model's consistency with these newly observed fine-grained features. Two natural concerns for any model that includes familial correlations would be that it is overparameterised or computationally inefficient in data fitting, but neither is the case for Cyton2. We demonstrate Cyton2's utility by challenging it with high-throughput flow cytometry data, which confirms the robustness of its parameter estimation as well as its ability to extract biological meaning from complex mixed stimulation experiments. Cyton2, therefore, offers an alternate mathematical model, one that is, more aligned to experimental observation, for drawing inferences on lymphocyte population dynamics.
Collapse
Affiliation(s)
- HoChan Cheon
- Hamilton Institute, Maynooth University, Maynooth, Ireland
| | - Andrey Kan
- Immunology Division, the Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, the University of Melbourne, Parkville, VIC, Australia
| | | | - Simone C. Oostindie
- Immunology Division, the Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, the University of Melbourne, Parkville, VIC, Australia
| | | | - Edwin D. Hawkins
- Department of Medical Biology, the University of Melbourne, Parkville, VIC, Australia
- Division of Inflammation, the Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Julia M. Marchingo
- Cell Signalling and Immunology Division, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Susanne Heinzel
- Immunology Division, the Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, the University of Melbourne, Parkville, VIC, Australia
| | - Ken R. Duffy
- Hamilton Institute, Maynooth University, Maynooth, Ireland
| | - Philip D. Hodgkin
- Immunology Division, the Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, the University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
15
|
Tackling cancer cell dormancy: Insights from immune models, and transplantation. Semin Cancer Biol 2021; 78:5-16. [PMID: 33582171 DOI: 10.1016/j.semcancer.2021.02.002] [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: 10/28/2020] [Revised: 01/06/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023]
Abstract
Disseminated non-dividing (dormant) cancer cells as well as those in equilibrium with the immune response remain the major challenge for successful treatment of cancer. The equilibrium between disseminated dormant cancer cells and the immune system is reminiscent of states that can occur during infection or allogeneic tissue and cell transplantation. We discuss here the major competing models of how the immune system achieves a self nonself discrimination (pathogen/danger patterns, quorum, and coinhibition/tuning models), and suggest that taking advantage of a combination of the proposed mechanisms in each model may lead to increased efficacy in tackling cancer cell dormancy.
Collapse
|
16
|
Kleef R, Nagy R, Baierl A, Bacher V, Bojar H, McKee DL, Moss R, Thoennissen NH, Szász M, Bakacs T. Low-dose ipilimumab plus nivolumab combined with IL-2 and hyperthermia in cancer patients with advanced disease: exploratory findings of a case series of 131 stage IV cancers - a retrospective study of a single institution. Cancer Immunol Immunother 2020; 70:1393-1403. [PMID: 33151369 PMCID: PMC8053148 DOI: 10.1007/s00262-020-02751-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
The 3-year overall survival (OS) rate of patients with previously treated or untreated stage III or IV melanoma has by now reached 63% using ipilimumab and nivolumab therapy. However, immune-related adverse events (irAEs) of grade 3 or 4 occurred in 59% of patients leading to discontinuation of therapy in 24.5% of patients and one death. Therapy with checkpoint inhibitors could be safer and more effective in combination with hyperthermia and fever inducing therapies. We conducted a retrospective analysis to test the safety and efficacy of a new combination immune therapy in 131 unselected stage IV solid cancer patients with 23 different histological types of cancer who exhausted all conventional treatments. Treatment consisted of locoregional- and whole-body hyperthermia, individually dose adapted interleukin 2 (IL-2) combined with low-dose ipilimumab (0.3 mg/kg) plus nivolumab (0.5 mg/kg). The objective response rate (ORR) was 31.3%, progression-free survival (PFS) was 10 months, survival probabilities at 6 months was 86.7% (95% CI, 81.0–92.8%), at 9 months was 73.5% (95% CI, 66.2–81.7%), at 12 months was 66.5% (95% CI, 58.6–75.4%), while at 24 months survival was 36.6% (95% CI:28.2%; 47.3%). irAEs of World Health Organization (WHO) Toxicity Scale grade 1, 2, 3, and 4 were observed in 23.66%, 16.03%, 6.11%, and 2.29% of patients, respectively. Our results suggest that the irAEs profile of the combined treatment is safer than that of the established protocols without compromising efficacy.
Collapse
Affiliation(s)
- R Kleef
- Immunology and Integrative Oncology, Auhofstraße 1, 1130, Vienna, Austria
| | - R Nagy
- Immunology and Integrative Oncology, Auhofstraße 1, 1130, Vienna, Austria
| | - A Baierl
- Department of Statistic and Operations Research, University of Vienna, Oskar-Morgenstern-Platz 1, 1090, Vienna, Austria
| | - V Bacher
- Immunology and Integrative Oncology, Auhofstraße 1, 1130, Vienna, Austria
| | - H Bojar
- NextGen Oncology, Dusseldorf, Germany
| | - D L McKee
- Integrative Cancer Consulting, Aptos, CA, USA
| | - R Moss
- Moss Reports, 104 Main Street, Unit 1422, Blue Hill, ME, 04614-1422, USA
| | - N H Thoennissen
- Oncology Center at Lenbachplatz, Ottostr. 3, 80333, Munich, Germany
| | - M Szász
- Cancer Centre, Semmelweis University, 1083, Budapest, Hungary
| | - T Bakacs
- PRET Therapeutics Ltd, 1124, Budapest, Hungary.
| |
Collapse
|
17
|
Pandey S, Gruenbaum A, Kanashova T, Mertins P, Cluzel P, Chevrier N. Pairwise Stimulations of Pathogen-Sensing Pathways Predict Immune Responses to Multi-adjuvant Combinations. Cell Syst 2020; 11:495-508.e10. [PMID: 33113356 DOI: 10.1016/j.cels.2020.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/29/2020] [Accepted: 09/30/2020] [Indexed: 12/31/2022]
Abstract
The immune system makes decisions in response to combinations of multiple microbial inputs. We do not understand the combinatorial logic governing how higher-order combinations of microbial signals shape immune responses. Here, using coculture experiments and statistical analyses, we discover a general property for the combinatorial sensing of microbial signals, whereby the effects of triplet combinations of microbial signals on immune responses can be predicted by combining the effects of single and pairs. Mechanistically, we find that singles and pairs dictate the information signaled by triplets in mouse and human DCs at the levels of transcription, chromatin, and protein secretion. We exploit this simplifying property to develop cell-based immunotherapies prepared with adjuvant combinations that trigger protective responses in mouse models of cancer. We conclude that the processing of multiple input signals by innate immune cells is governed by pairwise effects, which will inform the rationale combination of adjuvants to manipulate immunity.
Collapse
Affiliation(s)
- Surya Pandey
- Pritzker School of Molecular Engineering, the University of Chicago, Chicago, IL 60637, USA
| | - Adam Gruenbaum
- Pritzker School of Molecular Engineering, the University of Chicago, Chicago, IL 60637, USA
| | - Tamara Kanashova
- Proteomics Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Society, and Berlin Institute of Health, 13125 Berlin, Germany
| | - Philipp Mertins
- Proteomics Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Society, and Berlin Institute of Health, 13125 Berlin, Germany
| | - Philippe Cluzel
- School of Engineering and Applied Science & Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Nicolas Chevrier
- Pritzker School of Molecular Engineering, the University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
18
|
Verdon DJ, Mulazzani M, Jenkins MR. Cellular and Molecular Mechanisms of CD8 + T Cell Differentiation, Dysfunction and Exhaustion. Int J Mol Sci 2020; 21:ijms21197357. [PMID: 33027962 PMCID: PMC7582856 DOI: 10.3390/ijms21197357] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023] Open
Abstract
T cells follow a triphasic distinct pathway of activation, proliferation and differentiation before becoming functionally and phenotypically “exhausted” in settings of chronic infection, autoimmunity and in cancer. Exhausted T cells progressively lose canonical effector functions, exhibit altered transcriptional networks and epigenetic signatures and gain constitutive expression of a broad coinhibitory receptor suite. This review outlines recent advances in our understanding of exhausted T cell biology and examines cellular and molecular mechanisms by which a state of dysfunction or exhaustion is established, and mechanisms by which exhausted T cells may still contribute to pathogen or tumour control. Further, this review describes our understanding of exhausted T cell heterogeneity and outlines the mechanisms by which checkpoint blockade differentially engages exhausted T cell subsets to overcome exhaustion and recover T cell function.
Collapse
Affiliation(s)
- Daniel J. Verdon
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
| | - Matthias Mulazzani
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
| | - Misty R. Jenkins
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
- Institute of Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
- Correspondence:
| |
Collapse
|
19
|
Bhattacharyya ND, Feng CG. Regulation of T Helper Cell Fate by TCR Signal Strength. Front Immunol 2020; 11:624. [PMID: 32508803 PMCID: PMC7248325 DOI: 10.3389/fimmu.2020.00624] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/19/2020] [Indexed: 12/16/2022] Open
Abstract
T cells are critical in orchestrating protective immune responses to cancer and an array of pathogens. The interaction between a peptide MHC (pMHC) complex on antigen presenting cells (APCs) and T cell receptors (TCRs) on T cells initiates T cell activation, division, and clonal expansion in secondary lymphoid organs. T cells must also integrate multiple T cell-intrinsic and extrinsic signals to acquire the effector functions essential for the defense against invading microbes. In the case of T helper cell differentiation, while innate cytokines have been demonstrated to shape effector CD4+ T lymphocyte function, the contribution of TCR signaling strength to T helper cell differentiation is less understood. In this review, we summarize the signaling cascades regulated by the strength of TCR stimulation. Various mechanisms in which TCR signal strength controls T helper cell expansion and differentiation are also discussed.
Collapse
Affiliation(s)
- Nayan D Bhattacharyya
- Immunology and Host Defense Group, Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Tuberculosis Research Program, Centenary Institute, The University of Sydney, Sydney, NSW, Australia
| | - Carl G Feng
- Immunology and Host Defense Group, Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Tuberculosis Research Program, Centenary Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
20
|
Shissler SC, Singh NJ, Webb TJ. Thymic resident NKT cell subsets show differential requirements for CD28 co-stimulation during antigenic activation. Sci Rep 2020; 10:8218. [PMID: 32427927 PMCID: PMC7237672 DOI: 10.1038/s41598-020-65129-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 04/28/2020] [Indexed: 12/03/2022] Open
Abstract
Natural killer T (NKT) cells rapidly respond to antigenic stimulation with cytokine production and direct cytotoxicity. These innate-like characteristics arise from their differentiation into mature effector cells during thymic development. A subset of mature NKT cells remain thymic resident, but their activation and function remain poorly understood. We examined the roles of CD28 and CTLA-4 in driving the activation of thymic resident NKT cells. In contrast to studies with peripheral NKT cells, the proliferation of thymic NKT cells was significantly impaired when CD28 engagement was blocked, but unaffected by CTLA-4 activation or blockade. Within NKT subsets, however, stage 3 NKT cells, marked by higher NK1.1 expression, were significantly more sensitive to the loss of CD28 signals compared to NK1.1- stage 2 NKT cells. In good agreement, CD28 blockade suppressed NKT cell cytokine secretion, lowering the ratio of IFN-γ:IL-4 production by NK1.1+ NKT cells. Intriguingly, the activation-dependent upregulation of the master transcription factor PLZF did not require CD28-costimulation in either of the thymic NKT subsets, underlining a dichotomy between requirements for early activation vs subsequent proliferation and effector function by these cells. Collectively, our studies demonstrate the ability of CD28 co-stimulation to fine tune subset-specific responses by thymic resident NKT cells and contextually shape the milieu in this primary lymphoid organ.
Collapse
Affiliation(s)
- Susannah C Shissler
- Department of Microbiology and Immunology and the Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Nevil J Singh
- Department of Microbiology and Immunology and the Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Tonya J Webb
- Department of Microbiology and Immunology and the Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
21
|
Costa Del Amo P, Debebe B, Razavi-Mohseni M, Nakaoka S, Worth A, Wallace D, Beverley P, Macallan D, Asquith B. The Rules of Human T Cell Fate in vivo. Front Immunol 2020; 11:573. [PMID: 32322253 PMCID: PMC7156550 DOI: 10.3389/fimmu.2020.00573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/12/2020] [Indexed: 12/05/2022] Open
Abstract
The processes governing lymphocyte fate (division, differentiation, and death), are typically assumed to be independent of cell age. This assumption has been challenged by a series of elegant studies which clearly show that, for murine cells in vitro, lymphocyte fate is age-dependent and that younger cells (i.e., cells which have recently divided) are less likely to divide or die. Here we investigate whether the same rules determine human T cell fate in vivo. We combined data from in vivo stable isotope labeling in healthy humans with stochastic, agent-based mathematical modeling. We show firstly that the choice of model paradigm has a large impact on parameter estimates obtained using stable isotope labeling i.e., different models fitted to the same data can yield very different estimates of T cell lifespan. Secondly, we found no evidence in humans in vivo to support the model in which younger T cells are less likely to divide or die. This age-dependent model never provided the best description of isotope labeling; this was true for naïve and memory, CD4+ and CD8+ T cells. Furthermore, this age-dependent model also failed to predict an independent data set in which the link between division and death was explored using Annexin V and deuterated glucose. In contrast, the age-independent model provided the best description of both naïve and memory T cell dynamics and was also able to predict the independent dataset.
Collapse
Affiliation(s)
- Pedro Costa Del Amo
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Bisrat Debebe
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Milad Razavi-Mohseni
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Shinji Nakaoka
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Kawaguchi, Japan.,Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Andrew Worth
- The Jenner Institute Laboratories, University of Oxford, Oxford, United Kingdom
| | - Diana Wallace
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Peter Beverley
- TB Research Centre, National Heart and Lung Research Institute, Imperial College London, London, United Kingdom
| | - Derek Macallan
- Institute for Infection and Immunity, St. George's Hospital, University of London, London, United Kingdom
| | - Becca Asquith
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
22
|
Thiam LG, Aniweh Y, Quansah EB, Donkor JK, Gwira TM, Kusi KA, Niang M, Awandare GA. Cell trace far-red is a suitable erythrocyte dye for multi-color Plasmodium falciparum invasion phenotyping assays. Exp Biol Med (Maywood) 2020; 245:11-20. [PMID: 31903776 PMCID: PMC6987746 DOI: 10.1177/1535370219897393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/06/2019] [Indexed: 11/16/2022] Open
Abstract
Plasmodium falciparum erythrocyte invasion phenotyping assays are a very useful tool for assessing parasite diversity and virulence, and for characterizing the formation of ligand–receptor interactions. However, such assays need to be highly sensitive and reproducible, and the selection of labeling dyes for differentiating donor and acceptor erythrocytes is a critical factor. We investigated the suitability of cell trace far-red (CTFR) as a dye for P. falciparum invasion phenotyping assays. Using the dyes carboxyfluorescein diacetate succinimidyl ester (CFDA-SE) and dichloro dimethyl acridin one succinimidyl ester (DDAO-SE) as comparators, we used a dye-dilution approach to assess the limitations and specific staining procedures for the applicability of CTFR in P. falciparum invasion phenotyping assays. Our data show that CTFR effectively labels acceptor erythrocytes and provides a stable fluorescent intensity at relatively low concentrations. CTFR also yielded a higher fluorescence intensity relative to DDAO-SE and with a more stable fluorescence intensity over time. Furthermore, CTFR did not affect merozoites invasion of erythrocytes and was not toxic to the parasite’s intraerythrocytic development. Additionally, CTFR offers flexibility in the choice of combinations with several other DNA dyes, which broaden its usage for P. falciparum erythrocyte invasion assays, considering a wider range of flow cytometers with various laser settings.
Collapse
Affiliation(s)
- Laty G Thiam
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| | - Yaw Aniweh
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| | - Evelyn B Quansah
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| | - Jacob K Donkor
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| | - Theresa M Gwira
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| | - Kwadwo A Kusi
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
- Department of Immunology, Noguchi Memorial Institute for Medical
Research, University of Ghana, P. O. Box LG 581, Legon, Accra
| | - Makhtar Niang
- Unité d’Immunologie, Institut Pasteur de Dakar, Dakar 220,
Senegal
| | - Gordon A Awandare
- West African Centre for Cell Biology of Infectious Pathogens,
College of Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon,
Accra
- Department of Biochemistry, Cell and Molecular Biology, College of
Basic and Applied Sciences, University of Ghana, P. O. Box LG54, Legon, Accra
| |
Collapse
|
23
|
Abstract
Intercellular communication mediated by cytokines is the main mechanism by which cells of the immune system talk to each other. Many aspects of cytokine signalling in the immune system have been explored in great detail at the structural, biophysical, biochemical and cellular levels. However, a systematic understanding of the quantitative rules that govern cytokine-mediated cell-to-cell communication is still lacking. Here, we discuss recent efforts in the field of systems immunology to bring about a quantitative understanding of cytokine-mediated communication between leukocytes and to provide novel insights into the orchestration of immune responses and inflammation.
Collapse
|
24
|
Daneshpour H, Youk H. Modeling cell-cell communication for immune systems across space and time. ACTA ACUST UNITED AC 2019; 18:44-52. [PMID: 31922054 PMCID: PMC6941841 DOI: 10.1016/j.coisb.2019.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Communicating is crucial for cells to coordinate their behaviors. Immunological processes, involving diverse cytokines and cell types, are ideal for developing frameworks for modeling coordinated behaviors of cells. Here, we review recent studies that combine modeling and experiments to reveal how immune systems use autocrine, paracrine, and juxtacrine signals to achieve behaviors such as controlling population densities and hair regenerations. We explain that models are useful because one can computationally vary numerous parameters, in experimentally infeasible ways, to evaluate alternate immunological responses. For each model, we focus on the length-scales and time-scales involved and explain why integrating multiple length-scales and time-scales in a model remain challenging. We suggest promising modeling strategies for meeting this challenge and their practical consequences.
Collapse
Affiliation(s)
- Hirad Daneshpour
- Kavli Institute of Nanoscience, the Netherlands.,Department of Bionanoscience, Delft University of Technology, Delft, 2629HZ, the Netherlands
| | - Hyun Youk
- Kavli Institute of Nanoscience, the Netherlands.,Department of Bionanoscience, Delft University of Technology, Delft, 2629HZ, the Netherlands.,CIFAR, CIFAR Azrieli Global Scholars Program, Toronto, ON, M5G 1M1, Canada
| |
Collapse
|
25
|
Chevrier N. Decoding the Body Language of Immunity: Tackling the Immune System at the Organism Level. ACTA ACUST UNITED AC 2019; 18:19-26. [PMID: 32490290 DOI: 10.1016/j.coisb.2019.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The immune system is a dynamic mesh of molecules, cells and tissues spanning the entire organism. Despite a wealth of knowledge about the components of the immune system, little is known about the general rules governing the organismal circuitry of immunity. Deciphering the immune system at the scale of the whole organism is crucial to understanding fundamental problems in immunobiology and physiology, and to manipulate immunity for maintaining health and preventing disease. Here I discuss the emerging principles of inter-organ communications during immune responses by focusing on three common themes that are the regulation of the (i) composition, (ii) condition and (iii) coordination of communicating organs by molecular and cellular factors. Based on these common principles, I emphasize fundamental gaps in our knowledge of organismal immune processes and the outlook to tackle immunity at the scale of the whole organism.
Collapse
Affiliation(s)
- Nicolas Chevrier
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
26
|
Bakacs T, Moss RW, Kleef R, Szasz MA, Anderson CC. Exploiting autoimmunity unleashed by low-dose immune checkpoint blockade to treat advanced cancer. Scand J Immunol 2019; 90:e12821. [PMID: 31589347 DOI: 10.1111/sji.12821] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/31/2019] [Accepted: 08/26/2019] [Indexed: 12/15/2022]
Abstract
As a result of the cancer immunotherapy revolution, more than 2000 immuno-oncology agents are currently being tested or are in use to improve responses. Not unexpectedly, the 2018 Nobel Prize in Physiology or Medicine was awarded to James P. Allison and Tasuku Honjo for their development of cancer therapy by the blockade of co-inhibitory signals. Unfortunately, manipulation of the co-inhibitory receptors has also resulted in a safety issue: widespread iatrogenic immune-related adverse events (irAEs). Autoimmunity is emerging as the nemesis of immunotherapy. Originally, it was assumed that CTLA-4 blockade selectively targets T cells relevant to the antitumour immune response. However, an uncontrolled pan T cell activation was induced compromising tolerance to healthy self-tissues. The irAEs are very similar to that of a chronic graft-versus-host-disease (GVHD) reaction following allogeneic bone marrow transplantation (BMT). We hypothesized that ipilimumab induced a graft-versus-malignancy (GVM) effect, which eradicated metastatic melanoma in a minority of patients, but also involved an auto-GVHD reaction that resulted in widespread autoimmunity in the majority. Therefore, we argued for a profound theoretical point against the consensus of experts. The task is not to desperately put the genie back in the bottle by immune-suppressive treatments, but instead to harness the autoimmune forces. In this way, the same goal could be achieved by an antibody as by the adoptive transfer of alloreactive donor lymphocytes, but without severe GVHD. The proof-of-principle of a low-dose-combination immune checkpoint therapy, consisting only of approved drugs and treatments, was demonstrated in 111 stage IV cancer patients.
Collapse
Affiliation(s)
| | | | - Ralf Kleef
- Immunology & Integrative Oncology, Kleef Hyperthermie, Vienna, Austria
| | | | - Colin C Anderson
- Departments of Surgery and Medical Microbiology & Immunology, Alberta Diabetes Institute, Alberta Transplant Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
27
|
Abstract
Generating and maintaining a diverse repertoire of naive T cells is essential for protection against pathogens, and developing a mechanistic and quantitative description of the processes involved lies at the heart of our understanding of vertebrate immunity. Here, we review the biology of naive T cells from birth to maturity and outline how the integration of mathematical models and experiments has helped us to develop a full picture of their life histories.
Collapse
Affiliation(s)
- Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Hospital, London, UK
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| |
Collapse
|
28
|
Hennessy RJ, Pham K, Delconte R, Rautela J, Hodgkin PD, Huntington ND. Quantifying NK cell growth and survival changes in response to cytokines and regulatory checkpoint blockade helps identify optimal culture and expansion conditions. J Leukoc Biol 2019; 105:1341-1354. [PMID: 31079418 DOI: 10.1002/jlb.ma0718-296r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/01/2019] [Accepted: 04/28/2019] [Indexed: 01/02/2023] Open
Abstract
NK cells are innate lymphocytes critical for immune surveillance, particularly in eradication of metastatic cancer cells and acute antiviral responses. In contrast to T cells, NK cell-mediated immunity is rapid, with spontaneous cytotoxicity and cytokine/chemokine production upon pathogen detection. The renaissance in cancer immunology has cast NK cell biology back into the spotlight with an urgent need for deeper understanding of the regulatory networks that govern NK cell antitumor activity. To this end, we have adapted and refined a series of quantitative cellular calculus methods, previously applied to T and B lymphocytes, to dissect the biologic outcomes of NK cells following stimulation with cytokines (IL-15, IL-12, IL-18) or deletion of genes that regulate NK cell proliferation (Cish), survival (Bcl2l11), and activation-induced-cell-death (AICD; Fas). Our methodology is well suited to delineate effects on division rate, intrinsic apoptosis, and AICD, permitting variables such as population half-life, rate of cell division, and their combined influence on population numbers in response to stimuli to be accurately measured and modelled. Changes in these variables that result from gene deletion, concentration of stimuli, time, and cell density give insight into the dynamics of NK cell responses and serve as a platform to dissect the mechanism of action of putative checkpoints in NK cell activation and novel NK cell immunotherapy agents.
Collapse
Affiliation(s)
- Robert J Hennessy
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Kim Pham
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca Delconte
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Jai Rautela
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Philip D Hodgkin
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Nicholas D Huntington
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| |
Collapse
|
29
|
Dowling MR, Kan A, Heinzel S, Marchingo JM, Hodgkin PD, Hawkins ED. Regulatory T Cells Suppress Effector T Cell Proliferation by Limiting Division Destiny. Front Immunol 2018; 9:2461. [PMID: 30425712 PMCID: PMC6218578 DOI: 10.3389/fimmu.2018.02461] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/04/2018] [Indexed: 11/23/2022] Open
Abstract
Understanding how the strength of an effector T cell response is regulated is a fundamental problem in immunology with implications for immunity to pathogens, autoimmunity, and immunotherapy. The initial magnitude of the T cell response is determined by the sum of independent signals from antigen, co-stimulation and cytokines. By applying quantitative methods, the contribution of each signal to the number of divisions T cells undergo (division destiny) can be measured, and the resultant exponential increase in response magnitude accurately calculated. CD4+CD25+Foxp3+ regulatory T cells suppress self-reactive T cell responses and limit pathogen-directed immune responses before bystander damage occurs. Using a quantitative modeling framework to measure T cell signal integration and response, we show that Tregs modulate division destiny, rather than directly increasing the rate of death or delaying interdivision times. The quantitative effect of Tregs could be mimicked by modulating the availability of stimulatory co-stimuli and cytokines or through the addition of inhibitory signals. Thus, our analysis illustrates the primary effect of Tregs on the magnitude of effector T cell responses is mediated by modifying division destiny of responding cell populations.
Collapse
Affiliation(s)
- Mark R Dowling
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Andrey Kan
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Susanne Heinzel
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Julia M Marchingo
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Philip D Hodgkin
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Edwin D Hawkins
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
30
|
Khmelinskii I, Makarov VI. EPR hyperthermia of S. cerevisiae using superparamagnetic Fe 3O 4 nanoparticles. J Therm Biol 2018; 77:55-61. [PMID: 30196899 DOI: 10.1016/j.jtherbio.2018.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/22/2018] [Accepted: 08/06/2018] [Indexed: 11/24/2022]
Abstract
Presently we communicate a newly developed method of resonant radiofrequency heating, applicable in hyperthermal therapy. This method uses electron paramagnetic resonance (EPR) to transform the electromagnetic field energy into heat. We report the growth dynamics of the S. cerevisiae yeast cells exposed to EPR heating with superparamagnetic magnetite (Fe3O4) nanoparticles, with only 4% of yeast cells surviving hyperthermia. Given that EPR functions independently of type of the biologic species exposed, and produces spatially localized heating in conjunction with MRI hardware, it may be used in hyperthermal therapy of cancer and other diseases.
Collapse
Affiliation(s)
- I Khmelinskii
- Universidade do Algarve, FCT, DQF and CEOT, 8005-139 Faro, Portugal
| | - V I Makarov
- Department of Physics, University of Puerto Rico, Rio Piedras, PO Box 23343, San Juan, PR 00931-3343, USA.
| |
Collapse
|
31
|
Abstract
Problem-solving strategies in immunology currently utilize a series of ad hoc, qualitative variations on a foundation of Burnet's formulation of clonal selection theory. These modifications, including versions of two-signal theory, describe how signals regulate lymphocytes to make important decisions governing self-tolerance and changes to their effector and memory states. These theories are useful but are proving inadequate to explain the observable genesis and control of heterogeneity in cell types, the nonlinear passage of cell fate trajectories and how the input from multiple environmental signals can be integrated at different times and strengths. Here, I argue for a paradigm change to place immune theory on a firmer philosophical and quantitative foundation to resolve these difficulties. This change rejects the notion of identical cell subsets and substitutes the concept of a cell as comprised of autonomous functional mechanical components subject to stochastic variations in construction and operation. The theory aims to explain immunity in terms of cell population dynamics, dictated by the operation of cell machinery, such as randomizing elements, division counters, and fate timers. The effect of communicating signals alone and in combination within this system is determined with a cellular calculus. A series of models developed with these principles can resolve logical cell fate and signaling paradoxes and offer a reinterpretation for how self-non-self discrimination and immune response class are controlled.
Collapse
Affiliation(s)
- Philip D. Hodgkin
- Immunology DivisionThe Walter & Eliza Hall Institute of Medical ResearchParkvilleVic.Australia
- Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| |
Collapse
|
32
|
Smith NL, Patel RK, Reynaldi A, Grenier JK, Wang J, Watson NB, Nzingha K, Yee Mon KJ, Peng SA, Grimson A, Davenport MP, Rudd BD. Developmental Origin Governs CD8 + T Cell Fate Decisions during Infection. Cell 2018; 174:117-130.e14. [PMID: 29909981 DOI: 10.1016/j.cell.2018.05.029] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/03/2018] [Accepted: 05/11/2018] [Indexed: 12/22/2022]
Abstract
Heterogeneity is a hallmark feature of the adaptive immune system in vertebrates. Following infection, naive T cells differentiate into various subsets of effector and memory T cells, which help to eliminate pathogens and maintain long-term immunity. The current model suggests there is a single lineage of naive T cells that give rise to different populations of effector and memory T cells depending on the type and amounts of stimulation they encounter during infection. Here, we have discovered that multiple sub-populations of cells exist in the naive CD8+ T cell pool that are distinguished by their developmental origin, unique transcriptional profiles, distinct chromatin landscapes, and different kinetics and phenotypes after microbial challenge. These data demonstrate that the naive CD8+ T cell pool is not as homogeneous as previously thought and offers a new framework for explaining the remarkable heterogeneity in the effector and memory T cell subsets that arise after infection.
Collapse
Affiliation(s)
- Norah L Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Ravi K Patel
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Arnold Reynaldi
- Kirby Institute for Infection and Immunity, UNSW Australia, Sydney, NSW 2052, Australia
| | - Jennifer K Grenier
- RNA Sequencing Core, Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Jocelyn Wang
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Neva B Watson
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Kito Nzingha
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Kristel J Yee Mon
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Seth A Peng
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Miles P Davenport
- Kirby Institute for Infection and Immunity, UNSW Australia, Sydney, NSW 2052, Australia
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
33
|
Hammer Q, Rückert T, Borst EM, Dunst J, Haubner A, Durek P, Heinrich F, Gasparoni G, Babic M, Tomic A, Pietra G, Nienen M, Blau IW, Hofmann J, Na IK, Prinz I, Koenecke C, Hemmati P, Babel N, Arnold R, Walter J, Thurley K, Mashreghi MF, Messerle M, Romagnani C. Peptide-specific recognition of human cytomegalovirus strains controls adaptive natural killer cells. Nat Immunol 2018; 19:453-463. [PMID: 29632329 DOI: 10.1038/s41590-018-0082-6] [Citation(s) in RCA: 282] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 02/26/2018] [Indexed: 12/20/2022]
Abstract
Natural killer (NK) cells are innate lymphocytes that lack antigen-specific rearranged receptors, a hallmark of adaptive lymphocytes. In some people infected with human cytomegalovirus (HCMV), an NK cell subset expressing the activating receptor NKG2C undergoes clonal-like expansion that partially resembles anti-viral adaptive responses. However, the viral ligand that drives the activation and differentiation of adaptive NKG2C+ NK cells has remained unclear. Here we found that adaptive NKG2C+ NK cells differentially recognized distinct HCMV strains encoding variable UL40 peptides that, in combination with pro-inflammatory signals, controlled the population expansion and differentiation of adaptive NKG2C+ NK cells. Thus, we propose that polymorphic HCMV peptides contribute to shaping of the heterogeneity of adaptive NKG2C+ NK cell populations among HCMV-seropositive people.
Collapse
Affiliation(s)
- Quirin Hammer
- Innate Immunity, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
| | - Timo Rückert
- Innate Immunity, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
| | - Eva Maria Borst
- Institute for Virology, Hannover Medical School, Hannover, Germany
| | - Josefine Dunst
- Inflammation Biology, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
| | - André Haubner
- Innate Immunity, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
| | - Pawel Durek
- Cell Biology, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany.,Microbiota and Inflammation, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
| | - Frederik Heinrich
- Therapeutic Gene Regulation German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
| | - Gilles Gasparoni
- Department of Genetics, University of Saarland, Saarbrücken, Germany
| | - Marina Babic
- Innate Immunity, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
| | - Adriana Tomic
- Institute for Virology, Hannover Medical School, Hannover, Germany
| | - Gabriella Pietra
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Immunologia, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Mikalai Nienen
- Medical Clinic I, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| | - Igor Wolfgang Blau
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg Hofmann
- Institute of Virology Charité, Universitätsmedizin Berlin, Berlin, Germany.,Virology, Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
| | - Il-Kang Na
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christian Koenecke
- Institute of Immunology, Hannover Medical School, Hannover, Germany.,Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Philipp Hemmati
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nina Babel
- Medical Clinic I, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Renate Arnold
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jörn Walter
- Department of Genetics, University of Saarland, Saarbrücken, Germany
| | - Kevin Thurley
- Systems Biology of Inflammation, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
| | - Mir-Farzin Mashreghi
- Therapeutic Gene Regulation German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
| | - Martin Messerle
- Institute for Virology, Hannover Medical School, Hannover, Germany
| | - Chiara Romagnani
- Innate Immunity, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany. .,Medical Department I, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
34
|
Jones ZW, Leander R, Quaranta V, Harris LA, Tyson DR. A drift-diffusion checkpoint model predicts a highly variable and growth-factor-sensitive portion of the cell cycle G1 phase. PLoS One 2018; 13:e0192087. [PMID: 29432467 PMCID: PMC5809023 DOI: 10.1371/journal.pone.0192087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/17/2018] [Indexed: 11/28/2022] Open
Abstract
Even among isogenic cells, the time to progress through the cell cycle, or the intermitotic time (IMT), is highly variable. This variability has been a topic of research for several decades and numerous mathematical models have been proposed to explain it. Previously, we developed a top-down, stochastic drift-diffusion+threshold (DDT) model of a cell cycle checkpoint and showed that it can accurately describe experimentally-derived IMT distributions [Leander R, Allen EJ, Garbett SP, Tyson DR, Quaranta V. Derivation and experimental comparison of cell-division probability densities. J. Theor. Biol. 2014;358:129-135]. Here, we use the DDT modeling approach for both descriptive and predictive data analysis. We develop a custom numerical method for the reliable maximum likelihood estimation of model parameters in the absence of a priori knowledge about the number of detectable checkpoints. We employ this method to fit different variants of the DDT model (with one, two, and three checkpoints) to IMT data from multiple cell lines under different growth conditions and drug treatments. We find that a two-checkpoint model best describes the data, consistent with the notion that the cell cycle can be broadly separated into two steps: the commitment to divide and the process of cell division. The model predicts one part of the cell cycle to be highly variable and growth factor sensitive while the other is less variable and relatively refractory to growth factor signaling. Using experimental data that separates IMT into G1 vs. S, G2, and M phases, we show that the model-predicted growth-factor-sensitive part of the cell cycle corresponds to a portion of G1, consistent with previous studies suggesting that the commitment step is the primary source of IMT variability. These results demonstrate that a simple stochastic model, with just a handful of parameters, can provide fundamental insights into the biological underpinnings of cell cycle progression.
Collapse
Affiliation(s)
- Zack W. Jones
- Department of Mathematical Sciences, Middle Tennessee State University, Murfreesboro, TN 37132, United States of America
| | - Rachel Leander
- Department of Mathematical Sciences, Middle Tennessee State University, Murfreesboro, TN 37132, United States of America
| | - Vito Quaranta
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States of America
| | - Leonard A. Harris
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States of America
| | - Darren R. Tyson
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States of America
| |
Collapse
|
35
|
Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood 2018; 131:58-67. [PMID: 29118008 PMCID: PMC6317697 DOI: 10.1182/blood-2017-06-741033] [Citation(s) in RCA: 719] [Impact Index Per Article: 119.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/31/2017] [Indexed: 02/08/2023] Open
Abstract
CD28 and CTLA-4 are members of a family of immunoglobulin-related receptors that are responsible for various aspects of T-cell immune regulation. The family includes CD28, CTLA-4, and ICOS as well as other proteins, including PD-1, BTLA, and TIGIT. These receptors have both stimulatory (CD28, ICOS) and inhibitory roles (CTLA-4, PD-1, BTLA, and TIGIT) in T-cell function. Increasingly, these pathways are targeted as part of immune modulatory strategies to treat cancers, referred to generically as immune checkpoint blockade, and conversely to treat autoimmunity and CTLA-4 deficiency. Here, we focus on the biology of the CD28/CTLA-4 pathway as a framework for understanding the impacts of therapeutic manipulation of this pathway.
Collapse
Affiliation(s)
- Behzad Rowshanravan
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - Neil Halliday
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - David M Sansom
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
36
|
Tempany JC, Zhou JH, Hodgkin PD, Bryant VL. Superior properties of CellTrace Yellow™ as a division tracking dye for human and murine lymphocytes. Immunol Cell Biol 2017; 96:149-159. [PMID: 29363164 PMCID: PMC6446909 DOI: 10.1111/imcb.1020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/02/2017] [Accepted: 10/03/2017] [Indexed: 01/08/2023]
Abstract
The discovery of cell division tracking properties of 5‐(and‐6)‐carboxyfluorescein diacetate succinimidyl ester (CFSE) by Lyons and Parish in 1994 led to a broad range of new methods and numerous important biological discoveries. After labeling, CFSE is attached to free amine groups and intracellular proteins in the cytoplasm and nucleus of a cell, and halves in fluorescence intensity with each round of cell division, enabling enumeration of the number of divisions a cell has undergone. A range of popular division tracking dyes were subsequently developed, including CellTrace Violet (CTV), making available the green fluorescent channel previously occupied by CFSE. More recently, CellTrace Yellow (CTY) and CellTrace Far Red (CTFR), each with unique fluorescence properties, were introduced. In a comparison, we found that the fluorescence values of both dyes were well separated from autofluorescence, and enabled a greater number of divisions to be identified than CTV, before this limit was reached. These new dyes provided clear and well‐separated peaks for both murine and human B lymphocytes, and should find wide application. The range of excitation/emission spectra available for division tracking dyes now also facilitates multiplexing, that is, the labeling of cells with different combinations of dyes to give a unique fluorescence signature, allowing single cell in vitro and in vivo tracking. The combinatorial possibilities are significantly increased with these additional dyes.
Collapse
Affiliation(s)
- Jessica C Tempany
- Division of Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Jie Hs Zhou
- Division of Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Philip D Hodgkin
- Division of Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Vanessa L Bryant
- Division of Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
37
|
Regulatory T cells characterized by low Id3 expression are highly suppressive and accumulate during chronic infection. Oncotarget 2017; 8:102835-102851. [PMID: 29262527 PMCID: PMC5732693 DOI: 10.18632/oncotarget.22159] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 10/10/2017] [Indexed: 11/25/2022] Open
Abstract
Foxp3+ regulatory T (Treg) cells are broadly divided into naive-like and activated Treg cells, however recent studies suggest further Treg cell heterogeneity. Treg cells contribute to impaired T cell responses in chronic infections, but the role of specific Treg cell subpopulations in viral infections is not well defined. Here, we report that activated Treg cells are separated into two transcriptionally distinct subpopulations characterized by low or high expression of the transcriptional regulator Id3. Id3lo Treg cells are a highly suppressive Treg cell subpopulation, expressing elevated levels of immunomodulatory molecules and are capable of broadly targeting T cell responses. Viral infection and interleukin-2 promote the differentiation of Id3hi into Id3lo Treg cells and during chronic infection Id3lo Treg cells are the predominant Treg cell population. Thus, our report provides a framework, in which different activated Treg cell subpopulations specifically affect immune responses, possibly contributing to T cell dysfunction in chronic infections.
Collapse
|
38
|
Fleming EH, Ochoa EE, Nichols JE, O'Banion MK, Salkind AR, Roberts NJ. Reduced activation and proliferation of human lymphocytes exposed to respiratory syncytial virus compared to cells exposed to influenza virus. J Med Virol 2017; 90:26-33. [PMID: 28856681 DOI: 10.1002/jmv.24917] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 08/16/2017] [Indexed: 12/11/2022]
Abstract
Both respiratory syncytial virus (RSV) and influenza A virus (IAV) may infect human peripheral blood mononuclear leukocytes (PBMC) during the immune response to viral challenge as the cells are recruited to the respiratory tract. The current studies demonstrated differences in PBMC responses to the two viruses very early after exposure, including reduced fos protein and CD69 expression and IL-2 production by RSV-exposed T lymphocytes. Exposure to RSV resulted in reduced lymphocyte proliferation despite evidence of a virus-specific T lymphocyte frequency equivalent to that for influenza virus. Reduced RSV-induced proliferation was not due to apoptosis, which was itself reduced relative to that of influenza virus-exposed T lymphocytes. The data indicate that differential immune responses to RSV and influenza virus are determined early after exposure of human PBMC and support the concept that the anamnestic immune response that might prevent clinically evident reinfection is attenuated very soon after exposure to RSV. Thus, candidate RSV vaccines should be expected to reduce but not prevent clinical illness upon subsequent infection by RSV. Furthermore, effective therapeutic agents for RSV are likely to be needed, especially for high-risk populations, even after vaccine development.
Collapse
Affiliation(s)
- Elisa H Fleming
- Division of Infectious Diseases, Department of Internal Medicine, and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Eliana E Ochoa
- Division of Infectious Diseases, Department of Internal Medicine, and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Joan E Nichols
- Division of Infectious Diseases, Department of Internal Medicine, and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - M Kerry O'Banion
- Department of Neuroscience and Department of Neurology, University of Rochester School of Medicine, Rochester, New York
| | - Alan R Salkind
- Department of Medicine, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Norbert J Roberts
- Division of Infectious Diseases, Department of Internal Medicine, and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University School of Medicine, New York, New York
| |
Collapse
|
39
|
Dock J, Hultin L, Hultin P, Elliot J, Yang OO, Anton PA, Jamieson BD, Effros RB. Human immune compartment comparisons: Optimization of proliferative assays for blood and gut T lymphocytes. J Immunol Methods 2017; 445:77-87. [PMID: 28336395 PMCID: PMC5505254 DOI: 10.1016/j.jim.2017.03.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/30/2017] [Accepted: 03/17/2017] [Indexed: 11/20/2022]
Abstract
The accumulation of peripheral blood late-differentiated memory CD8 T cells with features of replicative (cellular) senescence, including inability to proliferate in vitro, has been extensively studied. Importantly, the abundance of these cells is directly correlated with increased morbidity and mortality in older persons. Of note, peripheral blood contains only 2% of the total body lymphocyte population. By contrast, the gut-associated lymphoid tissue (GALT) is the most extensive lymphoid organ, housing up to 60% of total body lymphocytes, but has never been assessed with respect to senescence profiles. We report here the development of a method for measuring and comparing proliferative capacity of peripheral blood and gut colorectal mucosa-derived CD8 T cells. The protocol involves a 5-day culture of mononuclear leukocyte populations, from blood and gut colorectal mucosa respectively, labeled with 5-(and 6)-carboxyfluorescein diacetate succinimidyl ester (CFSE) and 5-bromo-2'-deoxyuridine (BrdU) and stimulated with anti-CD2/3/28-linked microbeads. Variables tested and optimized as part of the protocol development include: mode of T cell stimulation, CFSE concentration, inclusion of a second proliferation marker, BrdU, culture duration, initial culture concentration, and inclusion of autologous irradiated feeder cells. Moving forward, this protocol demonstrates a significant advance in the ability of researchers to study compartment-specific differences of in vitro proliferative dynamics of CD8 T cells, as an indicator of replicative senescence and immunological aging. The study's two main novel contributions are (1) Optimization and adaptation of standard proliferative dynamics blood T cell protocols for T cells within the mucosal immune system. (2) Introduction of the novel technique of combining CFSE and BrdU staining to do so.
Collapse
Affiliation(s)
- Jeffrey Dock
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, United States
| | - Lance Hultin
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, United States; UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States
| | - Patricia Hultin
- Department of Epidemiology, Fielding School of Public Health, University of California-Los Angeles, Los Angeles, CA 90095, United States; UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States
| | - Julie Elliot
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, United States; UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States
| | - Otto O Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, United States; Department of Microbiology Immunology & Molecular Genetics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, United States; UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States; AIDS Healthcare Foundation, Los Angeles, CA 90028, United States
| | - Peter A Anton
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, United States; UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States
| | - Beth D Jamieson
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, United States; UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States
| | - Rita B Effros
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, United States; UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States.
| |
Collapse
|
40
|
Erez A, Altan-Bonnet G. Lymphocytic division clocked up by Myc. Immunol Cell Biol 2016; 95:119-120. [PMID: 27995905 DOI: 10.1038/icb.2016.115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Amir Erez
- National Cancer Institute, Bethesda, MD, USA
| | | |
Collapse
|
41
|
Hou D, Chen C, Seely EJ, Chen S, Song Y. High-Throughput Sequencing-Based Immune Repertoire Study during Infectious Disease. Front Immunol 2016; 7:336. [PMID: 27630639 PMCID: PMC5005336 DOI: 10.3389/fimmu.2016.00336] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 08/19/2016] [Indexed: 11/13/2022] Open
Abstract
The selectivity of the adaptive immune response is based on the enormous diversity of T and B cell antigen-specific receptors. The immune repertoire, the collection of T and B cells with functional diversity in the circulatory system at any given time, is dynamic and reflects the essence of immune selectivity. In this article, we review the recent advances in immune repertoire study of infectious diseases, which were achieved by traditional techniques and high-throughput sequencing (HTS) techniques. HTS techniques enable the determination of complementary regions of lymphocyte receptors with unprecedented efficiency and scale. This progress in methodology enhances the understanding of immunologic changes during pathogen challenge and also provides a basis for further development of novel diagnostic markers, immunotherapies, and vaccines.
Collapse
Affiliation(s)
- Dongni Hou
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University , Shanghai , China
| | - Cuicui Chen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University , Shanghai , China
| | - Eric John Seely
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California San Francisco , San Francisco, CA , USA
| | - Shujing Chen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University , Shanghai , China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University , Shanghai , China
| |
Collapse
|
42
|
Abstract
Co-stimulatory and co-inhibitory molecules direct the "second signal," which largely determines the outcome of the "first signal" generated by the interaction of T cell receptor (TCR) with cognate MHC-peptide complex. The co-stimulatory and co-inhibitory signals are key mechanistic contributors to the regulation of adaptive immunity, especially the T cell-mediated immune response. Regulatory T cells (Tregs) are a special population of T cells, which unlike other T cells function as "attenuators" to suppress T cell immunity. Dysregulation of either the "second signal" or Tregs leads to an unbalanced immune system, which can result in a range of immune-related disorders, including autoimmune diseases, chronic infections, and tumors. In contrast, precise manipulation of these two systems offers tremendous clinical opportunities to treat these same diseases. Co-stimulatory and co-inhibitory molecules modulate immunity at molecular level, whereas Tregs delicately control the immune response at cellular level. Accumulating evidence has demonstrated that these two regulatory strategies converge and synergize with each other. This review discusses recent progress on the roles of co-stimulatory and co-inhibitory signals in the context of Tregs.
Collapse
Affiliation(s)
- Weifeng Liu
- a Department of Biochemistry , Albert Einstein College of Medicine , Bronx , NY , USA.,b Department of Microbiology and Immunology , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Steven C Almo
- a Department of Biochemistry , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Xingxing Zang
- b Department of Microbiology and Immunology , Albert Einstein College of Medicine , Bronx , NY , USA.,c Department of Medicine , Albert Einstein College of Medicine , Bronx , NY , USA.,d Department of Urology , Albert Einstein College of Medicine , Bronx , NY , USA
| |
Collapse
|
43
|
Modeling the dynamics of neonatal CD8 + T-cell responses. Immunol Cell Biol 2016; 94:838-848. [PMID: 27142943 PMCID: PMC5069106 DOI: 10.1038/icb.2016.47] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/29/2016] [Accepted: 05/01/2016] [Indexed: 12/14/2022]
Abstract
Neonates are particularly susceptible to a number of infections, and the neonatal CD8+ T cell response demonstrates differences in both the phenotype and magnitude of responses to infection compared with adults. However, the underlying basis for these differences is unclear. We have used a mathematical modeling approach to analyze the dynamics of neonatal and adult CD8+ T cell responses following in vitro stimulation and in vivo infection, which allows us to dissect key cell-intrinsic differences in expansion, differentiation and memory formation. We found that neonatal cells started dividing 8 hrs earlier and proliferated at a faster rate (0.077 day−1 vs 0.105 day−1) than adult cells in vitro. In addition, neonatal cells also differentiated more rapidly, as measured by the loss in CD62L and Ly6C expression. We extended our mathematical modeling to analysis of neonatal and adult CD8+ T cells responding in vivo and demonstrated that neonatal cells divide more slowly than adult cells after day 4 post-infection. However, neonatal cells differentiate more rapidly, up-regulating more KLRG-1 per division than adult cells (20% vs. 5%). The dynamics of memory formation were also found to be different, with neonatal effector cells showing increased death (1.0 day−1 vs. 2.45 day−1). Comparison of the division of human cord blood and adult naïve cells stimulated in vitro showed more division in cord blood derived cells, consistent with the observations in mice. This work highlights differences of the cell-intrinsic division and differentiation program in neonatal CD8+ T cells.
Collapse
|
44
|
Fahl SP, Harris B, Coffey F, Wiest DL. Rpl22 Loss Impairs the Development of B Lymphocytes by Activating a p53-Dependent Checkpoint. THE JOURNAL OF IMMUNOLOGY 2016; 194:200-9. [PMID: 25416806 DOI: 10.4049/jimmunol.1402242] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although ribosomal proteins facilitate the ribosome’s core function of translation, emerging evidence suggests that some ribosomal proteins are also capable of performing tissue-restricted functions either from within specialized ribosomes or from outside of the ribosome. In particular, we have previously demonstrated that germline ablation of the gene encoding ribosomal protein Rpl22 causes a selective and p53-dependent arrest of ab T cell progenitors at the b-selection checkpoint. We have now identified a crucial role for Rpl22 during early B cell development. Germline ablation of Rpl22 results in a reduction in the absolute number of B-lineage progenitors in the bone marrow beginning at the pro–B cell stage. Although Rpl22-deficient pro–B cells are hyporesponsive to IL-7, a key cytokine required for early B cell development, the arrest of B cell development does not result from disrupted IL-7 signaling. Instead, p53 induction appears to be responsible for the developmental defects, as Rpl22 deficiency causes increased expression of p53 and activation of downstream p53 target genes, and p53 deficiency rescues the defect in B cell development in Rpl22-deficient mice. Interestingly, the requirement for Rpl22 in the B cell lineage appears to be developmentally restricted, because Rpl22-deficient splenic B cells proliferate normally in response to Ag receptor and Toll receptor stimuli and undergo normal class-switch recombination. These results indicate that Rpl22 performs a critical, developmentally restricted role in supporting early B cell development by preventing p53 induction.
Collapse
Affiliation(s)
- Shawn P Fahl
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Bryan Harris
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Francis Coffey
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - David L Wiest
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| |
Collapse
|
45
|
Cunningham CR, Champhekar A, Tullius MV, Dillon BJ, Zhen A, de la Fuente JR, Herskovitz J, Elsaesser H, Snell LM, Wilson EB, de la Torre JC, Kitchen SG, Horwitz MA, Bensinger SJ, Smale ST, Brooks DG. Type I and Type II Interferon Coordinately Regulate Suppressive Dendritic Cell Fate and Function during Viral Persistence. PLoS Pathog 2016; 12:e1005356. [PMID: 26808628 PMCID: PMC4726812 DOI: 10.1371/journal.ppat.1005356] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/01/2015] [Indexed: 12/21/2022] Open
Abstract
Persistent viral infections are simultaneously associated with chronic inflammation and highly potent immunosuppressive programs mediated by IL-10 and PDL1 that attenuate antiviral T cell responses. Inhibiting these suppressive signals enhances T cell function to control persistent infection; yet, the underlying signals and mechanisms that program immunosuppressive cell fates and functions are not well understood. Herein, we use lymphocytic choriomeningitis virus infection (LCMV) to demonstrate that the induction and functional programming of immunosuppressive dendritic cells (DCs) during viral persistence are separable mechanisms programmed by factors primarily considered pro-inflammatory. IFNγ first induces the de novo development of naive monocytes into DCs with immunosuppressive potential. Type I interferon (IFN-I) then directly targets these newly generated DCs to program their potent T cell immunosuppressive functions while simultaneously inhibiting conventional DCs with T cell stimulating capacity. These mechanisms of monocyte conversion are constant throughout persistent infection, establishing a system to continuously interpret and shape the immunologic environment. MyD88 signaling was required for the differentiation of suppressive DCs, whereas inhibition of stimulatory DCs was dependent on MAVS signaling, demonstrating a bifurcation in the pathogen recognition pathways that promote distinct elements of IFN-I mediated immunosuppression. Further, a similar suppressive DC origin and differentiation was also observed in Mycobacterium tuberculosis infection, HIV infection and cancer. Ultimately, targeting the underlying mechanisms that induce immunosuppression could simultaneously prevent multiple suppressive signals to further restore T cell function and control persistent infections. Persistent virus infections induce host derived immunosuppressive factors that attenuate the immune response and prevent control of infection. Although the mechanisms of T cell exhaustion are being defined, we know surprisingly little about the underlying mechanisms that induce the immunosuppressive state and the origin and functional programming of the cells that deliver these signals to the T cells. We recently demonstrated that type I interferon (IFN-I) signaling was responsible for many of the immune dysfunctions associated with persistent virus infection and in particular the induced expression of the suppressive factors IL-10 and PDL1 by dendritic cells (DCs). Yet, mechanistically how IFN-I signaling specifically generates and programs cells to become immunosuppressive is still unknown. Herein, we define the underlying mechanisms of IFN-I mediated immunosuppression and establish that the induction of factors and the generation of the DCs that express them are separable events integrally reliant on additional inflammatory factors. Further, we demonstrate a similar derivation of the suppressive DCs that emerge in other diseases associated with prolonged inflammation and immunosuppression, specifically in HIV infection, Mycobacterium tuberculosis, and cancer, indicating a conserved origin of immunosuppression and suggesting that targeting the pathways that underlie expression of immunosuppressive cells and factors could be beneficial to treat multiple chronic diseases.
Collapse
Affiliation(s)
- Cameron R. Cunningham
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Ameya Champhekar
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Michael V. Tullius
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Barbara Jane Dillon
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Anjie Zhen
- Division of Hematology and Oncology, Department of Medicine, UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Justin Rafael de la Fuente
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Jonathan Herskovitz
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Heidi Elsaesser
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Princess Margaret Cancer Center, Immune Therapy Program, University Health Network, Toronto, Ontario
| | - Laura M. Snell
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Princess Margaret Cancer Center, Immune Therapy Program, University Health Network, Toronto, Ontario
| | - Elizabeth B. Wilson
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Juan Carlos de la Torre
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Scott G. Kitchen
- Division of Hematology and Oncology, Department of Medicine, UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Marcus A. Horwitz
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Steven J. Bensinger
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Stephen T. Smale
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - David G. Brooks
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Princess Margaret Cancer Center, Immune Therapy Program, University Health Network, Toronto, Ontario
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
46
|
Ziraldo C, Gong C, Kirschner DE, Linderman JJ. Strategic Priming with Multiple Antigens can Yield Memory Cell Phenotypes Optimized for Infection with Mycobacterium tuberculosis: A Computational Study. Front Microbiol 2016; 6:1477. [PMID: 26779136 PMCID: PMC4701940 DOI: 10.3389/fmicb.2015.01477] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/08/2015] [Indexed: 12/16/2022] Open
Abstract
Lack of an effective vaccine results in 9 million new cases of tuberculosis (TB) every year and 1.8 million deaths worldwide. Although many infants are vaccinated at birth with BCG (an attenuated M. bovis), this does not prevent infection or development of TB after childhood. Immune responses necessary for prevention of infection or disease are still unknown, making development of effective vaccines against TB challenging. Several new vaccines are ready for human clinical trials, but these trials are difficult and expensive; especially challenging is determining the appropriate cellular response necessary for protection. The magnitude of an immune response is likely key to generating a successful vaccine. Characteristics such as numbers of central memory (CM) and effector memory (EM) T cells responsive to a diverse set of epitopes are also correlated with protection. Promising vaccines against TB contain mycobacterial subunit antigens (Ag) present during both active and latent infection. We hypothesize that protection against different key immunodominant antigens could require a vaccine that produces different levels of EM and CM for each Ag-specific memory population. We created a computational model to explore EM and CM values, and their ratio, within what we term Memory Design Space. Our model captures events involved in T cell priming within lymph nodes and tracks their circulation through blood to peripheral tissues. We used the model to test whether multiple Ag-specific memory cell populations could be generated with distinct locations within Memory Design Space at a specific time point post vaccination. Boosting can further shift memory populations to memory cell ratios unreachable by initial priming events. By strategically varying antigen load, properties of cellular interactions within the LN, and delivery parameters (e.g., number of boosts) of multi-subunit vaccines, we can generate multiple Ag-specific memory populations that cover a wide range of Memory Design Space. Given a set of desired characteristics for Ag-specific memory populations, we can use our model as a tool to predict vaccine formulations that will generate those populations.
Collapse
Affiliation(s)
- Cordelia Ziraldo
- Department of Chemical Engineering, University of Michigan, Ann ArborMI, USA; Department of Microbiology and Immunology, University of Michigan Medical School, Ann ArborMI, USA
| | - Chang Gong
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann ArborMI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann ArborMI, USA
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor MI, USA
| | - Jennifer J Linderman
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor MI, USA
| |
Collapse
|
47
|
Thurley K, Gerecht D, Friedmann E, Höfer T. Three-Dimensional Gradients of Cytokine Signaling between T Cells. PLoS Comput Biol 2015; 11:e1004206. [PMID: 25923703 PMCID: PMC4414419 DOI: 10.1371/journal.pcbi.1004206] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 02/17/2015] [Indexed: 11/20/2022] Open
Abstract
Immune responses are regulated by diffusible mediators, the cytokines, which act at sub-nanomolar concentrations. The spatial range of cytokine communication is a crucial, yet poorly understood, functional property. Both containment of cytokine action in narrow junctions between immune cells (immunological synapses) and global signaling throughout entire lymph nodes have been proposed, but the conditions under which they might occur are not clear. Here we analyze spatially three-dimensional reaction-diffusion models for the dynamics of cytokine signaling at two successive scales: in immunological synapses and in dense multicellular environments. For realistic parameter values, we observe local spatial gradients, with the cytokine concentration around secreting cells decaying sharply across only a few cell diameters. Focusing on the well-characterized T-cell cytokine interleukin-2, we show how cytokine secretion and competitive uptake determine this signaling range. Uptake is shaped locally by the geometry of the immunological synapse. However, even for narrow synapses, which favor intrasynaptic cytokine consumption, escape fluxes into the extrasynaptic space are expected to be substantial (≥20% of secretion). Hence paracrine signaling will generally extend beyond the synapse but can be limited to cellular microenvironments through uptake by target cells or strong competitors, such as regulatory T cells. By contrast, long-range cytokine signaling requires a high density of cytokine producers or weak consumption (e.g., by sparsely distributed target cells). Thus in a physiological setting, cytokine gradients between cells, and not bulk-phase concentrations, are crucial for cell-to-cell communication, emphasizing the need for spatially resolved data on cytokine signaling. The adaptive immune system fights pathogens through the activation of immune cell clones that specifically recognize a particular pathogen. Tight contacts, so-called immunological synapses, of immune cells with cells that present ‘digested’ pathogen molecules are pivotal for ensuring specificity. The discovery that immune responses are regulated by small diffusible proteins – the cytokines – has been surprising because cytokine diffusion to ‘bystander’ cells might compromise specificity. It has therefore been argued that cytokines are trapped in immunological synapses, whereas other authors have found that cytokines act on a larger scale through entire lymph nodes. Measurements of cytokine concentrations with fine spatial resolution have not been achieved. Here, we study the spatio-temporal dynamics of cytokines through mathematical analysis and three-dimensional numerical simulation and identify key parameters that control signaling range. We predict that even tight immunological synapses leak a substantial portion of the secreted cytokines. Nevertheless, rapid cellular uptake will render cytokine signals short-range and thus incidental activation of bystander cells can be limited. Long-range signals will only occur with multiple secreting cells or/and slow consumption by sparse target cells. Thus our study identifies key determinants of the spatial range of cytokine communication in realistic multicellular geometries.
Collapse
Affiliation(s)
- Kevin Thurley
- Division of Theoretical Systems Biology, German Cancer Research Center, Heidelberg, Germany
- Institute for Theoretical Biology, Charité-Universitätsmedizin, Berlin, Germany
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail: (KT); (DG); (EF); (TH)
| | - Daniel Gerecht
- Institute for Applied Mathematics, University of Heidelberg, Heidelberg, Germany
- * E-mail: (KT); (DG); (EF); (TH)
| | - Elfriede Friedmann
- Institute for Applied Mathematics, University of Heidelberg, Heidelberg, Germany
- * E-mail: (KT); (DG); (EF); (TH)
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center, Heidelberg, Germany
- Bioquant Center, University of Heidelberg, Heidelberg, Germany
- * E-mail: (KT); (DG); (EF); (TH)
| |
Collapse
|
48
|
Kinjyo I, Qin J, Tan SY, Wellard CJ, Mrass P, Ritchie W, Doi A, Cavanagh LL, Tomura M, Sakaue-Sawano A, Kanagawa O, Miyawaki A, Hodgkin PD, Weninger W. Real-time tracking of cell cycle progression during CD8+ effector and memory T-cell differentiation. Nat Commun 2015; 6:6301. [PMID: 25709008 PMCID: PMC4346633 DOI: 10.1038/ncomms7301] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 01/15/2015] [Indexed: 01/08/2023] Open
Abstract
The precise pathways of memory T-cell differentiation are incompletely understood. Here we exploit transgenic mice expressing fluorescent cell cycle indicators to longitudinally track the division dynamics of individual CD8+ T cells. During influenza virus infection in vivo, naive T cells enter a CD62Lintermediate state of fast proliferation, which continues for at least nine generations. At the peak of the anti-viral immune response, a subpopulation of these cells markedly reduces their cycling speed and acquires a CD62Lhi central memory cell phenotype. Construction of T-cell family division trees in vitro reveals two patterns of proliferation dynamics. While cells initially divide rapidly with moderate stochastic variations of cycling times after each generation, a slow-cycling subpopulation displaying a CD62Lhi memory phenotype appears after eight divisions. Phenotype and cell cycle duration are inherited by the progeny of slow cyclers. We propose that memory precursors cell-intrinsically modulate their proliferative activity to diversify differentiation pathways. CD8+ memory T cells appear during infection via a process of selection and differentiation that remains poorly understood. Using a fluorescent indicator of cell cycle progression, Kinjyo et al. show that slow-cycling memory precursors are derived from fast-cycling-activated T cells in influenza-infected mice.
Collapse
Affiliation(s)
- Ichiko Kinjyo
- Immune Imaging Program, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales 2042, Australia
| | - Jim Qin
- Immune Imaging Program, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales 2042, Australia
| | - Sioh-Yang Tan
- Immune Imaging Program, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales 2042, Australia
| | - Cameron J Wellard
- 1] Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Paulus Mrass
- Immune Imaging Program, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales 2042, Australia
| | - William Ritchie
- Immune Imaging Program, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales 2042, Australia
| | - Atsushi Doi
- Cell Innovator Co., Ltd., Fukuoka 812-8581, Japan
| | - Lois L Cavanagh
- Immune Imaging Program, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales 2042, Australia
| | - Michio Tomura
- Laboratory for Autoimmune Regulation, RIKEN Research Center for Allergy and Immunology, Yokohama 230-0045, Japan
| | - Asako Sakaue-Sawano
- Laboratory for Cell Function and Dynamics, Brain Science Institute, RIKEN, Saitama 351-0198, Japan
| | - Osami Kanagawa
- Laboratory for Autoimmune Regulation, RIKEN Research Center for Allergy and Immunology, Yokohama 230-0045, Japan
| | - Atsushi Miyawaki
- Laboratory for Cell Function and Dynamics, Brain Science Institute, RIKEN, Saitama 351-0198, Japan
| | - Philip D Hodgkin
- 1] Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Wolfgang Weninger
- 1] Immune Imaging Program, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales 2042, Australia [2] Discipline of Dermatology, Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia [3] Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia
| |
Collapse
|
49
|
Wang H, Yuan Z, Liu P, Zhou T. Division time-based amplifiers for stochastic gene expression. MOLECULAR BIOSYSTEMS 2015; 11:2417-28. [DOI: 10.1039/c5mb00391a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
While cell-to-cell variability is a phenotypic consequence of gene expression noise, sources of this noise may be complex – apart from intrinsic sources such as the random birth/death of mRNA and stochastic switching between promoter states, there are also extrinsic sources of noise such as cell division where division times are either constant or random.
Collapse
Affiliation(s)
- Haohua Wang
- School of Mathematics and Computational Science
- Sun Yat-Sen University
- Guangzhou 510275
- People's Republic of China
- Department of Mathematics College of Information Science and Technology Hainan University
| | - Zhanjiang Yuan
- School of Mathematics and Computational Science
- Sun Yat-Sen University
- Guangzhou 510275
- People's Republic of China
| | - Peijiang Liu
- School of Mathematics and Computational Science
- Sun Yat-Sen University
- Guangzhou 510275
- People's Republic of China
| | - Tianshou Zhou
- School of Mathematics and Computational Science
- Sun Yat-Sen University
- Guangzhou 510275
- People's Republic of China
| |
Collapse
|
50
|
Drozdenko G, Scheel T, Heine G, Baumgrass R, Worm M. Impaired T cell activation and cytokine production by calcitriol-primed human B cells. Clin Exp Immunol 2014; 178:364-72. [PMID: 24965738 DOI: 10.1111/cei.12406] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2014] [Indexed: 01/13/2023] Open
Abstract
The biologically active form of vitamin D3 , 1, 25-dihydroxyvitamin D3 (calcitriol), is a potent modulator of the immune response. We have shown previously that calcitriol modulates the immunoglobulin response in vitro and in vivo in mice and humans. To analyse the underlying molecular mechanisms we studied whether calcitriol-primed B cells modulate T cell activation and function. Human B cells were stimulated with anti-CD40 and interleukin (IL)-4 in the presence of increasing concentrations of calcitriol. After removal of calcitriol, primed B cells were co-cultured with autologous CD4(+) T cells; the B cell phenotype T cell activation and their consecutive cytokine production were also assessed. Naive T cells co-cultured with calcitriol-primed naive B cells showed a reduced expansion, nuclear factor of activated T cells, cytoplasmic 2 (NFATc2) expression and cytokine production upon restimulation. CD86 expression on B cells after calcitriol priming was identified as an underlying mechanism, as T cell activation and expansion was rescued by activating anti-CD28 antibodies. Our data indicate that calcitriol-primed B cells display an impaired capacity to activate T cells. Taken together, we identified a novel B cell-dependent vitamin D immune regulatory mechanism, namely by decreased co-stimulation of calcitriol-primed B cells.
Collapse
Affiliation(s)
- G Drozdenko
- Klinik für Dermatologie, Venerologie und Allergologie, Allergie-Centrum-Charité, CCM, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | |
Collapse
|