1
|
Bhamidipati K, Malleswara Rao Nakka N, Ahmed M, Javvaji K, Banerjee R, Puvvada N, Sesha Sainath AV, Chakravarty S. Enhancing cancer immunotherapy with mannose mimicking glycopolymer nanoparticles induced activation of Dendritic cells. Bioorg Chem 2024; 152:107711. [PMID: 39178706 DOI: 10.1016/j.bioorg.2024.107711] [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: 07/25/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024]
Abstract
Cancer immunotherapy leverages the immune system's inherent capacity to combat malignancies. However, effective stimulation of Dendritic cells (DCs) is challenging due to their limited distribution and the immune-suppressive tumor microenvironment. Thus, targeting mannose receptors, which are highly expressed on DCs, represents a promising strategy. This study investigates the development of mannose-based glycopolymer nanoparticles to induce activation of DCs through enhanced antigen presentation. A novel ABA-type triblock bioconjugated glycopolymer (PMn-b-PCL-b-PMn), which mimics mannose was synthesized. This polymer was further modified with Dihexadecyldimethylammonium bromide (DHDAB) to prepare cationic nanoparticles (CMNP) for gene delivery of pCMV-TRP2, an antigenic marker for both melanoma and glioblastoma. The immune response generated by CMNP and the CMNP-TRP2 polyplex was compared to an untreated control following subcutaneous injection in mice. Post-injection cytometric analysis revealed robust DC activation and increased T-cell populations in secondary lymphoid organs, including the spleen and lymph nodes. These findings suggest that CMNP can serve as a potent biomimicking vaccination vehicle against cancer, enhancing the immune response through targeted DCs activation.
Collapse
Affiliation(s)
- Keerti Bhamidipati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Naga Malleswara Rao Nakka
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India; Polymers and Functional Materials and Fluoro-Agrochemicals Department, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Mariam Ahmed
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Kalpana Javvaji
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Rajkumar Banerjee
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Nagaprasad Puvvada
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| | - Annadanam V Sesha Sainath
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India; Polymers and Functional Materials and Fluoro-Agrochemicals Department, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| | - Sumana Chakravarty
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
2
|
Ontiveros CO, Murray CE, Crossland G, Curiel TJ. Considerations and Approaches for Cancer Immunotherapy in the Aging Host. Cancer Immunol Res 2023; 11:1449-1461. [PMID: 37769157 PMCID: PMC11287796 DOI: 10.1158/2326-6066.cir-23-0121] [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: 02/08/2023] [Revised: 04/16/2023] [Accepted: 08/22/2023] [Indexed: 09/30/2023]
Abstract
Advances in cancer immunotherapy are improving treatment successes in many distinct cancer types. Nonetheless, most tumors fail to respond. Age is the biggest risk for most cancers, and the median population age is rising worldwide. Advancing age is associated with manifold alterations in immune cell types, abundance, and functions, rather than simple declines in these metrics, the consequences of which remain incompletely defined. Our understanding of the effects of host age on immunotherapy mechanisms, efficacy, and adverse events remains incomplete. A deeper understanding of age effects in all these areas is required. Most cancer immunotherapy preclinical studies examine young subjects and fail to assess age contributions, a remarkable deficit given the known importance of age effects on immune cells and factors mediating cancer immune surveillance and immunotherapy efficacy. Notably, some cancer immunotherapies are more effective in aged versus young hosts, while others fail despite efficacy in the young. Here, we review our current understanding of age effects on immunity and associated nonimmune cells, the tumor microenvironment, cancer immunotherapy, and related adverse effects. We highlight important knowledge gaps and suggest areas for deeper enquiries, including in cancer immune surveillance, treatment response, adverse event outcomes, and their mitigation.
Collapse
Affiliation(s)
- Carlos O. Ontiveros
- UT Health San Antonio Long School of Medicine and Graduate School of Biomedical Sciences, San Antonio, TX 78229
| | - Clare E. Murray
- UT Health San Antonio Long School of Medicine and Graduate School of Biomedical Sciences, San Antonio, TX 78229
| | - Grace Crossland
- Graduate School of Microbiology and Immunology, Dartmouth, Hanover, NH 03755
- The Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Tyler J. Curiel
- UT Health San Antonio Long School of Medicine and Graduate School of Biomedical Sciences, San Antonio, TX 78229
- Graduate School of Microbiology and Immunology, Dartmouth, Hanover, NH 03755
- The Geisel School of Medicine at Dartmouth, Hanover, NH 03755
- Dartmouth Health and Dartmouth Cancer Center, Lebanon, NH 03756
| |
Collapse
|
3
|
Sadasivam M, Jie C, Hamad ARA. Renal tubular epithelial cells are constitutive non-cognate stimulators of resident T cells. Cell Rep 2023; 42:113210. [PMID: 37796661 PMCID: PMC11259314 DOI: 10.1016/j.celrep.2023.113210] [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: 04/17/2023] [Revised: 08/06/2023] [Accepted: 09/18/2023] [Indexed: 10/07/2023] Open
Abstract
Understanding the roles of different cell types in regulating T cell homeostasis in various tissues is critical for understanding adaptive immunity. Here, we show that RTECs (renal tubular epithelial cells) are intrinsically programmed to polyclonally stimulate proliferation of kidney αβ T cells by a cell-cell contact mechanism that is major histocompatibility complex (MHC) independent and regulated by CD155, αVβ3-integrin, and vitronectin. Peripheral CD4 and CD8 are resistant to RTEC-mediated stimulation, while the minor subset of double-negative (DN) T cells are responsive. This functional property of RTEC is discovered by using a coculture system that recapitulates spontaneous in vivo polyclonal proliferation of kidney T cells, which are mainly comprised of central memory T (TCM) and effector memory T (TEM) cells. This robust cell-intrinsic stimulatory role of RTECs could be underlying the steady-state spontaneous proliferation of kidney T cells. The results have conceptual implications for understanding roles of different cell types in regulating systemic and organ-specific T cell homeostasis.
Collapse
Affiliation(s)
- Mohanraj Sadasivam
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 664G, Baltimore, MD 21205, USA
| | - Chunfa Jie
- Department of Biochemistry and Nutrition, Des Moines University, 3200 Grand Avenue, Ryan Hall 230, Des Moines, IA 50266, USA
| | - Abdel Rahim A Hamad
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 664G, Baltimore, MD 21205, USA; Department of Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 664G, Baltimore, MD 21205, USA.
| |
Collapse
|
4
|
von Essen MR, Chow HH, Holm Hansen R, Buhelt S, Sellebjerg F. Immune reconstitution following alemtuzumab therapy is characterized by exhausted T cells, increased regulatory control of proinflammatory T cells and reduced B cell control. Front Immunol 2023; 14:1249201. [PMID: 37744364 PMCID: PMC10512074 DOI: 10.3389/fimmu.2023.1249201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Alemtuzumab is a monoclonal antibody targeting CD52 on the surface of immune cells, approved for the treatment of active relapsing-remitting multiple sclerosis (RRMS). The purpose of this study was to analyze the repopulation of peripheral lymphocytes following alemtuzumab-induced lymphocyte depletion and investigate associations with disease activity and development of secondary autoimmunity. For this, blood samples were collected two years after initiation of alemtuzumab treatment and lymphocytes were subjected to a comprehensive flow cytometry analysis. Included in the study were 40 patients treated with alemtuzumab and 40 treatment-naïve patients with RRMS. Disease activity and development of secondary autoimmune disease was evaluated after three years of treatment. Our study confirms that alemtuzumab treatment profoundly alters the circulating lymphocyte phenotype and describes a reconstituted immune system characterized by T cell activation/exhaustion, an increased regulatory control of IL-17 producing effector T cells and CD20+ T cells, and a reduced control of B cells. There were no obvious associations between immune cell subsets and disease activity or development of secondary autoimmune disease during treatment with alemtuzumab. Our results indicate that the reconstituted immune response is skewed towards a more effective regulatory control of MS-associated proinflammatory T cell responses. Also, the enlarged pool of naïve B cells together with the apparent decrease in control of B cell activity may explain why alemtuzumab-treated patients retain the ability to mount a humoral immune response towards new antigens.
Collapse
Affiliation(s)
- Marina Rode von Essen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | | | | | | | | |
Collapse
|
5
|
Maenosono R. Sex difference and immunosenescence affect transplantation outcomes. FRONTIERS IN TRANSPLANTATION 2023; 2:1235740. [PMID: 38993850 PMCID: PMC11235384 DOI: 10.3389/frtra.2023.1235740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/09/2023] [Indexed: 07/13/2024]
Abstract
Kidney transplantation is a well-established alternative to renal replacement therapy. Although the number of patients with end-stage renal disease (ESRD) is increasing, the availability of kidney for transplantation is still insufficient to meet the needs. As age increases, the prevalence of ESRD increases; thus, the population of aged donors and recipients occupies large proportion. Accumulated senescent cells secrete pro-inflammatory factors and induce senescence. Additionally, it is gradually becoming clear that biological sex differences can influence aging and cause differences in senescence. Here, we review whether age-related sex differences affect organ transplant outcomes and what should be done in the future.
Collapse
Affiliation(s)
- Ryoichi Maenosono
- Department of Urology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| |
Collapse
|
6
|
Marques-Neto LM, Trentini MM, Kanno AI, Rodriguez D, Leite LCDC. Recombinant BCG expressing the LTAK63 adjuvant increased memory T cells and induced long-lasting protection against Mycobacterium tuberculosis challenge in mice. Front Immunol 2023; 14:1205449. [PMID: 37520577 PMCID: PMC10374402 DOI: 10.3389/fimmu.2023.1205449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/29/2023] [Indexed: 08/01/2023] Open
Abstract
Vaccine-induced protection against Mycobacterium tuberculosis (Mtb) is usually ascribed to the induction of Th1, Th17, and CD8+ T cells. However, protective immune responses should also involve other immune cell subsets, such as memory T cells. We have previously shown improved protection against Mtb challenge using the rBCG-LTAK63 vaccine (a recombinant BCG strain expressing the LTAK63 adjuvant, a genetically detoxified derivative of the A subunit from E. coli heat-labile toxin). Here we show that mice immunized with rBCG-LTAK63 exhibit a long-term (at least until 6 months) polyfunctional Th1/Th17 response in the draining lymph nodes and in the lungs. This response was accompanied by the increased presence of a diverse set of memory T cells, including central memory, effector memory and tissue-resident memory T cells. After the challenge, the T cell phenotype in the lymph nodes and lungs were characterized by a decrease in central memory T cells, and an increase in effector memory T cells and effector T cells. More importantly, when challenged 6 months after the immunization, this group demonstrated increased protection in comparison to BCG. In conclusion, this work provides experimental evidence in mice that the rBCG-LTAK63 vaccine induces a persistent increase in memory and effector T cell numbers until at least 6 months after immunization, which correlates with increased protection against Mtb. This improved immune response may contribute to enhance the long-term protection.
Collapse
|
7
|
Giannotta G, Murrone A, Giannotta N. COVID-19 mRNA Vaccines: The Molecular Basis of Some Adverse Events. Vaccines (Basel) 2023; 11:747. [PMID: 37112659 PMCID: PMC10145134 DOI: 10.3390/vaccines11040747] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
Each injection of any known vaccine results in a strong expression of pro-inflammatory cytokines. This is the result of the innate immune system activation, without which no adaptive response to the injection of vaccines is possible. Unfortunately, the degree of inflammation produced by COVID-19 mRNA vaccines is variable, probably depending on genetic background and previous immune experiences, which through epigenetic modifications could have made the innate immune system of each individual tolerant or reactive to subsequent immune stimulations.We hypothesize that we can move from a limited pro-inflammatory condition to conditions of increasing expression of pro-inflammatory cytokines that can culminate in multisystem hyperinflammatory syndromes following COVID-19 mRNA vaccines (MIS-V). We have graphically represented this idea in a hypothetical inflammatory pyramid (IP) and we have correlated the time factor to the degree of inflammation produced after the injection of vaccines. Furthermore, we have placed the clinical manifestations within this hypothetical IP, correlating them to the degree of inflammation produced. Surprisingly, excluding the possible presence of an early MIS-V, the time factor and the complexity of clinical manifestations are correlated to the increasing degree of inflammation: symptoms, heart disease and syndromes (MIS-V).
Collapse
Affiliation(s)
| | - Antonio Murrone
- Oncologia Territoriale, Hospice Cure Palliative ASUFC, 33030 Udine, Italy;
| | - Nicola Giannotta
- Medical and Surgery Sciences, Faculty of Medicine, Magna Græcia University, 88100 Catanzaro, Italy;
| |
Collapse
|
8
|
Yadav N, Patel H, Parmar R, Patidar M, Dalai SK. TCR-signals downstream adversely correlate with the survival signals of memory CD8 + T cells under homeostasis. Immunobiology 2023; 228:152354. [PMID: 36854249 DOI: 10.1016/j.imbio.2023.152354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/06/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023]
Abstract
The significance of self-peptide-MHC-I/TCR (SMT) interaction in the survival of CD8+ T cells during naïve- and developmental-stages is well documented. However, the same for the memory stage is contentious. Previous studies have attempted to address the issue using MHC-I or TCR deficient systems, but inconsistent findings with memory CD8+ T cells of different TCR specificities have complicated the interpretation. Differential presence and/or processing of TCR-signals downstream in memory CD8+ T cells of different TCR specificities could be thought of as a reason. In this study, we examined the TCR-signals downstream in memory CD8+ T cells and compared them to the presence of survival-related signals (Annexin-V, Bcl-2, and Ki-67). We categorically tracked foreign antigen-experienced memory CD8+ T (TM) cells generated after Plasmodium pre-erythrocytic-stage malaria infection in C57BL/6 mice. Interestingly, we found that memory CD8+ T cells had more TCR-signals downstream than naive cells. We reasoned and attributed the increased expression of cell adhesion molecules to the enhanced TCR-signaling. TCR-signals downstream correlate more closely with survival signals in naive CD8+ T cells than with death signals in TM cells. Further investigation using antigen-specific CD8+ T cells and diverse infection systems would aid in conceptualizing the findings.
Collapse
Affiliation(s)
- Naveen Yadav
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India; Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, USA.
| | - Hardik Patel
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Rajesh Parmar
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, USA
| | - Manoj Patidar
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India; Department of Zoology, Govt. College Manawar, Dhar, Madhya Pradesh, India
| | - Sarat K Dalai
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India.
| |
Collapse
|
9
|
Schaible P. Modifying enzyme replacement therapy - A perspective. J Cell Mol Med 2023; 27:165-173. [PMID: 36566487 PMCID: PMC9843529 DOI: 10.1111/jcmm.17653] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/26/2022] Open
Abstract
Several diseases are caused by the lack of functional proteins, including lysosomal storage diseases or haemophilia A and B. Patients suffering from one of these diseases are treated via enzyme replacement therapies to restore the missing protein. Although this treatment strategy prevents some disease symptoms, enzyme replacement therapies are very expensive and require very frequent infusions, which can cause infusion adverse reactions and massively impair the quality of life of the patients. This review proposes a technology to sustainably produce proteins within the patient to potentially make frequent protein-infusions redundant. This technology is based on blood circulating immune cells as producers of the needed therapeutic protein. To ensure a stable protein concentration over time the cells are equipped with a system, which induces cell proliferation when low therapeutic protein levels are detected and a system inhibiting cell proliferation when high therapeutic protein levels are detected.
Collapse
|
10
|
Oladipo OO, Adedeji BO, Adedokun SP, Gbadamosi JA, Salaudeen M. Regulation of effector and memory CD8 + T cell differentiation: a focus on orphan nuclear receptor NR4A family, transcription factor, and metabolism. Immunol Res 2022; 71:314-327. [PMID: 36571657 DOI: 10.1007/s12026-022-09353-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/16/2022] [Indexed: 12/27/2022]
Abstract
CD8 + T cells undergo rapid expansion followed by contraction and the development of memory cells after their receptors are activated. The development of immunological memory following acute infection is a complex phenomenon that involves several molecular, transcriptional, and metabolic mechanisms. As memory cells confer long-term protection and respond to secondary stimulation with strong effector function, understanding the mechanisms that influence their development is of great importance. Orphan nuclear receptors, NR4As, are immediate early genes that function as transcription factors and bind with the NBRE region of chromatin. Interestingly, the NBRE region of activated CD8 + T cells is highly accessible at the same time the expression of NR4As is induced. This suggests a potential role of NR4As in the early events post T cell activation that determines cell fate decisions. In this review, we will discuss the influence of NR4As on the differentiation of CD8 + T cells during the immune response to acute infection and the development of immunological memory. We will also discuss the signals, transcription factors, and metabolic mechanisms that control cell fate decisions. HIGHLIGHTS: Memory CD8 + T cells are an essential subset that mediates long-term protection after pathogen encounters. Some specific environmental cues, transcriptional factors, and metabolic pathways regulate the differentiation of CD8 + T cells and the development of memory cells. Orphan nuclear receptor NR4As are early genes that act as transcription factors and are highly expressed post-T cell receptor activation. NR4As influence the effector function and differentiation of CD8 + T cells and also control the development of immunological memory following acute infection.
Collapse
Affiliation(s)
- Oladapo O Oladipo
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
- College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| | - Bernard O Adedeji
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Department of Biochemistry and Nutrition, Nigerian Institute of Medical Research (NIMR), Yaba, Lagos, Nigeria
| | - Samson P Adedokun
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Jibriil A Gbadamosi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Marzuq Salaudeen
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
11
|
Soluble factors from TLR4- or TCR-activated cells contribute to stability of the resting phenotype and increase the expression of CXCR4 of human memory CD4 T cells. Immunol Res 2022; 71:388-403. [PMID: 36539634 DOI: 10.1007/s12026-022-09345-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022]
Abstract
It has been proposed that cytokines can induce activation of resting T cells in an antigen-independent manner. However, experimental conditions have included the use of fetal serum and nanogram concentrations of added cytokines. To evaluate the effect of cytokines and chemokines generated by activated immune cells on the phenotypic profile of human memory CD4 T cells, the cells were cultured in FBS-free conditions in the presence of IL-15 and 5% of hAB serum and incubated with conditioned medium (CM) obtained from PBMC activated through the TCR using anti-CD3/CD28/CD2 antibodies (TCR-CM) or through TLR4 using bacterial LPS (TLR4-CM). Cytokines and chemokines present in the CMs were evaluated by ProcartaPlex immunoassay. Cell viability, proliferation, and surface markers were determined by flow cytometry on day 2, 5, and 8 of culture. Cell viability was maintained by TLR4-CM plus IL-15 for 8 days but decreased in the presence of the TCR-CM plus IL-15. In combination with IL-15, the TLR4-CM, but not the TCR-CM, maintained the expression of CD3 and CD4 stable. Both conditions stabilized the expression of CD45RO and CCR5. Thus, the TLR4-CM better supported the viability and stability of the memory phenotype. None of the CMs induced proliferation or expression of activation markers; however, they induced an increased expression of CXCR4. This study indicates that resting memory CD4 T cells are not activated by, but may be sensitive to soluble factors produced by antigen or PAMP-stimulated cells, which may contribute to their homeostasis and favor the CXCR4 expression.
Collapse
|
12
|
Choi A, Jung YW, Choi H. The extrinsic factors important to the homeostasis of allergen-specific memory CD4 T cells. Front Immunol 2022; 13:1080855. [PMID: 36591273 PMCID: PMC9798121 DOI: 10.3389/fimmu.2022.1080855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Memory T cells, which are generated after the primary immune response to cognate antigens, possess unique features compared to naïve or effector T cells. These memory T cells are maintained for a long period of time and robustly reactivate in lymphoid or peripheral tissues where they re-encounter antigens. Environments surrounding memory T cells are importantly involved in the process of the maintenance and reactivation of these T cells. Although memory T cells are generally believed to be formed in response to acute infections, the pathogenesis and persistence of chronic inflammatory diseases, including allergic diseases, are also related to the effector functions of memory CD4 T cells. Thus, the factors involved in the homeostasis of allergen-specific memory CD4 T cells need to be understood to surmount these diseases. Here, we review the characteristics of allergen-specific memory CD4 T cells in allergic diseases and the importance of extrinsic factors for the homeostasis and reactivation of these T cells in the view of mediating persistence, recurrence, and aggravation of allergic diseases. Overall, this review provides a better understanding of memory CD4 T cells to devise effective therapeutic strategies for refractory chronic inflammatory diseases.
Collapse
Affiliation(s)
| | - Yong Woo Jung
- Department of Pharmacy, Korea University, Sejong-si, Republic of Korea
| | - Hanbyeul Choi
- Department of Pharmacy, Korea University, Sejong-si, Republic of Korea
| |
Collapse
|
13
|
Guan S, Suman S, Amann JM, Wu R, Carbone DP, Wang J, Dikov MM. Metabolic reprogramming by adenosine antagonism and implications in non-small cell lung cancer therapy. Neoplasia 2022; 32:100824. [PMID: 35914370 PMCID: PMC9344351 DOI: 10.1016/j.neo.2022.100824] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/24/2022] [Accepted: 07/11/2022] [Indexed: 11/26/2022]
Abstract
Non-small cell lung cancer (NSCLC) is a heterogeneous disease with genetic and environmental parameters that influence cell metabolism. Because of the complex interplay of environmental factors within the tumor microenvironment (TME) and the profound impact of these factors on the metabolic activities of tumor and immune cells, there is an emerging interest to advance the understanding of these diverse metabolic phenotypes in the TME. High levels of adenosine are characteristic of the TME, and adenosine can have a significant impact on both tumor cell growth and the immune response. Consistent with this, we showed in NSCLC data from TCGA that high expression of the A2BR leads to worse outcome and that expression of A2BR may be different for different mutation backgrounds. We then investigated the metabolic reprogramming of tumor cells and immune cells (T and dendritic cells) by adenosine. We used A2AR and A2BR antagonism or agonism as well as receptor knockout animals to explore whether these treatments altered specific immune compartments or conferred specific therapeutic vulnerabilities. Using the seahorse assay, we found that an A2BR antagonist modulates oxidative stress homeostasis in NSCLC cell lines. In addition, we found distinct metabolic roles of A2AR and A2BR receptors in T cell activation and dendritic cell maturation. These data suggest potential mechanisms and therapeutic benefits of A2 receptor antagonist therapy in NSCLC.
Collapse
Affiliation(s)
- Shuxiao Guan
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, United States.
| | - Shankar Suman
- The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, United States
| | - Joseph M Amann
- The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, United States
| | - Ruohan Wu
- The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, United States
| | - David P Carbone
- The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, United States
| | - Jie Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Mikhail M Dikov
- The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, United States
| |
Collapse
|
14
|
Tracking fluorescently labeled IL-15 and anti-PD-1 in the tumor microenvironment and draining lymph nodes. J Immunol Methods 2022; 505:113253. [PMID: 35358495 DOI: 10.1016/j.jim.2022.113253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/11/2022] [Accepted: 03/07/2022] [Indexed: 02/01/2023]
Abstract
Understanding the dynamics of the tumor microenvironment (TME) has become vital in discovering new targets for effective immunotherapies and enhancing current treatments. However, localization and distribution of immune cells and treatment biomolecules are poorly characterized to date. In this study, a murine Luminal B mammary adenocarcinoma model received a combinatorial treatment of fluorescently labeled anti-PD-1-Cy3 and IL-15 complex-Cy5 injected interperitoneally and intratumorally, respectively. Fluorescent labeling allowed for the visualization of the distribution of IL-15 complexes and anti-PD-1, as well as their localization to immune cells in the TME and tumor-draining lymph node. Using fluorescent microscopy and light sheet microscopy of whole-clarified tumors and draining lymph nodes, the localization of IL-15 complexes was found to be distributed around the periphery of the tumor at 4 h post injection and medially located at the center of the tumor at 24 h post injection, corresponding with high densities of CD8 cells in the tumor present at 48 h and 72 h post injection. Anti-PD-1 was distributed around the perimeter of the tumor and colocalized to IL-15 in the draining lymph nodes 24 h post injection. Colocalization of IL-15 was also established with NK cells, CD8+ T cells, and macrophages. This study develops a novel method to spatiotemporally track fluorescently labeled immunotherapeutic biomolecules in vivo, with implications for optimizing and further understanding the pharmacokinetics of clinical immunotherapies.
Collapse
|
15
|
Miron M, Meng W, Rosenfeld AM, Dvorkin S, Poon MML, Lam N, Kumar BV, Louzoun Y, Luning Prak ET, Farber DL. Maintenance of the human memory T cell repertoire by subset and tissue site. Genome Med 2021; 13:100. [PMID: 34127056 PMCID: PMC8204429 DOI: 10.1186/s13073-021-00918-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 06/01/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Immune-mediated protection is mediated by T cells expressing pathogen-specific T cell antigen receptors (TCR) that are maintained at diverse sites of infection as tissue-resident memory T cells (TRM) or that disseminate as circulating effector-memory (TEM), central memory (TCM), or terminal effector (TEMRA) subsets in blood and tissues. The relationship between circulating and tissue resident T cell subsets in humans remains elusive, and is important for promoting site-specific protective immunity. METHODS We analyzed the TCR repertoire of the major memory CD4+ and CD8+T cell subsets (TEM, TCM, TEMRA, and TRM) isolated from blood and/or lymphoid organs (spleen, lymph nodes, bone marrow) and lungs of nine organ donors, and blood of three living individuals spanning five decades of life. High-throughput sequencing of the variable (V) portion of individual TCR genes for each subset, tissue, and individual were analyzed for clonal diversity, expansion and overlap between lineage, T cell subsets, and anatomic sites. TCR repertoires were further analyzed for TRBV gene usage and CDR3 edit distance. RESULTS Across blood, lymphoid organs, and lungs, human memory, and effector CD8+T cells exhibit greater clonal expansion and distinct TRBV usage compared to CD4+T cell subsets. Extensive sharing of clones between tissues was observed for CD8+T cells; large clones specific to TEMRA cells were present in all sites, while TEM cells contained clones shared between sites and with TRM. For CD4+T cells, TEM clones exhibited the most sharing between sites, followed by TRM, while TCM clones were diverse with minimal sharing between sites and subsets. Within sites, TRM clones exhibited tissue-specific expansions, and maintained clonal diversity with age, compared to age-associated clonal expansions in circulating memory subsets. Edit distance analysis revealed tissue-specific biases in clonal similarity. CONCLUSIONS Our results show that the human memory T cell repertoire comprises clones which persist across sites and subsets, along with clones that are more restricted to certain subsets and/or tissue sites. We also provide evidence that the tissue plays a key role in maintaining memory T cells over age, bolstering the rationale for site-specific targeting of memory reservoirs in vaccines and immunotherapies.
Collapse
Affiliation(s)
- Michelle Miron
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
- Columbia Center for Translational Immunology, Columbia University, New York, NY, USA
| | - Wenzhao Meng
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aaron M Rosenfeld
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shirit Dvorkin
- Department of Mathematics, Bar Ilan University, Ramat Gan, Israel
| | - Maya Meimei Li Poon
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Nora Lam
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Brahma V Kumar
- Columbia Center for Translational Immunology, Columbia University, New York, NY, USA
| | - Yoram Louzoun
- Department of Mathematics, Bar Ilan University, Ramat Gan, Israel
| | - Eline T Luning Prak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA.
- Department of Surgery, Columbia University, New York, NY, USA.
| |
Collapse
|
16
|
Zhao Y, Cai C, Samir J, Palgen JL, Keoshkerian E, Li H, Bull RA, Luciani F, An H, Lloyd AR. Human CD8 T-stem cell memory subsets phenotypic and functional characterization are defined by expression of CD122 or CXCR3. Eur J Immunol 2021; 51:1732-1747. [PMID: 33844287 DOI: 10.1002/eji.202049057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/07/2021] [Accepted: 04/07/2021] [Indexed: 11/12/2022]
Abstract
Long-lived T-memory stem cells (TSCM ) are key to both naturally occurring and vaccine-conferred protection against infection. These cells are characterized by the CD45RA+ CCR7+ CD95+ phenotype. Significant heterogeneity within the TSCM population is recognized, but distinguishing surface markers and functional characterization of potential subsets are lacking. Human CD8 TSCM subsets were identified in healthy subjects who had been previously exposed to CMV or Influenza (Flu) virus in flow cytometry by expression of CD122 or CXCR3, and then characterized in proliferation, multipotency, self-renewal, and intracellular cytokine production (TNF-α, IL-2, IFN-γ), together with transcriptomic profiles. The TSCM CD122hi -expressing subset (versus CD122lo ) demonstrated greater proliferation, greater multipotency, and enhanced polyfunctionality with higher frequencies of triple positive (TNF-α, IL-2, IFN-γ) cytokine-producing cells upon exposure to recall antigen. The TSCM CXCR3lo subpopulation also had increased proliferation and polyfunctional cytokine production. Transcriptomic analysis further showed that the TSCM CD122hi population had increased expression of activation and homing molecules, such as Ccr6, Cxcr6, Il12rb, and Il18rap, and downregulated cell proliferation inhibitors, S100A8 and S100A9. These data reveal that the TSCM CD122hi phenotype is associated with increased proliferation, enhanced multipotency and polyfunctionality with an activated memory-cell like transcriptional profile, and hence, may be favored for induction by immunization and for adoptive immunotherapy.
Collapse
Affiliation(s)
- Yanran Zhao
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Curtis Cai
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Jerome Samir
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Jean-Louis Palgen
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Elizabeth Keoshkerian
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Hui Li
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Rowena A Bull
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Fabio Luciani
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia.,School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Hongyan An
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Andrew R Lloyd
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| |
Collapse
|
17
|
Heterodimeric IL-15 in Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13040837. [PMID: 33671252 PMCID: PMC7922495 DOI: 10.3390/cancers13040837] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The rapidly expanding field of cancer immunotherapy uses diverse technologies, including cytokines, T cells, and antibody administration, with the aim to induce effective immune responses leading to tumor control. Interleukin-15 (IL-15), a cytokine discovered in 1994, supports the homeostasis of cytotoxic immune cells and shows promise as an anti-tumor agent. Many studies have elucidated IL-15 synthesis, regulation and biological function and explored its therapeutic efficacy in preclinical cancer models. Escherichia coli-derived single-chain IL-15 was tested in the first in-human trial in cancer patients. Its effects were limited by the biology of IL-15, which in vivo comprises a complex of the IL-15 chain with the IL-15 receptor alpha (IL-15Rα) chain, together forming the IL-15 heterodimer (hetIL-15). Currently, single-chain IL-15 and several heterodimeric IL-15:IL-15Rα variants (hetIL-15, N-803 and RLI) are being tested in clinical trials. This review presents a summary of contemporary preclinical and clinical research on IL-15. Abstract Immunotherapy has emerged as a valuable strategy for the treatment of many cancer types. Interleukin-15 (IL-15) promotes the growth and function of cytotoxic CD8+ T and natural killer (NK) cells. It also enhances leukocyte trafficking and stimulates tumor-infiltrating lymphocytes expansion and activity. Bioactive IL-15 is produced in the body as a heterodimeric cytokine, comprising the IL-15 and the so-called IL-15 receptor alpha chain that are together termed “heterodimeric IL-15” (hetIL-15). hetIL-15, closely resembling the natural form of the cytokine produced in vivo, and IL-15:IL-15Rα complex variants, such as hetIL-15Fc, N-803 and RLI, are the currently available IL-15 agents. These molecules have showed favorable pharmacokinetics and biological function in vivo in comparison to single-chain recombinant IL-15. Preclinical animal studies have supported their anti-tumor activity, suggesting IL-15 as a general method to convert “cold” tumors into “hot”, by promoting tumor lymphocyte infiltration. In clinical trials, IL-15-based therapies are overall well-tolerated and result in the expansion and activation of NK and memory CD8+ T cells. Combinations with other immunotherapies are being investigated to improve the anti-tumor efficacy of IL-15 agents in the clinic.
Collapse
|
18
|
Wu J, Li G, Li L, Li D, Dong Z, Jiang P. Asparagine enhances LCK signalling to potentiate CD8 + T-cell activation and anti-tumour responses. Nat Cell Biol 2021; 23:75-86. [PMID: 33420490 DOI: 10.1038/s41556-020-00615-4] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 11/26/2020] [Indexed: 01/16/2023]
Abstract
Nutrient availability is central for T-cell functions and immune responses. Here we report that CD8+ T-cell activation and anti-tumour responses are strongly potentiated by the non-essential amino acid Asn. Increased Asn levels enhance CD8+ T-cell activation and effector functions against tumour cells in vitro and in vivo. Conversely, restriction of dietary Asn, ASNase administration or inhibition of the Asn transporter SLC1A5 impairs the activity and responses of CD8+ T cells. Mechanistically, Asn does not directly alter cellular metabolic fluxes; it instead binds the SRC-family protein tyrosine kinase LCK and orchestrates LCK phosphorylation at Tyr 394 and 505, thereby leading to enhanced LCK activity and T-cell-receptor signalling. Thus, our findings reveal a critical and metabolism-independent role for Asn in the direct modulation of the adaptive immune response by controlling T-cell activation and efficacy, and further uncover that LCK is a natural Asn sensor signalling Asn sufficiency to T-cell functions.
Collapse
Affiliation(s)
- Jun Wu
- Tsinghua-Peking Center for Life Sciences, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Gen Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Le Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Dan Li
- School of Medicine and Institute for Immunology, Tsinghua University, Beijing, China
| | - Zhongjun Dong
- School of Medicine and Institute for Immunology, Tsinghua University, Beijing, China
| | - Peng Jiang
- Tsinghua-Peking Center for Life Sciences, Beijing, China. .,School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
19
|
Hu C, Liu W, Xu N, Huang A, Zhang Z, Fan M, Ruan G, Wang Y, Xi T, Xing Y. Silk fibroin hydrogel as mucosal vaccine carrier: induction of gastric CD4+TRM cells mediated by inflammatory response induces optimal immune protection against Helicobacter felis. Emerg Microbes Infect 2020; 9:2289-2302. [PMID: 33000989 PMCID: PMC7594714 DOI: 10.1080/22221751.2020.1830719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tissue-resident memory T (TRM) cells, located in the epithelium of most peripheral tissues, constitute the first-line defense against pathogen infections. Our previous study reported that gastric subserous layer (GSL) vaccination induced a “pool” of protective tissue-resident memory CD4+T (CD4+TRM) cells in the gastric epithelium. However, the mechanistic details how CD4+TRM cells form in the gastric epithelium are unknown. Here, our results suggested that the vaccine containing CCF in combination with Silk fibroin hydrogel (SF) broadened the distribution of gastric intraepithelial CD4+TRM cells. It was revealed that the gastric intraepithelial TRM cells were even more important than circulating memory T cells against infection by Helicobacter felis. It was also shown that gastric-infiltrating neutrophils were involved as indispensable mediators which secreted CXCL10 to chemoattract CXCR3+CD4+T cells into the gastric epithelium. Blocking of CXCR3 or neutrophils significantly decreased the number of gastric intraepithelial CD4+TRM cells due to reduced recruitment of CD4+T cells. This study demonstrated the protective efficacy of gastric CD4+TRM cells against H. felis infection, and highlighted the influence of neutrophils on gastric intraepithelial CD4+TRM cells formation.
Collapse
Affiliation(s)
- Chupeng Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Wei Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ningyin Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, People's Republic of China
| | - An Huang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Zhenxing Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Menghui Fan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Guojing Ruan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yue Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
20
|
Singh J, Mohtashami M, Anderson G, Zúñiga-Pflücker JC. Thymic Engraftment by in vitro-Derived Progenitor T Cells in Young and Aged Mice. Front Immunol 2020; 11:1850. [PMID: 32973763 PMCID: PMC7462002 DOI: 10.3389/fimmu.2020.01850] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
T cells play a critical role in mediating antigen-specific and long-term immunity against viral and bacterial pathogens, and their development relies on the highly specialized thymic microenvironment. T cell immunodeficiency can be acquired in the form of inborn errors, or can result from perturbations to the thymus due to aging or irradiation/chemotherapy required for cancer treatment. Hematopoietic stem cell transplant (HSCT) from compatible donors is a cornerstone for the treatment of hematological malignancies and immunodeficiency. Although it can restore a functional immune system, profound impairments exist in recovery of the T cell compartment. T cells remain absent or low in number for many months after HSCT, depending on a variety of factors including the age of the recipient. While younger patients have a shorter refractory period, the prolonged T cell recovery observed in older patients can lead to a higher risk of opportunistic infections and increased predisposition to relapse. Thus, strategies for enhancing T cell recovery in aged individuals are needed to counter thymic damage induced by radiation and chemotherapy toxicities, in addition to naturally occurring age-related thymic involution. Preclinical results have shown that robust and rapid long-term thymic reconstitution can be achieved when progenitor T cells, generated in vitro from HSCs, are co-administered during HSCT. Progenitor T cells appear to rely on lymphostromal crosstalk via receptor activator of NF-κB (RANK) and RANK-ligand (RANKL) interactions, creating chemokine-rich niches within the cortex and medulla that likely favor the recruitment of bone marrow-derived thymus seeding progenitors. Here, we employed preclinical mouse models to demonstrate that in vitro-generated progenitor T cells can effectively engraft involuted aged thymuses, which could potentially improve T cell recovery. The utility of progenitor T cells for aged recipients positions them as a promising cellular therapy for immune recovery and intrathymic repair following irradiation and chemotherapy, even in a post-involution thymus.
Collapse
Affiliation(s)
| | | | - Graham Anderson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Juan Carlos Zúñiga-Pflücker
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
21
|
Collins N. Dietary Regulation of Memory T Cells. Int J Mol Sci 2020; 21:ijms21124363. [PMID: 32575427 PMCID: PMC7352243 DOI: 10.3390/ijms21124363] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Memory T cells are a fundamental component of immunological memory, providing rapid and potent host protection against secondary challenges. As such, memory T cells are key targets in the design of vaccination strategies and cancer immunotherapies, making it critical to understand the factors and mechanisms that regulate their biology. Diet is an environmental feature that impacts virtually all aspects of host physiology. However, the influence of specific dietary regiments and nutritional components on the immune system is only just starting to be uncovered. This article will review literature regarding the impact of diet and nutrition on memory T cell development, maintenance and function. It was recently shown that caloric restriction without undernutrition enhances memory T cell function, while diets high in fiber are also beneficial. However, memory T cell responses are dysfunctional in extreme nutritional states, such as undernutrition and diet-induced obesity. Therefore, diet and host nutritional status are major regulators of memory T cell biology and host fitness. To define the dietary balance required to promote optimal memory T cell responses could allow for the implementation of rational diet-based therapies that prevent or treat disease. Furthermore, that certain dietary regiments can enhance memory T cell function indicates the possibility of harnessing the underlying mechanisms in the design of novel vaccination strategies and cancer immunotherapies.
Collapse
Affiliation(s)
- Nicholas Collins
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Cline-Smith A, Axelbaum A, Shashkova E, Chakraborty M, Sanford J, Panesar P, Peterson M, Cox L, Baldan A, Veis D, Aurora R. Ovariectomy Activates Chronic Low-Grade Inflammation Mediated by Memory T Cells, Which Promotes Osteoporosis in Mice. J Bone Miner Res 2020; 35:1174-1187. [PMID: 31995253 PMCID: PMC8061311 DOI: 10.1002/jbmr.3966] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/23/2019] [Accepted: 01/16/2020] [Indexed: 12/27/2022]
Abstract
The loss of estrogen (E2 ) initiates a rapid phase of bone loss leading to osteoporosis in one-half of postmenopausal women, but the mechanism is not fully understood. Here, we show for the first time how loss of E2 activates low-grade inflammation to promote the acute phase of bone catabolic activity in ovariectomized (OVX) mice. E2 regulates the abundance of dendritic cells (DCs) that express IL-7 and IL-15 by inducing the Fas ligand (FasL) and apoptosis of the DC. In the absence of E2 , DCs become long-lived, leading to increased IL-7 and IL-15. We find that IL-7 and IL-15 together, but not alone, induced antigen-independent production of IL-17A and TNFα in a subset of memory T cells (TMEM ). OVX of mice with T-cell-specific ablation of IL15RA showed no IL-17A and TNFα expression, and no increase in bone resorption or bone loss, confirming the role of IL-15 in activating the TMEM and the need for inflammation. Our results provide a new mechanism by which E2 regulates the immune system, and how menopause leads to osteoporosis. The low-grade inflammation is likely to cause or contribute to other comorbidities observed postmenopause. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Anna Cline-Smith
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Ariel Axelbaum
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Elena Shashkova
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Mousumi Chakraborty
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Jessie Sanford
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Prabhjyot Panesar
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Macey Peterson
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Linda Cox
- Division of Bone and Mineral Diseases, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Angel Baldan
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Deborah Veis
- Division of Bone and Mineral Diseases, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Rajeev Aurora
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
23
|
Rodriguez-Barbosa JI, Schneider P, Graca L, Bühler L, Perez-Simon JA, del Rio ML. The Role of TNFR2 and DR3 in the In Vivo Expansion of Tregs in T Cell Depleting Transplantation Regimens. Int J Mol Sci 2020; 21:E3347. [PMID: 32397343 PMCID: PMC7247540 DOI: 10.3390/ijms21093347] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 01/05/2023] Open
Abstract
Regulatory T cells (Tregs) are essential for the maintenance of tolerance to self and non-self through cell-intrinsic and cell-extrinsic mechanisms. Peripheral Tregs survival and clonal expansion largely depend on IL-2 and access to co-stimulatory signals such as CD28. Engagement of tumor necrosis factor receptor (TNFR) superfamily members, in particular TNFR2 and DR3, contribute to promote peripheral Tregs expansion and sustain their survival. This property can be leveraged to enhance tolerance to allogeneic transplants by tipping the balance of Tregs over conventional T cells during the course of immune reconstitution. This is of particular interest in peri-transplant tolerance induction protocols in which T cell depletion is applied to reduce the frequency of alloreactive T cells or in conditioning regimens that allow allogeneic bone marrow transplantation. These conditioning regimens are being implemented to limit long-term side effects of continuous immunosuppression and facilitate the establishment of a state of donor-specific tolerance. Lymphopenia-induced homeostatic proliferation in response to cytoreductive conditioning is a window of opportunity to enhance preferential expansion of Tregs during homeostatic proliferation that can be potentiated by agonist stimulation of TNFR.
Collapse
Affiliation(s)
- Jose-Ignacio Rodriguez-Barbosa
- Transplantation Immunobiology, School of Biology and Biotechnology, Institute of Molecular Biology, Genomics and Proteomics, University of Leon, 24071 Leon, Spain;
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland;
| | - Luis Graca
- School of Medicine, Institute of Molecular Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal;
| | - Leo Bühler
- Faculty of Science and Medicine, Section of Medicine, University of Fribourg, 1700 Fribourg, Switzerland;
| | - Jose-Antonio Perez-Simon
- Department of Hematology, Institute of Biomedicine (IBIS/CSIC), University Hospital Virgen del Rocio, 41013 Sevilla, Spain;
| | - Maria-Luisa del Rio
- Transplantation Immunobiology, School of Biology and Biotechnology, Institute of Molecular Biology, Genomics and Proteomics, University of Leon, 24071 Leon, Spain;
| |
Collapse
|
24
|
Leth S, Jensen-Fangel S. Programmed cell death protein 1 (PD-1) in infection. APMIS 2020; 128:177-187. [PMID: 32304591 DOI: 10.1111/apm.13045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/08/2020] [Indexed: 12/13/2022]
Abstract
Exhausted and dysfunctional T cells triggered by infection and cancer render the immune system unable to eliminate these pathogens. Pharmacologic blockade of the surface receptors that inhibit T-cell function has shown remarkable success in patients with various malignancies. In this Review, we discuss the emerging evidence of inhibiting checkpoint pathways as a potential role in controlling or clearing infectious diseases. Though interesting tendencies, much work is still needed in order to develop safe strategies that can be translated into clinically relevant outcomes in patients with infections.
Collapse
Affiliation(s)
- Steffen Leth
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Jensen-Fangel
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
25
|
Memory CD4 + T Cells in Immunity and Autoimmune Diseases. Cells 2020; 9:cells9030531. [PMID: 32106536 PMCID: PMC7140455 DOI: 10.3390/cells9030531] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/20/2020] [Accepted: 02/20/2020] [Indexed: 12/26/2022] Open
Abstract
CD4+ T helper (Th) cells play central roles in immunity in health and disease. While much is known about the effector function of Th cells in combating pathogens and promoting autoimmune diseases, the roles and biology of memory CD4+ Th cells are complex and less well understood. In human autoimmune diseases such as multiple sclerosis (MS), there is a critical need to better understand the function and biology of memory T cells. In this review article we summarize current concepts in the field of CD4+ T cell memory, including natural history, developmental pathways, subsets, and functions. Furthermore, we discuss advancements in the field of the newly-described CD4+ tissue-resident memory T cells and of CD4+ memory T cells in autoimmune diseases, two major areas of important unresolved questions in need of answering to advance new vaccine design and development of novel treatments for CD4+ T cell-mediated autoimmune diseases.
Collapse
|
26
|
Lack of B Lymphocytes Enhances CD8 T Cell-Mediated Resistance against Respiratory Viral Infection but Compromises Memory Cell Formation. J Virol 2020; 94:JVI.01877-19. [PMID: 31723023 DOI: 10.1128/jvi.01877-19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 11/20/2022] Open
Abstract
Following a respiratory virus infection, CXCR3hi CX3CR1lo and CXCR3lo CX3CR1hi CD8 T cells localize to different compartments within the lung and play an important role in host resistance, but mechanisms governing their optimal generation are poorly defined. We serendipitously found that B cell-deficient (μMT-/-) mice were highly resistant to lethal infection with a virulent poxvirus strain and that depletion of CD8 T cells rendered these mice susceptible to infection. B cells were not required for the expansion of virus-specific CD8 T cells, but a greater proportion of activated CD8 T cells acquired an effector-like CXCR3lo CX3CR1hi phenotype in the absence of B cells. After recovery from infection, CD8 T cells in μMT-/- mice contracted normally but failed to survive and seed the memory cell pool in both the lungs and spleen. These findings reveal a previously unappreciated role for B cells in regulating the balance between CD8 T cell-mediated resistance against respiratory viral infection and memory cell development.IMPORTANCE B cells play critical role in host resistance against many respiratory viral infections. However, the role of B cells beyond antibody-producing cells is less well defined. In this study, we made a surprising observation that mice lacking B cells were more resistant to respiratory infection with vaccinia virus than wild-type mice. This enhanced resistance was mediated by CD8 T cells because when we depleted CD8 T cells in B cell-deficient mice, these mice were unable to survive the infection. Interestingly, CD8 T cells in B cell-deficient mice were skewed more toward effector phenotype and less toward memory phenotype, which resulted in severely compromised memory CD8 T cell development. Thus, our study shows a novel role of B cells as regulators of CD8 T cell-mediated host resistance and memory CD8 T cell formation during respiratory viral infection.
Collapse
|
27
|
Zacharias ZR, Legge KL. Chronic Ethanol Consumption Reduces Existing CD8 T Cell Memory and Is Associated with Lesions in Protection against Secondary Influenza A Virus Infections. THE JOURNAL OF IMMUNOLOGY 2019; 203:3313-3324. [PMID: 31712384 DOI: 10.4049/jimmunol.1900770] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/16/2019] [Indexed: 01/12/2023]
Abstract
Chronic alcohol consumption is associated with an increased incidence of disease severity during pulmonary infections. Our previous work in a mouse model of chronic alcohol consumption has detailed that the primary influenza A virus (IAV)-specific CD8 T cell response in mice that consumed ethanol (EtOH) had a reduced proliferative capacity as well as the ability to kill IAV target cells. Interestingly, recent studies have highlighted that human alcoholics have an increased susceptibility to IAV infections, even though they likely possess pre-existing immunity to IAV. However, the effects of chronic alcohol consumption on pre-existing immune responses (i.e., memory) to IAV have not been explored. Our results presented in this study show that IAV-immune mice that then chronically consumed alcohol (X31→EtOH) exhibited increased morbidity and mortality following IAV re-exposure compared with IAV-immune mice that had consumed water (X31→H2O). This increased susceptibility in X31→EtOH mice was associated with reduced IAV-specific killing of target cells and a reduction in the number of IAV-specific CD8 T cells within the lungs. Furthermore, upon IAV challenge, recruitment of the remaining memory IAV-specific CD8 T cells into the lungs is reduced in X31→EtOH mice. This altered recruitment is associated with a reduced pulmonary expression of CXCL10 and CXCL11, which are chemokines that are important for T cell recruitment to the lungs. Overall, these results demonstrate that chronic alcohol consumption negatively affects the resting memory CD8 T cell response and reduces the ability of memory T cells to be recruited to the site of infection upon subsequent exposures, therein contributing to an enhanced susceptibility to IAV infections.
Collapse
Affiliation(s)
- Zeb R Zacharias
- Interdisciplinary Immunology Graduate Program, University of Iowa, Iowa City, IA 52242.,Department of Pathology, University of Iowa, Iowa City, IA 52242; and
| | - Kevin L Legge
- Interdisciplinary Immunology Graduate Program, University of Iowa, Iowa City, IA 52242; .,Department of Pathology, University of Iowa, Iowa City, IA 52242; and.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
28
|
Silva PHL, Santos LN, Mendes MA, Nery JAC, Sarno EN, Esquenazi D. Involvement of TNF-Producing CD8 + Effector Memory T Cells with Immunopathogenesis of Erythema Nodosum Leprosum in Leprosy Patients. Am J Trop Med Hyg 2019; 100:377-385. [PMID: 30652669 DOI: 10.4269/ajtmh.18-0517] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Type 2 reaction (T2R) or erythema nodosum leprosum (ENL), a sudden episode of acute inflammation predominantly affecting lepromatous leprosy patients (LL), characterized by a reduced cellular immune response. This possibly indicates a close relationship between the onset of T2R and the altered frequency, and functional activity of T lymphocytes, particularly of memory subsets. This study performed ex vivo and in vitro characterizations of T cell blood subpopulations from LL patients with or without T2R. In addition, the evaluation of activity of these subpopulations was performed by analyzing the frequency of these cells producing IFN-γ, TNF, and IL-10 by flow cytometry. Furthermore, the expression of transcription factors, for the differentiation of T cells, were analyzed by quantitative real-time polymerase chain reaction. Our results showed an increased frequency of CD8+/TNF+ effector memory T cells (TEM) among T2Rs. Moreover, there was evidence of a reduced frequency of CD4 and CD8+ IFN-γ-producing cells in T2R, and a reduced expression of STAT4 and TBX21. Finally, a significant and positive correlation between bacteriological index (BI) of T2R patients and CD4+/TNF+ and CD4+/IFN-γ+ T cells was observed. Thus, negative correlation between BI and the frequency of CD4+/IL-10+ T cells was noted. These results suggest that CD8+/TNF+ TEM are primarily responsible for the transient alteration in the immune response to Mycobacterium leprae in ENL patients. Thus, our study improves our understanding of pathogenic mechanisms and might suggest new therapeutic approaches for leprosy.
Collapse
Affiliation(s)
- Pedro Henrique L Silva
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Luciana N Santos
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Mayara A Mendes
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - José A C Nery
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Euzenir N Sarno
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Danuza Esquenazi
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Department of Pathology and Laboratories, School of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
29
|
Wu C, Espinoza DA, Koelle SJ, Yang D, Truitt L, Schlums H, Lafont BA, Davidson-Moncada JK, Lu R, Kaur A, Hammer Q, Li B, Panch S, Allan DA, Donahue RE, Childs RW, Romagnani C, Bryceson YT, Dunbar CE. Clonal expansion and compartmentalized maintenance of rhesus macaque NK cell subsets. Sci Immunol 2019; 3:3/29/eaat9781. [PMID: 30389798 DOI: 10.1126/sciimmunol.aat9781] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/03/2018] [Indexed: 12/12/2022]
Abstract
Natural killer (NK) cells recognize and eliminate infected and malignant cells. Their life histories are poorly understood, particularly in humans, due to lack of informative models and endogenous clonal markers. Here, we apply transplantation of barcoded rhesus macaque hematopoietic cells to interrogate the landscape of NK cell production, expansion, and life histories at a clonal level long term and after proliferative challenge. We identify oligoclonal populations of rhesus CD56-CD16+ NK cells that are characterized by marked expansions and contractions over time yet remained long-term clonally uncoupled from other hematopoietic lineages, including CD56+CD16- NK cells. Individual or groups of CD56-CD16+ expanded clones segregated with surface expression of specific killer immunoglobulin-like receptors. These clonally distinct NK cell subpopulation patterns persisted for more than 4 years, including after transient in vivo anti-CD16-mediated depletion and subsequent regeneration. Profound and sustained interleukin-15-mediated depletion was required to generate new oligoclonal CD56-CD16+ NK cells. Together, our results indicate that linear NK cell production from multipotent hematopoietic progenitors or less mature CD56+CD16- cells is negligible during homeostasis and moderate proliferative stress. In such settings, peripheral compartmentalized self-renewal can maintain the composition of distinct, differentiated NK cell subpopulations.
Collapse
Affiliation(s)
- Chuanfeng Wu
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Diego A Espinoza
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samson J Koelle
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA.,Department of Statistics, University of Washington, Seattle, WA, USA
| | - Di Yang
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA.,Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lauren Truitt
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Heinrich Schlums
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Bernard A Lafont
- Viral Immunology Section, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Jan K Davidson-Moncada
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA.,Clinical Development and Translational Research, MacroGenics Inc. Rockville, MD, USA
| | - Rong Lu
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Amitinder Kaur
- Tulane National Primate Research Center, Covington, LA, USA
| | - Quirin Hammer
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Stockholm, Sweden.,Deutsches Rheuma-Forschungszentrum-A Leibnitz Institute, Charite Medical University, Berlin, Germany
| | - Brian Li
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA.,Department of Medicine, Beth Israel Hospital, Boston, MA, USA
| | - Sandhya Panch
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA.,Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - David A Allan
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Robert E Donahue
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Richard W Childs
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Chiara Romagnani
- Deutsches Rheuma-Forschungszentrum-A Leibnitz Institute, Charite Medical University, Berlin, Germany
| | - Yenan T Bryceson
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Stockholm, Sweden. .,Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Cynthia E Dunbar
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
30
|
Padrón Á, Hurez V, Gupta HB, Clark CA, Pandeswara SL, Yuan B, Svatek RS, Turk MJ, Drerup JM, Li R, Curiel TJ. Age effects of distinct immune checkpoint blockade treatments in a mouse melanoma model. Exp Gerontol 2018; 105:146-154. [DOI: 10.1016/j.exger.2017.12.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/26/2017] [Accepted: 12/28/2017] [Indexed: 02/08/2023]
|
31
|
Wang Q, Gao J, Wu X. Pseudoprogression and hyperprogression after checkpoint blockade. Int Immunopharmacol 2018; 58:125-135. [DOI: 10.1016/j.intimp.2018.03.018] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/10/2018] [Accepted: 03/19/2018] [Indexed: 12/25/2022]
|
32
|
Hurez V, Padrón Á, Svatek RS, Curiel TJ. Considerations for successful cancer immunotherapy in aged hosts. Exp Gerontol 2017; 107:27-36. [PMID: 28987644 DOI: 10.1016/j.exger.2017.10.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/30/2017] [Accepted: 10/03/2017] [Indexed: 12/22/2022]
Abstract
Improvements in understanding cancer immunopathogenesis have now led to unprecedented successes in immunotherapy to treat numerous cancers. Although aging is the most important risk factor for cancer, most pre-clinical cancer immunotherapy studies are undertaken in young hosts. This review covers age-related immune changes as they affect cancer immune surveillance, immunopathogenesis and immune therapy responses. Declining T cell function with age can impede efficacy of age-related cancer immunotherapies, but examples of successful approaches to breach this barrier have been reported. It is further recognized now that immune functions with age do not simply decline, but that they change in potentially detrimental ways. For example, detrimental immune cell populations can become predominant during aging (notably pro-inflammatory cells), the prevalence or function of suppressive cells can increase (notably myeloid derived suppressor cells), drugs can have age-specific effects on immune cells, and attributes of the aged microenvironment can impede or subvert immunity. Key advances in these and related areas will be reviewed as they pertain to cancer immunotherapy in the aged, and areas requiring additional study and some speculations on future research directions will be addressed. We prefer the term Age Related Immune Dysfunction (ARID) as most encompassing the totality of age-associated immune changes.
Collapse
Affiliation(s)
- Vincent Hurez
- Department of Medicine, University of Texas Health San Antonio, TX 78229, USA
| | - Álvaro Padrón
- Department of Medicine, University of Texas Health San Antonio, TX 78229, USA
| | - Robert S Svatek
- Department of Urology, University of Texas Health San Antonio, TX 78229, USA; The UT Health Cancer Center, University of Texas Health San Antonio, TX 78229, USA
| | - Tyler J Curiel
- Department of Medicine, University of Texas Health San Antonio, TX 78229, USA; The UT Health Cancer Center, University of Texas Health San Antonio, TX 78229, USA; Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, TX 78229, USA; The Barshop Institute for Aging and Longevity Studies, University of Texas Health San Antonio, TX 78229, USA.
| |
Collapse
|
33
|
Population mechanics: A mathematical framework to study T cell homeostasis. Sci Rep 2017; 7:9511. [PMID: 28842645 PMCID: PMC5573381 DOI: 10.1038/s41598-017-09949-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/17/2017] [Indexed: 12/01/2022] Open
Abstract
Unlike other cell types, T cells do not form spatially arranged tissues, but move independently throughout the body. Accordingly, the number of T cells in the organism does not depend on physical constraints imposed by the shape or size of specific organs. Instead, it is determined by competition for interleukins. From the perspective of classical population dynamics, competition for resources seems to be at odds with the observed high clone diversity, leading to the so-called diversity paradox. In this work we make use of population mechanics, a non-standard theoretical approach to T cell homeostasis that accounts for clone diversity as arising from competition for interleukins. The proposed models show that carrying capacities of T cell populations naturally emerge from the balance between interleukins production and consumption. These models also suggest remarkable functional differences in the maintenance of diversity in naïve and memory pools. In particular, the distribution of memory clones would be biased towards clones activated more recently, or responding to more aggressive pathogenic threats. In contrast, permanence of naïve T cell clones would be determined by their affinity for cognate antigens. From this viewpoint, positive and negative selection can be understood as mechanisms to maximize naïve T cell diversity.
Collapse
|
34
|
Sowell RT, Goldufsky JW, Rogozinska M, Quiles Z, Cao Y, Castillo EF, Finnegan A, Marzo AL. IL-15 Complexes Induce Migration of Resting Memory CD8 T Cells into Mucosal Tissues. THE JOURNAL OF IMMUNOLOGY 2017; 199:2536-2546. [PMID: 28814601 DOI: 10.4049/jimmunol.1501638] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 07/20/2017] [Indexed: 11/19/2022]
Abstract
IL-15 is an essential cytokine known to promote T cell survival and activate the effector function of memory phenotype CD8 T cells. Blocking IL-15 signals also significantly impacts tissue-specific effector and memory CD8 T cell formation. In this study, we demonstrate that IL-15 influences the generation of memory CD8 T cells by first promoting their accumulation into mucosal tissues and second by sustaining expression of Bcl-6 and T-bet. We show that the mechanism for this recruitment is largely dependent on mammalian target of rapamycin and its subsequent inactivation of FoxO1. Last, we show that IL-15 complexes delivered locally to mucosal tissues without reinfection is an effective strategy to enhance establishment of tissue resident memory CD8 T cells within mucosal tissues. This study provides mechanistic insight into how IL-15 controls the generation of memory CD8 T cells and influences their trafficking and ability to take up residence within peripheral tissues.
Collapse
Affiliation(s)
- Ryan T Sowell
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL 60612
| | - Josef W Goldufsky
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL 60612.,Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612; and
| | - Magdalena Rogozinska
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612; and
| | - Zurisaday Quiles
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612; and
| | - Yanxia Cao
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL 60612
| | - Eliseo F Castillo
- Department of Internal Medicine, Clinical Translational Science Center, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Alison Finnegan
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL 60612.,Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612; and
| | - Amanda L Marzo
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL 60612; .,Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612; and
| |
Collapse
|
35
|
Xu A, Freywald A, Xie Y, Li Z, Xiang J. CD8 + memory T-cell inflation renders compromised CD4 + T-cell-dependent CD8 + T-cell immunity via naïve T-cell anergy. Immunotargets Ther 2017; 6:39-49. [PMID: 28670575 PMCID: PMC5479263 DOI: 10.2147/itt.s131662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Whether inflation of CD8+ memory T (mT) cells, which is often derived from repeated prime-boost vaccinations or chronic viral infections in the elderly, would affect late CD8+ T-cell immunity is a long-standing paradox. We have previously established an animal model with mT-cell inflation by transferring ConA-stimulated monoclonal CD8+ T cells derived from Ova-specific T-cell-receptor transgenic OTI mice into irradiation-induced lymphopenic B6 mice. In this study, we also established another two animal models with mT-cell inflation by transferring, 1) ConA-stimulated monoclonal CD8+ T cells derived from lymphocytic choriomeningitis virus glycoprotein-specific T-cell-receptor transgenic P14 mice, and 2) ConA-stimulated polyclonal CD8+ T cells derived from B6.1 mice into B6 mice with irradiation-induced lymphopenia. We vaccinated these mice with recombinant Ova-expressing Listeria monocytogenes and Ova-pulsed dendritic cells, which stimulated CD4+ T cell-independent and CD4+ T-cell-dependent CD8+ T-cell responses, respectively, and assessed Ova-specific CD8+ T-cell responses by flow cytometry. We found that Ova-specific CD8+ T-cell responses derived from the latter but not the former vaccination were significantly reduced in mice with CD8+ mT-cell inflation compared to wild-type B6 mice. We determined that naïve CD8+ T cells purified from splenocytes of mice with mT-cell inflation had defects in cell proliferation upon stimulation in vitro and in vivo and upregulated T-cell anergy-associated Itch and GRAIL molecules. Taken together, our data reveal that CD8+ mT-cell inflation renders compromised CD4+ T-cell-dependent CD8+ T-cell immunity via naïve T-cell anergy, and thus show promise for the design of efficient vaccines for elderly patients with CD8+ mT-cell inflation.
Collapse
Affiliation(s)
- Aizhang Xu
- Cancer Research Cluster, Saskatchewan Cancer Agency.,Department of Oncology
| | - Andrew Freywald
- Department of Pathology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yufeng Xie
- Department of Oncology, First Affiliated Hospital, Soochow University, Suzhou
| | - Zejun Li
- Shanghai Veterinary Research Institute, Shanghai, China
| | - Jim Xiang
- Cancer Research Cluster, Saskatchewan Cancer Agency.,Department of Oncology
| |
Collapse
|
36
|
Hurez V, Padrón ÁS, Svatek RS, Curiel TJ. Considerations for successful cancer immunotherapy in aged hosts. Clin Exp Immunol 2016; 187:53-63. [PMID: 27690272 DOI: 10.1111/cei.12875] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2016] [Indexed: 12/22/2022] Open
Abstract
Immunotherapy is now experiencing unprecedented successes in treating various cancers based on new understandings of cancer immunopathogenesis. Nonetheless, although ageing is the biggest risk factor for cancer, the majority of cancer immunotherapy preclinical studies are conducted in young hosts. This review will explore age-related changes in immunity as they relate to cancer immune surveillance, immunopathogenesis and responses to immunotherapy. Although it is recognized that declining T cell function with age poses a great challenge to developing effective age-related cancer immunotherapies, examples of successful approaches to overcome this hurdle have been developed. Further, it is now recognized that immune functions do not simply decline with age, but rather change in ways than can be detrimental. For example, with age, specific immune cell populations with detrimental functions can become predominant (such as cells producing proinflammatory cytokines), suppressive cells can become more numerous or more suppressive (such as myeloid-derived suppressor cells), drugs can affect aged immune cells distinctly and the aged microenvironment is becoming recognized as a significant barrier to address. Key developments in these and other areas will be surveyed as they relate to cancer immunotherapy in aged hosts, and areas in need of more study will be assessed with some speculations for the future. We propose the term 'age-related immune dysfunction' (ARID) as best representative of age-associated changes in immunity.
Collapse
Affiliation(s)
- V Hurez
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Á S Padrón
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - R S Svatek
- Department of Urology, University of Texas Health Science Center, San Antonio, TX, USA.,Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, TX, USA
| | - T J Curiel
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA.,Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, TX, USA.,Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, TX, USA.,The Barshop Institute for Ageing and Longevity Studies, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
37
|
Geiger R, Rieckmann J, Wolf T, Basso C, Feng Y, Fuhrer T, Kogadeeva M, Picotti P, Meissner F, Mann M, Zamboni N, Sallusto F, Lanzavecchia A. L-Arginine Modulates T Cell Metabolism and Enhances Survival and Anti-tumor Activity. Cell 2016; 167. [PMID: 27745970 PMCID: PMC5075284 DOI: 10.1016/j.cell.2016.09.031 10.1016/j.cell.2016.09.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Metabolic activity is intimately linked to T cell fate and function. Using high-resolution mass spectrometry, we generated dynamic metabolome and proteome profiles of human primary naive T cells following activation. We discovered critical changes in the arginine metabolism that led to a drop in intracellular L-arginine concentration. Elevating L-arginine levels induced global metabolic changes including a shift from glycolysis to oxidative phosphorylation in activated T cells and promoted the generation of central memory-like cells endowed with higher survival capacity and, in a mouse model, anti-tumor activity. Proteome-wide probing of structural alterations, validated by the analysis of knockout T cell clones, identified three transcriptional regulators (BAZ1B, PSIP1, and TSN) that sensed L-arginine levels and promoted T cell survival. Thus, intracellular L-arginine concentrations directly impact the metabolic fitness and survival capacity of T cells that are crucial for anti-tumor responses.
Collapse
Affiliation(s)
- Roger Geiger
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona 6500, Switzerland,Institute of Microbiology, ETH Zurich, Zurich 8093, Switzerland,Corresponding author
| | - Jan C. Rieckmann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Tobias Wolf
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona 6500, Switzerland,Institute of Microbiology, ETH Zurich, Zurich 8093, Switzerland
| | - Camilla Basso
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona 6500, Switzerland
| | - Yuehan Feng
- Institute of Biochemistry, ETH Zurich, Zurich 8093, Switzerland
| | - Tobias Fuhrer
- Institute of Molecular Systems Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Maria Kogadeeva
- Institute of Molecular Systems Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Paola Picotti
- Institute of Biochemistry, ETH Zurich, Zurich 8093, Switzerland
| | - Felix Meissner
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona 6500, Switzerland,Center of Medical Immunology, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona 6500, Switzerland
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona 6500, Switzerland,Institute of Microbiology, ETH Zurich, Zurich 8093, Switzerland,Corresponding author
| |
Collapse
|
38
|
L-Arginine Modulates T Cell Metabolism and Enhances Survival and Anti-tumor Activity. Cell 2016; 167:829-842.e13. [PMID: 27745970 PMCID: PMC5075284 DOI: 10.1016/j.cell.2016.09.031] [Citation(s) in RCA: 1150] [Impact Index Per Article: 127.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/18/2016] [Accepted: 09/19/2016] [Indexed: 12/11/2022]
Abstract
Metabolic activity is intimately linked to T cell fate and function. Using high-resolution mass spectrometry, we generated dynamic metabolome and proteome profiles of human primary naive T cells following activation. We discovered critical changes in the arginine metabolism that led to a drop in intracellular L-arginine concentration. Elevating L-arginine levels induced global metabolic changes including a shift from glycolysis to oxidative phosphorylation in activated T cells and promoted the generation of central memory-like cells endowed with higher survival capacity and, in a mouse model, anti-tumor activity. Proteome-wide probing of structural alterations, validated by the analysis of knockout T cell clones, identified three transcriptional regulators (BAZ1B, PSIP1, and TSN) that sensed L-arginine levels and promoted T cell survival. Thus, intracellular L-arginine concentrations directly impact the metabolic fitness and survival capacity of T cells that are crucial for anti-tumor responses. Dataset on dynamic metabolome/proteome profiles of activated human naive T cells Intracellular L-arginine levels regulate several metabolic pathways in T cells T cells with increased L-arginine display enhanced survival and anti-tumor activity LiP-MS identified proteins that are structurally modified by high L-arginine levels
Collapse
|
39
|
A molecular threshold for effector CD8(+) T cell differentiation controlled by transcription factors Blimp-1 and T-bet. Nat Immunol 2016; 17:422-32. [PMID: 26950239 DOI: 10.1038/ni.3410] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/01/2016] [Indexed: 12/11/2022]
Abstract
T cell responses are guided by cytokines that induce transcriptional regulators, which ultimately control differentiation of effector and memory T cells. However, it is unknown how the activities of these molecular regulators are coordinated and integrated during the differentiation process. Using genetic approaches and transcriptional profiling of antigen-specific CD8(+) T cells, we reveal a common program of effector differentiation that is regulated by IL-2 and IL-12 signaling and the combined activities of the transcriptional regulators Blimp-1 and T-bet. The loss of both T-bet and Blimp-1 leads to abrogated cytotoxic function and ectopic IL-17 production in CD8(+) T cells. Overall, our data reveal two major overlapping pathways of effector differentiation governed by the availability of Blimp-1 and T-bet and suggest a model for cytokine-induced transcriptional changes that combine, quantitatively and qualitatively, to promote robust effector CD8(+) T cell differentiation.
Collapse
|
40
|
Suzuki T, Kishimoto H, Abe R. Requirement of interleukin 7 signaling for anti-tumor immune response under lymphopenic conditions in a murine lung carcinoma model. Cancer Immunol Immunother 2016; 65:341-54. [PMID: 26880265 PMCID: PMC11028809 DOI: 10.1007/s00262-016-1808-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 02/03/2016] [Indexed: 12/13/2022]
Abstract
Induction of lymphopenia before adoptive transfer of T cells was followed by lymphopenia-induced proliferation (LIP) and generated a potent anti-tumor immune response in rodents and in a clinical setting. Previously, we reported that CD28 signaling is essential for the differentiation of functional effector cytotoxic T lymphocytes (CTLs) under lymphopenic conditions and sequential LIP of T cells. In this study, to clarify the correlation between LIP and the anti-tumor effect, LIP was inhibited with interleukin 7 (IL7) receptor blockade at various stages, and the anti-tumor effect then assessed. We confirmed that IL7 signaling at the start of LIP is crucial for the anti-tumor immune response. In contrast, continuous IL7 signaling was not required for tumor regression, although LIP of naïve CD8+ T cells is usually regulated by IL7. The expansion and migration of CTLs in lymphopenic hosts depend on IL7 signaling during the induction phase. Here, we propose that IL7 signaling and subsequent LIP of T cells have distinct roles in the induction of T cell immunity during lymphopenia.
Collapse
Affiliation(s)
- Toshihiro Suzuki
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba, 278-0022, Japan
| | - Hidehiro Kishimoto
- Parasitology and Immunopathoetiology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Ryo Abe
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba, 278-0022, Japan.
| |
Collapse
|
41
|
Setoguchi R. IL-15 boosts the function and migration of human terminally differentiated CD8+T cells by inducing a unique gene signature. Int Immunol 2016; 28:293-305. [DOI: 10.1093/intimm/dxw004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/03/2016] [Indexed: 12/13/2022] Open
|
42
|
Panackal AA, Williamson KC, van de Beek D, Boulware DR, Williamson PR. Fighting the Monster: Applying the Host Damage Framework to Human Central Nervous System Infections. mBio 2016; 7:e01906-15. [PMID: 26814182 PMCID: PMC4742705 DOI: 10.1128/mbio.01906-15] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The host damage-response framework states that microbial pathogenesis is a product of microbial virulence factors and collateral damage from host immune responses. Immune-mediated host damage is particularly important within the size-restricted central nervous system (CNS), where immune responses may exacerbate cerebral edema and neurological damage, leading to coma and death. In this review, we compare human host and therapeutic responses in representative nonviral generalized CNS infections that induce archetypal host damage responses: cryptococcal menigoencephalitis and tuberculous meningitis in HIV-infected and non-HIV-infected patients, pneumococcal meningitis, and cerebral malaria. Consideration of the underlying patterns of host responses provides critical insights into host damage and may suggest tailored adjunctive therapeutics to improve disease outcome.
Collapse
Affiliation(s)
- Anil A Panackal
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kim C Williamson
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Diederik van de Beek
- Department of Neurology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - David R Boulware
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Peter R Williamson
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
43
|
Baz A, Groves P, Buttigieg K, Apte SH, Kienzle N, Kelso A. Quantitative assessment of the functional plasticity of memory CD8(+) T cells. Eur J Immunol 2016; 46:863-73. [PMID: 26799367 DOI: 10.1002/eji.201545726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 11/29/2015] [Accepted: 01/15/2016] [Indexed: 01/28/2023]
Abstract
While the functional plasticity of memory CD4(+) T cells has been studied extensively, less is known about this property in memory CD8(+) T cells. Here, we report the direct measurement of plasticity by paired daughter analysis of effector and memory OT-I CD8(+) T cells primed in vivo with ovalbumin. Naïve, effector, and memory OT-I cells were isolated and activated in single-cell culture; then, after the first division, their daughter cells were transferred to new cultures with and without IL-4; expression of IFN-γ and IL-4 mRNAs was measured 5 days later in the resultant subclones. Approximately 40% of clonogenic memory CD8(+) T cells were bipotential in this assay, giving rise to an IL-4(-) subclone in the absence of IL-4 and an IL-4(+) subclone in the presence of IL-4. The frequency of bipotential cells was lower among memory cells than naïve cells but markedly higher than among 8-day effectors. Separation based on high or low expression of CD62L, CD122, CD127, or Ly6C did not identify a phenotypic marker of the bipotential cells. Functional plasticity in memory CD8(+) T-cell populations can therefore reflect modulation at the level of a single memory cell and its progeny.
Collapse
Affiliation(s)
- Adriana Baz
- Cooperative Research Centre for Vaccine Technology, QIMR Berghofer Medical Research Institute, QLD, Australia
| | - Penny Groves
- Cooperative Research Centre for Vaccine Technology, QIMR Berghofer Medical Research Institute, QLD, Australia
| | - Kathy Buttigieg
- Cooperative Research Centre for Vaccine Technology, QIMR Berghofer Medical Research Institute, QLD, Australia
| | - Simon H Apte
- Cooperative Research Centre for Vaccine Technology, QIMR Berghofer Medical Research Institute, QLD, Australia
| | - Norbert Kienzle
- Cooperative Research Centre for Vaccine Technology, QIMR Berghofer Medical Research Institute, QLD, Australia
| | - Anne Kelso
- Cooperative Research Centre for Vaccine Technology, QIMR Berghofer Medical Research Institute, QLD, Australia
| |
Collapse
|
44
|
Johnson CB, Wrangle J, Mehrotra S, Li Z, Paulos CM, Cole DJ, Surh CD, Rubinstein MP. Harnessing the IL-7/IL-7Rα axis to improve tumor immunotherapy. Oncoimmunology 2016; 5:e1122865. [PMID: 27467935 DOI: 10.1080/2162402x.2015.1122865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 01/08/2023] Open
Abstract
IL-7 and IL-15 are critical for supporting T cells transferred into a lymphopenic environment. As activated CD8(+) T cells downregulate IL-7Rα, it is thought IL-15 is more important. However, we find that CD8(+) T cells activated with IL-12 have elevated IL-7Rα and rely on IL-7 for persistence and antitumor immunity.
Collapse
Affiliation(s)
- C Bryce Johnson
- Department of Surgery, Medical University of South Carolina , Charleston, SC
| | - John Wrangle
- Department of Microbiology & Immunology, Medical University of South Carolina , Charleston, SC
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina , Charleston, SC
| | - Zihai Li
- Department of Microbiology & Immunology, Medical University of South Carolina , Charleston, SC
| | - Chrystal M Paulos
- Department of Surgery, Medical University of South Carolina, Charleston, SC; Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
| | - David J Cole
- Department of Surgery, Medical University of South Carolina , Charleston, SC
| | - Charles D Surh
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, South Korea; Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea; Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Mark P Rubinstein
- Department of Surgery, Medical University of South Carolina, Charleston, SC; Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
45
|
Unbalanced recovery of regulatory and effector T cells after allogeneic stem cell transplantation contributes to chronic GVHD. Blood 2015; 127:646-57. [PMID: 26670634 DOI: 10.1182/blood-2015-10-672345] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 12/05/2015] [Indexed: 01/01/2023] Open
Abstract
The development and maintenance of immune tolerance after allogeneic hematopoietic stem cell transplantation (HSCT) requires the balanced reconstitution of donor-derived CD4 regulatory T cells (CD4Tregs) as well as effector CD4 (conventional CD4 T cells [CD4Tcons]) and CD8 T cells. To characterize the complex mechanisms that lead to unbalanced recovery of these distinct T-cell populations, we studied 107 adult patients who received T-replete stem cell grafts after reduced-intensity conditioning. Immune reconstitution of CD4Treg, CD4Tcon, and CD8 T cells was monitored for a 2-year period. CD3 T-cell counts gradually recovered to normal levels during this period but CD8 T cells recovered more rapidly than either CD4Tregs or CD4Tcons. Reconstituting CD4Tregs and CD4Tcons were predominantly central memory (CM) and effector memory (EM) cells and CD8 T cells were predominantly terminal EM cells. Thymic generation of naive CD4Tcon and CD8 T cells was maintained but thymic production of CD4Tregs was markedly decreased with little recovery during the 2-year study. T-cell proliferation was skewed in favor of CM and EM CD4Tcon and CD8 T cells, especially 6 to 12 months after HSCT. Intracellular expression of BCL2 was increased in CD4Tcon and CD8 T cells in the first 3 to 6 months after HSCT. Early recovery of naive and CM fractions within each T-cell population 3 months after transplant was also strongly correlated with the subsequent development of chronic graft-versus-host disease (GVHD). These dynamic imbalances favor the production, expansion, and persistence of effector T cells over CD4Tregs and were associated with the development of chronic GVHD.
Collapse
|
46
|
Trella E, Raafat N, Mengus C, Traunecker E, Governa V, Heidtmann S, Heberer M, Oertli D, Spagnoli GC, Zajac P. CD40 ligand-expressing recombinant vaccinia virus promotes the generation of CD8(+) central memory T cells. Eur J Immunol 2015; 46:420-31. [PMID: 26561341 DOI: 10.1002/eji.201545554] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 10/05/2015] [Accepted: 11/05/2015] [Indexed: 01/17/2023]
Abstract
Central memory CD8(+) T cells (TCM ) play key roles in the protective immunity against infectious agents, cancer immunotherapy, and adoptive treatments of malignant and viral diseases. CD8(+) TCM cells are characterized by specific phenotypes, homing, and proliferative capacities. However, CD8(+) TCM -cell generation is challenging, and usually requires CD4(+) CD40L(+) T-cell "help" during the priming of naïve CD8(+) T cells. We have generated a replication incompetent CD40 ligand-expressing recombinant vaccinia virus (rVV40L) to promote the differentiation of human naïve CD8(+) T cells into TCM specific for viral and tumor-associated antigens. Soluble CD40 ligand recombinant protein (sCD40L), and vaccinia virus wild-type (VV WT), alone or in combination, were used as controls. Here, we show that, in the absence of CD4(+) T cells, a single "in vitro" stimulation of naïve CD8(+) T cells by rVV40L-infected nonprofessional CD14(+) antigen presenting cells promotes the rapid generation of viral or tumor associated antigen-specific CD8(+) T cells displaying TCM phenotypic and functional properties. These observations demonstrate the high ability of rVV40L to fine tune CD8(+) mediated immune responses, and strongly support the use of similar reagents for clinical immunization and adoptive immunotherapy purposes.
Collapse
Affiliation(s)
- Emanuele Trella
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Nermin Raafat
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland.,Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Chantal Mengus
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Valeria Governa
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Michael Heberer
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Daniel Oertli
- Department of Surgery, University Hospital, Basel, Switzerland
| | - Giulio C Spagnoli
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Paul Zajac
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
47
|
Zhang X, Starnbach MN. An Excess of the Proinflammatory Cytokines IFN-γ and IL-12 Impairs the Development of the Memory CD8+ T Cell Response to Chlamydia trachomatis. THE JOURNAL OF IMMUNOLOGY 2015; 195:1665-75. [PMID: 26179901 DOI: 10.4049/jimmunol.1500457] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 06/12/2015] [Indexed: 12/13/2022]
Abstract
The obligate intracellular bacterium Chlamydia trachomatis is the most common cause of bacterial sexually transmitted disease in the United States and the leading cause of preventable blindness worldwide. Transfer of cultured Chlamydia-specific CD8(+) T cells or vaccination with recombinant virus expressing an MHC I-restricted Chlamydia Ag confers protection, yet surprisingly a protective CD8(+) T cell response is not stimulated following natural infection. In this study, we demonstrate that the presence of excess IL-12 and IFN-γ contributes to poor memory CD8(+) T cell development during C. trachomatis infection of mice. IL-12 is required for CD8(+) T cell expansion but drives effector CD8(+) T cells into a short-lived fate, whereas IFN-γ signaling impairs the development of effector memory cells. We show that transient blockade of IL-12 and IFN-γ during priming promotes the development of memory precursor effector CD8(+) T cells and increases the number of memory T cells that participate in the recall protection against subsequent infection. Overall, this study identifies key factors shaping memory development of Chlamydia-specific CD8(+) T cells that will inform future vaccine development against this and other pathogens.
Collapse
Affiliation(s)
- Xuqing Zhang
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Michael N Starnbach
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
48
|
Beloki L, Ciaurriz M, Mansilla C, Zabalza A, Perez-Valderrama E, Samuel ER, Lowdell MW, Ramirez N, Olavarria E. Assessment of the effector function of CMV-specific CTLs isolated using MHC-multimers from granulocyte-colony stimulating factor mobilized peripheral blood. J Transl Med 2015; 13:165. [PMID: 25990023 PMCID: PMC4458005 DOI: 10.1186/s12967-015-0515-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 05/04/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Adoptive transfer of CMV-specific T cells has shown promising results in preventing pathological effects caused by opportunistic CMV infection in immunocompromised patients following allogeneic hematopoietic stem cell transplantation. The majority of studies have used steady-state leukapheresis for CMV-reactive product manufacture, a collection obtained prior to or months after G-CSF mobilization, but the procurement of this additional sample is often not available in the unrelated donor setting. If the cellular product for adoptive immunotherapy could be generated from the same G-CSF mobilized collection, the problems associated with the additional harvest could be overcome. Despite the tolerogenic effects associated with G-CSF mobilization, recent studies described that CMV-primed T cells generated from mobilized donors remain functional. METHODS MHC-multimers are potent tools that allow the rapid production of antigen-specific CTLs. Therefore, in the present study we have assessed the feasibility and efficacy of CMV-specific CTL manufacture from G-CSF mobilized apheresis using MHC-multimers. RESULTS CMV-specific CTLs can be efficiently isolated from G-CSF mobilized samples with Streptamers and are able to express activation markers and produce cytokines in response to antigenic stimulation. However, this anti-viral functionality is moderately reduced when compared to non-mobilized products. CONCLUSIONS The translation of Streptamer technology for the isolation of anti-viral CTLs from G-CSF mobilized PBMCs into clinical practice would widen the number of patients that could benefit from this therapeutic strategy, although our results need to be taken into consideration before the infusion of antigen-specific T cells obtained from G-CSF mobilized samples.
Collapse
Affiliation(s)
- Lorea Beloki
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Miriam Ciaurriz
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Cristina Mansilla
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Amaya Zabalza
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Estela Perez-Valderrama
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Edward R Samuel
- Department of Haematology, University College London Medical School, University College London, London, UK.
| | - Mark W Lowdell
- Department of Haematology, University College London Medical School, University College London, London, UK.
| | - Natalia Ramirez
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Eduardo Olavarria
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain. .,Department of Haematology, Complejo Hospitalario de Navarra, Navarra Health Service, IDISNA (Navarra's Health Research Institute), Pamplona, Spain.
| |
Collapse
|
49
|
Arsenović-Ranin N, Kosec D, Nacka-Aleksić M, Pilipović I, Stojić-Vukanić Z, Djikić J, Bufan B, Leposavić G. Ovarian hormone level alterations during rat post-reproductive life-span influence CD8 + T-cell homeostasis. Exp Biol Med (Maywood) 2015; 240:1319-32. [PMID: 25716018 DOI: 10.1177/1535370215570817] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 12/21/2014] [Indexed: 01/01/2023] Open
Abstract
The study examined the putative role of ovarian hormones in shaping of rat peripheral T-cell compartment during post-reproductive period. In 20-month-old rats ovariectomized (Ox) at the very end of reproductive period, thymic output, cellularity and composition of major TCRαβ + peripheral blood lymphocyte and splenocyte subsets were analyzed. Ovariectomy led to the enlargement of CD8 + peripheral blood lymphocyte and splenocyte subpopulations. This reflected: (i) a more efficient thymic generation of CD8 + cells as indicated by increased number of CD4+CD8 + double positive and the most mature CD4-CD8+TCRαβ(high) thymocytes and CD8 + recent thymic emigrants (RTEs) in peripheral blood, but not in the spleen of Ox rats, and (ii) the expansion of CD8 + memory/activated peripheral blood lymphocytes and splenocytes. The latter was consistent with a greater frequency of proliferating cells among freshly isolated memory/activated CD8 + peripheral blood lymphocytes and splenocytes and increased proliferative response of CD8 + splenocytes to stimulation with plate-bound anti-CD3 antibody. The former could be related to the rise in splenic IL-7 and IL-15 mRNA expression. Although ovariectomy affected the overall number of CD4 + T cells in none of the examined compartments, it increased CD4+FoxP3 + peripheral blood lymphocyte and splenocyte counts by enhancing their generation in periphery. Collectively, the results suggest that ovariectomy-induced long-lasting disturbances in ovarian hormone levels (mirrored in diminished progesterone serum level in 20-month-old rats) affects both thymic CD8 + cell generation and peripheral homeostasis and leads to the expansion of CD4+FoxP3 + cells in the periphery, thereby enhancing autoreactive cell control on account of immune system efficacy to combat infections and tumors.
Collapse
Affiliation(s)
- Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 11221 Belgrade, Serbia
| | - Mirjana Nacka-Aleksić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 11221 Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia
| | - Jasmina Djikić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia
| | - Biljana Bufan
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia
| | - Gordana Leposavić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia
| |
Collapse
|
50
|
Lee PH, Yamada T, Park CS, Shen Y, Puppi M, Lacorazza HD. G0S2 modulates homeostatic proliferation of naïve CD8⁺ T cells and inhibits oxidative phosphorylation in mitochondria. Immunol Cell Biol 2015; 93:605-15. [PMID: 25666096 PMCID: PMC4531109 DOI: 10.1038/icb.2015.9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 12/24/2014] [Accepted: 01/15/2015] [Indexed: 11/16/2022]
Abstract
Since its discovery, diverse functions have been attributed to the G0/G1 switch gene 2 (G0S2), from lipid metabolism to control of cell proliferation. Our group showed for the first time that G0S2 promotes quiescence in hematopoietic stem cells by interacting with and retaining nucleolin around the nucleus. Herein, we report the role of G0S2 in the differentiation and function of CD8+ T cells examined in mice with an embryonic deletion of the G0s2 gene. G0S2 expression in naïve CD8+ T cells decreased immediately after T-cell receptor activation downstream of the MAPK, calcium/calmodulin, PI3K, and mTOR pathways. Surprisingly, G0S2-null naïve CD8+ T cells displayed increased basal and spare respiratory capacity that was not associated with increased mitochondrial biogenesis but with increased phosphorylation of AMPKα. Naïve CD8+ T cells showed increased proliferation in response to in vitro activation and in vivo lymphopenia; however, naïve CD8+ T cells expressing the OT-1 transgene exhibited normal differentiation of naïve cells to effector and memory CD8+ T cells upon infection with Listeria monocytogenes in a wild type or a G0s2-null environment, with increased circulating levels of free fatty acids. Collectively, our results suggest that G0S2 inhibits energy production by oxidative phosphorylation to fine-tune proliferation in homeostatic conditions.
Collapse
Affiliation(s)
- Ping-Hsien Lee
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Takeshi Yamada
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Chun Shik Park
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Ye Shen
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Monica Puppi
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - H Daniel Lacorazza
- 1] Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA [2] Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|