1
|
Pierce GF, Fong S, Long BR, Kaczmarek R. Deciphering conundrums of adeno-associated virus liver-directed gene therapy: focus on hemophilia. J Thromb Haemost 2024; 22:1263-1289. [PMID: 38103734 DOI: 10.1016/j.jtha.2023.12.005] [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: 09/15/2023] [Revised: 11/07/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
Adeno-associated virus gene therapy has been the subject of intensive investigation for monogenic disease gene addition therapy for more than 25 years, yet few therapies have been approved by regulatory agencies. Most have not progressed beyond phase 1/2 due to toxicity, lack of efficacy, or both. The liver is a natural target for adeno-associated virus since most serotypes have a high degree of tropism for hepatocytes due to cell surface receptors for the virus and the unique liver sinusoidal geometry facilitating high volumes of blood contact with hepatocyte cell surfaces. Recessive monogenic diseases such as hemophilia represent promising targets since the defective proteins are often synthesized in the liver and secreted into the circulation, making them easy to measure, and many do not require precise regulation. Yet, despite initiation of many disease-specific clinical trials, therapeutic windows are often nonexistent, resulting in excess toxicity and insufficient efficacy. Iterative progress built on these attempts is best illustrated by hemophilia, with the first regulatory approvals for factor IX and factor VIII gene therapies eventually achieved 25 years after the first gene therapy studies in humans. Although successful gene transfer may result in the production of sufficient transgenic protein to modify the disease, many emerging questions on durability, predictability, reliability, and variability of response have not been answered. The underlying biology accounting for these heterogeneous responses and the interplay between host and virus is the subject of intense investigation and the subject of this review.
Collapse
Affiliation(s)
- Glenn F Pierce
- World Federation of Hemophilia, Montreal, Quebec, Canada.
| | - Sylvia Fong
- BioMarin Pharmaceutical Inc, Research and Early Development, Novato, California, USA
| | - Brian R Long
- BioMarin Pharmaceutical Inc, Research and Early Development, Novato, California, USA
| | - Radoslaw Kaczmarek
- Department of Pediatrics, Indiana University School of Medicine, Wells Center for Pediatric Research, Indiana, USA; Laboratory of Glycobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| |
Collapse
|
2
|
Vaccination with Live or Heat-Killed Aspergillus fumigatus Δ sglA Conidia Fully Protects Immunocompromised Mice from Invasive Aspergillosis. mBio 2022; 13:e0232822. [PMID: 36066100 PMCID: PMC9600187 DOI: 10.1128/mbio.02328-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aspergillus fumigatus causes invasive aspergillosis (IA) in immunocompromised patients, resulting in high mortality rates. Currently, no vaccine formulations to promote immune protection in at-risk individuals have been developed. In this work, we deleted the sterylglucosidase-encoding gene, sglA, in Aspergillus fumigatus and investigated its role in fungal virulence and host vaccine protection. The ΔsglA mutant accumulated sterylglucosides (SGs), newly studied immunomodulatory glycolipids, and exhibited reduced hyphal growth and altered compositions of cell wall polysaccharides. Interestingly, the ΔsglA mutant was avirulent in two murine models of IA and was fully eliminated from the lungs. Both corticosteroid-induced immunosuppressed and cyclophosphamide-induced leukopenic mice vaccinated with live or heat-killed ΔsglA conidia were fully protected against a lethal wild-type A. fumigatus challenge. These results highlight the potential of SG-accumulating strains as safe and promising vaccine formulations against invasive fungal infections. IMPORTANCE Infections by Aspergillus fumigatus occur by the inhalation of environmental fungal spores called conidia. We found that live mutant conidia accumulating glycolipids named sterylglucosides are not able to cause disease when injected into the lung. Interestingly, these animals are now protected against a secondary challenge with live wild-type conidia. Remarkably, protection against a secondary challenge persists even with vaccination with heat-killed mutant conidia. These results will significantly advance the field of the research and development of a safe fungal vaccine for protection against the environmental fungus A. fumigatus.
Collapse
|
3
|
Normile TG, Del Poeta M. Three Models of Vaccination Strategies Against Cryptococcosis in Immunocompromised Hosts Using Heat-Killed Cryptococcus neoformans Δ sgl1. Front Immunol 2022; 13:868523. [PMID: 35615354 PMCID: PMC9124966 DOI: 10.3389/fimmu.2022.868523] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/11/2022] [Indexed: 12/16/2022] Open
Abstract
Vaccines are one of the greatest medical accomplishments to date, yet no fungal vaccines are currently available in humans mainly because opportunistic mycoses generally occur during immunodeficiencies necessary for vaccine protection. In previous studies, a live, attenuated Cryptococcus neoformans Δsgl1 mutant accumulating sterylglucosides was found to be avirulent and protected mice from a subsequent lethal infection even in absence of CD4+ T cells, a condition most associated with cryptococcosis (e.g., HIV). Here, we tested three strategies of vaccination against cryptococcosis. First, in our preventative model, protection was achieved even after a 3-fold increase of the vaccination window. Second, because live C. neoformans Δsgl1-vaccinated mice challenged more than once with WT strain had a significant decrease in lung fungal burden, we tested C. neoformans Δsgl1 as an immunotherapeutic. We found that therapeutic administrations of HK C. neoformans Δsgl1 post WT challenge significantly improves the lung fungal burden. Similarly, therapeutic administration of HK C. neoformans Δsgl1 post WT challenge resulted in 100% or 70% survival depending on the time of vaccine administration, suggesting that HK Δsgl1 is a robust immunotherapeutic option. Third, we investigated a novel model of vaccination in preventing reactivation from lung granuloma using C. neoformans Δgcs1. Remarkably, we show that administration of HK Δsgl1 prevents mice from reactivating Δgcs1 upon inducing immunosuppression with corticosteroids or by depleting CD4+ T cells. Our results suggest that HK Δsgl1 represents a clinically relevant, efficacious vaccine that confers robust host protection in three models of vaccination against cryptococcosis even during CD4-deficiency.
Collapse
Affiliation(s)
- Tyler G. Normile
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, United States
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, United States,Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, NY, United States,Veterans Administration Medical Center, Northport, NY, United States,*Correspondence: Maurizio Del Poeta,
| |
Collapse
|
4
|
Singh R, Anand A, Rawat AK, Saini S, Mahapatra B, Singh NK, Mishra AK, Singh S, Singh N, Kishore D, Kumar V, Das P, Singh RK. CD300a Receptor Blocking Enhances Early Clearance of Leishmania donovani From Its Mammalian Host Through Modulation of Effector Functions of Phagocytic and Antigen Experienced T Cells. Front Immunol 2022; 12:793611. [PMID: 35116028 PMCID: PMC8803664 DOI: 10.3389/fimmu.2021.793611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/27/2021] [Indexed: 12/17/2022] Open
Abstract
The parasites of the genus Leishmania survive and proliferate in the host phagocytic cells by taking control over their microbicidal functions. The parasite also promotes differentiation of antigen-specific anti-inflammatory cytokines producing effector T cells, which eventually results in disease pathogenesis. The mechanisms that parasites employ to dominate host adaptive immunity are largely unknown. For the first time, we report that L. donovani, which causes visceral leishmaniasis in the Indian subcontinent, upregulates the expression of an immune inhibitory receptor i.e., CD300a on antigen presenting and phagocytic cells to dampen their effector functions. The blocking of CD300a signals in leishmania antigens activated macrophages and dendritic cells enhanced the production of nitric oxide, pro-inflammatory cytokines along with MHCI/II genes expression, and reduced parasitic uptake. Further, the abrogation of CD300a signals in Leishmania infected mice benefited antigen-experienced, i.e., CD4+CD44+ and CD8+CD44+ T cells to acquire more pro-inflammatory cytokines producing phenotypes and helped in the early clearance of parasites from their visceral organs. The CD300a receptor blocking also enhanced the conversion of CD4+ T effectors cells to their memory phenotypes i.e., CCR7high CD62Lhigh up to 1.6 and 1.9 fold after 14 and 21 days post-infection, respectively. These findings implicate that CD300a is an important determinant of host phagocytic cells functions and T cells differentiation against Leishmania antigens.
Collapse
Affiliation(s)
- Rajan Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Anshul Anand
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Arun K. Rawat
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Shashi Saini
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Baishakhi Mahapatra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Naveen K. Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Alok K. Mishra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Samer Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Science, Banaras Hindu University, Varanasi, India
| | - Nisha Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Dhiraj Kishore
- Department of Medicine, Institute of Medical Science, Banaras Hindu University, Varanasi, India
| | - Vinod Kumar
- Department of Molecular Biology, Rajendra Memorial Research Institute, Patna, India
| | - Pradeep Das
- Department of Molecular Biology, Rajendra Memorial Research Institute, Patna, India
| | - Rakesh K. Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
- *Correspondence: Rakesh K. Singh,
| |
Collapse
|
5
|
Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, Sautès-Fridman C, Ma Y, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology 2021; 1:1323-1343. [PMID: 23243596 PMCID: PMC3518505 DOI: 10.4161/onci.22009] [Citation(s) in RCA: 187] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Solid tumors are constituted of a variety of cellular components, including bona fide malignant cells as well as endothelial, structural and immune cells. On one hand, the tumor stroma exerts major pro-tumorigenic and immunosuppressive functions, reflecting the capacity of cancer cells to shape the microenvironment to satisfy their own metabolic and immunological needs. On the other hand, there is a component of tumor-infiltrating leucocytes (TILs) that has been specifically recruited in the attempt to control tumor growth. Along with the recognition of the critical role played by the immune system in oncogenesis, tumor progression and response to therapy, increasing attention has been attracted by the potential prognostic and/or predictive role of the immune infiltrate in this setting. Data from large clinical studies demonstrate indeed that a robust infiltration of neoplastic lesions by specific immune cell populations, including (but not limited to) CD8+ cytotoxic T lymphocytes, Th1 and Th17 CD4+ T cells, natural killer cells, dendritic cells, and M1 macrophages constitutes an independent prognostic indicator in several types of cancer. Conversely, high levels of intratumoral CD4+CD25+FOXP3+ regulatory T cells, Th2 CD4+ T cells, myeloid-derived suppressor cells, M2 macrophages and neutrophils have frequently been associated with dismal prognosis. So far, only a few studies have addressed the true predictive potential of TILs in cancer patients, generally comforting the notion that—at least in some clinical settings—the immune infiltrate can reliably predict if a specific patient will respond to therapy or not. In this Trial Watch, we will summarize the results of clinical trials that have evaluated/are evaluating the prognostic and predictive value of the immune infiltrate in the context of solid malignancies.
Collapse
Affiliation(s)
- Laura Senovilla
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Orsay, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Behr FM, Beumer-Chuwonpad A, Kragten NAM, Wesselink TH, Stark R, van Gisbergen KPJM. Circulating memory CD8 + T cells are limited in forming CD103 + tissue-resident memory T cells at mucosal sites after reinfection. Eur J Immunol 2020; 51:151-166. [PMID: 32762051 DOI: 10.1002/eji.202048737] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/09/2020] [Accepted: 08/05/2020] [Indexed: 12/16/2022]
Abstract
Tissue-resident memory CD8+ T cells (TRM ) localize to barrier tissues and mediate local protection against reinvading pathogens. Circulating central memory (TCM ) and effector memory CD8+ T cells (TEM ) also contribute to tissue recall responses, but their potential to form mucosal TRM remains unclear. Here, we employed adoptive transfer and lymphocytic choriomeningitis virus reinfection models to specifically assess secondary responses of TCM and TEM at mucosal sites. Donor TCM and TEM exhibited robust systemic recall responses, but only limited accumulation in the small intestine, consistent with reduced expression of tissue-homing and -retention molecules. Murine and human circulating memory T cells also exhibited limited CD103 upregulation following TGF-β stimulation. Upon pathogen clearance, TCM and TEM readily gave rise to secondary TEM . TCM also formed secondary central memory in lymphoid tissues and TRM in internal tissues, for example, the liver. Both TCM and TEM failed to substantially contribute to resident mucosal memory in the small intestine, while activated intestinal TRM , but not liver TRM , efficiently reformed CD103+ TRM . Our findings demonstrate that circulating TCM and TEM are limited in generating mucosal TRM upon reinfection. This may pose important implications on cell therapy and vaccination strategies employing memory CD8+ T cells for protection at mucosal sites.
Collapse
Affiliation(s)
- Felix M Behr
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ammarina Beumer-Chuwonpad
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Natasja A M Kragten
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Thomas H Wesselink
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Regina Stark
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,BIH Center for Regenerative Therapies, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Tissue-resident memory CD8+ T cells shape local and systemic secondary T cell responses. Nat Immunol 2020; 21:1070-1081. [DOI: 10.1038/s41590-020-0723-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 06/01/2020] [Indexed: 01/01/2023]
|
8
|
Renkema KR, Huggins MA, Borges da Silva H, Knutson TP, Henzler CM, Hamilton SE. KLRG1 + Memory CD8 T Cells Combine Properties of Short-Lived Effectors and Long-Lived Memory. THE JOURNAL OF IMMUNOLOGY 2020; 205:1059-1069. [PMID: 32611727 DOI: 10.4049/jimmunol.1901512] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
Abstract
CD8 effector T cells with a CD127hi KLRG1- phenotype are considered precursors to the long-lived memory pool, whereas KLRG1+CD127low cells are viewed as short-lived effectors. Nevertheless, we and others have shown that a KLRG1+CD127low population persists into the memory phase and that these T cells (termed long-lived effector cells [LLEC]) display robust protective function during acute rechallenge with bacteria or viruses. Whether these T cells represent a true memory population or are instead a remnant effector cell population that failed to undergo initial contraction has remained unclear. In this study, we show that LLEC from mice express a distinct phenotypic and transcriptional signature that shares characteristics of both early effectors and long-lived memory cells. We also find that in contrast to KLRG1+ effector cells, LLEC undergo homeostatic proliferation and are not critically dependent on IL-15 for their maintenance. Furthermore, we find that LLEC are predominantly derived from KLRG1+ effector cells when isolated at day 12 of the response. Our work challenges the concept that the KLRG1+CD127low population is dominated by short-lived cells and shows that KLRG1 downregulation is not a prerequisite to become a long-lived protective memory T cell.
Collapse
Affiliation(s)
- Kristin R Renkema
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455; and
| | - Matthew A Huggins
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455; and
| | - Henrique Borges da Silva
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455; and
| | - Todd P Knutson
- Supercomputing Institute for Advanced Computational Research, University of Minnesota, Minneapolis, MN 55455
| | - Christy M Henzler
- Supercomputing Institute for Advanced Computational Research, University of Minnesota, Minneapolis, MN 55455
| | - Sara E Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455; .,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455; and
| |
Collapse
|
9
|
Bastos MF, Matias MDST, Alonso AC, Silva LCR, de Araújo AL, Silva PR, Benard G, Bocalini DS, Steven Baker J, Leme LEG. Moderate levels of physical fitness maintain telomere length in non-senescent T CD8+ cells of aged men. Clinics (Sao Paulo) 2020; 75:e1628. [PMID: 33174947 PMCID: PMC7605280 DOI: 10.6061/clinics/2020/e1628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 09/04/2020] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVES Immunosenescence is an age-associated change characterized by a decreased immune response. Although physical activity has been described as fundamental for maintaining the quality of life, few studies have evaluated the effects of different levels of exercise on telomere length in aged populations. The present study aimed to analyze the effects of different levels of physical activity, classified by the Maximal oxygen consumption (VO2 max) values, on the telomere length of memory Cluster of differentiation (CD) CD4+(CD45ROneg and CD45RO+), effector CD8+CD28neg, and CD8+CD28+ T cells in aged individuals. METHODS Fifty-three healthy elderly men (aged 65-85 years) were included in this study. Their fitness level was classified according to the American College of Sports Medicine (ACSM) for VO2 max (mL/kg/min). Blood samples were obtained from all participants to analyze the percentage of CD3, CD4, CD8, CD28+, naïve, and subpopulations of memory T cells by using flow cytometry. Furthermore, using the Flow-FISH methodology, the CD4+CD45RO+, CD4+CD45ROneg, CD8+CD28+, and CD8+CD28negT cell telomere lengths were measured. RESULTS There was a greater proportion of effector memory T CD4+ cells and longer telomeres in CD8+CD28+ T cells in the moderate physical fitness group than in the other groups. There was a higher proportion of terminally differentiated memory effector T cells in the low physical fitness group. CONCLUSION A moderate physical activity may positively influence the telomere shortening of CD28+CD8+T cells. However, additional studies are necessary to evaluate the importance of this finding with regard to immune function responses in older men.
Collapse
Affiliation(s)
- Marta Ferreira Bastos
- Programa de Pos graduacao Stricto sensu em Ciencias do Envelhecimento, Departamento de Pos graduacao e Pesquisa, Universidade Sao Judas Tadeu, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| | - Manuella de Sousa Toledo Matias
- Grupo Ortogeriatrico, Instituto de Ortopedia e Traumotologia, Escola de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, BR
| | - Angélica Castilho Alonso
- Programa de Pos graduacao Stricto sensu em Ciencias do Envelhecimento, Departamento de Pos graduacao e Pesquisa, Universidade Sao Judas Tadeu, Sao Paulo, SP, BR
- Grupo Ortogeriatrico, Instituto de Ortopedia e Traumotologia, Escola de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, BR
| | - Léia Cristina Rodrigues Silva
- Laboratorio de Dermatologia e Imunodeficiencias, Divisao de Dermatologia, Hospital das Clinicas (HCFMUSP), Escola de Medicina, Sao Paulo, SP, BR
| | - Adriana Ladeira de Araújo
- Laboratorio de Dermatologia e Imunodeficiencias, Divisao de Dermatologia, Hospital das Clinicas (HCFMUSP), Escola de Medicina, Sao Paulo, SP, BR
| | - Paulo Roberto Silva
- Grupo Ortogeriatrico, Instituto de Ortopedia e Traumotologia, Escola de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, BR
| | - Gil Benard
- Laboratorio de Dermatologia e Imunodeficiencias, Divisao de Dermatologia, Hospital das Clinicas (HCFMUSP), Escola de Medicina, Sao Paulo, SP, BR
| | - Danilo Sales Bocalini
- Laboratorio experimental de Fisiologia e Bioquimica, Centro de Esporte e Educacao Fisica da Universidade Federal do Espirito Santo, Vitoria, ES, BR
| | - Julien Steven Baker
- Department of Sport, Physical Education and Health, Hong Kong Baptist University, Hong Kong
| | - Luiz Eugênio Garcez Leme
- Grupo Ortogeriatrico, Instituto de Ortopedia e Traumotologia, Escola de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, BR
| |
Collapse
|
10
|
The ALFA-tag is a highly versatile tool for nanobody-based bioscience applications. Nat Commun 2019; 10:4403. [PMID: 31562305 PMCID: PMC6764986 DOI: 10.1038/s41467-019-12301-7] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 08/28/2019] [Indexed: 11/08/2022] Open
Abstract
Specialized epitope tags are widely used for detecting, manipulating or purifying proteins, but often their versatility is limited. Here, we introduce the ALFA-tag, a rationally designed epitope tag that serves a remarkably broad spectrum of applications in life sciences while outperforming established tags like the HA-, FLAG®- or myc-tag. The ALFA-tag forms a small and stable α-helix that is functional irrespective of its position on the target protein in prokaryotic and eukaryotic hosts. We characterize a nanobody (NbALFA) binding ALFA-tagged proteins from native or fixed specimen with low picomolar affinity. It is ideally suited for super-resolution microscopy, immunoprecipitations and Western blotting, and also allows in vivo detection of proteins. We show the crystal structure of the complex that enabled us to design a nanobody mutant (NbALFAPE) that permits efficient one-step purifications of native ALFA-tagged proteins, complexes and even entire living cells using peptide elution under physiological conditions. Epitope tags are widely used in various applications, but often lack versatility. Here, the authors introduce a small, alpha helical tag, which is recognized by a high affinity nanobody and can be used in a range of different applications, from protein purification to super-resolution imaging and in vivo detection of proteins.
Collapse
|
11
|
Zundler S, Becker E, Schulze LL, Neurath MF. Immune cell trafficking and retention in inflammatory bowel disease: mechanistic insights and therapeutic advances. Gut 2019; 68:1688-1700. [PMID: 31127023 DOI: 10.1136/gutjnl-2018-317977] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022]
Abstract
Intestinal immune cell trafficking has been identified as a central event in the pathogenesis of inflammatory bowel diseases (IBD). Intensive research on different aspects of the immune mechanisms controlling and controlled by T cell trafficking and retention has led to the approval of the anti-α4β7 antibody vedolizumab, the ongoing development of a number of further anti-trafficking agents (ATAs) such as the anti-β7 antibody etrolizumab or the anti-MAdCAM-1 antibody ontamalimab and the identification of potential future targets like G-protein coupled receptor 15. However, several aspects of the biology of immune cell trafficking and regarding the mechanism of action of ATAs are still unclear, for example, which impact these compounds have on the trafficking of non-lymphocyte populations like monocytes and how precisely these therapies differ with regard to their effect on immune cell subpopulations. This review will summarise recent advances of basic science in the field of intestinal immune cell trafficking and discuss these findings with regard to different pharmacological approaches from a translational perspective.
Collapse
Affiliation(s)
- Sebastian Zundler
- Department of Medicine 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Emily Becker
- Department of Medicine 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Lisa Lou Schulze
- Department of Medicine 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| |
Collapse
|
12
|
Vonderheid EC, Kantor GR, Telang GH, Bujanouskas P, Kadin ME. A histo-immunopathologic and prognostic study of erythrodermic cutaneous T-cell lymphoma. J Cutan Pathol 2019; 46:913-924. [PMID: 31403211 DOI: 10.1111/cup.13564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/23/2019] [Accepted: 08/05/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND Sézary syndrome (SS) and erythrodermic mycosis fungoides (E-MF) represent two expressions of erythrodermic cutaneous T-cell lymphoma (E-CTCL). METHODS Histopathologic features were compared on skin specimens from 41 patients with SS and 70 patients with E-MF. Immunopathologic findings were compared on 42 SS and 79 E-MF specimens. RESULTS Specimens of SS usually showed band-like dermal infiltrates with intermediate-sized lymphoid cells and few plasma cells; on the other hand E-MF more often had a perivascular infiltrative pattern, predominance of small/mixed lymphoid cells and eosinophils. SS also had lower numbers of CD8+ cells and higher numbers of CD62L+ cells compared to E-MF. For E-MF patients, the presence of large Pautrier collections, infiltrates with intermediate-sized cells, increased number of mitotic figures and ≥50% CD62L+ cells in the dermal infiltrate correlated with a relatively poor disease-specific survival. However, only the presence of mitotic figures retained prognostic significance with clinical stage as a covariate. CONCLUSIONS Clinical stage provides the most important prognostic information for patients with E-CTCL. However, mitotic activity for E-MF and CD8+ cells <20% for SS have additional value. We hypothesize that observed differences in plasma cell and eosinophil numbers may reflect the influence of CD62L+ central memory T-cells in the microenvironment.
Collapse
Affiliation(s)
- Eric C Vonderheid
- Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutes, Baltimore, Maryland
| | - Gary R Kantor
- Dermatopathologist, Dermpath Diagnostics, Institute for Dermatopathology, Newtown Square, Pennsylvania
| | - Gladys H Telang
- Department of Dermatology, The Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | | | - Marshall E Kadin
- Department of Dermatology, Boston University and Roger Williams Medical Center, Providence, Rhode Island
| |
Collapse
|
13
|
Longitudinal Changes in Cd4 +, Cd8 + T Cell Phenotype and Activation Marker Expression Following Antiretroviral Therapy Initiation among Patients with Cryptococcal Meningitis. J Fungi (Basel) 2019; 5:jof5030063. [PMID: 31319498 PMCID: PMC6787641 DOI: 10.3390/jof5030063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/18/2019] [Accepted: 06/26/2019] [Indexed: 11/26/2022] Open
Abstract
Despite improvement in the prognosis of HIV/AIDS (human immunodeficiency virus/acquired immune deficiency syndrome) patients on antiretroviral therapy (ART), cryptococcal meningitis (CM) still causes 10–15% mortality among HIV-infected patients. The immunological impact of ART on the CD4+ and CD8+ T cell repertoire during cryptococcal co-infection is unclear. We determined longitudinal phenotypic changes in T cell subsets among patients with CM after they initiated ART. We hypothesized that ART alters the clonotypic phenotype and structural composition of CD4+ and CD8+ T cells during CM co-infection. For this substudy, peripheral blood mononuclear cells (PBMC) were isolated at four time points from CM patients following ART initiation during the parent study (ClinicalTrials.gov number, NCT01075152). Phenotypic characterization of CD4+ and CD8+ T cells was done using T cell surface marker monoclonal antibodies by flow cytometry. There was variation in the expression of immunophenotypic markers defining central memory (CD27+CD45R0+), effector memory (CD45R0+CD27–), immune activation (CD38+ and Human Leucocyte Antigen DR (HLA-DR+), and exhaustion (Programmed cell death protein one (PD-1) in the CD4+ T cell subset. In comparison to the CD4+ T cell population, the CD8+ central memory subset declined gradually with minimal increase in the effector memory subset. Both CD4+ and CD8+ T cell immune exhaustion and activation markers remained elevated over 12 weeks. The relative surge and decline in the expression of T cell surface markers outlines a variation in the differentiation of CD4+ T cells during ART treatment during CM co-infection.
Collapse
|
14
|
Checkpoint Blockade Immunotherapy Induces Dynamic Changes in PD-1 -CD8 + Tumor-Infiltrating T Cells. Immunity 2019; 50:181-194.e6. [PMID: 30635236 DOI: 10.1016/j.immuni.2018.11.014] [Citation(s) in RCA: 420] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 08/16/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
Abstract
An improved understanding of the anti-tumor CD8+ T cell response after checkpoint blockade would enable more informed and effective therapeutic strategies. Here we examined the dynamics of the effector response of CD8+ tumor-infiltrating lymphocytes (TILs) after checkpoint blockade therapy. Bulk and single-cell RNA profiles of CD8+ TILs after combined Tim-3+PD-1 blockade in preclinical models revealed significant changes in the transcriptional profile of PD-1- TILs. These cells could be divided into subsets bearing characterstics of naive-, effector-, and memory-precursor-like cells. Effector- and memory-precursor-like TILs contained tumor-antigen-specific cells, exhibited proliferative and effector capacity, and expanded in response to different checkpoint blockade therapies across different tumor models. The memory-precursor-like subset shared features with CD8+ T cells associated with response to checkpoint blockade in patients and was compromised in the absence of Tcf7. Expression of Tcf7/Tcf1 was requisite for the efficacy of diverse immunotherapies, highlighting the importance of this transcriptional regulator in the development of effective CD8+ T cell responses upon immunotherapy.
Collapse
|
15
|
Iwamura C, Nakayama T. Role of CD1d- and MR1-Restricted T Cells in Asthma. Front Immunol 2018; 9:1942. [PMID: 30210497 PMCID: PMC6121007 DOI: 10.3389/fimmu.2018.01942] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 08/06/2018] [Indexed: 12/12/2022] Open
Abstract
Innate T lymphocytes are a group of relatively recently identified T cells that are not involved in either innate or adaptive immunity. Unlike conventional T cells, most innate T lymphocytes express invariant T cell receptor to recognize exogenous non-peptide antigens presented by a family of non-polymorphic MHC class I-related molecules, such as CD1d and MHC-related molecule-1 (MR1). Invariant natural killer T (iNKT) cells and mucosal-associated invariant T (MAIT) cells quickly respond to the antigens bound to CD1d and MR1 molecules, respectively, and immediately exert effector functions by secreting various cytokines and granules. This review describes the detrimental and beneficial roles of iNKT cells in animal models of asthma and in human asthmatic patients and also addresses the mechanisms through which iNKT cells are activated by environmental or extracellular factors. We also discuss the potential for therapeutic interventions of asthma by specific antibodies against NKT cells. Furthermore, we summarize the recent reports on the role of MAIT cells in allergic diseases.
Collapse
Affiliation(s)
- Chiaki Iwamura
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
16
|
Miller D, Romero R, Unkel R, Xu Y, Vadillo-Ortega F, Hassan SS, Gomez-Lopez N. CD71+ erythroid cells from neonates born to women with preterm labor regulate cytokine and cellular responses. J Leukoc Biol 2018; 103:761-775. [PMID: 29389020 DOI: 10.1002/jlb.5a0717-291rrr] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022] Open
Abstract
Neonatal CD71+ erythroid cells are thought to have immunosuppressive functions. Recently, we demonstrated that CD71+ erythroid cells from neonates born to women who underwent spontaneous preterm labor (PTL) are reduced to levels similar to those of term neonates; yet, their functional properties are unknown. Herein, we investigated the functionality of CD71+ erythroid cells from neonates born to women who underwent spontaneous preterm or term labor. CD71+ erythroid cells from neonates born to women who underwent PTL displayed a similar mRNA profile to that of those from term neonates. The direct contact between preterm or term neonatal CD71+ erythroid cells and maternal mononuclear immune cells, but not soluble products from these cells, induced the release of proinflammatory cytokines and a reduction in the release of TGF-β. Moreover, PTL-derived neonatal CD71+ erythroid cells (1) modestly altered CD8+ T cell activation; (2) inhibited conventional CD4+ and CD8+ T-cell expansion; (3) suppressed the expansion of CD8+ regulatory T cells; (4) regulated cytokine responses mounted by myeloid cells in the presence of a microbial product; and (5) indirectly modulated T-cell cytokine responses. In conclusion, neonatal CD71+ erythroid cells regulate neonatal T-cell and myeloid responses and their direct contact with maternal mononuclear cells induces a proinflammatory response. These findings provide insight into the biology of neonatal CD71+ erythroid cells during the physiologic and pathologic processes of labor.
Collapse
Affiliation(s)
- Derek Miller
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Immunology, Microbiology and Biochemistry, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Ronald Unkel
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yi Xu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Felipe Vadillo-Ortega
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Facultad de Medicina, UNAM, Branch at Instituto Nacional de Medicina Genomica, Mexico City, Mexico
| | - Sonia S Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Immunology, Microbiology and Biochemistry, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
17
|
DeLong JH, Hall AO, Konradt C, Coppock GM, Park J, Harms Pritchard G, Hunter CA. Cytokine- and TCR-Mediated Regulation of T Cell Expression of Ly6C and Sca-1. THE JOURNAL OF IMMUNOLOGY 2018; 200:1761-1770. [PMID: 29358280 DOI: 10.4049/jimmunol.1701154] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/21/2017] [Indexed: 02/05/2023]
Abstract
Ly6C and Sca-1 (Ly6A/E) are Ly6 family GPI-anchored surface molecules that are differentially expressed by multiple immune populations. Ly6C expression has been used to distinguish short-lived effector CD4+ T cells from memory precursor effector cells, whereas Sca-1 has been used in the identification of CD8+ memory stem cells. This study examines the expression patterns of these molecules and establishes that, in vitro, IL-27, type I IFN, and IFN-γ are potent inducers of Ly6C and Sca-1 in naive mouse CD4+ and CD8+ T cells, whereas TGF-β limits their expression. The induction of Ly6C and Sca-1 by IL-27 and IFN-γ is dependent on STAT1, but not STAT3 or T-bet. In mouse splenocytes, at homeostasis, Ly6C and Sca-1 expression was not restricted to effector cells, but was also found at various levels on naive and memory populations. However, in response to infection with Toxoplasma gondii, pathogen-specific T cells expressed high levels of these molecules and in this context, endogenous IL-27 and IFN-γ were required for the expression of Ly6C but not Sca-1. Together, these findings highlight the TCR-dependent and cytokine-mediated signals that modulate T cell expression of Ly6C and Sca-1 in vitro and in vivo during infection.
Collapse
Affiliation(s)
- Jonathan H DeLong
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Aisling O'Hara Hall
- Immunology Discovery Research, Janssen Research and Development, LLC, Spring House, PA 19002
| | - Christoph Konradt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Gaia M Coppock
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104.,Department of Nephrology, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Jeongho Park
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | | | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| |
Collapse
|
18
|
Xie A, Li R, Jiang T, Yan H, Zhang H, Yang Y, Yang L, Yechoor V, Chan L, Chen W. Anti-TCRβ mAb in Combination With Neurogenin3 Gene Therapy Reverses Established Overt Type 1 Diabetes in Female NOD Mice. Endocrinology 2017; 158:3140-3151. [PMID: 28977608 PMCID: PMC5659705 DOI: 10.1210/en.2016-1947] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 07/28/2017] [Indexed: 12/13/2022]
Abstract
Insulin-producing β cells in patients with type 1 diabetes (T1D) are destroyed by T lymphocytes. We investigated whether targeting the T-cell receptor (TCR) with a monoclonal antibody (mAb) abrogates T-cell response against residual and newly formed islets in overtly diabetic nonobese diabetic (NOD) mice. NOD mice with blood glucose levels of 250 to 350 mg/dL or 350 to 450 mg/dL were considered as new-onset or established overt diabetes, respectively. These diabetic NOD mice were transiently treated with an anti-TCR β chain (TCRβ) mAb, H57-597, for 5 days. Two weeks later, some NOD mice with established overt diabetes further received hepatic gene therapy using the islet-lineage determining gene Neurogenin3 (Ngn3), in combination with the islet growth factor gene betacellulin (Btc). We found that anti-TCRβ mAb (50 µg/d) reversed >80% new-onset diabetes in NOD mice for >14 weeks by reducing the number of effector T cells in the pancreas. However, anti-TCRβ mAb therapy alone reversed only ∼20% established overt diabetes in these mice. Among those overtly diabetic NOD mice whose diabetes was resistant to anti-TCRβ mAb treatment, ∼60% no longer had diabetes when they also received Ngn3-Btc hepatic gene transfer 2 weeks after initial anti-TCRβ mAb treatment. This combination of Ngn3-Btc gene therapy and anti-TCRβ mAb treatment induced the sustained formation of periportal insulin-producing cells in the liver of overtly diabetic mice. Therefore, directly targeting TCRβ with a mAb potently reverses new-onset T1D in NOD mice and protects residual and newly formed gene therapy-induced hepatic neo-islets from T-cell‒mediated destruction in mice with established overt diabetes.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/therapeutic use
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Betacellulin/genetics
- Combined Modality Therapy
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/therapy
- Female
- Genetic Therapy/methods
- Immunotherapy/methods
- Insulin-Secreting Cells/cytology
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/physiology
- Islets of Langerhans/cytology
- Islets of Langerhans/immunology
- Liver/cytology
- Mice
- Mice, Inbred NOD
- Nerve Tissue Proteins/genetics
- Receptors, Antigen, T-Cell, alpha-beta/antagonists & inhibitors
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Aini Xie
- Center for Immunobiology and Transplantation Research, Department of Surgery, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas 77030
- Division of Diabetes, Endocrinology & Metabolism, Diabetes & Endocrinology Research Center, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rongying Li
- Division of Diabetes, Endocrinology & Metabolism, Diabetes & Endocrinology Research Center, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Tao Jiang
- Center for Immunobiology and Transplantation Research, Department of Surgery, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas 77030
| | - Hui Yan
- Center for Immunobiology and Transplantation Research, Department of Surgery, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas 77030
| | - Hedong Zhang
- Center for Immunobiology and Transplantation Research, Department of Surgery, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas 77030
| | - Yisheng Yang
- Division of Diabetes, Endocrinology & Metabolism, Diabetes & Endocrinology Research Center, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Lina Yang
- Division of Diabetes, Endocrinology & Metabolism, Diabetes & Endocrinology Research Center, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Vijay Yechoor
- Division of Diabetes, Endocrinology & Metabolism, Diabetes & Endocrinology Research Center, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Lawrence Chan
- Division of Diabetes, Endocrinology & Metabolism, Diabetes & Endocrinology Research Center, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Wenhao Chen
- Center for Immunobiology and Transplantation Research, Department of Surgery, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas 77030
- Division of Diabetes, Endocrinology & Metabolism, Diabetes & Endocrinology Research Center, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
- Department of Surgery, Weill Cornell Medical College of Cornell University, New York, New York 10065
| |
Collapse
|
19
|
White MJ, Beaver CM, Goodier MR, Bottomley C, Nielsen CM, Wolf ASFM, Boldrin L, Whitmore C, Morgan J, Pearce DJ, Riley EM. Calorie Restriction Attenuates Terminal Differentiation of Immune Cells. Front Immunol 2017; 7:667. [PMID: 28127296 PMCID: PMC5226962 DOI: 10.3389/fimmu.2016.00667] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/19/2016] [Indexed: 12/20/2022] Open
Abstract
Immune senescence is a natural consequence of aging and may contribute to frailty and loss of homeostasis in later life. Calorie restriction increases healthy life-span in C57BL/6J (but not DBA/2J) mice, but whether this is related to preservation of immune function, and how it interacts with aging, is unclear. We compared phenotypic and functional characteristics of natural killer (NK) cells and T cells, across the lifespan, of calorie-restricted (CR) and control C57BL/6 and DBA/2 mice. Calorie restriction preserves a naïve T cell phenotype and an immature NK cell phenotype as mice age. The splenic T cell populations of CR mice had higher proportions of CD11a-CD44lo cells, lower expression of TRAIL, KLRG1, and CXCR3, and higher expression of CD127, compared to control mice. Similarly, splenic NK cells from CR mice had higher proportions of less differentiated CD11b-CD27+ cells and correspondingly lower proportions of highly differentiated CD11b+CD27-NK cells. Within each of these subsets, cells from CR mice had higher expression of CD127, CD25, TRAIL, NKG2A/C/E, and CXCR3 and lower expression of KLRG1 and Ly49 receptors compared to controls. The effects of calorie restriction on lymphoid cell populations in lung, liver, and lymph nodes were identical to those seen in the spleen, indicating that this is a system-wide effect. The impact of calorie restriction on NK cell and T cell maturation is much more profound than the effect of aging and, indeed, calorie restriction attenuates these age-associated changes. Importantly, the effects of calorie restriction on lymphocyte maturation were more marked in C57BL/6 than in DBA/2J mice indicating that delayed lymphocyte maturation correlates with extended lifespan. These findings have implications for understanding the interaction between nutritional status, immunity, and healthy lifespan in aging populations.
Collapse
Affiliation(s)
- Matthew J White
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine , London , UK
| | - Charlotte M Beaver
- UCL Institute of Healthy Ageing, University College London , London , UK
| | - Martin R Goodier
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine , London , UK
| | - Christian Bottomley
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine , London , UK
| | - Carolyn M Nielsen
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine , London , UK
| | - Asia-Sophia F M Wolf
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine , London , UK
| | - Luisa Boldrin
- Dubowitz Neuromuscular Centre, Developmental Neurosciences Programme, Molecular Neurosciences Section, Institute of Child Health, University College London , London , UK
| | - Charlotte Whitmore
- Dubowitz Neuromuscular Centre, Developmental Neurosciences Programme, Molecular Neurosciences Section, Institute of Child Health, University College London , London , UK
| | - Jennifer Morgan
- Dubowitz Neuromuscular Centre, Developmental Neurosciences Programme, Molecular Neurosciences Section, Institute of Child Health, University College London , London , UK
| | - Daniel J Pearce
- UCL Institute of Healthy Ageing, University College London , London , UK
| | - Eleanor M Riley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine , London , UK
| |
Collapse
|
20
|
Zheng M, Sun R, Wei H, Tian Z. NK Cells Help Induce Anti-Hepatitis B Virus CD8+ T Cell Immunity in Mice. THE JOURNAL OF IMMUNOLOGY 2016; 196:4122-31. [PMID: 27183639 DOI: 10.4049/jimmunol.1500846] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 03/16/2016] [Indexed: 12/12/2022]
Abstract
Although recent clinical studies demonstrate that NK cell function is impaired in hepatitis B virus (HBV)-persistent patients, whether or how NK cells play a role in anti-HBV adaptive immunity remains to be explored. Using a mouse model mimicking acute HBV infection by hydrodynamic injection of an HBV plasmid, we observed that although serum hepatitis B surface Ag and hepatitis B envelope Ag were eliminated within 3 to 4 wk, HBV might persist for >8 wk in CD8(-/-) mice and that adoptive transfer of anti-HBV CD8(+) T cells restored the ability to clear HBV in HBV-carrier Rag1(-/-) mice. These results indicate that CD8(+) T cells are critical in HBV elimination. Furthermore, NK cells increased IFN-γ production after HBV plasmid injection, and NK cell depletion led to significantly increased HBV persistence along with reduced frequency of hepatitis B core Ag-specific CD8(+) T cells. Adoptive transfer of IFN-γ-sufficient NK cells restored donor CD8(+) T cell function, indicating that NK cells positively regulated CD8(+) T cells via secreting IFN-γ. We also observed that NK cell depletion correlated with decreased effector memory CD8(+) T cell frequencies. Importantly, adoptive transfer experiments showed that NK cells were involved in anti-HBV CD8(+) T cell recall responses. Moreover, DX5(+)CD49a(-) conventional, but not DX5(-)CD49a(+) liver-resident, NK cells were involved in improving CD8(+) T cell responses against HBV. Overall, the current study reveals that NK cells, especially DX5(+)CD49a(-) conventional NK cells, promote the antiviral activity of CD8(+) T cell responses via secreting IFN-γ in a mouse model mimicking acute HBV infection.
Collapse
Affiliation(s)
- Meijuan Zheng
- Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; and
| | - Rui Sun
- Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China;
| | - Haiming Wei
- Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Zhigang Tian
- Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
21
|
Modulation of Autoimmune T-Cell Memory by Stem Cell Educator Therapy: Phase 1/2 Clinical Trial. EBioMedicine 2015; 2:2024-36. [PMID: 26844283 PMCID: PMC4703710 DOI: 10.1016/j.ebiom.2015.11.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 10/29/2015] [Accepted: 11/03/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a T cell-mediated autoimmune disease that causes a deficit of pancreatic islet β cells. The complexities of overcoming autoimmunity in T1D have contributed to the challenges the research community faces when devising successful treatments with conventional immune therapies. Overcoming autoimmune T cell memory represents one of the key hurdles. METHODS In this open-label, phase 1/phase 2 study, Caucasian T1D patients (N = 15) received two treatments with the Stem Cell Educator (SCE) therapy, an approach that uses human multipotent cord blood-derived multipotent stem cells (CB-SCs). SCE therapy involves a closed-loop system that briefly treats the patient's lymphocytes with CB-SCs in vitro and returns the "educated" lymphocytes (but not the CB-SCs) into the patient's blood circulation. This study is registered with ClinicalTrials.gov, NCT01350219. FINDINGS Clinical data demonstrated that SCE therapy was well tolerated in all subjects. The percentage of naïve CD4(+) T cells was significantly increased at 26 weeks and maintained through the final follow-up at 56 weeks. The percentage of CD4(+) central memory T cells (TCM) was markedly and constantly increased at 18 weeks. Both CD4(+) effector memory T cells (TEM) and CD8(+) TEM cells were considerably decreased at 18 weeks and 26 weeks respectively. Additional clinical data demonstrated the modulation of C-C chemokine receptor 7 (CCR7) expressions on naïve T, TCM, and TEM cells. Following two treatments with SCE therapy, islet β-cell function was improved and maintained in individuals with residual β-cell function, but not in those without residual β-cell function. INTERPRETATION Current clinical data demonstrated the safety and efficacy of SCE therapy in immune modulation. SCE therapy provides lasting reversal of autoimmune memory that could improve islet β-cell function in Caucasian subjects. FUNDING Obra Social "La Caixa", Instituto de Salud Carlos III, Red de Investigación Renal, European Union FEDER Funds, Principado de Asturias, FICYT, and Hackensack University Medical Center Foundation.
Collapse
Key Words
- AIRE, autoimmune regulator
- Autoimmunity
- CB-SCs, human cord blood-derived multipotent stem cells
- CCR7, C–C chemokine receptor 7
- Cord blood stem cell
- HLA, human leukocyte antigen
- HbA1C, glycated hemoglobin
- IL, interleukin
- Immune modulation
- M2, muscarinic acetylcholine receptor 2
- MLR, mixed leukocyte reactions
- MNC, mononuclear cells
- Memory T cells
- OGTT, oral glucose tolerance test
- PBMC, peripheral blood mononuclear cells
- R, responder
- S, stimulator
- SCE, Stem Cell Educator
- T1D, type 1 diabetes
- TCM, central memory T cells
- TCR, T-cell receptor
- TEM, effector memory T cells
- TGF-β1, transforming growth factor-β1
- Th, helper T cell
- Tregs, regulatory T cells
- Type 1 diabetes
Collapse
|
22
|
Longbrake EE, Ramsbottom MJ, Cantoni C, Ghezzi L, Cross AH, Piccio L. Dimethyl fumarate selectively reduces memory T cells in multiple sclerosis patients. Mult Scler 2015; 22:1061-1070. [PMID: 26459150 DOI: 10.1177/1352458515608961] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 09/07/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Dimethyl fumarate (DMF) alters the phenotype of circulating immune cells and causes lymphopenia in a subpopulation of treated multiple sclerosis (MS) patients. OBJECTIVE To phenotypically characterize circulating leukocytes in DMF-treated MS patients. METHODS Cross-sectional observational comparisons of peripheral blood from DMF-treated MS patients (n = 17 lymphopenic and n = 24 non-lymphopenic), untreated MS patients (n = 17) and healthy controls (n = 23); immunophenotyped using flow cytometry. Longitudinal samples were analyzed for 13 DMF-treated patients. RESULTS Lymphopenic DMF-treated patients had significantly fewer circulating CD8(+) and CD4(+) T cells, CD56(dim) natural killer (NK) cells, CD19(+) B cells and plasmacytoid dendritic cells when compared to controls. CXCR3(+) and CCR6(+) expression was disproportionately reduced among CD4(+) T cells, while the proportion of T-regulatory (T-reg) cells was unchanged. DMF did not affect circulating CD56(hi) NKcells, monocytes or myeloid dendritic cells. Whether lymphopenic or not, DMF-treated patients had a lower proportion of circulating central and effector memory T cells and concomitant expansion of naïve T cells compared to the controls. CONCLUSIONS DMF shifts the immunophenotypes of circulating T cells, causing a reduction of memory cells and a relative expansion of naïve cells, regardless of the absolute lymphocyte count. This may represent one mechanism of action of the drug. Lymphopenic patients had a disproportionate loss of CD8(+) T-cells, which may affect their immunocompetence.
Collapse
Affiliation(s)
- E E Longbrake
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
| | - M J Ramsbottom
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
| | - C Cantoni
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
| | - L Ghezzi
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri.,Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, Fondazione Cà Granda, IRCCS Ospedale Policlinico, Milan, Italy
| | - A H Cross
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
| | - L Piccio
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
23
|
Khan SH, Martin MD, Starbeck-Miller GR, Xue HH, Harty JT, Badovinac VP. The Timing of Stimulation and IL-2 Signaling Regulate Secondary CD8 T Cell Responses. PLoS Pathog 2015; 11:e1005199. [PMID: 26431533 PMCID: PMC4592272 DOI: 10.1371/journal.ppat.1005199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 09/09/2015] [Indexed: 11/19/2022] Open
Abstract
Memory CD8 T cells provide protection to immune hosts by eliminating pathogen-infected cells during re-infection. While parameters influencing the generation of primary (1°) CD8 T cells are well established, the factors controlling the development of secondary (2°) CD8 T cell responses remain largely unknown. Here, we address the mechanisms involved in the generation and development of 2° memory (M) CD8 T cells. We observed that the time at which 1° M CD8 T cells enter into immune response impacts their fate and differentiation into 2° M CD8 T cells. Late-entry of 1° M CD8 T cells into an immune response (relative to the onset of infection) not only facilitated the expression of transcription factors associated with memory formation in 2° effector CD8 T cells, but also influenced the ability of 2° M CD8 T cells to localize within the lymph nodes, produce IL-2, and undergo Ag-driven proliferation. The timing of stimulation of 1° M CD8 T cells also impacted the duration of expression of the high-affinity IL-2 receptor (CD25) on 2° effector CD8 T cells and their sensitivity to IL-2 signaling. Importantly, by blocking or enhancing IL-2 signaling in developing 2° CD8 T cells, we provide direct evidence for the role of IL-2 in controlling the differentiation of Ag-driven 2° CD8 T cell responses. Thus, our data suggest that the process of 1° M to 2° M CD8 T cell differentiation is not fixed and can be manipulated, a notion with relevance for the design of future prime-boost vaccination approaches.
Collapse
Affiliation(s)
- Shaniya H. Khan
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Matthew D. Martin
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Gabriel R. Starbeck-Miller
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Hai-Hui Xue
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
| | - John T. Harty
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Vladimir P. Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
24
|
Abstract
Memory CD4 T cells are strategically positioned at mucosal surfaces to initiate a robust adaptive immune response. The detection of specific antigen via the T-cell receptor causes these memory T cells to unleash a potent antimicrobial response that includes rousing local innate immune populations for tissue-specific defense. Paradoxically, these same memory T cells can also be stimulated by nonantigen-specific signals that are generated by the activity of local innate immune cells. This versatility of mucosal memory T cells in both the initiation and the sensing of local innate immunity could be a vitally important asset during pathogen defense but alternatively could be responsible for initiating and maintaining chronic inflammation in sensitive mucosal tissues.
Collapse
|
25
|
A single dose of inactivated hepatitis A vaccine promotes HAV-specific memory cellular response similar to that induced by a natural infection. Vaccine 2015; 33:3813-20. [DOI: 10.1016/j.vaccine.2015.06.099] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 11/22/2022]
|
26
|
|
27
|
Abstract
Tissue-resident memory T (Trm) cells constitute a recently identified lymphocyte lineage that occupies tissues without recirculating. They provide a first response against infections reencountered at body surfaces, where they accelerate pathogen clearance. Because Trm cells are not present within peripheral blood, they have not yet been well characterized, but are transcriptionally, phenotypically, and functionally distinct from recirculating central and effector memory T cells. In this review, we will summarize current knowledge of Trm cell ontogeny, regulation, maintenance, and function and will highlight technical considerations for studying this population.
Collapse
Affiliation(s)
- Jason M Schenkel
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - David Masopust
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
28
|
Yuzefpolskiy Y, Baumann FM, Penny LA, Studzinski GP, Kalia V, Sarkar S. Vitamin D receptor signals regulate effector and memory CD8 T cell responses to infections in mice. J Nutr 2014; 144:2073-82. [PMID: 25320188 DOI: 10.3945/jn.114.202895] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Vitamin D insufficiency is associated with broad-ranging human disease sequelae such as bone disease, cancer, cardiovascular disease, allergy, autoimmune disorders, diabetes, and infectious diseases. Disease risk and severity of a large proportion of the nonskeletal disorders heavily involve the cytotoxic cluster of differentiation (CD) 8 T lymphocyte (CTL) arm of cellular adaptive immunity. Considering the importance of vitamin D in CTL-dependent diseases, there is a critical need for systematic in-depth explorations into the role of vitamin D deficiency in generation and maintenance of CTL immunity during infections and vaccinations. OBJECTIVE With the use of wild-type (WT) vitamin D-sufficient mice and the vitamin D receptor knockout (Vdr(-/-)) mouse model of in vivo deficiency of vitamin D signaling, we systematically analyzed the impact of vitamin D deficiency on antigen-specific effector and memory CD8 T cell responses to acute viral and bacterial infections. METHODS WT and Vdr(-/-) mice were infected with lymphocytic choriomeningitis virus, a natural mouse pathogen, and antigen-specific CTL responses were analyzed during priming, expansion, contraction, and memory phases. Magnitude, breadth, cytokine production, and localization of antiviral effector and memory CTLs to lymphoid and nonlymphoid tissues were specifically assessed. RESULTS The absence of vitamin D signals led to 1) aberrant CD8 T cell effector differentiation (∼2-fold lower granzyme B and reduced B cell lymphoma 2; P ≤ 0.05) and enhanced contraction (∼15% increase; P ≤ 0.05) in antigen-specific CTLs; 2) a significantly restricted (P ≤ 0.05) breadth of the antigen-specific CD8 T cell effector and memory repertoire; and 3) preferential localization of effector (∼2.5-fold increase; P ≤ 0.01) and memory (∼5-fold increase; P ≤ 0.001) CD8 T cells to the lymph nodes compared to nonlymphoid tissues. CONCLUSION Our data show a previously unrecognized impact of vitamin D deficiency on the quantity, quality, breadth, and location of CD8 T cell immunity to acute viral and bacterial infections.
Collapse
Affiliation(s)
- Yevgeniy Yuzefpolskiy
- Center for Molecular Immunology and Infectious Diseases, Department of Veterinary and Biomedical Sciences, and The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA; and
| | - Florian M Baumann
- Center for Molecular Immunology and Infectious Diseases, Department of Veterinary and Biomedical Sciences, and The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA; and
| | - Laura A Penny
- Center for Molecular Immunology and Infectious Diseases, Department of Veterinary and Biomedical Sciences, and The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA; and
| | - George P Studzinski
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
| | - Vandana Kalia
- Center for Molecular Immunology and Infectious Diseases, Department of Veterinary and Biomedical Sciences, and The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA; and
| | - Surojit Sarkar
- Center for Molecular Immunology and Infectious Diseases, Department of Veterinary and Biomedical Sciences, and The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA; and
| |
Collapse
|
29
|
Strbo N, Garcia-Soto A, Schreiber TH, Podack ER. Secreted heat shock protein gp96-Ig: next-generation vaccines for cancer and infectious diseases. Immunol Res 2014; 57:311-25. [PMID: 24254084 DOI: 10.1007/s12026-013-8468-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Over the past decade, our laboratory has developed a secreted heat shock protein (HSP), chaperone gp96, cell-based vaccine that generates effective anti-tumor and anti-infectious immunity in vivo. Gp96-peptide complexes were identified as an extremely efficient stimulator of MHC I-mediated antigen cross-presentation, generating CD8 cytotoxic T-lymphocyte responses detectable in blood, spleen, gut and reproductive tract to femto-molar concentrations of antigen. These studies provided the first evidence that cell-based gp96-Ig-secreting vaccines may serve as a potent modality to induce both systemic and mucosal immunity. This approach takes advantage of the combined adjuvant and antigen delivery capacity of gp96 for the generation of cytotoxic immunity against a wide range of antigens in both anti-vial and anti-cancer vaccination. Here, we review the vaccine design that utilizes the unique property/ability of endoplasmic HSP gp96 to bind antigenic peptides and deliver them to antigen-presenting cells.
Collapse
Affiliation(s)
- Natasa Strbo
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, RMSB 3008, 1600 NW 10th Ave, Miami, FL, 33136, USA,
| | | | | | | |
Collapse
|
30
|
Hosking MP, Flynn CT, Whitton JL. Antigen-specific naive CD8+ T cells produce a single pulse of IFN-γ in vivo within hours of infection, but without antiviral effect. THE JOURNAL OF IMMUNOLOGY 2014; 193:1873-85. [PMID: 25015828 DOI: 10.4049/jimmunol.1400348] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In vitro studies have shown that naive CD8(+) T cells are unable to express most of their effector proteins until after at least one round of cell division has taken place. We have reassessed this issue in vivo and find that naive CD8(+) T cells mount Ag-specific responses within hours of infection, before proliferation has commenced. Newly activated naive Ag-specific CD8(+) T cells produce a rapid pulse of IFN-γ in vivo and begin to accumulate granzyme B and perforin. Later, in vivo cytolytic activity is detectable, coincident with the initiation of cell division. Despite the rapid development of these functional attributes, no antiviral effect was observed early during infection, even when the cells are present in numbers similar to those of virus-specific memory cells. The evolutionary reason for the pulse of IFN-γ synthesis by naive T cells is uncertain, but the lack of antiviral impact suggests that it may be regulatory.
Collapse
Affiliation(s)
- Martin P Hosking
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Claudia T Flynn
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - J Lindsay Whitton
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
31
|
Rai D, Martin MD, Badovinac VP. The longevity of memory CD8 T cell responses after repetitive antigen stimulations. THE JOURNAL OF IMMUNOLOGY 2014; 192:5652-9. [PMID: 24829415 DOI: 10.4049/jimmunol.1301063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In experimental models in which the Ag-stimulation history of memory CD8 T cell populations was clearly defined (adoptive transfer of a known number of TCR-transgenic memory CD8 T cells), all facets of the ensuing CD8 T cell responses, including proliferative expansion, duration and extent of contraction, diversification of memory CD8 T cell transcriptomes, and life-long survival, were dependent on the number of prior Ag encounters. However, the extent to which sequential adoptive-transfer models reflect the physiological scenario in which memory CD8 T cells are generated by repetitive Ag challenges of individual hosts (no adoptive transfer involved) is not known. Direct comparison of endogenous memory CD8 T cell responses generated in repetitively infected hosts revealed that recurrent homologous boosting was required to preserve the numbers and increase the phenotypic and functional complexity of the developing memory CD8 T cell pool. Although life-long survival of the memory CD8 T cells was not impacted, phenotype (i.e., upregulation of CD62L) and function (i.e., homeostatic turnover, Ag-stimulated IL-2 production) of repeatedly stimulated memory CD8 T cells were dependent on time after last Ag encounter. Therefore, repetitive Ag challenges of individual hosts can substantially influence the numerical and functional attributes of polyclonal memory CD8 T cells, a notion with important implications for the design of future vaccination strategies aimed at increasing the number of protective memory CD8 T cells.
Collapse
Affiliation(s)
- Deepa Rai
- Department of Pathology, University of Iowa, Iowa City, IA 52242; and
| | - Matthew D Martin
- Department of Pathology, University of Iowa, Iowa City, IA 52242; and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA 52242; and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
32
|
Abstract
The activation of T cells is a tightly regulated process that has evolved to maximize protective immune responses to pathogens while minimizing damage to self-tissues. A delicate balance of cell-intrinsic, costimulatory, and transcriptional pathways as well as micro-environmental cues such as local cytokines controls the magnitude and nature of T-cell responses in vivo. The discovery of functional small noncoding RNAs called micro-RNAs (miRNAs) has introduced new mechanisms that contribute to the regulation of protein translation and cellular responses to stimuli. miRNAs are short (approximately 22 bp) RNA species, which bind to mRNAs and suppress translation. Due to their short length and imperfect base pairing requirements, each miRNA has the potential to regulate various pathways through the translational inhibition of multiple mRNAs. The human and mouse genomes each encode hundreds of miRNAs, and studying the function of miRNAs has led to the realization that they play important roles in diverse biological processes from development and cancer to immunity. This review focuses on the function of mir-155 in T cells and the impact of this miRNA on autoimmunity, tumor immunity, and pathogen-induced immunity.
Collapse
|
33
|
Santos-Gomes GM, Rodrigues A, Teixeira F, Carreira J, Alexandre-Pires G, Carvalho S, Santos-Mateus D, Martins C, Vale-Gato I, Marques C, Tomás AM. Immunization with the Leishmania infantum recombinant cyclophilin protein 1 confers partial protection to subsequent parasite infection and generates specific memory T cells. Vaccine 2014; 32:1247-53. [PMID: 24486368 DOI: 10.1016/j.vaccine.2014.01.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 11/13/2013] [Accepted: 01/14/2014] [Indexed: 12/15/2022]
Abstract
Control of zoonotic visceral leishmaniosis can be achieved using several available drugs. These drugs present high toxicity and require longer treatment regimens which complicate compliance to the treatment. Other control measures directed to the vector or the reservoirs are useful tools to restrain the spreading of this disease but the effects are transitory. A safe, affordable and efficient vaccine conferring long lasting immunity should be the most cost effective way of controlling zoonotic visceral leishmaniosis. The present study aims at characterizing a cyclophilin protein 1 of Leishmania infantum (LiCyP1) and investigating whether recombinant LiCyP1 (LirCyP1) is able to confer protection against infection by evaluating viable parasite load and the generation of specific CD4(+) and CD8(+) effector and central memory T cells in rodent model. LiCyP1 is present in the cytoplasm of L. infantum amastigotes and promastigotes. Immunization of BALB/c mice with LirCyP1 confers high protection to L. infantum infection, causing a marked reduction in parasite replication in the liver and spleen. Furthermore, helper and cytotoxic memory T cell subsets able to specifically recognize parasite antigens expanded in immunized and in challenged mice. CD4(+) T cell subpopulation of intermediate phenotype (CD62L(high)CD127(low)) of challenging mice also presented an accentuated expansion after the recall. This study demonstrated that LirCyP1 confers partial protection to L. infantum infection, promoting the generation of a desired long lasting immunity. LirCyP1 can be considered a potential candidate for the design of a vaccine against zoonotic visceral leishmaniosis.
Collapse
Affiliation(s)
- G M Santos-Gomes
- Unidade de Ensino e Investigação de Parasitologia Médica, Centro de Malária e Outras Doenças Tropicais, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008 Lisboa, Portugal.
| | - A Rodrigues
- Unidade de Ensino e Investigação de Parasitologia Médica, Centro de Malária e Outras Doenças Tropicais, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - F Teixeira
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal; ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Largo Prof. Abel Salazar 2, 4099-003 Porto, Portugal
| | - J Carreira
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal; ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Largo Prof. Abel Salazar 2, 4099-003 Porto, Portugal
| | - G Alexandre-Pires
- CIISA, Faculdade de Medicina Veterinária, Universidade Técnica de Lisboa, Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - S Carvalho
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal; ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Largo Prof. Abel Salazar 2, 4099-003 Porto, Portugal
| | - D Santos-Mateus
- Unidade de Ensino e Investigação de Parasitologia Médica, Centro de Malária e Outras Doenças Tropicais, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - C Martins
- Departamento de Imunologia, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo dos Mártires de Pátria, Lisboa, Portugal
| | - I Vale-Gato
- Unidade de Ensino e Investigação de Parasitologia Médica, Centro de Malária e Outras Doenças Tropicais, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - C Marques
- Unidade de Ensino e Investigação de Parasitologia Médica, Centro de Malária e Outras Doenças Tropicais, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - A M Tomás
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal; ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Largo Prof. Abel Salazar 2, 4099-003 Porto, Portugal
| |
Collapse
|
34
|
Starbeck-Miller GR, Badovinac VP, Barber DL, Harty JT. Cutting edge: Expression of FcγRIIB tempers memory CD8 T cell function in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:35-9. [PMID: 24285839 PMCID: PMC3874719 DOI: 10.4049/jimmunol.1302232] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
During reinfection, high-affinity IgG Abs form complexes with both soluble Ag and Ag displayed on the surface of infected cells. These interactions regulate cellular activation of both innate cells and B cells, which express specific combinations of activating FcγRs (FcγRI, FcγRIII, FcγRIV) and/or the inhibitory FcγR (FcγRIIB). Direct proof for functional expression of FcγR by Ag-specific CD8 T cells is lacking. In this article, we show that the majority of memory CD8 T cells generated by bacterial or viral infection express only FcγRIIB, and that FcγRIIB could be detected on previously activated human CD8 T cells. Of note, FcγR stimulation during in vivo Ag challenge not only inhibited the cytotoxicity of memory CD8 T cells against peptide-loaded or virus-infected targets, but FcγRIIB blockade during homologous virus challenge enhanced the secondary CD8 T cell response. Thus, memory CD8 T cells intrinsically express a functional FcγRIIB, permitting Ag-Ab complexes to regulate secondary CD8 T cell responses.
Collapse
Affiliation(s)
| | - Vladimir P. Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Daniel L. Barber
- T Lymphocyte Biology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John T. Harty
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
35
|
Starbeck-Miller GR, Xue HH, Harty JT. IL-12 and type I interferon prolong the division of activated CD8 T cells by maintaining high-affinity IL-2 signaling in vivo. ACTA ACUST UNITED AC 2013; 211:105-20. [PMID: 24367005 PMCID: PMC3892973 DOI: 10.1084/jem.20130901] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The signal 3 cytokines interleukin-12 and type I interferon sustain CD8 T cell division by prolonging expression of CD25 in vivo. TCR ligation and co-stimulation induce cellular division; however, optimal accumulation of effector CD8 T cells requires direct inflammatory signaling by signal 3 cytokines, such as IL-12 or type I IFNs. Although in vitro studies suggest that IL-12/type I IFN may enhance T cell survival or early proliferation, the mechanisms underlying optimal accumulation of CD8 T cells in vivo are unknown. In particular, it is unclear if disparate signal 3 cytokines optimize effector CD8 T cell accumulation by the same mechanism and how these inflammatory cytokines, which are transiently produced early after infection, affect T cell accumulation many days later at the peak of the immune response. Here, we show that transient exposure of CD8 T cells to IL-12 or type I IFN does not promote survival or confer an early proliferative advantage in vivo, but rather sustains surface expression of CD25, the high-affinity IL-2 receptor. This prolongs division of CD8 T cells in response to basal IL-2, through activation of the PI3K pathway and expression of FoxM1, a positive regulator of cell cycle progression genes. Thus, signal 3 cytokines use a common pathway to optimize effector CD8 T cell accumulation through a temporally orchestrated sequence of cytokine signals that sustain division rather than survival.
Collapse
Affiliation(s)
- Gabriel R Starbeck-Miller
- Interdisciplinary Graduate Program in Immunology, 2 Department of Microbiology, and 3 Department of Pathology, University of Iowa, Iowa City, IA 52242
| | | | | |
Collapse
|
36
|
Kim MV, Ouyang W, Liao W, Zhang MQ, Li MO. The transcription factor Foxo1 controls central-memory CD8+ T cell responses to infection. Immunity 2013; 39:286-97. [PMID: 23932570 DOI: 10.1016/j.immuni.2013.07.013] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 05/03/2013] [Indexed: 11/19/2022]
Abstract
Memory T cells protect hosts from pathogen reinfection, but how these cells emerge from a pool of antigen-experienced T cells is unclear. Here, we show that mice lacking the transcription factor Foxo1 in activated CD8+ T cells have defective secondary, but not primary, responses to Listeria monocytogenes infection. Compared to short-lived effector T cells, memory-precursor T cells expressed higher amounts of Foxo1, which promoted their generation and maintenance. Chromatin immunoprecipitation sequencing revealed the transcription factor Tcf7 and the chemokine receptor Ccr7 as Foxo1-bound target genes, which have critical functions in central-memory T cell differentiation and trafficking. These findings demonstrate that Foxo1 is selectively incorporated into the genetic program that regulates memory CD8+ T cell responses to infection.
Collapse
Affiliation(s)
- Myoungjoo V Kim
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
37
|
Umansky V, Sevko A. Ret transgenic mouse model of spontaneous skin melanoma: focus on regulatory T cells. Pigment Cell Melanoma Res 2013; 26:457-63. [PMID: 23560814 DOI: 10.1111/pcmr.12104] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 03/29/2013] [Indexed: 01/07/2023]
Abstract
Ret transgenic mouse model of skin malignant melanoma is characterized by the overexpression of the human ret transgene in melanin-containing cells. Transgenic mice spontaneously develop skin tumors with metastases in lymph nodes, lungs, liver, brain, and the bone marrow. Tumor lesions show typical melanoma morphology and express melanoma-associated antigens. Although transgenic mice demonstrate an accumulation of melanoma antigen-specific memory and effector T cells, their anti-tumor effects could be blocked by highly immunosuppressive leukocytes enriched in the tumor microenvironment and in the periphery. Here, we discuss the role of one of the most potent immunosuppressive subset, regulatory T cells, in the melanoma progression in this model.
Collapse
Affiliation(s)
- Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany.
| | | |
Collapse
|
38
|
Hombach AA, Chmielewski M, Rappl G, Abken H. Adoptive Immunotherapy with Redirected T Cells Produces CCR7− Cells That Are Trapped in the Periphery and Benefit from Combined CD28-OX40 Costimulation. Hum Gene Ther 2013; 24:259-69. [DOI: 10.1089/hum.2012.247] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Andreas A. Hombach
- Center for Molecular Medicine Cologne (CMMC) and Department I Internal Medicine, University of Cologne, 50931 Cologne, Germany
| | - Markus Chmielewski
- Center for Molecular Medicine Cologne (CMMC) and Department I Internal Medicine, University of Cologne, 50931 Cologne, Germany
| | - Gunter Rappl
- Center for Molecular Medicine Cologne (CMMC) and Department I Internal Medicine, University of Cologne, 50931 Cologne, Germany
| | - Hinrich Abken
- Center for Molecular Medicine Cologne (CMMC) and Department I Internal Medicine, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
39
|
Synergistic Induction of Macrophage Inflammatory Protein-3α;/CCL20 Production by Interleukin-17A and Tumor Necrosis Factor-α; in Nasal Polyp Fibroblasts. World Allergy Organ J 2013; 2:218-23. [PMID: 23283206 PMCID: PMC3651001 DOI: 10.1097/wox.0b013e3181bdd219] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Accumulation of T cells and immature dendritic cells (DCs) is one of the characteristic features of nasal polyps. However, the question remains why these cells accumulate in nasal polyp tissue. Macrophage inflammatory protein-3α (MIP-3α/CCL20) is a chemokine involved in the migration of T cells and immature DCs into inflammatory tissue sites. Fibroblasts are a rich source of cytokines and chemokines. The objective of this study was to demonstrate the expression of MIP-3α/CCL20 in nasal polyp fibroblasts after stimulation with proinflammatory cytokines such as interleukin-17 (IL-17) and tumor necrosis factor-α (TNF-α). Methods Fibroblast lines were established from nasal polyps. MIP-3α/CCL20 mRNA expression was evaluated by real-time reverse transcription-polymerase chain reaction (real-time RT-PCR). The amount of MIP-3α/CCL20 in the supernatants was measured by enzyme-linked immunosorbent assay (ELISA). Results IL-17A and TNF-α synergistically induced MIP-3α/CCL20 production by nasal polyp fibroblasts in a dose- and time-dependent manner. This synergy was observed by stimulation with TNF-α plus IL-17A or IL-17F, but not IL-17E. Conclusions Nasal polyp fibroblasts, by producing MIP-3α/CCL20, may play an important role in the recruitment of T cells and DCs in upper airway inflammatory lesions such as nasal polyps.
Collapse
|
40
|
Regulation of memory CD4 T-cell pool size and function by natural killer T cells in vivo. Proc Natl Acad Sci U S A 2012; 109:16992-7. [PMID: 23027937 DOI: 10.1073/pnas.1203494109] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
To develop more effective vaccines and strategies to regulate chronic inflammatory diseases, it is important to understand the mechanisms of immunological memory. Factors regulating memory CD4(+) T helper (Th)-cell pool size and function remain unclear, however. We show that activation of type I invariant natural killer T (iNKT) cells with glycolipid ligands and activation of type II natural killer T (NKT) cells with the endogenous ligand sulfatide induced dramatic proliferation and expansion of memory, but not naïve, CD4 T cells. NKT cell-induced proliferation of memory Th1 and Th2 cells was dependent largely on the production of IL-2, with Th2-cell proliferation also affected by loss of IL-4. Type II NKT cells were also required for efficient maintenance of memory CD4 T cells in vivo. Activation of iNKT cells resulted in up-regulation of IFN-γ expression by memory Th2 cells. These IFN-γ-producing memory Th2 cells showed a decreased capability to induce Th2 cytokines and eosinophilic airway inflammation. Thus, activated NKT cells directly regulate memory CD4 T-cell pool size and function via the production of cytokines in vivo.
Collapse
|
41
|
Wiesel M, Oxenius A. From crucial to negligible: functional CD8⁺ T-cell responses and their dependence on CD4⁺ T-cell help. Eur J Immunol 2012; 42:1080-8. [PMID: 22539281 DOI: 10.1002/eji.201142205] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
CD8(+) T cells play an important role in controlling pathogenic infections and are therefore key players in the immune response. It has been shown that among other factors CD4(+) T cells can shape the magnitude as well as the quality of primary and/or secondary CD8(+) T-cell responses. However, due to the complexity and the differences among diverse immunization or infection models, the overall requirement, the time points, as well as the specific mechanism(s) of CD4(+) T-cell help may differ substantially. Here, we summarize current knowledge about the differential requirement of CD4(+) T-cell help in promoting primary CD8(+) T-cell responses as well as establishing functional memory CD8(+) T cells in various experimental settings.
Collapse
Affiliation(s)
- Melanie Wiesel
- Institute for Microbiology, ETH Zürich, Zürich, Switzerland
| | | |
Collapse
|
42
|
Skin infection generates non-migratory memory CD8+ T(RM) cells providing global skin immunity. Nature 2012; 483:227-31. [PMID: 22388819 PMCID: PMC3437663 DOI: 10.1038/nature10851] [Citation(s) in RCA: 664] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 01/12/2012] [Indexed: 02/07/2023]
|
43
|
Endo Y, Iwamura C, Kuwahara M, Suzuki A, Sugaya K, Tumes DJ, Tokoyoda K, Hosokawa H, Yamashita M, Nakayama T. Eomesodermin controls interleukin-5 production in memory T helper 2 cells through inhibition of activity of the transcription factor GATA3. Immunity 2012; 35:733-45. [PMID: 22118525 DOI: 10.1016/j.immuni.2011.08.017] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 06/06/2011] [Accepted: 08/23/2011] [Indexed: 12/24/2022]
Abstract
The regulation of memory CD4(+) helper T (Th) cell function, such as polarized cytokine production, remains unclear. Here we show that memory T helper 2 (Th2) cells are divided into four subpopulations by CD62L and CXCR3 expression. All four subpopulations produced interleukin-4 (IL-4) and IL-13, whereas only the CD62L(lo)CXCR3(lo) population produced IL-5 accompanied by increased H3-K4 methylation at the Il5 gene locus. The transcription factor Eomesodermin (encoded by Eomes) was highly expressed in memory Th2 cells, whereas its expression was selectively downregulated in the IL-5-producing cells. Il5 expression was enhanced in Eomes-deficient cells, and Eomesodermin was shown to interact with the transcription factor GATA3, preventing GATA3 binding to the Il5 promoter. Memory Th2 cell-dependent airway inflammation was attenuated in the absence of the CD62L(lo)CXCR3(lo) population but was enhanced by Eomes-deficient memory Th2 cells. Thus, IL-5 production in memory Th2 cells is regulated by Eomesodermin via the inhibition of GATA3 activity.
Collapse
Affiliation(s)
- Yusuke Endo
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana Chuo-ku, Chiba 260-8670, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Martin MD, Condotta SA, Harty JT, Badovinac VP. Population dynamics of naive and memory CD8 T cell responses after antigen stimulations in vivo. THE JOURNAL OF IMMUNOLOGY 2011; 188:1255-65. [PMID: 22205031 DOI: 10.4049/jimmunol.1101579] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The extent to which the progeny of one primary memory CD8 T cell differs from the progeny of one naive CD8 T cell of the same specificity remains an unresolved question. To explore cell-autonomous functional differences between naive and memory CD8 T cells that are not influenced by differences in the priming environment, an experimental model has been developed in which physiological numbers of both populations of cells were cotransferred into naive hosts before Ag stimulation. Interestingly, naive CD8 T cells undergo greater expansion in numbers than do primary memory CD8 T cells after various infections or immunizations. The intrinsic ability of one naive CD8 T cell to give rise to more effector CD8 T cells than one memory CD8 T cell is independent of the number and quality of primary memory CD8 T cells present in vivo. The sustained proliferation of newly activated naive CD8 T cells contributed to their greater magnitude of expansion. Additionally, longitudinal analyses of primary and secondary CD8 T cell responses revealed that on a per-cell basis naive CD8 T cells generate higher numbers of long-lived memory cells than do primary memory CD8 T cells. This enhanced "memory generation potential" of responding naive CD8 T cells occurred despite the delayed contraction of secondary CD8 T cell responses. Taken together, the data in this study revealed previously unappreciated differences between naive and memory CD8 T cells and will help further define the functional potential for both cell types.
Collapse
Affiliation(s)
- Matthew D Martin
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
45
|
Hombach AA, Abken H. Costimulation by chimeric antigen receptors revisited the T cell antitumor response benefits from combined CD28-OX40 signalling. Int J Cancer 2011; 129:2935-44. [PMID: 22030616 DOI: 10.1002/ijc.25960] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Accepted: 12/10/2010] [Indexed: 12/14/2022]
Abstract
The therapeutic success of adoptive therapy with chimeric antigen receptor (CAR) engineered T cells depends on the appropriate costimulation of CD3ζ to induce full T cell activation. Costimulatory endodomains of the CD28 family are therefore fused with CD3ζ in a dual signalling CAR. Serious adverse events in two most recent trials; however, highlight the need to analyse in more detail the impact of each costimulatory endodomain on individual effector functions of redirected T cells. We therefore performed a thoroughly controlled side-by-side comparison of the most frequently used endodomains with respect to their impact on CD4(+) and CD8(+) T cell effector functions. CD28 reinforced T cell proliferation and is mandatory to induce IL-2. In the absence of added IL-2, CD28 and OX40 (CD137) but not 4-1BB (CD134) enhanced specific cytolysis. While CD28, 4-1BB and OX40 similarly improved pro-inflammatory cytokine secretion, OX40 most efficiently prevented activation induced cell death of CD62L(-) effector memory T cells. CD28 was superior to initiate the T cell response, OX40 and 4-1BB sustained the response in long term with OX40 being most effective. We consequently combined the beneficial functions in a 3rd generation CD28-OX40 CAR which substantially improved the antitumor response without loosing specificity.
Collapse
Affiliation(s)
- Andreas A Hombach
- Clinic I Internal Medicine Tumorgenetics, University Hospital Cologne, Cologne, Germany
| | | |
Collapse
|
46
|
Deng C, Qi H, Wang X, Zhou H, Deng S, Li F. Role of T and Dendritic Cells in Mouse Islet Allografts Treated With Anti-CD45RB Monoclonal Antibodies. Transplant Proc 2011; 43:2721-7. [DOI: 10.1016/j.transproceed.2011.05.049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 03/29/2011] [Accepted: 05/11/2011] [Indexed: 01/09/2023]
|
47
|
Crawley AM, Angel JB. Expression of γ-chain cytokine receptors on CD8+ T cells in HIV infection with a focus on IL-7Rα (CD127). Immunol Cell Biol 2011; 90:379-87. [PMID: 21863001 DOI: 10.1038/icb.2011.66] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
When interleukin-2 (IL-2) receptor γ-chain (γ(C))-sharing cytokine receptors on T cells bind their specific ligands (IL-2, -4, -7, -9, -15 or -21), they initiate a variety of cell signals that promote survival, differentiation or antiviral or antitumor cytolytic functions. Although expression of the γ(C) is constitutive across T-cell subsets, the varying expression of other receptor complex components can regulate cytokine signalling and function. Impaired γ(C) cytokine activity in HIV infection, and the role of γ(C) cytokines in CD8(+) T-cell function and homeostasis, implicates these molecules among potential contributors to the observed decline of cytolytic activity (CTL) in HIV disease. In particular, this review will be highlighting information about the IL-7 receptor (IL-7R) complex, which is composed of the γ(C) and the IL-7Rα (CD127) chains. There has been an abundance of HIV-related CD127 research and its important role in CD8(+) T-cell survival and function. The expression of CD127 undergoes dramatic changes throughout the course of T-cell responses in HIV infection. The expression of CD127 is significantly decreased in progressive HIV disease, whereas effective antiretroviral therapy results in its recovery. Observations of impaired IL-7 activity in HIV(+) individuals have suggested that CD127 has an important role in HIV immunopathogenesis. In addition, a soluble form of CD127 (sCD127) is upregulated in the plasma of HIV(+) individuals. Hence, CD127 is being increasingly considered as a marker of disease prognosis, and related information may provide insight into understanding the expression and role of other γ(C) receptors in HIV disease and contribute to the development of novel cytokine-based therapeutics.
Collapse
Affiliation(s)
- Angela M Crawley
- Department of Chronic Disease, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | |
Collapse
|
48
|
Martin MD, Wirth TC, Lauer P, Harty JT, Badovinac VP. The impact of pre-existing memory on differentiation of newly recruited naive CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:2923-31. [PMID: 21832161 DOI: 10.4049/jimmunol.1100698] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
One goal of immunization is to generate memory CD8 T cells of sufficient quality and quantity to confer protection against infection. It has been shown that memory CD8 T cell differentiation in vivo is controlled, at least in part, by the amount and duration of infection, Ag, and inflammatory cytokines present early after the initiation of the response. In this study, we used models of anti-vectorial immunity to investigate the impact of pre-existing immunity on the development and differentiation of vector-induced primary CD8 T cell responses. We showed that existing CD8 T cell memory influences the magnitude of naive CD8 T cell responses. However, the differentiation of newly recruited (either TCR-transgenic or endogenous) primary CD8 T cells into populations with the phenotype (CD62L(hi), CD27(hi), KLRG-1(low)) and function (tissue distribution, Ag-driven proliferation, cytokine production) of long-term memory was facilitated when they were primed in the presence of vector-specific memory CD8 T cells of the same or unrelated specificity. Therefore, these data suggested that the presence of anti-vectorial immunity impacts the rate of differentiation of vector-induced naive CD8 T cells, a notion with important implications for the design of future vaccination strategies.
Collapse
Affiliation(s)
- Matthew D Martin
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
49
|
Yang S, Liu F, Wang QJ, Rosenberg SA, Morgan RA. The shedding of CD62L (L-selectin) regulates the acquisition of lytic activity in human tumor reactive T lymphocytes. PLoS One 2011; 6:e22560. [PMID: 21829468 PMCID: PMC3145643 DOI: 10.1371/journal.pone.0022560] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 06/27/2011] [Indexed: 01/13/2023] Open
Abstract
CD62L/L-selectin is a marker found on naïve T cells and further distinguishes central memory (Tcm, CD62L+) from effector memory (Tem, CD62L-) T cells. The regulation of CD62L plays a pivotal role in controlling the traffic of T lymphocytes to and from peripheral lymph nodes. CD62L is shed from the cell membrane following T cell activation, however, the physiological significance of this event remains to be elucidated. In this study, we utilized in vitro generated anti-tumor antigen T cells and melanoma lines as a model to evaluate the dynamics of CD62L shedding and expression of CD107a as a marker of lytic activity. Upon encounter, with matched tumor lines, antigen reactive T cells rapidly lose CD62L expression and this was associated with the acquisition of CD107a. By CD62L ELISA, we confirmed that this transition was mediated by the shedding of CD62L when T cells encountered specific tumor antigen. The introduction of a shedding resistant mutant of CD62L into the tumor antigen-reactive T cell line JKF6 impaired CD107a acquisition following antigen recognition and this was correlated with decreased lytic activity as measured by (51)Cr release assays. The linkage of the shedding of CD62L from the surface of anti-tumor T cells and acquisition of lytic activity, suggests a new function for CD62L in T cell effector functions and anti-tumor activity.
Collapse
Affiliation(s)
- Shicheng Yang
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fang Liu
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Qiong J. Wang
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Steven A. Rosenberg
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Richard A. Morgan
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
50
|
Homeostatic cytokines orchestrate the segregation of CD4 and CD8 memory T-cell reservoirs in mice. Blood 2011; 118:3039-50. [PMID: 21791416 DOI: 10.1182/blood-2011-04-349746] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Memory T cells (T(M)s) have been detected in many tissues but their quantitative distribution remains largely undefined. We show that in mice there is a remarkably biased accumulation of long-term CD4 T(M)s into mucosal sites (mainly gut, especially Peyer patches), and CD8 T(M)s into lymph nodes and spleen (in particular, peripheral lymph nodes [PLNs]). This distinction correlates with their differentiated expression of PLN- and gut-homing markers. CD8 and CD4 T(M)s selectively require the expression of PLN-homing marker CCR7 or gut-homing marker α4β7 for maintenance. PLNs and gut supply CD8 and CD4 T(M)s with their individually favored homeostatic cytokine, IL-15, or IL-7. Cytokine stimulation in turn regulates the different gut-homing marker expression on CD4 and CD8 T(M)s. IL-15 plays a major role in vivo regulating CD8 T(M)s homing to PLNs. Thus, the reservoir segregation of CD4 and CD8 T(M)s meets their individual needs for homeostatic cytokines and is under feedback control of cytokine stimulation.
Collapse
|