1
|
Leon C, Tokarev A, Bouchnita A, Volpert V. Modelling of the Innate and Adaptive Immune Response to SARS Viral Infection, Cytokine Storm and Vaccination. Vaccines (Basel) 2023; 11:vaccines11010127. [PMID: 36679972 PMCID: PMC9861811 DOI: 10.3390/vaccines11010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023] Open
Abstract
In this work, we develop mathematical models of the immune response to respiratory viral infection, taking into account some particular properties of the SARS-CoV infections, cytokine storm and vaccination. Each model consists of a system of ordinary differential equations that describe the interactions of the virus, epithelial cells, immune cells, cytokines, and antibodies. Conventional analysis of the existence and stability of stationary points is completed by numerical simulations in order to study the dynamics of solutions. The behavior of the solutions is characterized by large peaks of virus concentration specific to acute respiratory viral infections. At the first stage, we study the innate immune response based on the protective properties of interferon secreted by virus-infected cells. Viral infection down-regulates interferon production. This competition can lead to the bistability of the system with different regimes of infection progression with high or low intensity. After that, we introduce the adaptive immune response with antigen-specific T- and B-lymphocytes. The resulting model shows how the incubation period and the maximal viral load depend on the initial viral load and the parameters of the immune response. In particular, an increase in the initial viral load leads to a shorter incubation period and higher maximal viral load. The model shows that a deficient production of antibodies leads to an increase in the incubation period and even higher maximum viral loads. In order to study the emergence and dynamics of cytokine storm, we consider proinflammatory cytokines produced by cells of the innate immune response. Depending on the parameters of the model, the system can remain in the normal inflammatory state specific for viral infections or, due to positive feedback between inflammation and immune cells, pass to cytokine storm characterized by the excessive production of proinflammatory cytokines. Finally, we study the production of antibodies due to vaccination. We determine the dose-response dependence and the optimal interval of vaccine dose. Assumptions of the model and obtained results correspond to the experimental and clinical data.
Collapse
Affiliation(s)
- Cristina Leon
- Interdisciplinary Center for Mathematical Modelling in Biomedicine, S.M. Nikol’skii Mathematical Institute, Peoples Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St., 117198 Moscow, Russia
- M&S Decisions, 5 Naryshkinskaya Alley, 125167 Moscow, Russia
- Department of Foreign Languages No. 2, Plekhanov Russian University of Economics, 36 Stremyanny Lane, 115093 Moscow, Russia
- Correspondence:
| | - Alexey Tokarev
- Interdisciplinary Center for Mathematical Modelling in Biomedicine, S.M. Nikol’skii Mathematical Institute, Peoples Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St., 117198 Moscow, Russia
- Semenov Institute of Chemical Physics, 4 Kosygin St., 119991 Moscow, Russia
- Bukhara Engineering Technological Institute, 15 Murtazoyeva Street, Bukhara 200100, Uzbekistan
| | - Anass Bouchnita
- Department of Mathematical Sciences, The University of Texas at El Paso, El Paso, TX 79902, USA
| | - Vitaly Volpert
- Interdisciplinary Center for Mathematical Modelling in Biomedicine, S.M. Nikol’skii Mathematical Institute, Peoples Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St., 117198 Moscow, Russia
- Institut Camille Jordan, UMR 5208 CNRS, University Lyon 1, 69622 Villeurbanne, France
| |
Collapse
|
2
|
Nainu F, Ophinni Y, Shiratsuchi A, Nakanishi Y. Apoptosis and Phagocytosis as Antiviral Mechanisms. Subcell Biochem 2023; 106:77-112. [PMID: 38159224 DOI: 10.1007/978-3-031-40086-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Viruses are infectious entities that make use of the replication machinery of their hosts to produce more progenies, causing disease and sometimes death. To counter viral infection, metazoan hosts are equipped with various defense mechanisms, from the rapid-evoking innate immune responses to the most advanced adaptive immune responses. Previous research demonstrated that cells in fruit flies and mice infected with Drosophila C virus and influenza, respectively, undergo apoptosis, which triggers the engulfment of apoptotic virus-infected cells by phagocytes. This process involves the recognition of eat-me signals on the surface of virus-infected cells by receptors of specialized phagocytes, such as macrophages and neutrophils in mice and hemocytes in fruit flies, to facilitate the phagocytic elimination of virus-infected cells. Inhibition of phagocytosis led to severe pathologies and death in both species, indicating that apoptosis-dependent phagocytosis of virus-infected cells is a conserved antiviral mechanism in multicellular organisms. Indeed, our understanding of the mechanisms underlying apoptosis-dependent phagocytosis of virus-infected cells has shed a new perspective on how hosts defend themselves against viral infection. This chapter explores the mechanisms of this process and its potential for developing new treatments for viral diseases.
Collapse
Affiliation(s)
- Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia.
| | - Youdiil Ophinni
- Division of Clinical Virology, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan
- Laboratory of Host Defense, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Akiko Shiratsuchi
- Center for Medical Education, Sapporo Medical University, Sapporo, Japan
- Division of Biological Function and Regulation, Graduate School of Medicine, Sapporo Medical University, Sapporo, Japan
| | | |
Collapse
|
3
|
Therapeutic potential of viral vectors that express venom peptides for neurological diseases. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
4
|
Kupffer Cells Survive Plasmodium berghei Sporozoite Exposure and Respond with a Rapid Cytokine Release. Pathogens 2018; 7:pathogens7040091. [PMID: 30477234 PMCID: PMC6313776 DOI: 10.3390/pathogens7040091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 01/08/2023] Open
Abstract
The liver stage of the Plasmodium life cycle features sporozoite traversal of the liver sinusoidal barrier through Kupffer cells (KCs) followed by invasion of hepatocytes. Little is known about the interaction of Plasmodium sporozoites with KCs, the liver-resident macrophages. Previous reports suggest KCs do not mount a pro-inflammatory response and undergo cell death following this interaction. Our work explores this interaction using primary rat KCs (PRKCs) and Plasmodium berghei sporozoites. We analyzed PRKC culture supernatants for markers of an immunological response through cytokine arrays. Additionally, cell wounding and death were assessed by monitoring lactate dehydrogenase (LDH) levels in these supernatants and by live/dead cell imaging. We found that PRKCs mount an immunological response to P. berghei sporozoites by releasing a diverse set of both pro- and anti-inflammatory cytokines, including IFNγ, IL-12p70, Mip-3α, IL-2, RANTES, IL-1α, IL-4, IL-5, IL-13, EPO, VEGF, IL-7, and IL-17α. We also observed no difference in LDH level or live/dead staining upon sporozoite exposure, suggesting that the KCs are not deeply wounded or dying. Overall, our data suggest that sporozoites may be actively modulating the KC's reaction to their presence and altering the way the innate immune system is triggered by KCs.
Collapse
|
5
|
Jarrett KE, Lee C, De Giorgi M, Hurley A, Gillard BK, Doerfler AM, Li A, Pownall HJ, Bao G, Lagor WR. Somatic Editing of Ldlr With Adeno-Associated Viral-CRISPR Is an Efficient Tool for Atherosclerosis Research. Arterioscler Thromb Vasc Biol 2018; 38:1997-2006. [PMID: 30026278 PMCID: PMC6202188 DOI: 10.1161/atvbaha.118.311221] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/27/2018] [Indexed: 01/23/2023]
Abstract
Objective- Atherosclerosis studies in Ldlr knockout mice require breeding to homozygosity and congenic status on C57BL6/J background, a process that is both time and resource intensive. We aimed to develop a new method for generating atherosclerosis through somatic deletion of Ldlr in livers of adult mice. Approach and Results- Overexpression of PCSK9 (proprotein convertase subtilisin/kexin type 9) is currently used to study atherosclerosis, which promotes degradation of LDLR (low-density lipoprotein receptor) in the liver. We sought to determine whether CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats-associated 9) could also be used to generate atherosclerosis through genetic disruption of Ldlr in adult mice. We engineered adeno-associated viral (AAV) vectors expressing Staphylococcus aureus Cas9 and a guide RNA targeting the Ldlr gene (AAV-CRISPR). Both male and female mice received either (1) saline, (2) AAV-CRISPR, or (3) AAV-hPCSK9 (human PCSK9)-D374Y. A fourth group of germline Ldlr-KO mice was included for comparison. Mice were placed on a Western diet and followed for 20 weeks to assess plasma lipids, PCSK9 protein levels, atherosclerosis, and editing efficiency. Disruption of Ldlr with AAV-CRISPR was robust, resulting in severe hypercholesterolemia and atherosclerotic lesions in the aorta. AAV-hPCSK9 also produced hypercholesterolemia and atherosclerosis as expected. Notable sexual dimorphism was observed, wherein AAV-CRISPR was superior for Ldlr removal in male mice, while AAV-hPCSK9 was more effective in female mice. Conclusions- This all-in-one AAV-CRISPR vector targeting Ldlr is an effective and versatile tool to model atherosclerosis with a single injection and provides a useful alternative to the use of germline Ldlr-KO mice.
Collapse
Affiliation(s)
- Kelsey E. Jarrett
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ciaran Lee
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Marco De Giorgi
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ayrea Hurley
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Baiba K. Gillard
- Houston Methodist Research Institute, Houston, TX 77030, USA
- Weill Cornell Medicine, New York, NY 10065, USA
| | - Alexandria M. Doerfler
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ang Li
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Henry J. Pownall
- Houston Methodist Research Institute, Houston, TX 77030, USA
- Weill Cornell Medicine, New York, NY 10065, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - William R. Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
6
|
Virus Infection and Death Receptor-Mediated Apoptosis. Viruses 2017; 9:v9110316. [PMID: 29077026 PMCID: PMC5707523 DOI: 10.3390/v9110316] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 02/07/2023] Open
Abstract
Virus infection can trigger extrinsic apoptosis. Cell-surface death receptors of the tumor necrosis factor family mediate this process. They either assist persistent viral infection or elicit the elimination of infected cells by the host. Death receptor-mediated apoptosis plays an important role in viral pathogenesis and the host antiviral response. Many viruses have acquired the capability to subvert death receptor-mediated apoptosis and evade the host immune response, mainly by virally encoded gene products that suppress death receptor-mediated apoptosis. In this review, we summarize the current information on virus infection and death receptor-mediated apoptosis, particularly focusing on the viral proteins that modulate death receptor-mediated apoptosis.
Collapse
|
7
|
Grakoui A, Crispe IN. Presentation of hepatocellular antigens. Cell Mol Immunol 2016; 13:293-300. [PMID: 26924525 PMCID: PMC4856799 DOI: 10.1038/cmi.2015.109] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/24/2015] [Accepted: 11/25/2015] [Indexed: 12/31/2022] Open
Abstract
The liver is an organ in which antigen-specific T-cell responses manifest a bias toward immune tolerance. This is clearly seen in the rejection of allogeneic liver transplants, and multiple other phenomena suggest that this effect is more general. These include tolerance toward antigens introduced via the portal vein, immune failure to several hepatotropic viruses, the lack of natural liver-stage immunity to malaria parasites, and the frequent metastasis of cancers to the liver. Here we review the mechanisms by which T cells engage with hepatocellular antigens, the context in which such encounters occur, and the mechanisms that act to suppress a full T-cell response. While many mechanisms play a role, we will argue that two important processes are the constraints on the cross-presentation of hepatocellular antigens, and the induction of negative feedback inhibition driven by interferons. The constant exposure of the liver to microbial products from the intestine may drive innate immunity, rendering the local environment unfavorable for specific T-cell responses through this mechanism. Nevertheless, tolerance toward hepatocellular antigens is not monolithic and under specific circumstances allows both effective immunity and immunopathology.
Collapse
Affiliation(s)
- Arash Grakoui
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine and Yerkes National Primate Research Center, Atlanta, GA, USA
| | | |
Collapse
|
8
|
Rogge M, Yin XT, Godfrey L, Lakireddy P, Potter CA, Del Rosso CR, Stuart PM. Therapeutic Use of Soluble Fas Ligand Ameliorates Acute and Recurrent Herpetic Stromal Keratitis in Mice. Invest Ophthalmol Vis Sci 2016; 56:6377-86. [PMID: 26444718 DOI: 10.1167/iovs.15-16588] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE The present study was designed to test the therapeutic value of soluble FasL (sFasL) in an acute model of herpetic stromal keratitis (HSK) and, more importantly, a recurrent model of HSK using BALB/c, BALB-lpr, and National Institutes of Health (NIH) mice. METHODS Mice were infected either acutely with the KOS strain of herpes simplex virus 1 (HSV-1) or latently with the McKrae strain of HSV-1. Acutely infected mice as well as ultraviolet-B (UV-B) reactivated mice (recurrent infection) were treated with sFasL, or soluble TNF-related apoptosis inducing ligand (sTRAIL), or BSA daily or 3 times/wk by using either a combination of subconjunctival injection and topical ointment, or with topical ointment alone. These mice then were evaluated for corneal opacity and neovascularization for 6 weeks. RESULTS Following acute and recurrent HSV-1 infection, wild-type BALB/c mice treated with sFasL displayed significantly reduced incidence of corneal opacity and neovascularization compared to the control animals. However, BALB-lpr mice, which are deficient in Fas+ inflammatory cells, displayed no such differences in ocular disease, as expected. Latently infected NIH mice treated with sFasL displayed similar results. Flow cytometric analysis revealed that the corneal inflammatory infiltrate in those treated with sFasL was significantly less than in sTRAIL- or BSA-treated mice. Furthermore, corneas from sFasL-treated mice displayed relatively more cells undergoing apoptosis. CONCLUSIONS This study provides evidence that sFasL treatment has potential therapeutic benefit in reducing inflammatory infiltrate and neovascularization in primary and recurrent forms of HSK, and that it does so by augmenting the restriction of Fas+ inflammatory cells mediated by membrane FasL.
Collapse
|
9
|
Regulation of tissue-dependent differences in CD8+ T cell apoptosis during viral infection. J Virol 2014; 88:9490-503. [PMID: 24942579 DOI: 10.1128/jvi.01223-14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Virus-specific CD8+ T cells in the lymphoid organs contract at the resolution of virus infections by apoptosis or by dissemination into peripheral tissues, and those residing in nonlymphoid organs, including the peritoneal cavity and fat pads, are more resistant to apoptosis than those in the spleen and lymph nodes. This stability of memory T cells in the nonlymphoid tissues may enhance protection to secondary challenges. Here, we show that lymphocytic choriomeningitis virus (LCMV)-specific CD8+ T cells in nonlymphoid tissues were enriched for memory precursors (expressing high levels of interleukin-7 receptor and low levels of killer cell lectin-like receptor G1 [IL-7Rhi KLRG1lo]) and had higher expression of CD27, CXCR3, and T cell factor-1 (TCF-1), each a marker that is individually correlated with decreased apoptosis. CD8+ T cells in the peritoneal cavity of TCF-1-deficient mice had decreased survival, suggesting a role for TCF-1 in promoting survival in the nonlymphoid tissues. CXCR3+ CD8+ T cells resisted apoptosis and accumulated in the lymph nodes of mice treated with FTY720, which blocks the export of lymph node cells into peripheral tissue. The peritoneal exudate cells (PEC) expressed increased amounts of CXCR3 ligands, CXCL9 and CXCL10, which may normally recruit these nonapoptotic cells from the lymph nodes. In addition, adoptive transfer of splenic CD8+ T cells into PEC or spleen environments showed that the peritoneal environment promoted survival of CD8+ T cells. Thus, intrinsic stability of T cells which are present in the nonlymphoid tissues along with preferential migration of apoptosis-resistant CD8+ T cells into peripheral sites and the availability of tissue-specific factors that enhance memory cell survival may collectively account for the tissue-dependent apoptotic differences. IMPORTANCE Most infections are initiated at nonlymphoid tissue sites, and the presence of memory T cells in nonlymphoid tissues is critical for protective immunity in various viral infection models. Virus-specific CD8+ T cells in the nonlymphoid tissues are more resistant to apoptosis than those in lymphoid organs during the resolution and memory phase of the immune response to acute LCMV infection. Here, we investigated the mechanisms promoting stability of T cells in the nonlymphoid tissues. This increased resistance to apoptosis of virus-specific CD8+ T cells in nonlymphoid tissues was due to several factors. Nonlymphoid tissues were enriched in memory phenotype CD8+ T cells, which were intrinsically resistant to apoptosis irrespective of the tissue environment. Furthermore, apoptosis-resistant CD8+ T cells preferentially migrated into the nonlymphoid tissues, where the availability of tissue-specific factors may enhance memory cell survival. Our findings are relevant for the generation of long-lasting vaccines providing protection at peripheral infection sites.
Collapse
|
10
|
The role of apoptosis in immune hyporesponsiveness following AAV8 liver gene transfer. Mol Ther 2013; 21:2227-35. [PMID: 24126962 DOI: 10.1038/mt.2013.94] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 03/28/2013] [Indexed: 12/13/2022] Open
Abstract
Gene therapy provides a significant opportunity to treat a variety of inherited and acquired diseases. However, adverse immune responses toward the adeno-associated virus (AAV) antigens may limit its success. The mechanisms responsible for immunity or tolerance toward AAV-encoded transgene products remain poorly defined. Studies in mice demonstrate that AAV2/8 gene transfer to liver is associated with immunological hyporesponsiveness toward both AAV vector and antigenic transgene product. To evaluate the role of activation-induced cell death (AICD) and cytokine withdrawal (intrinsic cell death) in the deletion of mature T lymphocytes, we compared immunological responses in hepatic AAV2/8 transfer in murine recipients lacking the Fas receptor, and recipients overexpressing Bcl-xL, to WT murine counterparts. Prolonged transgene expression was dependent on both Fas signaling and Bcl-xL-regulated apoptosis in T cells. Abrogation of intrinsic cell death enhanced Th1 responses, whereas AICD functioned to limit neutralizing antibody production toward AAV2/8. In addition, immune hyporesponsiveness and stable transgene expression was dependent on upregulation of FasL expression on transduced hepatocytes and a corresponding apoptosis of infiltrating Fas (+) cells. These data provide evidence that both AICD and apoptosis due to cytokine withdrawal of lymphocytes are essential for immune hyporesponsiveness toward hepatic AAV2/8-encoded transgene product in the setting of liver gene transfer.
Collapse
|
11
|
Viswanathan K, Bot I, Liu L, Dai E, Turner PC, Togonu-Bickersteth B, Richardson J, Davids JA, Williams JM, Bartee MY, Chen H, van Berkel TJC, Biessen EAL, Moyer RW, Lucas AR. Viral cross-class serpin inhibits vascular inflammation and T lymphocyte fratricide; a study in rodent models in vivo and human cell lines in vitro. PLoS One 2012; 7:e44694. [PMID: 23049756 PMCID: PMC3458838 DOI: 10.1371/journal.pone.0044694] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 08/10/2012] [Indexed: 12/25/2022] Open
Abstract
Poxviruses express highly active inhibitors, including serine proteinase inhibitors (serpins), designed to target host immune defense pathways. Recent work has demonstrated clinical efficacy for a secreted, myxomaviral serpin, Serp-1, which targets the thrombotic and thrombolytic proteases, suggesting that other viral serpins may have therapeutic application. Serp-2 and CrmA are intracellular cross-class poxviral serpins, with entirely distinct functions from the Serp-1 protein. Serp-2 and CrmA block the serine protease granzyme B (GzmB) and cysteine proteases, caspases 1 and 8, in apoptotic pathways, but have not been examined for extracellular anti-inflammatory activity. We examined the ability of these cross-class serpins to inhibit plaque growth after arterial damage or transplant and to reduce leukocyte apoptosis. We observed that purified Serp-2, but not CrmA, given as a systemic infusion after angioplasty, transplant, or cuff-compression injury markedly reduced plaque growth in mouse and rat models in vivo. Plaque growth was inhibited both locally at sites of surgical trauma, angioplasty or transplant, and systemically at non-injured sites in ApoE-deficient hyperlipidemic mice. With analysis in vitro of human cells in culture, Serp-2 selectively inhibited T cell caspase activity and blocked cytotoxic T cell (CTL) mediated killing of T lymphocytes (termed fratricide). Conversely, both Serp-2 and CrmA inhibited monocyte apoptosis. Serp-2 inhibitory activity was significantly compromised either in vitro with GzmB antibody or in vivo in ApoE/GzmB double knockout mice. Conclusions The viral cross-class serpin, Serp-2, that targets both apoptotic and inflammatory pathways, reduces vascular inflammation in a GzmB-dependent fashion in vivo, and inhibits human T cell apoptosis in vitro. These findings indicate that therapies targeting Granzyme B and/or T cell apoptosis may be used to inhibit T lymphocyte apoptosis and inflammation in response to arterial injury.
Collapse
Affiliation(s)
| | - Ilze Bot
- Division of Biopharmaceutics, Leiden/Amsterdam Center for Drug Research, Leiden, The Netherlands
- University of Maastracht, Maastracht, The Netherlands
| | - Liying Liu
- Vascular Biology Research Group, Robarts' Research Institute, London, Canada
- Department of Medicine, Divisions of Cardiovascular Medicine and Rheumatology, University of Florida, Gainesville, Florida, United States of America
| | - Erbin Dai
- Vascular Biology Research Group, Robarts' Research Institute, London, Canada
- Department of Medicine, Divisions of Cardiovascular Medicine and Rheumatology, University of Florida, Gainesville, Florida, United States of America
| | - Peter C. Turner
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Babajide Togonu-Bickersteth
- Vascular Biology Research Group, Robarts' Research Institute, London, Canada
- Department of Medicine, Divisions of Cardiovascular Medicine and Rheumatology, University of Florida, Gainesville, Florida, United States of America
| | - Jakob Richardson
- Vascular Biology Research Group, Robarts' Research Institute, London, Canada
| | - Jennifer A. Davids
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Jennifer M. Williams
- Department of Medicine, Divisions of Cardiovascular Medicine and Rheumatology, University of Florida, Gainesville, Florida, United States of America
| | - Mee Y. Bartee
- Department of Medicine, Divisions of Cardiovascular Medicine and Rheumatology, University of Florida, Gainesville, Florida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Hao Chen
- Department of Medicine, Divisions of Cardiovascular Medicine and Rheumatology, University of Florida, Gainesville, Florida, United States of America
| | - Theo J. C. van Berkel
- Division of Biopharmaceutics, Leiden/Amsterdam Center for Drug Research, Leiden, The Netherlands
- University of Maastracht, Maastracht, The Netherlands
| | - Erik A. L. Biessen
- Division of Biopharmaceutics, Leiden/Amsterdam Center for Drug Research, Leiden, The Netherlands
- University of Maastracht, Maastracht, The Netherlands
| | - Richard W. Moyer
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Alexandra R. Lucas
- Vascular Biology Research Group, Robarts' Research Institute, London, Canada
- Department of Medicine, Divisions of Cardiovascular Medicine and Rheumatology, University of Florida, Gainesville, Florida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
12
|
Morris JE, Zobell S, Yin XT, Zakeri H, Summers BC, Leib DA, Stuart PM. Mice with mutations in Fas and Fas ligand demonstrate increased herpetic stromal keratitis following corneal infection with HSV-1. THE JOURNAL OF IMMUNOLOGY 2011; 188:793-9. [PMID: 22156346 DOI: 10.4049/jimmunol.1102251] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
HSV-1 infection of the cornea leads to a potentially blinding immunoinflammatory lesion of the cornea, termed herpetic stromal keratitis. It has also been shown that one of the factors limiting inflammation of the cornea is the presence of Fas ligand (FasL) on corneal epithelium and endothelium. In this study, the role played by FasL expression in the cornea following acute infection with HSV-1 was determined. Both BALB/c and C57BL/6 (B6) mice with HSV-1 infection were compared with their lpr and gld counterparts. Results indicated that mice bearing mutations in the Fas Ag (lpr) displayed the most severe disease, whereas the FasL-defective gld mouse displayed an intermediate phenotype. It was further demonstrated that increased disease was due to lack of Fas expression on bone marrow-derived cells. Of interest, although virus persisted slightly longer in the corneas of mice bearing lpr and gld mutations, the persistence of infectious virus in the trigeminal ganglia was the same for all strains infected. Further, B6 mice bearing lpr and gld mutations were also more resistant to virus-induced mortality than were wild-type B6 mice. Thus, neither disease nor mortality correlated with viral replication in these mice. Collectively, the findings indicate that the presence of FasL on the cornea restricts the entry of Fas(+) bone marrow-derived inflammatory cells and thus reduces the severity of HSK.
Collapse
Affiliation(s)
- Jessica E Morris
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
|
14
|
Lee MJ, Jin YH, Kim K, Choi Y, Kim HC, Park S. Expression of hepatitis B virus x protein in hepatocytes suppresses CD8 T cell activity. Immune Netw 2010; 10:126-34. [PMID: 20844737 PMCID: PMC2939357 DOI: 10.4110/in.2010.10.4.126] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 07/20/2010] [Accepted: 07/23/2010] [Indexed: 01/07/2023] Open
Abstract
Background CD8+ T cells contribute to the clearance of Hepatitis B virus (HBV) infection and an insufficient CD8+ T cell response may be one of the major factors leading to chronic HBV infection. Since the HBx antigen of HBV can up-regulate cellular expression of several immunomodulatory molecules, we hypothesized that HBx expression in hepatocytes might affect CD8+ T cell activity. Methods We analyzed the activation and apoptosis of CD8+ T cells co-cultured with primary hepatocytes rendered capable of expressing HBx by recombinant baculovirus infection. Results Expression of HBx in hepatocytes induced low production of interferon-γ and apoptosis of CD8+ T cells, with no effect on CD8 T cell proliferation. However, transcriptional levels of H-2K, ICAM-1 and PD-1 ligand did not correlate with HBx expression in hepatocytes. Conclusion Our results suggest that HBx may inhibit CD8+ T cell response by regulation of interferon-γ production and apoptosis.
Collapse
Affiliation(s)
- Mi Jin Lee
- Department of Microbiology and Immunology, Ajou University School of Medicine, Suwon 442-721, Korea
| | | | | | | | | | | |
Collapse
|
15
|
Holst PJ, Ørskov C, Thomsen AR, Christensen JP. Quality of the transgene-specific CD8+ T cell response induced by adenoviral vector immunization is critically influenced by virus dose and route of vaccination. THE JOURNAL OF IMMUNOLOGY 2010; 184:4431-9. [PMID: 20212099 DOI: 10.4049/jimmunol.0900537] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adenoviral vectors have been widely used for experimental gene therapy and vaccination, yet there is a surprising lack of knowledge connecting the route and dose of adenovirus administration to the induced transgene-specific immune response. We have recently demonstrated polyfunctional CD8(+) T cells and protective memory responses using adenoviral vectors, which seem to contrast with recent reports suggesting that an exhausted CD8(+) T cell phenotype is induced by inoculation with adenoviral vectors. Accordingly, we investigated the route and dose interrelationship for transgene-specific CD8(+) T cells using adenoviral vectors encoding beta-galactosidase applied either s.c. or i.v. Irrespective of the route of inoculation, most of the adenoviral inoculum was found to disseminate systemically as the dose was raised beyond 10(9) particles. The number of transgene-specific CD8(+) T cells correlated positively with dissemination, whereas the functional capacity of the generated T cells correlated inversely with vector dissemination. A comparison of the immune response to s.c. or i.v. administration at moderate doses revealed that inoculation by both routes induced a transient peak of IFN-gamma-producing CD8(+) T cells 2 to 3 wk postinfection, but following i.v. administration, these cells were only detected in the liver. Two to four months after systemic, but not peripheral, immunization, dysfunctional transgene-specific CD8(+) T cells impaired in both cytokine production and important in vivo effector functions, accumulated in the spleen. These findings indicate that the localization of the adenoviral inoculum and not the total Ag load determines the quality of the CD8(+) T cell response induced with adenoviral vaccines.
Collapse
Affiliation(s)
- Peter Johannes Holst
- Department of International Health, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
16
|
Hepatic microenvironment programs hematopoietic progenitor differentiation into regulatory dendritic cells, maintaining liver tolerance. Blood 2008; 112:3175-85. [PMID: 18669892 DOI: 10.1182/blood-2008-05-159921] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The liver has been generally considered an organ prone to tolerance induction and maintenance. However, whether and how the unique liver microenvironment contributes to tolerance maintenance is largely unknown. Here, we used liver fibroblastic stromal cells to mimic the liver microenvironment and found that liver stroma could induce Lin(-)CD117(+) progenitors to differentiate into dendritic cells (DCs) with low CD11c, MHC II but high CD11b expression, high IL-10, but low IL-12 secretion. Such regulatory DCs could inhibit T-cell proliferation in vitro and in vivo, induce apoptosis of the activated T cells, and alleviate the damage of autoimmune hepatitis. Furthermore, liver stroma-derived macrophage colony-stimulating factor (M-CSF) was found to contribute to the generation of such regulatory DCs. Regulatory DC-derived PGE2 and T cell-derived IFN-gamma were responsible for the regulatory function. The natural counterpart of regulatory DCs was phenotypically and functionally identified in the liver. Importantly, Lin(-)CD117(+) progenitors could be differentiated into regulatory DCs in the liver once transferred into the liver. Infusion with liver regulatory DCs alleviated experimental autoimmune hepatitis. Therefore, we demonstrate that the liver microenvironment is highly important to program progenitors to differentiate into regulatory DCs in situ, which contributes to the maintenance of liver tolerance.
Collapse
|
17
|
Xia CQ, Qiu Y, Peng RH, Lo-Dauer J, Clare-Salzler MJ. Infusion of UVB-treated splenic stromal cells induces suppression of beta cell antigen-specific T cell responses in NOD mice. J Autoimmun 2008; 30:283-92. [PMID: 18226498 DOI: 10.1016/j.jaut.2007.11.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Revised: 11/28/2007] [Accepted: 11/29/2007] [Indexed: 02/02/2023]
Abstract
Our previous study has demonstrated that transfusion of UVB-irradiation-induced apoptotic beta cells effectively prevents type 1 diabetes (T1D) in non-obese diabetic (NOD) mice. However, the limitation of beta cell source would preclude the clinical application of this approach. Therefore, in the present study, we have attempted to establish a more practical approach by utilizing apoptotic non-beta cells to prevent T1D. We find that apoptotic splenic stromal cells significantly suppress beta cell antigen-reactive T cell proliferation in vitro and in vivo. Moreover, beta cell antigen-specific T cells primed by beta cell antigens in the presence of apoptotic stromal cells have markedly reduced responsiveness to the re-stimulation of the same beta cell antigen. We also find that beta cell antigen-specific IL-10-producing CD4+ T cells are induced in the presence of apoptotic splenic stromal cells. As expected, transfusion of apoptotic stromal cells effectively protected NOD mice from developing T1D. Furthermore, the proliferation of adoptively transferred beta cell antigen-specific TCR-transgenic T cells in pancreatic draining lymph nodes is markedly suppressed in UVB-stroma-treated mice, indicating that UVB-stroma treatment induces immune tolerance to multiple beta cell antigens. This study provides an effective and convenient approach for managing T1D by utilizing apoptotic non-beta cells.
Collapse
Affiliation(s)
- Chang-Qing Xia
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, P.O. Box 100275, 1600 SW Archer Road, Gainesville, FL 32610, United States.
| | | | | | | | | |
Collapse
|
18
|
Goldberg AC, Bittencourt PL, Oliveira LC, Ramasawmy R, Marin MLC, Palacios SA, Kalil J, Porta G. Autoimmune hepatitis in Brazil: an overview. Scand J Immunol 2007; 66:208-16. [PMID: 17635798 DOI: 10.1111/j.1365-3083.2007.01967.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autoimmune hepatitis is an immune cell-mediated chronic liver disease of unknown cause that leads, when untreated, to cirrhosis and liver failure. Importantly, this disease affects not only adults but children as well. Genetic susceptibility is clearly important and the major susceptibility factor identified up to now is the HLA-DRB1 locus, but other genes may play a role as well. HLA-DRB1 alleles present in South American patients differ from those found in patients in other parts of the world. In addition, we have recently identified two chromosomal regions where additional susceptibility factors may be found in Brazilian patients, namely, the class III MHC region and the 5q31 region where the IL-4 and IL-13 genes are located. This review discusses the current knowledge of the pathogenesis of this autoimmune disease occurring in the setting of an immune-privileged organ, the liver, and compares the data on gene polymorphisms studied in Brazil and in other parts of the world.
Collapse
Affiliation(s)
- A C Goldberg
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Polakos NK, Klein I, Richter MV, Zaiss DM, Giannandrea M, Crispe IN, Topham DJ. Early intrahepatic accumulation of CD8+ T cells provides a source of effectors for nonhepatic immune responses. THE JOURNAL OF IMMUNOLOGY 2007; 179:201-10. [PMID: 17579039 DOI: 10.4049/jimmunol.179.1.201] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Interactions between the liver and CD8+ T cells can lead to tolerance, due in part to CD8+ T cell death. To test whether this was the case in an extrahepatic infection, we investigated the fate and effector capacity of intrahepatic CD8+ T cells during lung-restricted influenza infection in mice. Virus-specific T cells accumulated in livers without detectable intrahepatic presentation of viral Ags, and this accumulation was not restricted to the contraction phase, but was apparent as early as day 5. Intrahepatic influenza-specific cells were functionally similar to those recovered from the bronchioalveolar lavage, based on ex vivo cytokine production and specific target lysis. Both adoptive transfer of liver lymphocytes and orthotopic liver transplant of organs containing accumulated effector T cells revealed that activated CD8s from the liver were viable, expanded during reinfection, and generated a memory population that trafficked to lymphoid organs. Thus, intrahepatic CD8+ T cells re-enter circulation and generate functional memory, indicating that the liver does not uniformly incapacitate activated CD8+ T cells. Instead, it constitutes a substantial reservoir of usable Ag-specific effector CD8+ T cells involved in both acute and recall immune responses.
Collapse
Affiliation(s)
- Noelle K Polakos
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Holst PJ, Orskov C, Qvortrup K, Christensen JP, Thomsen AR. CCR5 and CXCR3 are dispensable for liver infiltration, but CCR5 protects against virus-induced T-cell-mediated hepatic steatosis. J Virol 2007; 81:10101-12. [PMID: 17626099 PMCID: PMC2045423 DOI: 10.1128/jvi.01242-07] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
CCR5 and CXCR3 are important molecules in regulating the migration of activated lymphocytes. Thus, the majority of tissue-infiltrating T cells found in the context of autoimmune conditions and viral infections express CCR5 and CXCR3, and the principal chemokine ligands are expressed within inflamed tissues. Accordingly, intervention studies have pointed to nonredundant roles of these receptors in models of allograft rejection, viral infection, and autoimmunity. In spite of this, considerable controversy exists, with many studies failing to support a role for CCR5 or CXCR3 in disease pathogenesis. One possible explanation is that different chemokine receptors may take over in the absence of any individual receptor, thus rendering individual receptors redundant. We have attempted to address this issue by analyzing CCR5(-/-), CXCR3(-/-), and CCR5/CXCR3(-/-) mice with regard to virus-induced liver inflammation, generation and recruitment of effector cells, virus control, and immunopathology. Our results indicate that CCR5 and CXCR3 are largely dispensable for tissue infiltration and virus control. In contrast, the T-cell response is accelerated in CCR5(-/-) and CCR5/CXCR3(-/-) mice and the absence of CCR5 is associated with the induction of CD8(+) T-cell-mediated immunopathology consisting of marked hepatic microvesicular steatosis.
Collapse
Affiliation(s)
- P J Holst
- Institute of Medical Microbiology and Immunology, University of Copenhagen, The Panum Institute, 3C Blegdamsvej, DK-2200 Copenhagen N, Denmark
| | | | | | | | | |
Collapse
|
21
|
Holst PJ, Bartholdy C, Stryhn A, Thomsen AR, Christensen JP. Rapid and sustained CD4+ T-cell-independent immunity from adenovirus-encoded vaccine antigens. J Gen Virol 2007; 88:1708-1716. [PMID: 17485530 DOI: 10.1099/vir.0.82727-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Many novel vaccine strategies rely on recombinant viral vectors for antigen delivery, and adenovirus vectors have emerged among the most potent of these. In this report, we have compared the immune response induced through priming with adenovirus vector-encoded full-length viral protein to that elicited with an adenovirus-encoded minimal epitope covalently linked to β
2-microglobulin. We demonstrate that the β
2-microglobulin-linked epitope induced an accelerated and augmented CD8+ T-cell response. Furthermore, the immunity conferred by vaccination with β
2-microglobulin-linked lymphocytic choriomeningitis virus (LCMV)-derived epitopes was long-lived and protective. Notably, in contrast to full-length protein, the response elicited with the β
2-microglobulin-linked LCMV-derived epitope was CD4+ T-cell independent. Furthermore, virus-specific CD8+ T cells primed in the absence of CD4+ T-cell help were sustained in the long term and able to expand and control a secondary challenge with LCMV. Our results demonstrate that modifications to the antigen used in adenovirus vaccines may be used to improve the induced T-cell response. Such a strategy for CD4+ T-cell-independent immunity from adenovirus vectors offers prospects for vaccination against opportunistic pathogens in AIDS patients and possibly immunotherapy in chronic virus infections.
Collapse
Affiliation(s)
- Peter J Holst
- Institute of Medical Microbiology and Immunology, University of Copenhagen, The Panum Institute Building 22.5, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| | - Christina Bartholdy
- Institute of Medical Microbiology and Immunology, University of Copenhagen, The Panum Institute Building 22.5, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| | - Annette Stryhn
- Institute of Medical Microbiology and Immunology, University of Copenhagen, The Panum Institute Building 22.5, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| | - Allan R Thomsen
- Institute of Medical Microbiology and Immunology, University of Copenhagen, The Panum Institute Building 22.5, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| | - Jan P Christensen
- Institute of Medical Microbiology and Immunology, University of Copenhagen, The Panum Institute Building 22.5, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
22
|
Keating R, Yue W, Rutigliano JA, So J, Olivas E, Thomas PG, Doherty PC. Virus-specific CD8+ T cells in the liver: armed and ready to kill. THE JOURNAL OF IMMUNOLOGY 2007; 178:2737-45. [PMID: 17312116 DOI: 10.4049/jimmunol.178.5.2737] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Influenza A virus infection of C57BL/6 mice is a well-characterized model for studying CD8+ T cell-mediated immunity. Analysis of primary and secondary responses showed that the liver is highly enriched for CD8+ T cells specific for the immunodominant H2D(b)NP(366-374) (D(b)NP(366)) epitope. Functional analysis established that these liver-derived virus-specific CD8+ T cells are fully competent cytotoxic effectors and IFN-gamma secretors. In addition, flow cytometric analysis of early apoptotic cells showed that these influenza-specific CD8+ T cells from liver are as viable as those in the spleen, bronchoalveolar lavage, mediastinal lymph nodes, or lung. Moreover, cytokine profiles of the influenza-specific CD8+ T cells recovered from different sites were consistent with the bronchoalveolar lavage, rather than liver population, being the most susceptible to activation-induced cell death. Importantly, adoptively transferred influenza virus-specific CD8+ T cells from the liver survived and were readily recalled after virus challenge. Together, these results show clearly that the liver is not a "graveyard" for influenza virus-specific CD8+ T cells.
Collapse
Affiliation(s)
- Rachael Keating
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Kupffer cells (KC) constitute 80-90% of the tissue macrophages present in the body. They reside within the lumen of the liver sinusoids, and are therefore constantly exposed to gut-derived bacteria, microbial debris and bacterial endotoxins, known to activate macrophages. Upon activation KC release various products, including cytokines, prostanoides, nitric oxide and reactive oxygen species. These factors regulate the phenotype of KC themselves, and the phenotypes of neighboring cells, such as hepatocytes, stellate cells, endothelial cells and other immune cells that traffic through the liver. Therefore, KC are intimately involved in the liver's response to infection, toxins, ischemia, resection and other stresses. This review summarizes established basic concepts of KC function as well as their role in the pathogenesis of various liver diseases.
Collapse
Affiliation(s)
- Manfred Bilzer
- Department of Medicine II, University of Munich, Klinikum Grosshadern, Munich, Germany.
| | | | | |
Collapse
|
24
|
Kennedy R, Celis E. T helper lymphocytes rescue CTL from activation-induced cell death. THE JOURNAL OF IMMUNOLOGY 2006; 177:2862-72. [PMID: 16920921 PMCID: PMC1594817 DOI: 10.4049/jimmunol.177.5.2862] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
T cell activation is characterized by a vast expansion of Ag-specific T cells followed by an equally extensive reduction in T cell numbers. This decline is due, in part, to activation-induced apoptosis of the responding T cells during repeated encounter with Ag. In the current study, we used solid-phase MHC class I/peptide monomers to cause activation-induced cell death (AICD) of previously activated CD8 T cells in an Ag-specific manner. AICD occurred rapidly and was mediated primarily by Fas-FasL interactions. Most interestingly, we observed that Th cells could provide survival signals to CTL significantly reducing the level of AICD. Both Th1 and Th2 subsets were capable of protecting CTL from AICD, and a major role for soluble factors in this protection was ruled out, as cell-to-cell contact was an essential component of this Th-mediated protection. Upon encounter with Ag-expressing tumor cells, CTL underwent significant apoptosis. However, in the presence of Th cells, the CTL not only were protected against death, but also had significantly greater lytic ability. In vivo tumor protection studies using peptide immunization showed that the activation of Ag-specific Th cells was crucial for optimal protection, but did not affect the magnitude of the CTL response in the lymphoid tissues. In this study, we examine the type of help that CD4 T cells may provide and propose a model of Th cell-CTL interaction that reduces CTL death. Our results show a novel role for Th cells in the maintenance of CTL responses.
Collapse
Affiliation(s)
- Richard Kennedy
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | |
Collapse
|
25
|
Liu MS, Wang L, Zhao ZF, Zhang GY, Zhang Y, Feng JN. Construction of recombinant plasmid series with two Fas-targeted short hairpin RNAs. Shijie Huaren Xiaohua Zazhi 2006; 14:2174-2179. [DOI: 10.11569/wcjd.v14.i22.2174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct the recombinant plasmid expressing two Fas-targeted short hairpin RNA (shRNA) by pEGFP6-1 and pGenesil-1 plasmids vector.
METHODS: Two pairs of DNA sequences were designed, and then synthesized into complementary chains by annealing, respectively. Then the obtained products containing short hairpin structure were inserted into plasmid vector pEGFP6-1/pGenesil-1 with U6 promoter. The recombinant plasmids were transformed into Escherichia coli strain DH5a for screening and amplifying. The sequence analysis of the plasmids identified by restriction enzyme were carried out. The two plasmids were first digested with SacⅠ, and then with SalⅠ. A long segment from pEGFP6-1-siFas1 and a short segment from pGenesil-1-siFas2 were reclaimed, and connected with T4 DNA Ligase. The recombinant plasmid series was identified by restriction endonuclease digestion.
RESULTS: The two Fas-targeted shRNAs were successfully inserted into the plasmid vector pEGFP6-1, and the coding sequences of the obtained shRNAs were consistent with the designed fragments.
CONCLUSION: The recombinant plasmid series of two Fas-targeted shRNAs is successfully constructed in one pEGFP6-1 plasmid.
Collapse
|
26
|
Aloysius MM, Takhar A, Robins A, Eremin O. Dendritic cell biology, dysfunction and immunotherapy in gastrointestinal cancers. Surgeon 2006; 4:195-210. [PMID: 16892837 DOI: 10.1016/s1479-666x(06)80061-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Gastrointestinal (GI) cancers make up a significant proportion of newly diagnosed malignant disease. The five-year survival for these GI cancers is poor. Anti-cancer host defences are thought to play a role in these cancers, albeit they are suboptimal. Novel immunotherapies are being introduced to treat such patients. This review describes basic cell biology of dendritic cells, as they are thoughtto play a key role in generating effective anti-tumour responses. Dendritic cell dysfunction in patients with various cancers is documented and immunotherapy using dendritic cells in a range of GI cancers is described and discussed
Collapse
Affiliation(s)
- M M Aloysius
- Section of Surgery, University Hospital Nottingham, Queen's Medical Centre, Nottingham UK.
| | | | | | | |
Collapse
|
27
|
Abstract
Apoptosis or programmed cell death occurs in the liver as in other organs. In the normal state it is not a frequent mode of hepatic cell destruction. Morphological and biochemical characteristics of liver cell apoptosis do not differ from what is observed in other cells. The Fas receptor pathway, a frequent hepatic apoptotic pathway among various others, involves intra-cellular signals amplified by mitochondria. Although hepatic apoptosis may occur by following several others pathways, Fas, which is abundantly expressed in the plasma membrane of hepatocytes, is very often involved in hepatocyte demise during B or C viral hepatitis irrespective of their clinical form, alcoholic hepatitis, cholestasis due to accumulation of hepatic biliary salts, or certain types of drug-induced hepatitis. Fas is also probably responsible for the death of biliary cells in primary biliary cirrhosis. In contrast one of the causes of resistance to apoptosis of hepatic cancerous cells could be related to an alteration of the Fas receptor. This is why much experimental work is presently performed to achieve inhibition of the Fas receptor either at the mRNA level or at the level of Fas-inductible proteolytic enzymes called caspases. One perspective is a specific treatment of apoptosis as an adjuvant treatment of liver diseases.
Collapse
Affiliation(s)
- Gérard Feldmann
- INSERM U 773, Faculté de Médecine Xavier Bichat, Université Paris 7- Denis Diderot, Paris.
| |
Collapse
|
28
|
Dobrzynski E, Fitzgerald JC, Cao O, Mingozzi F, Wang L, Herzog RW. Prevention of cytotoxic T lymphocyte responses to factor IX-expressing hepatocytes by gene transfer-induced regulatory T cells. Proc Natl Acad Sci U S A 2006; 103:4592-7. [PMID: 16537361 PMCID: PMC1450216 DOI: 10.1073/pnas.0508685103] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Indexed: 11/18/2022] Open
Abstract
Treatment of genetic disease such as the bleeding disorder hemophilia B [deficiency in blood coagulation factor IX (F.IX)] by gene replacement therapy is hampered by the risk of immune responses to the therapeutic gene product and to the gene transfer vector. Immune competent mice of two different strains were tolerized to human F.IX by hepatic gene transfer mediated by adenoassociated viral vector. These animals were subsequently challenged by systemic administration of an E1/E3-deleted adenoviral vector, which is known to induce a cytotoxic T lymphocyte response to the transgene product. Immune tolerance prevented cytotoxic T lymphocyte activation to F.IX and CD8(+) cellular infiltrates in the liver. Moreover, a sustained and substantial increase in hepatic F.IX expression from the adenoviral vector was achieved despite in vitro T cell responses to adenoviral antigens. Cytolytic responses to therapeutic and to viral vector-derived antigens had been prevented in vivo by activation of regulatory CD4(+) T cells, which mediated suppression of inflammatory lymphocyte responses to the liver. This result suggests that augmentation of regulatory T cell activation should provide new means to avoid destructive immune responses in gene transfer.
Collapse
Affiliation(s)
- Eric Dobrzynski
- *Department of Pediatrics, The Children’s Hospital of Philadelphia and University of Pennsylvania Medical School, Philadelphia, PA 19104; and
| | - Julie C. Fitzgerald
- *Department of Pediatrics, The Children’s Hospital of Philadelphia and University of Pennsylvania Medical School, Philadelphia, PA 19104; and
| | - Ou Cao
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Alachua, FL 32615
| | - Federico Mingozzi
- *Department of Pediatrics, The Children’s Hospital of Philadelphia and University of Pennsylvania Medical School, Philadelphia, PA 19104; and
| | - Lixin Wang
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Alachua, FL 32615
| | - Roland W. Herzog
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Alachua, FL 32615
| |
Collapse
|
29
|
Chen J, Zajac AJ, McPherson SA, Hsu HC, Yang P, Wu Q, Xu X, Wang X, Fujihashi K, Curiel DT, Mountz JD. Primary adenovirus-specific cytotoxic T lymphocyte response occurs after viral clearance and liver enzyme elevation. Gene Ther 2005; 12:1079-88. [PMID: 15815704 DOI: 10.1038/sj.gt.3302494] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The virus-specific cytotoxic T lymphocyte (CTL) response is a major obstacle to effective delivery of adenovirus gene therapy. However, its relative role in viral clearance, transgene elimination and hepatotoxicity remains unclear. In this paper, we present an analysis of viral clearance and liver toxicity in relation to the induction of the virus-specific CD8 T-cell response revealed by an MHC class I tetramer. A surprisingly high number of tetramer+ CD8 T cells were found in the liver and lung and reached peak values at days 8 and 10, respectively, post-infection. Nearly 100% of these tetramer+ CD8 T cells expressed high levels of granzyme B and IFNgamma. Remarkably, liver viral load and liver enzyme elevation peaked early, at days 2 and 4, respectively, post-infection, before the specific CTL response was detectable. After generation of CTLs, there was only minimal liver damage or further decrease in virus titer. These results indicated that the primary peak response of tetramer+ CTLs does not correlate with the elimination of adenovirus or liver cytotoxic response.
Collapse
Affiliation(s)
- J Chen
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bosque A, Pardo J, Martínez-Lorenzo MJ, Lasierra P, Larrad L, Marzo I, Naval J, Anel A. Human CD8+ T cell blasts are more sensitive than CD4+ T cell blasts to regulation by APO2L/TRAIL. Eur J Immunol 2005; 35:1812-21. [PMID: 15884050 DOI: 10.1002/eji.200526046] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The mechanisms responsible for the down-modulation of the activation of separated CD4(+) or CD8(+) human T cell blasts were studied using cells obtained from healthy donors. In the presence of IL-2, human CD8(+) T cell blasts were more sensitive than CD4(+) T cell blasts to regulation by APO2 ligand/TNF-related apoptosis-inducing ligand (APO2L/TRAIL), while both T cell subsets were equally sensitive to Fas/CD95 regulation. This regulation was defined as inhibition of IL-2-dependent T cell growth in the absence of cell death induction, characterized by cell cycle arrest in G(2)/M. The physiological validity of these observations was corroborated by the demonstration of intracellular FasL and APO2L/TRAIL expression in CD4(+) and CD8(+) T cell blasts, which were secreted in their bioactive form into the supernatant upon PHA, CD3 or CD59 reactivation. Additionally, the inhibition of IL-2-dependent CD4(+) or CD8(+) T cell blast growth upon CD3 or CD59 ligation was dependent, at least partially, on FasL and/or APO2L/TRAIL. These data precisely define the role of APO2L/TRAIL in the regulation of human T cell activation.
Collapse
Affiliation(s)
- Alberto Bosque
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Sosa L, Vidlak D, Strachota JM, Pavlik J, Jerrells TR. Rescue of in vivo FAS-induced apoptosis of hepatocytes by corticosteroids either associated with alcohol consumption by mice or provided exogenously. Int Immunopharmacol 2005; 5:301-14. [PMID: 15652761 DOI: 10.1016/j.intimp.2004.09.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2004] [Accepted: 09/30/2004] [Indexed: 12/20/2022]
Abstract
The pathogenic effects of many hepatic viral infections are mediated, at least in part, by the immune response to the infected hepatocyte. The immune response in the infected liver involves the interaction of cytotoxic T cells (CTL) with the hepatocytes through the interaction of FAS-ligand on the CTL and FAS on the hepatocyte. The initial hypothesis for this study was that alcohol consumption by mice would sensitize the liver to apoptosis induced by ligation of FAS. C57Bl/6 mice fed ethanol in a liquid diet did show an increased percentage of apoptotic cells 2 h after injection with anti-FAS as compared with the percentage in the control mice. However, 4 and 6 h after anti-FAS injection, control mice showed high percentages of apoptotic cells (20% to 41%) compared with 5% and 4% apoptotic cells in the ethanol-fed mice. The decreased apoptosis of ethanol-fed mice correlated closely with corticosterone levels in the sera. This was confirmed by the finding that adrenalectomized (ADX) mice provided a high level of corticosterone in drinking water were protected against FAS-induced hepatocyte apoptosis. Ethanol-fed mice showed a significant elevation of serum alanine aminotransferase (ALT) levels indicating the development of hepatitis in spite of the relatively low proportion of apoptotic cells in the liver. In conclusion, high levels of corticosterone protect hepatocytes from FAS-mediated apoptosis, but do not prevent the ultimate development of liver damage. In experiments where mice were provided ethanol chronically in drinking water, where stress is minimal, higher levels of ALT were noted in animals in the ethanol group as compared with animals in the control group. These data support the suggestion that ethanol increases hepatocyte sensitivity to FAS-mediated damage.
Collapse
Affiliation(s)
- Laura Sosa
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | | | | | | | | |
Collapse
|
32
|
Yarovinsky TO, Mohning MP, Bradford MA, Monick MM, Hunninghake GW. Increased sensitivity to staphylococcal enterotoxin B following adenoviral infection. Infect Immun 2005; 73:3375-84. [PMID: 15908364 PMCID: PMC1111844 DOI: 10.1128/iai.73.6.3375-3384.2005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcal enterotoxin B induces toxic shock and is a major virulence factor of staphylococcal diseases. We examined the effects of systemic adenoviral infection on responses to staphylococcal enterotoxin B in a murine model. We found that adenoviral infection markedly increases the severity of liver injury following exposure to staphylococcal enterotoxin B without d-galactosamine sensitization. In adenovirus-infected mice, staphylococcal enterotoxin B triggered a more profound hypothermia and increased apoptosis in the liver. Consistent with these observations, we also found that adenoviral infection primed for an increased production of gamma interferon in vivo and in vitro following stimulation with staphylococcal enterotoxin B. Gamma-interferon-knockout mice did not show increased sensitivity to staphylococcal enterotoxin B following adenoviral infection. These data suggest that a preexisting viral infection primes mice for subsequent staphylococcal enterotoxin B exposure, possibly via a gamma-interferon-mediated mechanism.
Collapse
Affiliation(s)
- Timur O Yarovinsky
- Division of Pulmonary, Critical Care, and Occupational Medicine, 100 EMRB, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA.
| | | | | | | | | |
Collapse
|
33
|
Ferreira PG, Costa-E-Silva A, Oliveira MJR, Monteiro E, Aguas AP. Leukocyte-hepatocyte interaction in calicivirus infection: differences between rabbits that are resistant or susceptible to rabbit haemorrhagic disease (RHD). Vet Immunol Immunopathol 2005; 103:217-21. [PMID: 15621308 DOI: 10.1016/j.vetimm.2004.09.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2003] [Revised: 09/06/2004] [Accepted: 09/17/2004] [Indexed: 11/16/2022]
Abstract
Calicivirus infection is lethal for adult rabbits, whereas young rabbits (less than 8-weeks-old) are resistant to the same infectious agent. The virus replicates in the liver and causes a fulminant hepatitis in adult rabbits leading to rabbit haemorrhagic disease (RHD); this is in contrast with the mild and transient hepatitis observed in infected young rabbits. We have used electron microscopy to compare liver leukocyte infiltrates between young (resistant) and adult (susceptible) rabbits, 36-48 h after inoculation of the animals with caliciviruses. In adult rabbits, liver infiltrates were made up mostly of heterophils, and they were located near hepatocytes showing severe cellular damage. In contrast, liver leukocyte infiltrates of RHD-resistant young rabbits were dominated by lymphocytes that depicted membrane contacts with the cell surface of undamaged hepatocytes. We conclude that: (i) the cellular inflammatory response of the liver to calicivirus infection is different in rabbits that are susceptible (adult) or resistant (young) to RHD; (ii) leukocyte infiltration of the adult liver by heterophils is probably directed at the removal of dead hepatocytes, whereas the liver lymphocytic infiltration of young rabbits suggests the expression of viral antigens on the surface of liver cells of the RHD-resistant animals.
Collapse
Affiliation(s)
- P G Ferreira
- Department of Anatomy, Abel Salazar Institute for Biomedical Science (ICBAS), Unit for Multidisciplinary for Biomedical Research (UMIB), Largo Prof. Abel Salazar, 2, 4099-003 Porto, Portugal.
| | | | | | | | | |
Collapse
|
34
|
Burra P, Samuel D, Wendon J, Pietrangelo A, Gupta S. Strategies for liver support: from stem cells to xenotransplantation. J Hepatol 2004; 41:1050-9. [PMID: 15582142 DOI: 10.1016/j.jhep.2004.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Patrizia Burra
- Department of Surgical and Gastroenterological Sciences, University Hospital, Padova, Italy.
| | | | | | | | | |
Collapse
|
35
|
Engelmark MT, Renkema KY, Gyllensten UB. No evidence of the involvement of the Fas -670 promoter polymorphism in cervical cancer in situ. Int J Cancer 2004; 112:1084-5. [PMID: 15316939 DOI: 10.1002/ijc.20515] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
36
|
Murray DA, Crispe IN. TNF-α Controls Intrahepatic T Cell Apoptosis and Peripheral T Cell Numbers. THE JOURNAL OF IMMUNOLOGY 2004; 173:2402-9. [PMID: 15294953 DOI: 10.4049/jimmunol.173.4.2402] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
At the end of an immune response, activated lymphocyte populations contract, leaving only a small memory population. The deletion of CD8(+) T cells from the periphery is associated with an accumulation of CD8(+) T cells in the liver, resulting in both CD8(+) T cell apoptosis and liver damage. After adoptive transfer and in vivo activation of TCR transgenic CD8(+) T cells, an increased number of activated CD8(+) T cells was observed in the lymph nodes, spleen, and liver of mice treated with anti-TNF-alpha. However, caspase activity was decreased only in CD8(+) T cells in the liver, not in those in the lymphoid organs. These results indicate that TNF-alpha is responsible for inducing apoptosis in the liver and suggest that CD8(+) T cells escaping this mechanism of deletion can recirculate into the periphery.
Collapse
Affiliation(s)
- Debbie A Murray
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY 14642, USA
| | | |
Collapse
|
37
|
Cao Q, Xia Y, Azadniv M, Crispe IN. The E2F-1 Transcription Factor Promotes Caspase-8 and Bid Expression, and Enhances Fas Signaling in T Cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:1111-7. [PMID: 15240700 DOI: 10.4049/jimmunol.173.2.1111] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The immune system depends on the extensive proliferation of rare Ag-specific precursor T lymphocytes, followed by their differentiation, the delivery of effector function, and finally death by apoptosis. T cells that lack the E2F-1 transcription factor, which is activated as cells pass the restriction point and enter S phase, show defects in activation-induced cell death. We now report that E2F-1 increases the activity of an apoptotic pathway that is important in murine primary T cells. Thus, E2F-1 promotes the transcription of Bid, a molecule that links death receptor signaling to the activation of apoptotic mechanisms in mitochondria. It also promotes the transcription of caspase-8, the enzyme that cleaves and activates Bid. Enforced expression of Bid can partially restore apoptosis in E2F-1-deficient T cells. Thus, E2F-1 integrates cell cycle progression with apoptosis.
Collapse
Affiliation(s)
- Qingyu Cao
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
38
|
Dong H, Zhu G, Tamada K, Flies DB, van Deursen JMA, Chen L. B7-H1 determines accumulation and deletion of intrahepatic CD8(+) T lymphocytes. Immunity 2004; 20:327-36. [PMID: 15030776 DOI: 10.1016/s1074-7613(04)00050-0] [Citation(s) in RCA: 318] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2003] [Revised: 01/16/2004] [Accepted: 02/04/2004] [Indexed: 02/06/2023]
Abstract
Upon systemic activation by antigens, CD8(+), but not CD4(+), T cells selectively accumulate and undergo apoptosis in the liver, a mechanism associated with the induction of hepatic tolerance and chronic infection. The molecular basis for CD8(+) T cell preference in this process is unknown. We prepared B7-H1-deficient mice by gene targeting and found spontaneous accumulation of CD8(+) T cells in the liver while CD4(+) T cell levels remained normal. Moreover, antigen-driven CD8(+) T cells proliferated normally while apoptotic levels during the contraction phase was selectively impaired in the liver, leading to accelerated hepatocyte damage in experimental autoimmune hepatitis. Therefore, B7-H1 is a key protein selectively regulating the accumulation and deletion of intrahepatic CD8(+) T cells and may also contribute to inflammation, autoimmune diseases, and tolerance in the liver.
Collapse
Affiliation(s)
- Haidong Dong
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
39
|
Tai KF, Chen DS, Hwang LH. Curative potential of GM-CSF-secreting tumor cell vaccines on established orthotopic liver tumors: Mechanisms for the superior antitumor activity of live tumor cell vaccines. J Biomed Sci 2004; 11:228-38. [PMID: 14966373 DOI: 10.1007/bf02256566] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2003] [Accepted: 10/28/2003] [Indexed: 11/28/2022] Open
Abstract
In preclinical studies, tumor cells genetically engineered to secrete cytokines, hereafter referred to as tumor cell vaccines, can often generate systemic antitumor immunity. This study investigated the therapeutic effects of live or irradiated tumor cell vaccines that secrete granulocyte-macrophage colony-stimulating factor (GM-CSF) on established orthotopic liver tumors. Experimental results indicated that two doses (3 x 10(7) cells per dose) of irradiated tumor cell vaccines were therapeutically ineffective, whereas one dose (3 x 10(6) cells) of live tumor cell vaccines caused complete tumor regression. In vivo depletion of CD8+ T cells, but not natural killer cells, restored tumor formation in the live vaccine-treated animals. Additionally, the treatment of cells with live vaccine induced markedly higher levels of cytotoxic T lymphocyte activity than the irradiated vaccines in the draining lymph nodes. The higher levels of cytokine and antigen loads could partly explain the superior antitumor activity of live tumor cell vaccines, but other unidentified mechanisms could also play a role in the early T cell activation in the lymph nodes. A protocol using multiple and higher dosages of irradiated tumor cell vaccines also caused significant regression of liver tumors. These results suggest that the GM-CSF-secreting tumor cell vaccines are highly promising for orthotopic liver tumors if higher levels of immune responses are elicited during early tumor development.
Collapse
Affiliation(s)
- Kuo-Feng Tai
- Graduate Institute of Microbiology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | | | | |
Collapse
|
40
|
Ruggieri A, Murdolo M, Rapicetta M. Induction of FAS ligand expression in a human hepatoblastoma cell line by HCV core protein. Virus Res 2004; 97:103-10. [PMID: 14602201 DOI: 10.1016/j.virusres.2003.08.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Tumour cells and virus infected cells expressing Fas ligand (FasL) can evade immune surveillance by inducing apoptosis in T cells expressing Fas. In order to characterise a possible role of hepatitis C virus (HCV) core protein in similar mechanisms during HCV infection, we investigated Fas ligand expression and activity in a human hepatoblastoma cell line (HepG2) constitutively expressing this protein. Strong FasL induction was detected by immunoblotting and flow cytometry analysis in the core expressing cell lines Hep39. In contrast, vector transfected cells or cell lines expressing HCV E1-E2 proteins did not show FasL expression. Co-cultivation experiments of Hep39 cells with a Fas-sensitive T cell line indicated that FasL induced by the core protein had apoptotic activity toward target cells. Effect of the core protein on induction of FasL promoter was further examined by co-transfection of HepG2 cells with core-bearing plasmid and a vector in which luciferase gene expression is driven by human FasL promoter. Results of the luciferase assay indicated a positive regulation of FasL promoter by the core protein. In conclusion, HCV core protein plays a role in the induction of functional FasL in hepatoblastoma cell line and apoptosis in a target T cell line expressing Fas. Similar mechanisms may contribute, in vivo, to establishment of chronic infection and development of hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
- A Ruggieri
- Laboratory of Virology, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | | | | |
Collapse
|
41
|
Pham BN, Martinot-Peignoux M, Valla D, Dubois S, Degott C, Mosnier JF. Differential expression of perforin and granzyme B in the liver of patients with chronic hepatitis C. Hum Pathol 2003; 34:770-7. [PMID: 14506637 DOI: 10.1016/s0046-8177(03)00244-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
T lymphocytes have been reported to be the predominant inflammatory cells in the liver of patients with chronic hepatitis C. On activation, CD8+ T lymphocytes can exert their cytolytic activity by releasing their granule components, notably perforin and granzyme B. The aim of the present study was to assess whether the granule cytolytic pathway was used by liver-infiltrating CD8+ T lymphocytes. Immunostaining for perforin and granzyme B was performed in 25 patients with chronic hepatitis C, according to the disease activity and their virologic status. Cells stained for perforin and for granzyme B represented 0.15% and 0.10% of the total liver-infiltrating CD8+ T lymphocytes, respectively. Perforin was expressed mainly by activated CD8+ T lymphocytes located in liver lobules. In contrast, granzyme B was expressed mainly by activated CD8+ T lymphocytes located in interface hepatitis and portal tracts. The results were similar in the different groups of patients, whatever the disease activity. In conclusion, this is the first study showing a differential expression of granule components of CD8+ T lymphocytes in the same tissue in vivo. Perforin and granzyme B may be differently expressed by liver-infiltrating CD8+ T lymphocytes, according to their localization in the different specific compartments of the liver, in patients with chronic hepatitis C.
Collapse
Affiliation(s)
- Bach-Nga Pham
- Service d'Hématologie et Immunologie, Unité de Recherche INSERM U481, Clichy, France
| | | | | | | | | | | |
Collapse
|
42
|
Wang XZ, Stepp SE, Brehm MA, Chen HD, Selin LK, Welsh RM. Virus-specific CD8 T cells in peripheral tissues are more resistant to apoptosis than those in lymphoid organs. Immunity 2003; 18:631-42. [PMID: 12753740 DOI: 10.1016/s1074-7613(03)00116-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
CD8 T cells persist at high frequencies in peripheral organs after resolution of an immune response, and their presence in the periphery is important for resistance to secondary challenge. We show here that LCMV-specific T cells in peripheral tissue (peritoneal cavity, lung, fat pads) reacted much less with the apoptotic marker Annexin-V than those in spleen and lymph nodes. This was not due to a TCR-based selection. In comparison to lymphoid tissue, T cells in the periphery expressed lower levels of Fas and Fas ligand and were resistant to activation-induced cell death in vitro. This may contribute to the survival of nondividing peripheral memory T cells, enabling them to efficiently function without being driven into apoptosis.
Collapse
Affiliation(s)
- Xiaoting Z Wang
- Department of Pathology, University of Massachusetts Medical School, Worcester 01655, USA
| | | | | | | | | | | |
Collapse
|
43
|
Lai HC, Sytwu HK, Sun CA, Yu MH, Yu CP, Liu HS, Chang CC, Chu TY. Single nucleotide polymorphism at Fas promoter is associated with cervical carcinogenesis. Int J Cancer 2003; 103:221-5. [PMID: 12455036 DOI: 10.1002/ijc.10800] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The causal role of human papillomavirus (HPV) in cervical carcinogenesis is beyond reasonable questioning. The progression from HPV infection, squamous intraepithelial lesions (SIL) to squamous cell carcinomata (SCC), however, is very uncommon and inefficient. Host genetic factors that may confer the susceptibility of disease progression are largely unknown. Apoptosis is an important fail-safe check for tumor development, in which Fas/FasL interaction contributes substantially. The purpose of our study is to test the hypothesis that an A/G polymorphism at -670 of Fas promoter with different transcriptional activity is associated with the risk for cervical neoplasia. A hospital-based case-control study was conducted, in which 104 patients of low grade SIL (LSIL), 131 high grade SIL (HSIL) and 176 SCC as well as age-matched, 1:1 controls were tested for Fas polymorphism by PCR-RFLP. HPV genotypes were determined in case groups by MY PCR-reverse line blot. The frequency of A allele was significantly (p = 0.006) higher in SCC than in control, conferring an odd ratio of 1.5 (95% CI = 1.1-2.0). The distribution of Fas (-670) genotypes also differed significantly between HSIL, SCC and each of their control (p = 0.017 and 0.03, respectively), with the A/A genotype conferring an OR of 1.3 (95% CI = 1.1-1.6) and 1.6 (95% CI = 1.0-2.5), respectively. Remarkably, the frequency of A allele and A/A genotype increased gradually in accordance with the multi-step carcinogenesis from LSIL, HSIL to SCC (p(test for trend) = 0.0066 and 0.0007, respectively). In addition, there was no difference of Fas genotypes between HPV (+) and HPV (-) cases. Fas genotypes, however, differed in LSIL infected with different HPV types (p = 0.033). The present study demonstrated an association between Fas polymorphism and cervical carcinogenesis. We deduced a possible effect of apoptosis of immune cells in this virus-induced cancer.
Collapse
Affiliation(s)
- Hung-Cheng Lai
- Graduate Institutes of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
The T-cell biology of the liver is unlike that of any other organ. The local lymphocyte population is enriched in natural killer (NK) and NKT cells, which might have crucial roles in the recruitment of circulating T cells. A large macrophage population and the efficient trafficking of dendritic cells from sinusoidal blood to lymph promote antigen trapping and T-cell priming, but the local presentation of antigen causes T-cell inactivation, tolerance and apoptosis. These local mechanisms might result from the need to maintain immunological silence to harmless antigenic material in food. The overall bias of intrahepatic T-cell responses towards tolerance might account for the survival of liver allografts and for the persistence of some liver pathogens.
Collapse
Affiliation(s)
- Ian Nicholas Crispe
- The David H Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, The University of Rochester, Rochester, New York 14642, USA.
| |
Collapse
|
45
|
Klugewitz K, Blumenthal-Barby F, Schrage A, Knolle PA, Hamann A, Crispe IN. Immunomodulatory effects of the liver: deletion of activated CD4+ effector cells and suppression of IFN-gamma-producing cells after intravenous protein immunization. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:2407-13. [PMID: 12193708 DOI: 10.4049/jimmunol.169.5.2407] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The liver is tolerogenic in many situations, including as an allograft and during the response to allogeneic MHC expressed on hepatocytes. The majority of data that address this issue focus on endogenous Ags. Little is known about CD4(+) T cells and their fate under tolerizing conditions, especially with respect to fully differentiated CD4(+) effector T cells. In this study, we used the adoptive transfer of populations of TCR-transgenic CD4(+) T cells, skewed toward the Th1 or Th2 phenotype, to test whether either apoptotic or immune deviation mechanisms apply to cytokine-producing CD4(+) T cells that enter the liver. After transfer, Th1 and Th2 cells could be detected up to 25 days in lymphoid organs and the liver. Intravenous high dose Ag application resulted in accumulation, proliferation, and subsequent deletion of effector cells within the liver. Th1 cells lost their capacity to produce cytokines, whereas IL-4 expression was sustained within Th2 cells from the liver. However, there was no evidence for a deviation of Th1-programmed cells toward a Th2 (IL-4) or regulatory T cell (IL-10) pattern of cytokine expression. We used isolated populations of liver-derived APCs to test whether the liver had the capacity to impose a bias toward IL-4 expression in T cells. These experiments showed that liver sinusoidal endothelial cells selectively suppress the expansion of IFN-gamma-producing cells, yet they promote the outgrowth of IL-4-expressing Th2 cells, creating an immune suppressive milieu within this organ. These data suggest that presentation of Ags in the liver leads to modulation of immune response in terms of quantity and quality.
Collapse
Affiliation(s)
- Katja Klugewitz
- Section of Immunobiology, Yale University Medical School, New Haven, CT 06520, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Gregory SH, Wing EJ. Neutrophil‐Kupffer cell interaction: a critical component of host defenses to systemic bacterial infections. J Leukoc Biol 2002. [DOI: 10.1189/jlb.72.2.239] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Stephen H. Gregory
- Department of Medicine, Rhode Island Hospital and Brown University School of Medicine, Providence
| | - Edward J. Wing
- Department of Medicine, Rhode Island Hospital and Brown University School of Medicine, Providence
| |
Collapse
|
47
|
Zuñiga E, Motran CC, Montes CL, Yagita H, Gruppi A. Trypanosoma cruzi infection selectively renders parasite-specific IgG+ B lymphocytes susceptible to Fas/Fas ligand-mediated fratricide. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3965-73. [PMID: 11937553 DOI: 10.4049/jimmunol.168.8.3965] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The control of B cell expansion has been thought to be solely regulated by T lymphocytes. We show in this study that Trypanosoma cruzi infection induces up-regulation of both Fas and Fas ligand (FasL) molecules on B cells and renders them susceptible to B cell-B cell killing (referred to as fratricide throughout this paper) mediated via Fas/FasL. Moreover, by in vivo administration of anti-FasL blocking mAb we demonstrate that Fas-mediated B cell apoptosis is an ongoing process during this parasitic infection. We also provide evidence that B cells that have switched to IgG isotype are the preferential targets of B cell fratricide. More strikingly, this death pathway selectively affects IgG(+) B cells reactive to parasite but not self Ags. Parasite-specific but not self-reactive B cells triggered during this response are rescued after either in vitro or in vivo FasL blockade. Fratricide among parasite-specific IgG(+) B lymphocytes could impair the immune control of T. cruzi and possibly other chronic protozoan parasites. Our results raise the possibility that the blockade of Fas/FasL interaction in the B cell compartment of T. cruzi-infected mice may provide a means for enhancing antiparasitic humoral immune response without affecting host tolerance.
Collapse
Affiliation(s)
- Elina Zuñiga
- Department of Clinical Biochemistry, Faculty of Chemical Science, National University of Cordoba, Cordoba, Argentina
| | | | | | | | | |
Collapse
|
48
|
Jendro MC, Köhler L, Kuipers JG, Zeidler H. Microbe-induced T cell apoptosis: subversion of the host defense system? FEMS Microbiol Lett 2002; 207:121-6. [PMID: 11958928 DOI: 10.1111/j.1574-6968.2002.tb11039.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Cells of multicellular organisms are equipped with a self destruction program called apoptosis to ensure homeostasis of the organism. Contraction of the lymphocyte compartment following recovery from an infection is controlled by this mechanism. But apoptosis of lymphocytes might be an Achilles tendon accessible to microbes to subvert the immune system. Evidence is cumulating that microbes use this mechanism to destroy microbe-specific T cells. We present an overview of microbe-induced T cell apoptosis discussing the consequences for the pathogenesis of microbial infection. The conventional role of lymphocytes during infection is to impose apoptotic threat to infected cells, the subject of this review highlights the opposite, lymphocytes as targets of microbe-induced death.
Collapse
Affiliation(s)
- Michael C Jendro
- Department of Rheumatology, Medical School Hannover, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | | | | | | |
Collapse
|
49
|
Abstract
The liver has emerged as an organ with distinct immunological properties. In this review, we summarize evidence that shows that the liver can remove apoptotic, or non-apoptotic but activated, CD8+ T cells from the circulation and induce apoptosis in these activated T cells by either active or passive mechanisms. Hepatitis viruses, particularly hepatitis C virus, often establish persistent infection. We review evidence that suggests that these viruses exploit intrahepatic tolerance mechanisms to protect themselves from immune attack.
Collapse
Affiliation(s)
- Sun Park
- The David H Smith Center for VaccineBiology and Immunology, The University of Rochester, Rochester, New York 14642, USA
| | | | | | | |
Collapse
|
50
|
Liu ZX, Nishida H, He JW, Lai MMC, Feng N, Dennert G. Hepatitis C virus genotype 1b core protein does not exert immunomodulatory effects on virus-induced cellular immunity. J Virol 2002; 76:990-7. [PMID: 11773374 PMCID: PMC135789 DOI: 10.1128/jvi.76.3.990-997.2002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The hepatitis C virus (HCV) core protein is among the most conserved proteins in HCV and is known to induce sensitization of cytotoxic T lymphocytes (CTL). Therefore, it is a prime candidate for a component of a potential HCV vaccine. The HCV core protein has, however, been reported to exert multiple effects on cell functions, raising questions as to its suitability for this purpose. This question was investigated here with mice into which replication-deficient adenoviruses expressing core protein of an HCV genotype 1b isolate were injected. We show that induction of cytokines in response to the infection, infiltration of lymphocytes into the infected liver, priming of virus-specific CTL, and liver injury are not modulated by expression of the core protein in the liver. Moreover, no changes in the sensitivity to tumor necrosis factor alpha- or Fas-mediated liver injury are demonstrable. A similar lack of demonstrable effects of the core protein on immune functions has also been obtained using transgenic mice expressing another HCV genotype 1b core protein. It is concluded that the HCV core protein of genotype 1b has no modulatory effects on induction of virus-specific immune responses and may therefore be a suitable component of an HCV vaccine.
Collapse
Affiliation(s)
- Zhang-Xu Liu
- Department of Molecular Microbiology and Immunology, USC/Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, USA
| | | | | | | | | | | |
Collapse
|