1
|
Roe JM, Seely K, Bussard CJ, Eischen Martin E, Mouw EG, Bayles KW, Hollingsworth MA, Brooks AE, Dailey KM. Hacking the Immune Response to Solid Tumors: Harnessing the Anti-Cancer Capacities of Oncolytic Bacteria. Pharmaceutics 2023; 15:2004. [PMID: 37514190 PMCID: PMC10384176 DOI: 10.3390/pharmaceutics15072004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Oncolytic bacteria are a classification of bacteria with a natural ability to specifically target solid tumors and, in the process, stimulate a potent immune response. Currently, these include species of Klebsiella, Listeria, Mycobacteria, Streptococcus/Serratia (Coley's Toxin), Proteus, Salmonella, and Clostridium. Advancements in techniques and methodology, including genetic engineering, create opportunities to "hijack" typical host-pathogen interactions and subsequently harness oncolytic capacities. Engineering, sometimes termed "domestication", of oncolytic bacterial species is especially beneficial when solid tumors are inaccessible or metastasize early in development. This review examines reported oncolytic bacteria-host immune interactions and details the known mechanisms of these interactions to the protein level. A synopsis of the presented membrane surface molecules that elicit particularly promising oncolytic capacities is paired with the stimulated localized and systemic immunogenic effects. In addition, oncolytic bacterial progression toward clinical translation through engineering efforts are discussed, with thorough attention given to strains that have accomplished Phase III clinical trial initiation. In addition to therapeutic mitigation after the tumor has formed, some bacterial species, referred to as "prophylactic", may even be able to prevent or "derail" tumor formation through anti-inflammatory capabilities. These promising species and their particularly favorable characteristics are summarized as well. A complete understanding of the bacteria-host interaction will likely be necessary to assess anti-cancer capacities and unlock the full cancer therapeutic potential of oncolytic bacteria.
Collapse
Affiliation(s)
- Jason M Roe
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA
| | - Kevin Seely
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA
| | - Caleb J Bussard
- College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80130, USA
| | | | - Elizabeth G Mouw
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA
| | - Kenneth W Bayles
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Michael A Hollingsworth
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Amanda E Brooks
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA
- College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80130, USA
- Office of Research & Scholarly Activity, Rocky Vista University, Ivins, UT 84738, USA
| | - Kaitlin M Dailey
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
2
|
Tian L, Zhou W, Wu X, Hu Z, Qiu L, Zhang H, Chen X, Zhang S, Lu Z. CTLs: Killers of intracellular bacteria. Front Cell Infect Microbiol 2022; 12:967679. [PMID: 36389159 PMCID: PMC9645434 DOI: 10.3389/fcimb.2022.967679] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/12/2022] [Indexed: 09/10/2023] Open
Abstract
Many microbial pathogens have evolved a range of capabilities to evade host immune defense mechanisms and to survive and multiply in host cells. The presence of host intracellular bacteria makes it difficult for specific antibodies to function. After the intracellular bacteria escape the attack of the innate immune system, such as phagocytes, they survive in cells, and then adaptive immunity comes into play. Cytotoxic T lymphocytes (CTLs) play an important role in eliminating intracellular bacteria. The regulation of key transcription factors could promote CD4+/CD8+ T cells to acquire cytolytic ability. The TCR-CD3 complex transduces activation signals generated by TCR recognition of antigen and promotes CTLs to generate multiple pathways to kill intracellular bacteria. In this review, the mechanism of CD4/CD8 CTLs differentiation and how CD4/CD8 CTLs kill intracellular bacteria are introduced. In addition, their application and prospects in the treatment of bacterial infections are discussed.
Collapse
Affiliation(s)
- Li Tian
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Zhou
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xianwei Wu
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhuannan Hu
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Qiu
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huiyong Zhang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xue Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Shaoyan Zhang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenhui Lu
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Alam MS, Cavanaugh C, Pereira M, Babu U, Williams K. Susceptibility of aging mice to listeriosis: Role of anti-inflammatory responses with enhanced Treg-cell expression of CD39/CD73 and Th-17 cells. Int J Med Microbiol 2020; 310:151397. [PMID: 31974050 DOI: 10.1016/j.ijmm.2020.151397] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 11/14/2019] [Accepted: 12/18/2019] [Indexed: 12/22/2022] Open
Abstract
Foodborne Listeria monocytogenes (Lm) causes serious illness and death in immunosuppressed hosts, including the elderly population. We investigated Lm susceptibility and inflammatory cytokines in geriatric mice. Young-adult and old mice were gavaged with a Lm strain Lmo-InlAm. Tissues were assayed for Lm burden and splenocytes were analyzed for Th1/Th2/Th17/Treg responses and expression of CD39 and CD73. Old Lm-infected mice lost body-weight dose-dependently, had higher Lm colonization, and showed higher inflammatory responses than Lm-infected young-adult mice. After infection, IL-17 levels increased significantly in old mice whereas IFN-γ levels were unchanged. Levels of IL-10 and Treg cells were increased in infected old mice as compared to infected young-adult mice. Age-dependent enhanced expression of CD39/CD73 was observed in purified Treg prior to infection, suggesting increased baseline adenosine production in old mice. Lm lysate-treated splenocytes from older mice produced significantly higher levels of IL-10, IL17, and IL-1β, produced less IFN-γ and IL-2, and proliferated less than splenocytes from young-adult mice. Data suggests that older mice maybe more susceptible to Lm infection due to an imbalance of Th cell responses with disproportionate and persistent anti-inflammatory responses. Lm infection enhanced differentiation of proinflammatory Th17 cells, which may also exacerbate pathological responses during listeriosis.
Collapse
Affiliation(s)
- M Samiul Alam
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA.
| | - Christopher Cavanaugh
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| | - Marion Pereira
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| | - Uma Babu
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| | - Kristina Williams
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| |
Collapse
|
4
|
D'Orazio SEF. Innate and Adaptive Immune Responses during Listeria monocytogenes Infection. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0065-2019. [PMID: 31124430 PMCID: PMC11086964 DOI: 10.1128/microbiolspec.gpp3-0065-2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Indexed: 12/15/2022] Open
Abstract
It could be argued that we understand the immune response to infection with Listeria monocytogenes better than the immunity elicited by any other bacteria. L. monocytogenes are Gram-positive bacteria that are genetically tractable and easy to cultivate in vitro, and the mouse model of intravenous (i.v.) inoculation is highly reproducible. For these reasons, immunologists frequently use the mouse model of systemic listeriosis to dissect the mechanisms used by mammalian hosts to recognize and respond to infection. This article provides an overview of what we have learned over the past few decades and is divided into three sections: "Innate Immunity" describes how the host initially detects the presence of L. monocytogenes and characterizes the soluble and cellular responses that occur during the first few days postinfection; "Adaptive Immunity" discusses the exquisitely specific T cell response that mediates complete clearance of infection and immunological memory; "Use of Attenuated Listeria as a Vaccine Vector" highlights the ways that investigators have exploited our extensive knowledge of anti-Listeria immunity to develop cancer therapeutics.
Collapse
Affiliation(s)
- Sarah E F D'Orazio
- University of Kentucky, Microbiology, Immunology & Molecular Genetics, Lexington, KY 40536-0298
| |
Collapse
|
5
|
Two cases of listeria rhombencephalitis. IDCases 2017; 11:22-25. [PMID: 29255676 PMCID: PMC5726744 DOI: 10.1016/j.idcr.2017.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/01/2017] [Accepted: 12/01/2017] [Indexed: 12/20/2022] Open
Abstract
Listeria rhombencephalitis (LRE) is a rare encephalitis of the hindbrain that can present with a variety of neurological symptoms. It is a diagnostic challenge, but prompt antimicrobial therapy is important to prevent high rates of mortality and morbidity. We report two cases of LRE, with several contrasting clinical features and different disease courses. Despite being rare, it is important to consider listeria in patients with possible meningoencephalitis, even if cultures are negative. Empirical treatment of meningoencephalitis should provide coverage for listeria, especially if the patient is at risk of listeriosis or there is a potential history of listeria exposure.
Collapse
|
6
|
Uchiyama R, Yonehara S, Taniguchi S, Ishido S, Ishii KJ, Tsutsui H. Inflammasome and Fas-Mediated IL-1β Contributes to Th17/Th1 Cell Induction in Pathogenic Bacterial Infection In Vivo. THE JOURNAL OF IMMUNOLOGY 2017; 199:1122-1130. [DOI: 10.4049/jimmunol.1601373] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 05/31/2017] [Indexed: 12/25/2022]
|
7
|
Yang X, Zhang X, Sun Y, Tu T, Fu ML, Miller M, Fu YX. A BTLA-mediated bait and switch strategy permits Listeria expansion in CD8α(+) DCs to promote long-term T cell responses. Cell Host Microbe 2015; 16:68-80. [PMID: 25011109 DOI: 10.1016/j.chom.2014.05.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 02/09/2014] [Accepted: 05/30/2014] [Indexed: 12/31/2022]
Abstract
Listeria monocytogenes infected CD8α(+) DCs in the spleen are essential for CD8(+) T cell generation. CD8α(+) DCs are also necessary for Listeria expansion and dissemination within the host. The mechanisms that regulate CD8α(+) DCs to allow Listeria expansion are unclear. We find that activating the B and T lymphocyte attenuator (BTLA), a coinhibitory receptor for T cells, suppresses, while blocking BTLA enhances, both the primary and memory CD8 T cell responses against Listeria. Btla(-/-) mice have lower effector and memory CD8(+) T cells while paradoxically also being more resistant to Listeria. Although bacterial entry into Btla(-/-) CD8α(+) DCs is unaffected, Listeria fails to expand within these cells. BTLA signaling limits Fas/FasL-mediated suppression of Listeria expansion within CD8α(+) DCs to more effectively alert adaptive immune cells. This study uncovers a BTLA-mediated strategy used by the host that permits Listeria proliferation to enable increasing T cell responses for long-term protection.
Collapse
Affiliation(s)
- Xuanming Yang
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Xunmin Zhang
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA; International joint Cancer Institute, The Second Military Medical University, Shanghai, 200433, China
| | - Yonglian Sun
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Tony Tu
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - May Lynne Fu
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Mendy Miller
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.
| | - Yang-Xin Fu
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
8
|
Porturas TP, Sun H, Buchlis G, Lou Y, Liang X, Cathopoulis T, Fayngerts S, Johnson DS, Wang Z, Chen YH. Crucial roles of TNFAIP8 protein in regulating apoptosis and Listeria infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:5743-50. [PMID: 25948813 DOI: 10.4049/jimmunol.1401987] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 03/30/2015] [Indexed: 01/01/2023]
Abstract
TNF-α-induced protein 8 (TNFAIP8 or TIPE) is a newly described regulator of cancer and infection. However, its precise roles and mechanisms of actions are not well understood. We report in this article that TNFAIP8 regulates Listeria monocytogenes infection by controlling pathogen invasion and host cell apoptosis in a RAC1 GTPase-dependent manner. TNFAIP8-knockout mice were resistant to lethal L. monocytogenes infection and had reduced bacterial load in the liver and spleen. TNFAIP8 knockdown in murine liver HEPA1-6 cells increased apoptosis, reduced bacterial invasion into cells, and resulted in dysregulated RAC1 activation. TNFAIP8 could translocate to plasma membrane and preferentially associate with activated RAC1-GTP. The combined effect of reduced bacterial invasion and increased sensitivity to TNF-α-induced clearance likely protected the TNFAIP8-knockout mice from lethal listeriosis. Thus, by controlling bacterial invasion and the death of infected cells through RAC1, TNFAIP8 regulates the pathogenesis of L. monocytogenes infection.
Collapse
Affiliation(s)
- Thomas P Porturas
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Honghong Sun
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - George Buchlis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Yunwei Lou
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and Department of Immunology, Shandong University School of Medicine, Ji'nan 250012, People's Republic of China
| | - Xiaohong Liang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and Department of Immunology, Shandong University School of Medicine, Ji'nan 250012, People's Republic of China
| | - Terry Cathopoulis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Svetlana Fayngerts
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Derek S Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Zhaojun Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| | - Youhai H Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104; and
| |
Collapse
|
9
|
Jensen S, Steffensen MA, Jensen BAH, Schlüter D, Christensen JP, Thomsen AR. Adenovirus-Based Vaccine againstListeria monocytogenes: Extending the Concept of Invariant Chain Linkage. THE JOURNAL OF IMMUNOLOGY 2013; 191:4152-64. [DOI: 10.4049/jimmunol.1301290] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
10
|
Uchiyama R, Yonehara S, Tsutsui H. Fas-mediated inflammatory response in Listeria monocytogenes infection. THE JOURNAL OF IMMUNOLOGY 2013; 190:4245-54. [PMID: 23509366 DOI: 10.4049/jimmunol.1203059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The molecular mechanisms of Fas (CD95/Apo-1)-mediated apoptosis are increasingly understood. However, the role of Fas-mediated production of proinflammatory cytokines such as IL-18 and IL-1β in bacterial infection is unclear. We demonstrate the importance of Fas-mediated signaling in IL-18/IL-1β production postinfection with Listeria monocytogenes without the contribution of caspase-1 inflammasome. IL-18/IL-1β production in L. monocytogenes-infected peritoneal exudate cells from Fas-deficient mice was lower than those from wild type mice, indicating that Fas signaling contributes to cytokine production. L. monocytogenes infection induced Fas ligand expression on NK cells, which stimulates Fas expressed on the infected macrophages, leading to the production of IL-18/IL-1β. This was independent of caspase-1, caspase-11, and nucleotide-binding domain and leucine-rich repeat-containing receptors (NLRs) such as Nlrp3 and Nlrc4, but dependent on apoptosis-associated speck-like protein containing a caspase recruitment domain. Wild type cells exhibited caspase-8 activation, whereas Fas-deficient cells did not. L. monocytogenes-induced caspase-8 activation was abrogated by inhibitor for intracellular reactive oxygen species, N-acetyl-L-cysteine. L. monocytogenes-infected macrophages produced type-I IFNs such as IFN-β1, which was required for Il18 gene expression. Thus, Fas signaling regulates innate inflammatory cytokine production in L. monocytogenes infection.
Collapse
Affiliation(s)
- Ryosuke Uchiyama
- Department of Microbiology, Hyogo College of Medicine, Nishinomiya, 663-8501, Japan.
| | | | | |
Collapse
|
11
|
Guillermo LVC, Pereira WF, De Meis J, Ribeiro-Gomes FL, Silva EM, Kroll-Palhares K, Takiya CM, Lopes MF. Targeting caspases in intracellular protozoan infections. Immunopharmacol Immunotoxicol 2010; 31:159-73. [PMID: 18785049 DOI: 10.1080/08923970802332164] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Caspases are cysteine aspartases acting either as initiators (caspases 8, 9, and 10) or executioners (caspases 3, 6, and 7) to induce programmed cell death by apoptosis. Parasite infections by certain intracellular protozoans increase host cell life span by targeting caspase activation. Conversely, caspase activation, followed by apoptosis of lymphocytes and other cells, prevents effective immune responses to chronic parasite infection. Here we discuss how pharmacological inhibition of caspases might affect the immunity to protozoan infections, by either blocking or delaying apoptosis.
Collapse
Affiliation(s)
- Landi V C Guillermo
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Abstract
Cellular apoptosis induced by T cells is mainly mediated by two pathways. One, granule exocytosis utilizes perforin/granzymes. The other involves signaling through death receptors of the TNF-alpha R super-family, especially FasL. Perforin plays a central role in apoptosis induced by granzymes. However, the mechanisms of perforin-mediated cytotoxicity are still not elucidated completely. Perforin is not only a pore-forming protein, but also performs multiple biological functions or perforin performs one biological function (cytolysis), but has multiple biological implications in the cellular immune responses, including regulation of proliferation of CD8+ CTLs.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Neurology, 300 Jefferson Hospital for Neurosciences Building, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA. [corrected]
| |
Collapse
|
13
|
Decreased susceptibility of mice to infection with Listeria monocytogenes in the absence of interleukin-18. Infect Immun 2008; 76:3881-90. [PMID: 18573894 DOI: 10.1128/iai.01651-07] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The induction of proinflammatory cytokines such as gamma interferon (IFN-gamma) and tumor necrosis factor alpha is crucial for the early control of bacterial infections. Since interleukin-18 (IL-18) acts as a potent inducer of IFN-gamma, it might play an important role in the induction of a protective immune response in listeriosis. We used a murine model of systemic Listeria monocytogenes infection to study the immune response to these intracellular bacteria in the absence of IL-18. For this purpose, IL-18-deficient mice and mice treated with anti-IL-18 neutralizing antibody were infected with L. monocytogenes, and their innate and adaptive immune responses were compared to those of control mice. Unexpectedly, we found that mice deficient in IL-18 were partially resistant to primary infection with L. monocytogenes. At day 3 after infection, the numbers of listeriae in the livers and spleens of control mice were up to 500 times higher than those in IL-18-deficient or anti-IL-18 antibody-treated mice. In addition, the level of proinflammatory cytokines was markedly reduced in IL-18-deficient mice. Enhanced resistance to L. monocytogenes infection in IL-18-deficient mice was accompanied by increased numbers of leukocytes and reduced apoptosis in the spleen 48 to 72 h after infection. In contrast, control and IL-18-deficient mice showed no significant differences in their abilities to mount a protective L. monocytogenes-specific T-cell response.
Collapse
|
14
|
|
15
|
Narni-Mancinelli E, Campisi L, Bassand D, Cazareth J, Gounon P, Glaichenhaus N, Lauvau G. Memory CD8+ T cells mediate antibacterial immunity via CCL3 activation of TNF/ROI+ phagocytes. J Exp Med 2007; 204:2075-87. [PMID: 17698589 PMCID: PMC2118695 DOI: 10.1084/jem.20070204] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Accepted: 08/01/2007] [Indexed: 11/12/2022] Open
Abstract
Cytolysis, interferon gamma and tumor necrosis factor (TNF) alpha secretion are major effector mechanisms of memory CD8+ T cells that are believed to be required for immunological protection in vivo. By using mutants of the intracellular bacterium Listeria monocytogenes, we found that none of these effector activities is sufficient to protect against secondary infection with wild-type (WT) bacteria. We demonstrated that CCL3 derived from reactivated memory CD8+ T cells is required for efficient killing of WT bacteria. CCL3 induces a rapid TNF-alpha secretion by innate inflammatory mononuclear phagocytic cells (MPCs), which further promotes the production of radical oxygen intermediates (ROIs) by both MPCs and neutrophils. ROI generation is the final bactericidal mechanism involved in L. monocytogenes clearance. These results therefore uncover two levels of regulation of the antibacterial secondary protective response: (a) an antigen-dependent phase in which memory CD8+ T cells are reactivated and control the activation of the innate immune system, and (b) an antigen-independent phase in which the MPCs coordinate innate immunity and promote the bactericidal effector activities. In this context, CCL3-secreting memory CD8+ T cells are able to mediate "bystander" killing of an unrelated pathogen upon antigen-specific reactivation, a mechanism that may be important for the design of therapeutic vaccines.
Collapse
Affiliation(s)
- Emilie Narni-Mancinelli
- Institut National de la Santé et de la Recherche Médicale E-344, Groupe Avenir, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | | | | | | | | | | | | |
Collapse
|
16
|
Willberg CB, Ward SM, Clayton RF, Naoumov NV, McCormick C, Proto S, Harris M, Patel AH, Klenerman P. Protection of hepatocytes from cytotoxic T cell mediated killing by interferon-alpha. PLoS One 2007; 2:e791. [PMID: 17726521 PMCID: PMC1949144 DOI: 10.1371/journal.pone.0000791] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Accepted: 07/25/2007] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cellular immunity plays a key role in determining the outcome of hepatitis C virus (HCV) infection, although the majority of infections become persistent. The mechanisms behind persistence are still not clear; however, the primary site of infection, the liver, may be critical. We investigated the ability of CD8+ T-cells (CTL) to recognise and kill hepatocytes under cytokine stimulation. METHODS/PRINCIPLE FINDINGS Resting hepatocytes cell lines expressed low levels of MHC Class I, but remained susceptible to CTL cytotoxicity. IFN-alpha treatment, in vitro, markedly increased hepatocyte MHC Class I expression, however, reduced sensitivity to CTL cytotoxicity. IFN-alpha stimulated hepatocyte lines were still able to present antigen and induce IFN-gamma expression in interacting CTL. Resistance to killing was not due to the inhibition of the FASL/FAS- pathway, as stimulated hepatocytes were still susceptible to FAS-mediated apoptosis. In vitro stimulation with IFN-alpha, or the introduction of a subgenomic HCV replicon into the HepG2 line, upregulated the expression of the granzyme-B inhibitor-proteinase inhibitor 9 (PI-9). PI-9 expression was also observed in liver tissue biopsies from patients with chronic HCV infection. CONCLUSION/SIGNIFICANCE IFN-alpha induces resistance in hepatocytes to perforin/granzyme mediate CTL killing pathways. One possible mechanism could be through the expression of the PI-9. Hindrance of CTL cytotoxicity could contribute to the chronicity of hepatic viral infections.
Collapse
Affiliation(s)
- Christian B. Willberg
- Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
- Division of Experimental Medicine, University of California at San Francisco, San Francisco, California, United States of America
| | - Scott M. Ward
- Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
- Princess Alexandra Hospital, Woolloongabba, Australia
| | | | - Nikolai V. Naoumov
- The University College London Institute of Hepatology, University College London, London, United Kingdom
| | - Christopher McCormick
- Faculty of Biological Sciences, Institute of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
- Molecular Microbiology and Infection, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Sandra Proto
- The University College London Institute of Hepatology, University College London, London, United Kingdom
| | - Mark Harris
- Faculty of Biological Sciences, Institute of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Arvind H. Patel
- Medical Research Council Virology Unit, Glasgow, United Kingdom
| | - Paul Klenerman
- Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| |
Collapse
|
17
|
Ben Moshe T, Barash H, Kang TB, Kim JC, Kovalenko A, Gross E, Schuchmann M, Abramovitch R, Galun E, Wallach D. Role of caspase-8 in hepatocyte response to infection and injury in mice. Hepatology 2007; 45:1014-24. [PMID: 17385212 DOI: 10.1002/hep.21495] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UNLABELLED Caspase-8 has been implicated in signaling for apoptotic cell death and for certain nonapoptotic functions. However, knowledge of actual physiological or pathophysiological processes to which this enzyme contributes is lacking. Using a mouse model and employing the conditional knockout approach to delete the caspase-8 gene specifically in the liver, we found that caspase-8 deficiency in hepatocytes facilitates infection of the liver by Listeria monocytogenes, attenuates the hepatocyte proliferation wave during the first 48 hours after partial hepatectomy and, depending on the genetic background of the mice, prompts a chronic inflammatory response to the hepatectomy, as a result of which the proliferation of hepatocytes, although initially suppressed, might later be persistently enhanced, resulting in significant hepatomegaly. CONCLUSION These findings indicate that caspase-8 participates in regulation of the cellular response to infection and injury and that it does so by affecting various cellular functions, including cell death, cell proliferation, and induction of inflammation.
Collapse
Affiliation(s)
- Tehila Ben Moshe
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Ulett GC, Adderson EE. Regulation of Apoptosis by Gram-Positive Bacteria: Mechanistic Diversity and Consequences for Immunity. CURRENT IMMUNOLOGY REVIEWS 2006; 2:119-141. [PMID: 19081777 PMCID: PMC2600511 DOI: 10.2174/157339506776843033] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Apoptosis, or programmed cell death (PCD), is an important physiological mechanism, through which the human immune system regulates homeostasis and responds to diverse forms of cellular damage. PCD may also be involved in immune counteraction to microbial infection. Over the past decade, the amount of research on bacteria-induced PCD has grown tremendously, and the implications of this mechanism on immunity are being elucidated. Some pathogenic bacteria actively trigger the suicide response in critical lineages of leukocytes that orchestrate both the innate and adaptive immune responses; other bacteria proactively prevent PCD to benefit their own survival and persistence. Currently, the microbial virulence factors, which represent the keys to unlocking the suicide response in host cells, are a primary focus of this field. In this review, we discuss these bacterial "apoptosis regulatory molecules" and the apoptotic events they either trigger or prevent, the host target cells of this regulatory activity, and the possible ramifications for immunity to infection. Gram-positive pathogens including Staphylococcus, Streptococcus, Bacillus, Listeria, and Clostridia species are discussed as important agents of human infection that modulate PCD pathways in eukaryotic cells.
Collapse
Affiliation(s)
- Glen C Ulett
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105-2794, USA
| | | |
Collapse
|
19
|
Cassataro J, Velikovsky CA, de la Barrera S, Estein SM, Bruno L, Bowden R, Pasquevich KA, Fossati CA, Giambartolomei GH. A DNA vaccine coding for the Brucella outer membrane protein 31 confers protection against B. melitensis and B. ovis infection by eliciting a specific cytotoxic response. Infect Immun 2005; 73:6537-46. [PMID: 16177328 PMCID: PMC1230944 DOI: 10.1128/iai.73.10.6537-6546.2005] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The development of an effective subunit vaccine against brucellosis is a research area of intense interest. The outer membrane proteins (Omps) of Brucella spp. have been extensively characterized as potential immunogenic and protective antigens. This study was conducted to evaluate the immunogenicity and protective efficacy of the B. melitensis Omp31 gene cloned in the pCI plasmid (pCIOmp31). Immunization of BALB/c mice with pCIOmp31 conferred protection against B. ovis and B. melitensis infection. Mice vaccinated with pCIOmp31 developed a very weak humoral response, and in vitro stimulation of their splenocytes with recombinant Omp31 did not induced the secretion of gamma interferon. Splenocytes from Omp31-vaccinated animals induced a specific cytotoxic-T-lymphocyte activity, which leads to the in vitro lysis of Brucella-infected macrophages. pCIOmp31 immunization elicited mainly CD8(+) T cells, which mediate cytotoxicity via perforins, but also CD4(+) T cells, which mediate lysis via the Fas-FasL pathway. In vivo depletion of T-cell subsets showed that the pCIOmp31-induced protection against Brucella infection is mediated predominantly by CD8(+) T cells, although CD4(+)T cells also contribute. Our results demonstrate that the Omp31 DNA vaccine induces cytotoxic responses that have the potential to contribute to protection against Brucella infection. The protective response could be related to the induction of CD8(+) T cells that eliminate Brucella-infected cells via the perforin pathway.
Collapse
Affiliation(s)
- Juliana Cassataro
- Laboratorio de Inmunogenética, Hospital de Clínicas José de San Martín, Facultad de Medicina, Universidad de Buenos Aires, Argentina.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
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
|
21
|
Hehlgans T, Pfeffer K. The intriguing biology of the tumour necrosis factor/tumour necrosis factor receptor superfamily: players, rules and the games. Immunology 2005; 115:1-20. [PMID: 15819693 PMCID: PMC1782125 DOI: 10.1111/j.1365-2567.2005.02143.x] [Citation(s) in RCA: 579] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2004] [Revised: 01/17/2004] [Accepted: 01/25/2005] [Indexed: 11/28/2022] Open
Abstract
The members of the tumour necrosis factor (TNF)/tumour necrosis factor receptor (TNFR) superfamily are critically involved in the maintenance of homeostasis of the immune system. The biological functions of this system encompass beneficial and protective effects in inflammation and host defence as well as a crucial role in organogenesis. At the same time, members of this superfamily are responsible for host damaging effects in sepsis, cachexia, and autoimmune diseases. This review summarizes recent progress in the immunobiology of the TNF/TNFR superfamily focusing on results obtained from animal studies using gene targeted mice. The different modes of signalling pathways affecting cell proliferation, survival, differentiation, apoptosis, and immune organ development as well as host defence are reviewed. Molecular and cellular mechanisms that demonstrate a therapeutic potential by targeting individual receptors or ligands for the treatment of chronic inflammatory or autoimmune diseases are discussed.
Collapse
Affiliation(s)
- Thomas Hehlgans
- Institute of Medical Microbiology, University of Düsseldorf, Germany.
| | | |
Collapse
|
22
|
Zheng SJ, Jiang J, Shen H, Chen YH. Reduced Apoptosis and Ameliorated Listeriosis in TRAIL-Null Mice. THE JOURNAL OF IMMUNOLOGY 2004; 173:5652-8. [PMID: 15494516 DOI: 10.4049/jimmunol.173.9.5652] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Listeriosis is an infectious disease caused by the bacterium Listeria monocytogenes. Although it is well recognized that apoptosis plays a critical role in the pathogenesis of the disease, the molecular mechanisms of cell death in listeriosis remain to be established. We report in this study that mice deficient in TRAIL were partially resistant to primary listeriosis, and blocking TRAIL with a soluble death receptor 5 markedly ameliorated the disease. The numbers of Listeria in the liver and spleen of TRAIL+/+ mice were 10-100 times greater than those in TRAIL-/- mice following primary Listeria infection. This was accompanied by a significant increase in the survival rate of TRAIL-/- mice. Lymphoid and myeloid cell death was significantly inhibited in TRAIL-/- mice, which led to marked enlargement of the spleen. These results establish a critical role for TRAIL in apoptosis during listeriosis.
Collapse
Affiliation(s)
- Shi-Jun Zheng
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
23
|
Abstract
Listeria monocytogenes is a Gram-positive bacterium that is often used to study the mammalian immune response to infection because it is easy to culture, is relatively safe to work with and causes a highly predictable infection in laboratory mice. The broad application of this mouse model has resulted in a torrent of studies characterizing the contributions of different cytokines, receptors, adaptors and effector molecules to resistance against infection with Listeria monocytogenes. These studies, which are yielding one of the most comprehensive pictures of the 'battle' between host and microorganism, are reviewed here.
Collapse
Affiliation(s)
- Eric G Pamer
- Infectious Disease Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, Immunology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10021, USA.
| |
Collapse
|
24
|
Menaker RJ, Jones NL. Fascination with bacteria-triggered cell death: the significance of Fas-mediated apoptosis during bacterial infection in vivo. Microbes Infect 2004; 5:1149-58. [PMID: 14554257 DOI: 10.1016/j.micinf.2003.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increasing evidence indicates that bacterial pathogens have developed mechanisms to modulate the apoptotic signaling cascade of host cells and thereby cause disease. The Fas death receptor pathway is one of the most extensively investigated apoptotic signaling pathways. In this review we discuss the role of Fas signaling during the interplay between bacterial pathogens and the host in vivo.
Collapse
Affiliation(s)
- Rena J Menaker
- Research Institute, Rm. 8409, Hospital for Sick Children, 555 University Avenue, Toronto, Ont., Canada M5G 1X8
| | | |
Collapse
|
25
|
Hamilton SE, Porter BB, Messingham KAN, Badovinac VP, Harty JT. MHC class Ia-restricted memory T cells inhibit expansion of a nonprotective MHC class Ib (H2-M3)-restricted memory response. Nat Immunol 2004; 5:159-68. [PMID: 14745446 DOI: 10.1038/ni1026] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2003] [Accepted: 11/28/2003] [Indexed: 11/09/2022]
Abstract
Listeria monocytogenes infection generates major histocompatibility complex (MHC) class Ia-restricted and MHC class Ib-(H2-M3)-restricted effector and memory CD8+ T cells. However, only MHC class Ia-restricted memory cells expand after rechallenge, and it is unknown if MHC class Ib-restricted memory CD8+ T cells generated by vaccination are protective. We show here that H2-M3-restricted memory CD8+ T cells were capable of secondary expansion but, in contrast to primary H2-M3-restricted effector cells, failed to provide protective immunity. In lm-immune mice, MHC class Ia-restricted memory CD8+ T cells prevented the expansion of H2-M3-restricted memory T cell populations by limiting dendritic cell antigen presentation. Thus, protective immunity by H2-M3-restricted T cells is limited to primary infection, indicating that memory MHC class Ia-restricted T cells prevent nonessential immune responses during secondary infection.
Collapse
Affiliation(s)
- Sara E Hamilton
- Interdisciplinary Program in Immunology, The University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
26
|
Messingham KAN, Badovinac VP, Harty JT. Deficient anti-listerial immunity in the absence of perforin can be restored by increasing memory CD8+ T cell numbers. THE JOURNAL OF IMMUNOLOGY 2004; 171:4254-62. [PMID: 14530349 DOI: 10.4049/jimmunol.171.8.4254] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Compared with wild-type (WT) mice, Listeria monocytogenes (LM)-vaccinated perforin-deficient (PKO) mice have elevated levels of CD8(+) T cell memory, but exhibit reduced levels of protection against virulent LM. In this study, Ag-specific CD8(+) T cells from LM-vaccinated WT and PKO mice were used in adoptive transfer assays to determine the contribution of perforin-dependent cytolysis in protective immunity to LM. Perforin deficiency resulted in an approximately 5-fold reduction in the per-cell protective capacity of Ag-specific memory CD8(+) T cells that was not caused by differences in memory cell quality as measured by CD62L/CD27 expression, TCR repertoire use, functional avidity, differences in expansion of Ag-specific cells upon infection, or maintenance of memory levels over time. However, perforin-deficient CD8(+) T cells exhibited reduced in vivo cytotoxic function compared to WT CD8(+) T cells. Consistent with the existence of perforin-independent effector pathways, double-vaccinated PKO mice were as resistant to challenge with LM as single-vaccinated WT mice. Thus, increasing the number of memory CD8(+) T cells can overcome diminished per-cell protective immunity in the absence of perforin.
Collapse
|
27
|
Vanden Bush TJ, Rosenbusch RF. Characterization of the immune response to Mycoplasma bovis lung infection. Vet Immunol Immunopathol 2003; 94:23-33. [PMID: 12842609 DOI: 10.1016/s0165-2427(03)00056-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To better understand the interaction between Mycoplasma bovis and its bovine host, we have characterized the immune response generated during an experimental lung infection with M. bovis. Proliferation ([3H]-thymidine blastogenesis) and Th1/Th2 cytokine production were used to monitor peripheral cellular immune responses. Flow cytometry analysis was used to determine T-cell subset activity by CD25 expression. Humoral immune response was monitored by the identification of antigen-specific IgG1 and IgG2 isotypes over time. Herein, we show that M. bovis antigen stimulates activation of CD4+ and CD8+ cells in vitro in a manner consistent with memory, and that gammadelta-T cells are activated by antigen in a manner consistent with innate immunity. In addition, the percentage of cells producing IFN-gamma during recall response is equal to that of IL-4 producing cells. Serological analysis shows M. bovis stimulates increased production of antigen-specific IgG1 while very little IgG2 is produced. We therefore submit that experimental lung infection of cattle with M. bovis results in a Th2-skewed immune response.
Collapse
Affiliation(s)
- Tony J Vanden Bush
- Department of Veterinary Microbiology and Preventive Medicine, Veterinary Medical Research Institute, College of Veterinary Medicine, Iowa State University, Building #5, Ames, IA 50011, USA
| | | |
Collapse
|
28
|
de Saint Basile G, Fischer A. Defective cytotoxic granule-mediated cell death pathway impairs T lymphocyte homeostasis. Curr Opin Rheumatol 2003; 15:436-45. [PMID: 12819472 DOI: 10.1097/00002281-200307000-00011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Hemophagocytic syndrome is a severe and often fatal syndrome resulting from excessive activation and proliferation of T lymphocytes and macrophages. Onset of a hemophagocytic syndrome characterized the course of several human inherited immune disorders, all of them resulting from molecular defects of the perforin-dependent cytotoxic process exerted by both T and Natural Killer (NK) lymphocytes. These disorders highlight the determinant role of this lytic pathway in the control of lymphocyte expansion and homeostasis. New effectors of this secretory pathway have been thus identified.
Collapse
|
29
|
Badovinac VP, Hamilton SE, Harty JT. Viral infection results in massive CD8+ T cell expansion and mortality in vaccinated perforin-deficient mice. Immunity 2003; 18:463-74. [PMID: 12705850 DOI: 10.1016/s1074-7613(03)00079-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Perforin-mediated cytotoxicity is essential for clearance of primary LCMV infection. BALB/c-perforin-deficient (PKO) mice survived LCMV infection by deleting NP(118)-specific CD8(+) T cells whereas vaccination of PKO mice with Listeria expressing NP(118) generated a stable memory CD8(+) T cell population. However, >85% of vaccinated BALB/c-PKO mice died after LCMV infection. Mortality was associated with enormous expansion of NP(118)-specific CD8(+) T cells in both lymphoid and nonlymphoid tissues and aberrant CD8(+) T cell cytokine production. Depletion of CD8(+) T cells or treatment with anti-IFNgamma antibody rescued vaccinated mice from mortality. Thus, perforin was essential for resistance to secondary LCMV infection, and, in the absence of perforin, vaccination resulted in lethal disease mediated by dysregulated CD8(+) T cell expansion and cytokine production.
Collapse
|
30
|
Lin CW, Lee JY, Tsao YP, Shen CP, Lai HC, Chen SL. Oral vaccination with recombinant Listeria monocytogenes expressing human papillomavirus type 16 E7 can cause tumor growth in mice to regress. Int J Cancer 2002; 102:629-37. [PMID: 12448006 DOI: 10.1002/ijc.10759] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Listeria monocytogenes is a Gram-positive, facultative intracellular bacterium with the ability to present secreted proteins to the major histocompatibility complex class I pathway to stimulate cell-mediated immune response. In our study, we constructed the recombinant L. monocytogenes encoding human papillomavirus type 16 E7 gene (rLM-E7). When orally administered to syngeneic mice, rLM-E7 could induce a cytotoxic T-lymphocyte (CTL) response. Furthermore, in vitro flow cytometric assay and in vivo immune deficiency assays showed that rLM-E7 could prevent and eradicate tumor growth via CD8+-dependent CTLs. Hence, the potency of rLM-E7 as a therapeutic vaccine for cervical cancer is the result of the induction E7-specific cell-mediated immunity by L. monocytogenes. In addition to potency, this vaccine also offers ease of administration and reduced cost of production compared with other vaccines formulated for injection. Thus, L. monocytogenes encoding HPV-16 E7 may be a useful oral vaccine for cervical cancer treatment.
Collapse
Affiliation(s)
- Chih-Wei Lin
- Department of Microbiology & Immunology, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | | | | | | | | | | |
Collapse
|
31
|
San Mateo LR, Chua MM, Weiss SR, Shen H. Perforin-mediated CTL cytolysis counteracts direct cell-cell spread of Listeria monocytogenes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:5202-8. [PMID: 12391238 DOI: 10.4049/jimmunol.169.9.5202] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The immune system has evolved various effector cells and functions to combat diverse infectious agents equipped with different virulence strategies. CD8 T cells play a critical role in protective immunity to Listeria monocytogenes (Lm), a bacterium that grows within the host cell cytosol and spreads directly into neighboring cells. The importance of CD8 T cells during Lm infection is currently attributed to the cytosolic niche of this organism, which allows it to evade many aspects of immune surveillance. CTL lysis of infected cells is believed to be an essential protective mechanism, presumably functioning to release intracellular bacteria, although its precise role remains to be fully defined. In this study, we examined the contribution of perforin-mediated CTL cytolysis to protective immunity against recombinant Lm capable of or defective in cell-cell spread. We found that CTL cytolysis is critical for protective immunity to Lm capable of cell-cell spread while protective immunity against spread-defective Lm is largely independent of CTL cytolysis. These results demonstrate that an important function of CTL cytolysis is to counter the microbial virulence strategy of direct cell-cell spread. We propose a model that advances the current view of the role of CTL cytolysis in immunity to intracellular pathogens.
Collapse
MESH Headings
- Animals
- Antigens, Viral/immunology
- Cytotoxicity, Immunologic/genetics
- Epitopes, T-Lymphocyte/immunology
- Glycoproteins/immunology
- Immunity, Cellular/genetics
- Immunologic Memory/genetics
- Listeria monocytogenes/genetics
- Listeria monocytogenes/immunology
- Listeria monocytogenes/pathogenicity
- Listeriosis/genetics
- Listeriosis/immunology
- Listeriosis/microbiology
- Lymphocytic choriomeningitis virus/immunology
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Peptide Fragments/immunology
- Perforin
- Pore Forming Cytotoxic Proteins
- Recombination, Genetic
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/microbiology
- T-Lymphocytes, Cytotoxic/virology
- Viral Proteins/immunology
- Virulence
Collapse
Affiliation(s)
- Lani R San Mateo
- Department of Microbiology, School of Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | |
Collapse
|
32
|
Abstract
Virtually all of the measurable cell-mediated cytotoxicity delivered by cytotoxic T lymphocytes and natural killer cells comes from either the granule exocytosis pathway or the Fas pathway. The granule exocytosis pathway utilizes perforin to traffic the granzymes to appropriate locations in target cells, where they cleave critical substrates that initiate DNA fragmentation and apoptosis; granzymes A and B induce death via alternate, nonoverlapping pathways. The Fas/FasL system is responsible for activation-induced cell death but also plays an important role in lymphocyte-mediated killing under certain circumstances. The interplay between these two cytotoxic systems provides opportunities for therapeutic interventions to control autoimmune diseases and graft vs. host disease, but oversuppression of these pathways may also lead to increased viral susceptibility and/or decreased tumor cell killing.
Collapse
Affiliation(s)
- John H Russell
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | |
Collapse
|
33
|
Abstract
Expansion and contraction of antigen-specific CD8(+) T cells after infection or vaccination results in stable memory. Recent evidence demonstrates that anti-microbial effector molecules such as perforin and IFN-gamma regulate specific aspects of CD8(+) T cell homeostasis by mechanisms that may be dependent or independent of pathogen clearance.
Collapse
Affiliation(s)
- John T Harty
- Department of Microbiology, and Interdisciplinary Graduate Program in Immunology, University of Iowa, 3-512 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242, USA
| | | |
Collapse
|
34
|
Jones NL, Day AS, Jennings H, Shannon PT, Galindo-Mata E, Sherman PM. Enhanced disease severity in Helicobacter pylori-infected mice deficient in Fas signaling. Infect Immun 2002; 70:2591-2597. [PMID: 11953400 PMCID: PMC127915 DOI: 10.1128/iai.70.5.2591-2597.2002] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2001] [Revised: 11/28/2001] [Accepted: 01/22/2002] [Indexed: 11/20/2022] Open
Abstract
Recent evidence suggests that immune-mediated gastric epithelial cell apoptosis through Fas-Fas ligand interactions participates in Helicobacter pylori disease pathogenesis. To define the role of Fas signaling in vivo, H. pylori strain SS1 infection in C57BL/6 mice was compared to that in mice deficient in the Fas ligand (gld). gld mice had a degree of gastritis similar to that of C57BL/6 mice after 6 weeks (gastritis score, 5.2 +/- 0.6 [mean +/- standard error] versus 3.5 +/- 0.8) and 12 weeks (4.0 +/- 0.7 versus 3.4 +/- 0.5) of infection. Bacterial colonization was comparable in each group of mice at 12 weeks of infection (2.1 +/- 0.3 versus 1.6 +/- 0.3 for gld and C57BL/6, respectively; the difference is not significant). Sixty-seven percent of H. pylori-infected gld mice displayed atrophic changes in the gastric mucosa, compared with 37% of infected C57BL/6 mice, at 12 weeks. In addition, atrophic changes were more severe in H. pylori-infected gld mice (P < 0.05). Splenocytes isolated from H. pylori-infected C57BL/6 mice had a twofold increase in production of the Th1 cytokine gamma interferon (IFN-gamma) in response to H. pylori antigens at both 6 and 12 weeks compared to controls (143 +/- 65 versus 69 +/-26 pg/ml and 336 +/- 73 versus 172 +/- 60, respectively). In contrast, there was a lack of detectable IFN-gamma in gld mice infected with the bacterium. H. pylori-infected C57BL/6 mice had increased epithelial cell apoptosis compared with sham-infected C57BL/6 mice (35.0 +/- 8.9 versus 12.3 +/- 6.9; P < 0.05). Epithelial cell apoptosis did not differ between H. pylori-infected and control gld mice (5.2 +/- 1.6 versus 6.5 +/- 2.9 [not significant]). These data demonstrate that mice with mutations in the Fas ligand develop more severe premalignant mucosal changes in response to infection with H. pylori in association with both an impaired gastric epithelial cell apoptotic response and IFN-gamma production. The Fas death pathway modulates disease pathophysiology following murine infection with H. pylori. Deregulation of the Fas pathway could be involved in the transition from gastritis to gastric cancers during H. pylori infection.
Collapse
Affiliation(s)
- Nicola L Jones
- Department of Physiology, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| | | | | | | | | | | |
Collapse
|
35
|
Wing EJ, Gregory SH. Listeria monocytogenes: clinical and experimental update. J Infect Dis 2002; 185 Suppl 1:S18-24. [PMID: 11865436 DOI: 10.1086/338465] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Listeria monocytogenes, a small gram-positive bacillus, causes sepsis and meningitis in immunocompromised patients and a devastating maternal/fetal infection in pregnant women. Recent outbreaks demonstrated that L. monocytogenes can cause gastroenteritis in otherwise healthy individuals and more severe invasive disease in immunocompromised patients. Centralized processing in the food industry may be the cause of these large-scale listeriosis outbreaks. The mouse model of listeriosis, which was developed in the 1960s, has been extraordinarily useful for studying T cell-mediated immunity. Contrary to the original concept that macrophages are the principal effector cells in listeriosis, we found that immigrating neutrophils play the predominant role in early liver defenses. At later time points, CD8(+) T cells lyse infected hepatocytes by both perforin- and Fas-L/Fas--dependent mechanisms. Of interest, nonclassical major histocompatibility complex (MHC) class Ib--restricted cytolytic activity is expressed early during primary infection, whereas MHC class Ia--restricted activity is predominant through late primary and secondary infections.
Collapse
Affiliation(s)
- Edward J Wing
- Department of Medicine, Brown University Medical School, Rhode Island Hospital, Providence, Rhode Island 02903, USA.
| | | |
Collapse
|
36
|
Matthews AE, Weiss SR, Shlomchik MJ, Hannum LG, Gombold JL, Paterson Y. Antibody is required for clearance of infectious murine hepatitis virus A59 from the central nervous system, but not the liver. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:5254-63. [PMID: 11673540 DOI: 10.4049/jimmunol.167.9.5254] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Intracerebral inoculation with mouse hepatitis virus strain A59 results in viral replication in the CNS and liver. To investigate whether B cells are important for controlling mouse hepatitis virus strain A59 infection, we infected muMT mice who lack membrane-bound IgM and therefore mature B lymphocytes. Infectious virus peaked and was cleared from the livers of muMT and wild-type mice. However, while virus was cleared from the CNS of wild-type mice, virus persisted in the CNS of muMT mice. To determine how B cells mediate viral clearance, we first assessed CD4(+) T cell activation in the absence of B cells as APC. CD4(+) T cells express wild-type levels of CD69 after infection in muMT mice. IFN-gamma production in response to viral Ag in muMT mice was also normal during acute infection, but was decreased 31 days postinfection compared with that in wild-type mice. The role of Ab in viral clearance was also assessed. In wild-type mice plasma cells appeared in the CNS around the time that virus is cleared. The muMT mice that received A59-specific Ab had decreased virus, while mice with B cells deficient in Ab secretion did not clear virus from the CNS. Viral persistence was not detected in FcR or complement knockout mice. These data suggest that clearance of infectious mouse hepatitis virus strain A59 from the CNS requires Ab production and perhaps B cell support of T cells; however, virus is cleared from the liver without the involvement of Abs or B cells.
Collapse
Affiliation(s)
- A E Matthews
- Microbiology Department, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | | | | | | | | |
Collapse
|
37
|
Auerbuch V, Lenz LL, Portnoy DA. Development of a competitive index assay to evaluate the virulence of Listeria monocytogenes actA mutants during primary and secondary infection of mice. Infect Immun 2001; 69:5953-7. [PMID: 11500481 PMCID: PMC98721 DOI: 10.1128/iai.69.9.5953-5957.2001] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2001] [Accepted: 06/05/2001] [Indexed: 11/20/2022] Open
Abstract
We developed a competitive index assay for murine listeriosis that tests the virulence of Listeria monocytogenes strains in different organs and at various times postinoculation. Studies presented here demonstrate the reproducibility of this assay during primary and secondary infection of inbred and outbred mice. We verified the validity of this assay by performing competitive index analysis of a well-characterized strain of L. monocytogenes lacking the actA gene. In addition, we found that while L. monocytogenes strains unable to recruit vasodilator-stimulated phosphoprotein (VASP) to their surface exhibit a 10-fold virulence attenuation in the livers of naive animals, they display a 50-fold survival defect in the liver during secondary listeriosis.
Collapse
Affiliation(s)
- V Auerbuch
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3202, USA
| | | | | |
Collapse
|
38
|
Le LQ, Kabarowski JH, Weng Z, Satterthwaite AB, Harvill ET, Jensen ER, Miller JF, Witte ON. Mice lacking the orphan G protein-coupled receptor G2A develop a late-onset autoimmune syndrome. Immunity 2001; 14:561-71. [PMID: 11371358 DOI: 10.1016/s1074-7613(01)00145-5] [Citation(s) in RCA: 156] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mice with a targeted disruption of the gene encoding a lymphoid-expressed orphan G protein-coupled receptor, G2A, demonstrate a normal pattern of T and B lineage differentiation through young adulthood. As G2A-deficient animals age, they develop secondary lymphoid organ enlargement associated with abnormal expansion of both T and B lymphocytes. Older G2A-deficient mice (>1 year) develop a slowly progressive wasting syndrome, characterized by lymphocytic infiltration into various tissues, glomerular immune complex deposition, and anti-nuclear autoantibodies. G2A-deficient T cells are hyperresponsive to TCR stimulation, exhibiting enhanced proliferation and a lower threshold for activation. Our findings demonstrate that G2A plays a critical role in controlling peripheral lymphocyte homeostasis and that its ablation results in the development of a novel, late-onset autoimmune syndrome.
Collapse
Affiliation(s)
- L Q Le
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Deckert M, Soltek S, Geginat G, Lütjen S, Montesinos-Rongen M, Hof H, Schlüter D. Endogenous interleukin-10 is required for prevention of a hyperinflammatory intracerebral immune response in Listeria monocytogenes meningoencephalitis. Infect Immun 2001; 69:4561-71. [PMID: 11402000 PMCID: PMC98533 DOI: 10.1128/iai.69.7.4561-4571.2001] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2001] [Accepted: 04/03/2001] [Indexed: 11/20/2022] Open
Abstract
To analyze the role of interleukin-10 (IL-10) in bacterial cerebral infections, we studied cerebral listeriosis in IL-10-deficient (IL-10(-/-)) and wild-type (WT) mice, the latter of which express high levels of IL-10 in both primary and secondary cerebral listeriosis. IL-10(-/-) mice succumbed to primary as well as secondary listeriosis, whereas WT mice were significantly protected from secondary listeriosis by prior intraperitoneal immunization with Listeria monocytogenes. Meningoencephalitis developed in both strains; however, in IL-10(-/-) mice the inflammation was more severe and associated with increased brain edema and multiple intracerebral hemorrhages. IL-10(-/-) mice recruited significantly increased numbers of leukocytes, in particular granulocytes, to the brain, and the intracerebral cytokine (tumor necrosis factor, IL-1, IL-12, gamma interferon, and inducible nitric oxide synthase) and chemokine (crg2/IP-10, RANTES, MuMig, macrophage inflammatory protein 1alpha [MIP-1alpha], and MIP-1beta) transcription was enhanced compared to that in WT mice. Despite this prominent hyperinflammation, the frequencies of intracerebral L. monocytogenes-specific CD8(+) T cells were reduced and the intracerebral bacterial load was not reduced in IL-10(-/-) mice compared to WT mice. Following intraperitoneal infection, IL-10(-/-) mice exhibited hepatic hyperinflammation without better bacterial clearance; however, in contrast to the mice with cerebral listeriosis, they did not succumb, illustrating that intrinsic factors of the target organ have a strong impact on the course and outcome of the infection.
Collapse
Affiliation(s)
- M Deckert
- Institut für Neuropathologie, Universitätsklinken Bonn, Bonn, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Yamada K, Yoshino K, Sekikawa K, Madarame H, Yagita H, Nakane A. Effect of a matrix metalloproteinase inhibitor on host resistance against Listeria monocytogenes infection. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 2000; 29:187-94. [PMID: 11064265 DOI: 10.1111/j.1574-695x.2000.tb01522.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hydroxy acid-based matrix metalloproteinase (MMP) inhibitors have been shown to inhibit tumor infiltration and growth, endotoxin shock, and acute graft-versus-host disease. Blockade of the release of soluble tumor necrosis factor-alpha (TNF-alpha) and CD95 ligand (CD95L; FasL) from cell-associated forms is reportedly involved in the mechanism of the drug effect. We investigated the effect of a MMP inhibitor, KB-R7785, on host resistance against Listeria monocytogenes infection, in which TNF-alpha is essentially required for the defense, in mice. The administration of KB-R7785 exacerbated listeriosis, while the drug prevented lethal shock induced by lipopolysaccharide and D-galactosamine. KB-R7785 inhibited soluble TNF-alpha production in spleen cell cultures stimulated by heat-killed L. monocytogenes and the drug treatment reduced serum TNF-alpha levels in infected mice, whereas the compound was ineffective on the modulation of interferon-gamma and interleukin-10 production. The effect of KB-R7785 was considered to be dependent on TNF-alpha because the drug failed to affect L. monocytogenes infection in anti-TNF-alpha monoclonal antibody-treated mice and TNF-alpha knockout mice. Anti-CD95L monoclonal antibody was also ineffective on the infection. These results suggest that induction of infectious diseases, to which TNF-alpha is critical in host resistance, should be considered in MMP inhibitor-treated hosts.
Collapse
Affiliation(s)
- K Yamada
- Department of Bacteriology, Hirosaki University School of Medicine, Zaifu-cho 5, Hirosaki, Aomori 036-8562, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Based on T cell subset depletion studies and the analysis of gene knockout mice, it is evident that CD8(+) T cells contribute to resistance against intracellular infections with certain viral, protozoan, and bacterial pathogens. Although they are known primarily for their capacity to kill infected cells, CD8(+) T cells elaborate a variety of effector mechanisms with the potential to defend against infection. Microbes use multiple strategies to cause infection, and the nature of the pathogenhost interaction may determine which CD8(+) T cell effector mechanisms are required for immunity. In this review, we summarize our current understanding of the effector functions used by CD8(+) T cells in resistance to pathogens. Analyses of mice deficient in perforin and/or Fas demonstrate that cytolysis is critical for immunity against some, but not all, infections and also reveal the contribution of cytolysis to the pathogenesis of disease. The role of CD8(+) T cell-derived cytokines in resistance to infection has been analyzed by systemic treatment with neutralizing antibodies and cytokine gene knockout mice. These studies are complicated by the fact that few, if any, cytokines are uniquely produced by CD8(+) T cells. Thus, the requirement for CD8(+) T cell- derived cytokines in resistance against most pathogens remains to be defined. Finally, recent studies of human CD8(+) T cells reveal the potential for novel effector mechanisms in resistance to infection.
Collapse
Affiliation(s)
- J T Harty
- Department of Microbiology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, USA.
| | | | | |
Collapse
|
42
|
Wing EJ, Gregory SH. From hot dogs to CD8+ T cells: Listeria monocytogenes. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2000; 111:76-84. [PMID: 10881333 PMCID: PMC2194371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Affiliation(s)
- E J Wing
- Department of Medicine, Brown University, Providence, Rhode Island, USA
| | | |
Collapse
|
43
|
Harty JT, White D. A knockout approach to understanding CD8+ cell effector mechanisms in adaptive immunity to Listeria monocytogenes. Immunobiology 1999; 201:196-204. [PMID: 10631568 DOI: 10.1016/s0171-2985(99)80059-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
In the described experimental approach, we use an attenuated LM strain to evoke LM specific CD8+ T cell responses. In this fashion, we can immunize immunocompromised gene knockout mice, that would succumb to low level infection with virulent LM. We then generate antigen matched, LM-specific CD8+ T cell lines from wild-type and gene knockout mice, and compare their capacity to provide immunity to LM infection in vivo. To date, our results demonstrate that CD8+ T cell-derived IFN-gamma and TNF are not required effector functions. Perforin deficiency has an impact on CD8+ T cell immunity but our studies provide strong evidence for the existence of perforin independent pathways of CD8+ T cell immunity to LM. To assess the potential for redundancy in effector mechanisms, we have generated mice deficient in both perforin and IFN-gamma and are developing mice deficient in perforin and TNF. By removing the major CD8+ T cell effector mechanisms, singly and in combination, we will eventually determine whether immunity to LM can be provided by redundant effector pathways or if novel effector mechanisms exist beyond our current knowledge. The generation of MHC matched, single and double knockout mice, will also aid in continuing studies to analyze the role of these molecules in resistance to in vivo infection.
Collapse
Affiliation(s)
- J T Harty
- Department of Microbiology, University of Iowa, Iowa City, USA. john-harty@uiowa-edu
| | | |
Collapse
|
44
|
Abstract
Many exciting advances in our understanding of T cell mediated immunity to bacterial infection have occurred in the past several years. T cell responses have been more fully characterized, due in part to the development of MHC class I tetramers. The importance of cytokines and various effector molecules in defense against infection has come to light. Finally, intracellular bacteria are being exploited to deliver antigens and DNA in an effort to induce immunity to pathogens.
Collapse
Affiliation(s)
- K M Kerksiek
- Sections of Infectious Diseases and Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | |
Collapse
|
45
|
Perry LL, Feilzer K, Hughes S, Caldwell HD. Clearance of Chlamydia trachomatis from the murine genital mucosa does not require perforin-mediated cytolysis or Fas-mediated apoptosis. Infect Immun 1999; 67:1379-85. [PMID: 10024585 PMCID: PMC96471 DOI: 10.1128/iai.67.3.1379-1385.1999] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The molecular mechanisms of resistance to genital infection with the mouse pneumonitis (MoPn) strain of Chlamydia trachomatis are unknown. A role for major histocompatibility complex class II-restricted, interleukin-12-dependent CD4(+) T cells has been established, but the functional activity of these cells does not depend on secretion of gamma interferon. Here we examined the potential contribution of T-cell-mediated cytotoxicity and apoptosis to mucosal clearance of MoPn by using mice deficient in the molecular mediators of target cell lysis. Animals lacking perforin, Fas, Fas ligand, or both perforin and Fas ligand were infected genitally with C. trachomatis MoPn and monitored for expression of immunity to chlamydial antigens and clearance of MoPn from the genital mucosa. In each case, the profile of spleen cytokine production, the magnitude of the host antibody response, and the kinetics of chlamydial clearance were similar to those of genetically intact controls. Compensatory overproduction of tumor necrosis factor alpha, an alternate mediator of apoptosis in certain cell types, did not appear to account for the ability of mutant mice to resolve Chlamydia infections. These results fail to support CD4(+) T-cell-mediated apoptosis or CD8(+) T-cell-mediated cytotoxicity as being critical to the clearance of C. trachomatis MoPn urogenital infections.
Collapse
Affiliation(s)
- L L Perry
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA
| | | | | | | |
Collapse
|
46
|
White DW, MacNeil A, Busch DH, Pilip IM, Pamer EG, Harty JT. Perforin-Deficient CD8+ T Cells: In Vivo Priming and Antigen-Specific Immunity Against Listeria monocytogenes. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.2.980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
CD8+ T cells require perforin to mediate immunity against some, but not all, intracellular pathogens. Previous studies with H-2b MHC perforin gene knockout (PO) mice revealed both perforin-dependent and perforin-independent pathways of CD8+ T cell-mediated immunity to Listeria monocytogenes (LM). In this study, we address two previously unresolved issues regarding the requirement for perforin in antilisterial immunity: 1) Is CD8+ T cell-mediated, perforin-independent immunity specific for a single Ag or generalizable to multiple Ags? 2) Is there a deficiency in the priming of the CD8+ T cell compartment of PO mice following an immunizing challenge with LM? We used H-2d MHC PO mice to generate CD8+ T cell lines individually specific for three known Ags expressed by a recombinant strain of virulent LM. Adoptive transfer experiments into BALB/c host mice revealed that immunity can be mediated by PO CD8+ T cells specific for all Ags examined, indicating that perforin-independent immunity is not limited to CD8+ T cells that recognize listeriolysin O. Analysis of epitope-specific CD8+ T cell expansion by MHC class I tetramer staining and ELISPOT revealed no deficiency in either the primary or secondary response to LM infection in PO mice. These results demonstrate that the perforin-independent pathway of antilisterial resistance mediated by CD8+ T cells is generalizable to multiple epitopes. Furthermore, the results show that reduced antilisterial resistance observed with polyclonal PO CD8+ T cells is a consequence of a deficiency in effector function and not a result of suboptimal CD8+ T cell priming.
Collapse
Affiliation(s)
| | - Adam MacNeil
- †Department of Microbiology, University of Iowa, Iowa City, IA 52242; and
| | - Dirk H. Busch
- ‡Sections of Infectious Diseases and Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Ingrid M. Pilip
- ‡Sections of Infectious Diseases and Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Eric G. Pamer
- ‡Sections of Infectious Diseases and Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - John T. Harty
- *Interdisciplinary Graduate Program in Immunology and
- †Department of Microbiology, University of Iowa, Iowa City, IA 52242; and
| |
Collapse
|