1
|
Pereira-Dutra FS, Souza EK, Souza TS, Goltara-Gomes TC, Ferraro-Moreira F, Palhinha L, Cunha-Fernandes T, Rajão MA, Silva AR, Bozza PT. Accumulation of lipid droplets induced by Listeria monocytogenes in macrophages: implications for survival and evasion of innate immunity. J Leukoc Biol 2024; 116:1364-1371. [PMID: 38727078 DOI: 10.1093/jleuko/qiae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 11/28/2024] Open
Abstract
Listeriosis, caused by Listeria monocytogenes (L.m.), poses a significant public health concern as one of the most severe foodborne diseases. The pathogenesis of L.m. involves critical steps such as phagosome rupture and escape upon internalization. Throughout infection, L.m. influences various host processes, including lipid metabolism pathways, yet the role of lipid droplets (LDs) remains unclear. Here, we reported a rapid, time-dependent increase in LD formation in macrophages induced by L.m. LD biogenesis was found to be dependent on L.m. viability and virulence genes, particularly on the activity of the pore-forming protein listeriolysin O (LLO). The prevention of LD formation by inhibiting diacylglycerol O-acyltransferase 1 (DGAT1) and cytosolic phospholipase A2 (cPLA2) significantly reduced intracellular bacterial survival, impaired prostaglandin E2 synthesis, and decreased interleukin-10 production. Additionally, inhibiting LD formation led to increased levels of tumor necrosis factor α and interferon β. Collectively, our data suggest a role for LDs in promoting L.m. cell survival and evasion within macrophages.
Collapse
Affiliation(s)
- Filipe S Pereira-Dutra
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Ellen K Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Tamyris S Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Taynná C Goltara-Gomes
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Felipe Ferraro-Moreira
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Lohanna Palhinha
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Tamires Cunha-Fernandes
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Matheus A Rajão
- Program of Immunology and Tumor Biology, National Cancer Institute, INCA, André Cavalcanti St, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Adriana R Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| |
Collapse
|
2
|
Pal S, Sheff S, Al-Kuhlani M, Ojcius DM, de la Maza LM. Role of TRAIL-R in Primary and Secondary Genital and Respiratory Chlamydia muridarum Infections in Mice. Microbiol Spectr 2022; 10:e0161722. [PMID: 35876584 PMCID: PMC9431660 DOI: 10.1128/spectrum.01617-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/01/2022] [Indexed: 11/20/2022] Open
Abstract
The tumor necrosis factor (TNF)-related apoptosis-inducing ligand receptor (TRAIL-R) suppresses inflammation and could therefore affect the course of Chlamydia infections and their long-term sequelae. Wild-type (WT) and TRAIL-R-/- C57BL/6 mice were inoculated vaginally with Chlamydia muridarum; the course of the infection was followed with vaginal cultures and the presence of hydrosalpinx determined. To evaluate the role of TRAIL-R following a secondary infection, the mice were vaginally reinfected. WT and TRAIL-R-/- male mice were also infected and reinfected in the respiratory tract, and the course of the diseases and the infections were followed. Following the primary and secondary vaginal infection, no significant differences in vaginal shedding or hydrosalpinx formation were observed between the WT and TRAIL-R-/- mice. The WT and TRAIL-R-/- mice mounted antibody responses in serum and vaginal washes that were not significantly different. After the primary and secondary intranasal infections of the male mice, changes in body weight were determined, and no significant differences were observed between the WT and TRAIL-R-/- mice. Ten days after the primary and the secondary infections, the weight of the lungs and number of C. muridarum inclusion forming units (IFU) were determined. The lungs of the WT mice weighed less compared with the TRAIL-R-/- mice following a primary infection but not after a secondary infection. No differences in the number of C. muridarum IFU in the lungs were observed between the two groups of mice. In conclusion, despite playing a role in inflammation cell-signaling pathways in vitro, TRAIL-R does not appear to play a major role in the susceptibility, clinical outcomes, or long-term sequelae of C. muridarum infections in vivo. IMPORTANCE TNF-related apoptosis-inducing ligand receptor (TRAIL-R) is involved in suppressing inflammatory responses. Bacterial pathogens such as Chlamydia spp. elicit inflammatory responses in humans following genital, ocular, and respiratory infections. The inflammatory responses are important to control the spread of Chlamydia. However, in certain instances, these inflammatory responses can produce long-term sequelae, including fibrosis. Fibrosis, or scarring, in the genital tract, eye, and respiratory system results in functional deficiencies, including infertility, blindness, and chronic obstructive lung disease, respectively. The goal of this study was to determine if mice deficient in TRAIL-R infected in the genital and respiratory tracts with Chlamydia spp. suffer more or less severe infections, infertility, or lung diseases than wild-type mice. Our results show no differences between the immune responses, infection severity, and long-term sequelae between TRAIL-R knockout and wild-type animals following a genital or a respiratory infection with Chlamydia.
Collapse
Affiliation(s)
- Sukumar Pal
- Department of Pathology and Laboratory Medicine, Medical Sciences I, University of California, Irvine, Irvine, California, USA
| | - Sydni Sheff
- Department of Pathology and Laboratory Medicine, Medical Sciences I, University of California, Irvine, Irvine, California, USA
| | - Mufadhal Al-Kuhlani
- Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, California, USA
- Life Science Department, Fresno City College, Fresno, California, USA
| | - David M. Ojcius
- Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, California, USA
| | - Luis M. de la Maza
- Department of Pathology and Laboratory Medicine, Medical Sciences I, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
3
|
Xia X, Chen Y, Xu J, Yu C, Chen W. SRC-3 deficiency protects host from Listeria monocytogenes infection through increasing ROS production and decreasing lymphocyte apoptosis. Int Immunopharmacol 2021; 96:107625. [PMID: 33857803 DOI: 10.1016/j.intimp.2021.107625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 10/21/2022]
Abstract
Listeria monocytogenes is the third major cause of death among food poisoning. Our previous studies have demonstrated that steroid receptor coactivator 3 (SRC-3) plays a critical protective role in host defense against extracellular bacterial pathogens such as Escherichia coli and Citrobacter rodentium. However, its role involved in intracellular bacterial pathogen infection remains unclear. Herein, we found that SRC-3-/- mice are more resistant to L. monocytogenes infection after tail intravenous injection with L. monocytogenes compared with wild-type mice. After infecting with L. monocytogenes, SRC-3-/- mice exhibited decreased mortality rate, decreased bacterial load, less body weight loss, less proinflammatory cytokines and less severe tissue damage compared with wild-type mice. SRC-3-/- mice produced more ROS and decreased L. monocytogenes-induced lymphocyte apoptosis. Mechanically, SRC-3-/- mice displayed decreased expressions of negative regulator of ROS (NRROS) and interferon (IFN)-β and its target genes such as Daxx, Mx1 and TRAIL associated with apoptosis. Taken together, SRC-3 deficiency can protect host from L. monocytogenes infection through increasing ROS production and decreasing lymphocyte apoptosis via affecting the expressions of NRROS and IFN-β.
Collapse
Affiliation(s)
| | - Yuan Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China.
| | - Wenbo Chen
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China.
| |
Collapse
|
4
|
Sag D, Ayyildiz ZO, Gunalp S, Wingender G. The Role of TRAIL/DRs in the Modulation of Immune Cells and Responses. Cancers (Basel) 2019; 11:cancers11101469. [PMID: 31574961 PMCID: PMC6826877 DOI: 10.3390/cancers11101469] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/09/2019] [Accepted: 09/20/2019] [Indexed: 12/26/2022] Open
Abstract
Expression of TRAIL (tumor necrosis factor–related apoptosis–inducing ligand) by immune cells can lead to the induction of apoptosis in tumor cells. However, it becomes increasingly clear that the interaction of TRAIL and its death receptors (DRs) can also directly impact immune cells and influence immune responses. Here, we review what is known about the role of TRAIL/DRs in immune cells and immune responses in general and in the tumor microenvironment in particular.
Collapse
Affiliation(s)
- Duygu Sag
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Zeynep Ozge Ayyildiz
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Sinem Gunalp
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Gerhard Wingender
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Biomedicine and Health Technologies, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| |
Collapse
|
5
|
Detrimental Type I Interferon Signaling Dominates Protective AIM2 Inflammasome Responses during Francisella novicida Infection. Cell Rep 2019; 22:3168-3174. [PMID: 29562174 DOI: 10.1016/j.celrep.2018.02.096] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 01/29/2018] [Accepted: 02/23/2018] [Indexed: 01/18/2023] Open
Abstract
Interferons (IFNs) and inflammasomes are essential mediators of anti-microbial immunity. Type I IFN signaling drives activation of the AIM2 inflammasome in macrophages; however, the relative contribution of IFNs and inflammasome responses in host defense is less understood. We report intact AIM2 inflammasome responses in mice lacking type I IFN signaling during infection with F. novicida. Lack of type I IFN signaling conferred protection to F. novicida infection in contrast to the increased susceptibility in AIM2-deficient mice. Mice lacking both AIM2 and IFNAR2 were protected against the infection. The detrimental effects of type I IFN signaling were due to its ability to induce activation of apoptotic caspases and cell death. These results demonstrate the contrasting effects of type I IFN signaling and AIM2 during F. novicida infection in vivo and indicate a dominant role for type I IFNs in mediating detrimental responses despite the protective AIM2 inflammasome responses.
Collapse
|
6
|
McDougal CE, Sauer JD. Listeria monocytogenes: The Impact of Cell Death on Infection and Immunity. Pathogens 2018; 7:pathogens7010008. [PMID: 29324702 PMCID: PMC5874734 DOI: 10.3390/pathogens7010008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/28/2017] [Accepted: 01/03/2018] [Indexed: 02/07/2023] Open
Abstract
Listeria monocytogenes has evolved exquisite mechanisms for invading host cells and spreading from cell-to-cell to ensure maintenance of its intracellular lifecycle. As such, it is not surprising that loss of the intracellular replication niche through induction of host cell death has significant implications on the development of disease and the subsequent immune response. Although L. monocytogenes can activate multiple pathways of host cell death, including necrosis, apoptosis, and pyroptosis, like most intracellular pathogens L. monocytogenes has evolved a series of adaptations that minimize host cell death to promote its virulence. Understanding how L. monocytogenes modulates cell death during infection could lead to novel therapeutic approaches. In addition, as L. monocytogenes is currently being developed as a tumor immunotherapy platform, understanding how cell death pathways influence the priming and quality of cell-mediated immunity is critical. This review will focus on the mechanisms by which L. monocytogenes modulates cell death, as well as the implications of cell death on acute infection and the generation of adaptive immunity.
Collapse
Affiliation(s)
- Courtney E McDougal
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
7
|
Margaroli C, Oberle S, Lavanchy C, Scherer S, Rosa M, Strasser A, Pellegrini M, Zehn D, Acha-Orbea H, Ehirchiou D. Role of proapoptotic BH3-only proteins inListeria monocytogenesinfection. Eur J Immunol 2016; 46:1427-37. [DOI: 10.1002/eji.201545857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 01/25/2016] [Accepted: 03/29/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Camilla Margaroli
- Department of Biochemistry CIIL; University of Lausanne; Epalinges Switzerland
| | - Susanne Oberle
- Swiss Vaccine Research Institute; Centre des laboratoires d'Epalinges; Epalinges Switzerland
- Division of Immunology and Allergy; Department of Medicine; Lausanne University Hospital; Lausanne Switzerland
| | - Christine Lavanchy
- Department of Biochemistry CIIL; University of Lausanne; Epalinges Switzerland
| | - Stefanie Scherer
- Swiss Vaccine Research Institute; Centre des laboratoires d'Epalinges; Epalinges Switzerland
- Division of Immunology and Allergy; Department of Medicine; Lausanne University Hospital; Lausanne Switzerland
| | - Muriel Rosa
- Department of Biochemistry CIIL; University of Lausanne; Epalinges Switzerland
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research; Melbourne Australia
- The Department of Medical Biology; University of Melbourne; Melbourne Australia
| | - Marc Pellegrini
- The Walter and Eliza Hall Institute of Medical Research; Melbourne Australia
- The Department of Medical Biology; University of Melbourne; Melbourne Australia
| | - Dietmar Zehn
- Technische Universität München; Weihenstephaner Berg 3; 85354 Freising-Weihenstephan Germany
| | - Hans Acha-Orbea
- Department of Biochemistry CIIL; University of Lausanne; Epalinges Switzerland
| | - Driss Ehirchiou
- Department of Biochemistry CIIL; University of Lausanne; Epalinges Switzerland
| |
Collapse
|
8
|
Gyurkovska V, Ivanovska N. Distinct roles of TNF-related apoptosis-inducing ligand (TRAIL) in viral and bacterial infections: from pathogenesis to pathogen clearance. Inflamm Res 2016; 65:427-37. [PMID: 26943649 DOI: 10.1007/s00011-016-0934-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 02/03/2016] [Accepted: 02/24/2016] [Indexed: 02/02/2023] Open
Abstract
INTRODUCTION Apoptotic death of different cells observed during infection is thought to limit overwhelming inflammation in response to microbial challenge. However, the underlying apoptotic death mechanisms have not been well defined. Tumor necrosis factor (TNF) related apoptosis-inducing ligand (TRAIL) is a type II transmembrane protein belonging to the TNF superfamily, which is involved not only in tumor growth suppression but in infection control and also in the regulation of both innate and adaptive immune responses. FINDINGS In this review, we have summarized data of recent studies on the influence of the TRAIL/TRAIL receptor (TRAIL-R) system on the development of viral and bacterial infections. TRAIL may have a dual function in the immune system being able to kill infected cells and also to participate in the pathogenesis of multiple infections. Moreover, many pathogens have evolved mechanisms to manipulate TRAIL signaling thus increasing pathogen replication. CONCLUSION Present data highlight an essential role for the TRAIL/TRAIL-R system in the regulation and modulation of apoptosis and show that TRAIL has distinct roles in pathogenesis and pathogen elimination. Knowledge of the factors that determine whether TRAIL is helpful or harmful supposes its potential therapeutic implications that are only beginning to be explored.
Collapse
Affiliation(s)
- Valeriya Gyurkovska
- Institute of Microbiology, Department of Immunology, 26 G. Bonchev Str., 1113, Sofia, Bulgaria
| | - Nina Ivanovska
- Institute of Microbiology, Department of Immunology, 26 G. Bonchev Str., 1113, Sofia, Bulgaria.
| |
Collapse
|
9
|
Mondal TK, Emeny RT, Gao D, Ault JG, Kasten-Jolly J, Lawrence DA. A physical/psychological and biological stress combine to enhance endoplasmic reticulum stress. Toxicol Appl Pharmacol 2015; 289:313-22. [PMID: 26391182 DOI: 10.1016/j.taap.2015.09.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/07/2015] [Accepted: 09/16/2015] [Indexed: 12/29/2022]
Abstract
The generation of an immune response against infectious and other foreign agents is substantially modified by allostatic load, which is increased with chemical, physical and/or psychological stressors. The physical/psychological stress from cold-restraint (CR) inhibits host defense against Listeria monocytogenes (LM), due to early effects of the catecholamine norepinephrine (NE) from sympathetic nerves on β1-adrenoceptors (β1AR) of immune cells. Although CR activates innate immunity within 2h, host defenses against bacterial growth are suppressed 2-3 days after infection (Cao and Lawrence 2002). CR enhances inducible nitric oxide synthase (iNOS) expression and NO production. The early innate activation leads to cellular reduction-oxidation (redox) changes of immune cells. Lymphocytes from CR-treated mice express fewer surface thiols. Splenic and hepatic immune cells also have fewer proteins with free thiols after CR and/or LM, and macrophages have less glutathione after the in vivo CR exposure or exposure to NE in vitro. The early induction of CR-induced oxidative stress elevates endoplasmic reticulum (ER) stress, which could interfere with keeping phagocytized LM within the phagosome or re-encapsuling LM by autophagy once they escape from the phagosome. ER stress-related proteins, such as glucose-regulated protein 78 (GRP78), have elevated expression with CR and LM. The results indicate that CR enhances the unfolded protein response (UPR), which interferes with host defenses against LM. Thus, it is postulated that increased stress, as exists with living conditions at low socioeconomic conditions, can lower host defenses against pathogens because of oxidative and ER stress processes.
Collapse
Affiliation(s)
| | - Rebecca T Emeny
- Wadsworth Center, New York State Department of Health, Albany, NY 12201
| | - Donghong Gao
- Wadsworth Center, New York State Department of Health, Albany, NY 12201
| | - Jeffrey G Ault
- Wadsworth Center, New York State Department of Health, Albany, NY 12201
| | - Jane Kasten-Jolly
- Wadsworth Center, New York State Department of Health, Albany, NY 12201
| | - David A Lawrence
- Wadsworth Center, New York State Department of Health, Albany, NY 12201.
| |
Collapse
|
10
|
Amarante-Mendes GP, Griffith TS. Therapeutic applications of TRAIL receptor agonists in cancer and beyond. Pharmacol Ther 2015; 155:117-31. [PMID: 26343199 DOI: 10.1016/j.pharmthera.2015.09.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
TRAIL/Apo-2L is a member of the TNF superfamily first described as an apoptosis-inducing cytokine in 1995. Similar to TNF and Fas ligand, TRAIL induces apoptosis in caspase-dependent manner following TRAIL death receptor trimerization. Because tumor cells were shown to be particularly sensitive to this cytokine while normal cells/tissues proved to be resistant along with being able to synthesize and release TRAIL, it was rapidly appreciated that TRAIL likely served as one of our major physiologic weapons against cancer. In line with this, a number of research laboratories and pharmaceutical companies have attempted to exploit the ability of TRAIL to kill cancer cells by developing recombinant forms of TRAIL or TRAIL receptor agonists (e.g., receptor-specific mAb) for therapeutic purposes. In this review article we will describe the biochemical pathways used by TRAIL to induce different cell death programs. We will also summarize the clinical trials related to this pathway and discuss possible novel uses of TRAIL-related therapies. In recent years, the physiological importance of TRAIL has expanded beyond being a tumoricidal molecule to one critical for a number of clinical settings - ranging from infectious disease and autoimmunity to cardiovascular anomalies. We will also highlight some of these conditions where modulation of the TRAIL/TRAIL receptor system may be targeted in the future.
Collapse
Affiliation(s)
- Gustavo P Amarante-Mendes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, SP, Brazil; Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia, Brazil.
| | - Thomas S Griffith
- Department of Urology, Masonic Cancer Center, Center for Immunology, University of Minnesota, Minneapolis, MN, USA; Minneapolis VA Health Care System, Minneapolis, MN 55417, USA.
| |
Collapse
|
11
|
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
|
12
|
Calame DG, Mueller-Ortiz SL, Morales JE, Wetsel RA. The C5a anaphylatoxin receptor (C5aR1) protects against Listeria monocytogenes infection by inhibiting type 1 IFN expression. THE JOURNAL OF IMMUNOLOGY 2014; 193:5099-107. [PMID: 25297874 DOI: 10.4049/jimmunol.1401750] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Listeria monocytogenes is a major cause of mortality resulting from food poisoning in the United States. In mice, C5 has been genetically linked to host resistance to listeriosis. Despite this genetic association, it remains poorly understood how C5 and its activation products, C5a and C5b, confer host protection to this Gram-positive intracellular bacterium. In this article, we show in a systemic infection model that the major receptor for C5a, C5aR1, is required for a normal robust host immune response against L. monocytogenes. In comparison with wild-type mice, C5aR1(-/-) mice had reduced survival and increased bacterial burden in their livers and spleens. Infected C5aR1(-/-) mice exhibited a dramatic reduction in all major subsets of splenocytes, which was associated with elevated caspase-3 activity and increased TUNEL staining. Because type 1 IFN has been reported to impede the host response to L. monocytogenes through the promotion of splenocyte death, we examined the effect of C5aR1 on type 1 IFN expression in vivo. Indeed, serum levels of IFN-α and IFN-β were significantly elevated in L. monocytogenes-infected C5aR1(-/-) mice. Similarly, the expression of TRAIL, a type 1 IFN target gene and a proapoptotic factor, was elevated in NK cells isolated from infected C5aR1(-/-) mice. Treatment of C5aR1(-/-) mice with a type 1 IFNR blocking Ab resulted in near-complete rescue of L. monocytogenes-induced mortality. Thus, these findings reveal a critical role for C5aR1 in host defense against L. monocytogenes through the suppression of type 1 IFN expression.
Collapse
Affiliation(s)
- Daniel G Calame
- The Brown Foundation Institute of Molecular Medicine, Research Center for Immunology and Autoimmune Diseases, The University of Texas Health Science Center at Houston, Houston, TX 77030; M.D./Ph.D. Program, University of Texas Medical School at Houston/The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030; and
| | - Stacey L Mueller-Ortiz
- The Brown Foundation Institute of Molecular Medicine, Research Center for Immunology and Autoimmune Diseases, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - John E Morales
- The Brown Foundation Institute of Molecular Medicine, Research Center for Immunology and Autoimmune Diseases, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Rick A Wetsel
- The Brown Foundation Institute of Molecular Medicine, Research Center for Immunology and Autoimmune Diseases, The University of Texas Health Science Center at Houston, Houston, TX 77030; Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, TX 77030
| |
Collapse
|
13
|
Eshleman EM, Lenz LL. Type I interferons in bacterial infections: taming of myeloid cells and possible implications for autoimmunity. Front Immunol 2014; 5:431. [PMID: 25309533 PMCID: PMC4161047 DOI: 10.3389/fimmu.2014.00431] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/23/2014] [Indexed: 01/13/2023] Open
Abstract
Type I interferons (IFNs) were first described for their ability to protect the host from viral infections and may also have beneficial effects under specific conditions within some bacterial infections. Yet, these pleiotropic cytokines are now known to exacerbate infections by numerous life-threatening bacteria, including the intracellular pathogens Listeria monocytogenes and Mycobacterium tuberculosis. The evidence that such detrimental effects occur during bacterial infections in both animals and humans argues for selective pressure. In this review, we summarize the evidence demonstrating a pro-bacterial role for type I IFNs and discuss possible mechanisms that have been proposed to explain such effects. The theme emerges that type I IFNs act to suppress myeloid cell immune responses. The evolutionary conservation of such anti-inflammatory effects, particularly in the context of infections, suggests they may be important for limiting chronic inflammation. Given the effectiveness of type I IFNs in treatment of certain autoimmune diseases, their production may also act to raise the threshold for activation of immune responses to self-antigens.
Collapse
Affiliation(s)
- Emily M Eshleman
- Department of Immunology and Microbiology, University of Colorado School of Medicine , Aurora, CO , USA
| | - Laurel L Lenz
- Department of Immunology and Microbiology, University of Colorado School of Medicine , Aurora, CO , USA ; Department of Biomedical Research, National Jewish Health , Denver, CO , USA
| |
Collapse
|
14
|
Perez DA, Vago JP, Athayde RM, Reis AC, Teixeira MM, Sousa LP, Pinho V. Switching off key signaling survival molecules to switch on the resolution of inflammation. Mediators Inflamm 2014; 2014:829851. [PMID: 25136148 PMCID: PMC4127222 DOI: 10.1155/2014/829851] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 06/02/2014] [Accepted: 07/01/2014] [Indexed: 01/07/2023] Open
Abstract
Inflammation is a physiological response of the immune system to injury or infection but may become chronic. In general, inflammation is self-limiting and resolves by activating a termination program named resolution of inflammation. It has been argued that unresolved inflammation may be the basis of a variety of chronic inflammatory diseases. Resolution of inflammation is an active process that is fine-tuned by the production of proresolving mediators and the shutdown of intracellular signaling molecules associated with cytokine production and leukocyte survival. Apoptosis of leukocytes (especially granulocytes) is a key element in the resolution of inflammation and several signaling molecules are thought to be involved in this process. Here, we explore key signaling molecules and some mediators that are crucial regulators of leukocyte survival in vivo and that may be targeted for therapeutic purposes in the context of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Denise Alves Perez
- Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Juliana Priscila Vago
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Sinalização inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Rayssa Maciel Athayde
- Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Alesandra Corte Reis
- Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Mauro Martins Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Lirlândia Pires Sousa
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Sinalização inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Vanessa Pinho
- Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| |
Collapse
|
15
|
Zhu J, Chen L, Shi J, Liu S, Liu Y, Zheng D. TRAIL receptor deficiency sensitizes mice to dextran sodium sulphate-induced colitis and colitis-associated carcinogenesis. Immunology 2014; 141:211-21. [PMID: 24117005 DOI: 10.1111/imm.12181] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 10/02/2013] [Accepted: 10/04/2013] [Indexed: 12/21/2022] Open
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and its receptor (TRAIL-R) play important roles in immune regulation and cancer cell death. Although TRAIL has been shown to induce chemokine release in various tumour cells, the function of TRAIL-R in the development of colitis and colitis-associated carcinogenesis has not been explored. In this study, we found that TRAIL-R-deficient mice exhibited a higher incidence of colitis and colitis-associated cancer than that of wild-type (WT) mice, and TRAIL-R expression was down-regulated in WT mice that were fed dextran sulphate sodium. Chemokines, including CCL2 and CXCL1, were highly expressed in the serum and inflammatory colon tissues of TRAIL-R(-/-) mice compared with WT mice, and TRAIL-R(-/-) mice showed a marked infiltration of immune cells during colitis. Hyperactivation of Janus kinase and nuclear factor-κB in colon epithelial cells was also observed, which correlated with the severity of colonic inflammation in TRAIL-R(-/-) mice. These data suggest that TRAIL-R plays a protective role in chemical-induced colon injury and negatively regulates mucosal immune responses.
Collapse
Affiliation(s)
- Jieqing Zhu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | | | | | | | | | | |
Collapse
|
16
|
Abstract
Although type I interferons (IFN-I) were initially defined as potent antiviral agents, they can also cause decreased host resistance to some bacterial and viral infections. The many antiviral functions of the IFN-I include direct suppression of viral replication and activation of the immune response against viruses. In addition to their antiviral effects, IFN-I are also protective against several extracellular bacterial infections, in part, by promoting the induction of TNF-α and nitric oxide. In contrast, there is a negative effect of IFN-I on host resistance during chronic infection with lymphocytic choriomeningitis virus (LCMV) and acute infections with intracellular bacteria. In the case of LCMV, chronic IFN-I signaling induces adaptive immune system suppression. Blockade of IFN-I signaling removes the suppression and allows CD4 T-cell- and IFN-γ-mediated resolution of the infection. During acute intracellular bacterial infection, IFN-I suppress innate immunity by at least two defined mechanisms. During Francisella infection, IFN-I prevent IL-17 upregulation on γδ T cells and neutrophil recruitment. Following Listeria infection, IFN-I promote the cell death of macrophages and lymphocytes, which leads to innate immune suppression. These divergent findings for the role of IFN-I on pathogen control emphasize the complexity of the interferons system and force more mechanistic evaluation of its role in pathogenesis. This review evaluates IFN-I during infection with an emphasis on work carried out IFN-I-receptor-deficient mice.
Collapse
Affiliation(s)
- Javier Antonio Carrero
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
17
|
Kearney S, Delgado C, Lenz LL. Differential effects of type I and II interferons on myeloid cells and resistance to intracellular bacterial infections. Immunol Res 2013; 55:187-200. [PMID: 22983898 DOI: 10.1007/s12026-012-8362-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The type I and II interferons (IFNs) play important roles in regulating immune responses during viral and bacterial infections and in the context of autoimmune and neoplastic diseases. These two IFN types bind to distinct cell surface receptors that are expressed by nearly all cells to trigger signal transduction events and elicit diverse cellular responses. In some cases, type I and II IFNs trigger similar cellular responses, while in other cases, the IFNs have unique or antagonistic effects on host cells. Negative regulators of IFN signaling also modulate cellular responses to the IFNs and play important roles in maintaining immunological homeostasis. In this review, we provide an overview of how IFNs stimulate cellular responses. We discuss the disparate effects of type I and II IFNs on host resistance to certain intracellular bacterial infections and provide an overview of models that have been proposed to account for these disparate effects. Mechanisms of antagonistic cross talk between type I and II IFNs are also introduced.
Collapse
Affiliation(s)
- Staci Kearney
- Integrated Department of Immunology, University of Colorado School of Medicine, Aurora, CO, USA
| | | | | |
Collapse
|
18
|
Telieps T, Ewald F, Gereke M, Annemann M, Rauter Y, Schuster M, Ueffing N, von Smolinski D, Gruber AD, Bruder D, Schmitz I. Cellular-FLIP, Raji isoform (c-FLIP R) modulates cell death induction upon T-cell activation and infection. Eur J Immunol 2013; 43:1499-510. [PMID: 23505065 DOI: 10.1002/eji.201242819] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 01/25/2013] [Accepted: 03/07/2013] [Indexed: 12/25/2022]
Abstract
Dysregulation of apoptosis caused by an imbalance of pro- and anti-apoptotic protein expression can lead to cancer, neurodegenerative, and autoimmune diseases. Cellular-FLIP (c-FLIP) proteins inhibit apoptosis directly at the death-inducing signaling complex of death receptors, such as CD95, and have been linked to apoptosis regulation during immune responses. While the isoforms c-FLIPL and c-FLIPS are well characterized, the function of c-FLIPR remains poorly understood. Here, we demonstrate the induction of endogenous murine c-FLIPR in activated lymphocytes for the first time. To analyze c-FLIPR function in vivo, we generated transgenic mice expressing murine c-FLIPR specifically in hematopoietic cells. As expected, lymphocytes from c-FLIPR transgenic mice were protected against CD95-induced apoptosis in vitro. In the steady state, transgenic mice had normal cell numbers and unaltered frequencies of B cells and T-cell subsets in lymphoid organs. However, when challenged with Listeria monocytogenes, c-FLIPR transgenic mice showed less liver necrosis and better bacterial clearance compared with infected wild-type mice. We conclude that c-FLIPR expression in hematopoietic cells supports an efficient immune response against bacterial infections.
Collapse
Affiliation(s)
- Tanja Telieps
- Laboratory of Systems-Oriented Immunology and Inflammation Research, Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University, Magdeburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Age-dependent differences in systemic and cell-autonomous immunity to L. monocytogenes. Clin Dev Immunol 2013; 2013:917198. [PMID: 23653659 PMCID: PMC3638699 DOI: 10.1155/2013/917198] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Accepted: 03/07/2013] [Indexed: 12/13/2022]
Abstract
Host defense against infection can broadly be categorized into systemic immunity and cell-autonomous immunity. Systemic immunity is crucial for all multicellular organisms, increasing in importance with increasing cellular complexity of the host. The systemic immune response to Listeria monocytogenes has been studied extensively in murine models; however, the clinical applicability of these findings to the human newborn remains incompletely understood. Furthermore, the ability to control infection at the level of an individual cell, known as “cell-autonomous immunity,” appears most relevant following infection with L. monocytogenes; as the main target, the monocyte is centrally important to innate as well as adaptive systemic immunity to listeriosis. We thus suggest that the overall increased risk to suffer and die from L. monocytogenes infection in the newborn period is a direct consequence of age-dependent differences in cell-autonomous immunity of the monocyte to L. monocytogenes. We here review what is known about age-dependent differences in systemic innate and adaptive as well as cell-autonomous immunity to infection with Listeria monocytogenes.
Collapse
|
20
|
Dimberg LY, Anderson CK, Camidge R, Behbakht K, Thorburn A, Ford HL. On the TRAIL to successful cancer therapy? Predicting and counteracting resistance against TRAIL-based therapeutics. Oncogene 2013; 32:1341-50. [PMID: 22580613 PMCID: PMC4502956 DOI: 10.1038/onc.2012.164] [Citation(s) in RCA: 224] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 03/19/2012] [Accepted: 03/21/2012] [Indexed: 12/11/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and agonistic antibodies against TRAIL death receptors (DR) kill tumor cells while causing virtually no damage to normal cells. Several novel drugs targeting TRAIL receptors are currently in clinical trials. However, TRAIL resistance is a common obstacle in TRAIL-based therapy and limits the efficiency of these drugs. In this review article we discuss different mechanisms of TRAIL resistance, and how they can be predicted and therapeutically circumvented. In addition, we provide a brief overview of all TRAIL-based clinical trials conducted so far. It is apparent that although the effects of TRAIL therapy are disappointingly modest overall, a small subset of patients responds very well to TRAIL. We argue that the true potential of targeting TRAIL DRs in cancer can only be reached when we find efficient ways to select for those patients that are most likely to benefit from the treatment. To achieve this, it is crucial to identify biomarkers that can help us predict TRAIL sensitivity.
Collapse
Affiliation(s)
- L Y Dimberg
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | | | | | | | | | | |
Collapse
|
21
|
Abstract
Ware and Benedict discuss the pros and cons of TRAIL signaling in the context of bacterial infection and vascular disease. Since the discovery of TNF-related apoptosis-inducing ligand (TRAIL) and its network of receptors, the majority of attention has focused on the clinical potential of manipulating this pathway in cancer therapy. However, the widespread expression of TRAIL under inflammatory conditions and the ability to induce both apoptotic and prosurvival signaling pathways has suggested that TRAIL plays broader roles in regulating immune processes. Two new studies now show that expression of TRAIL by neutrophils in the lung facilitates defenses against bacterial pathogens, whereas expression of TRAIL by cells within arterioles exacerbates vascular disease. These differentiating results highlight that the context of TRAIL signaling can determine whether the outcome is beneficial or pathogenic for the host.
Collapse
Affiliation(s)
- Chris A Benedict
- the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | | |
Collapse
|
22
|
Steinwede K, Henken S, Bohling J, Maus R, Ueberberg B, Brumshagen C, Brincks EL, Griffith TS, Welte T, Maus UA. TNF-related apoptosis-inducing ligand (TRAIL) exerts therapeutic efficacy for the treatment of pneumococcal pneumonia in mice. ACTA ACUST UNITED AC 2012; 209:1937-52. [PMID: 23071253 PMCID: PMC3478925 DOI: 10.1084/jem.20120983] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Apoptotic death of alveolar macrophages observed during lung infection with Streptococcus pneumoniae is thought to limit overwhelming lung inflammation in response to bacterial challenge. However, the underlying apoptotic death mechanism has not been defined. Here, we examined the role of the TNF superfamily member TNF-related apoptosis-inducing ligand (TRAIL) in S. pneumoniae-induced macrophage apoptosis, and investigated the potential benefit of TRAIL-based therapy during pneumococcal pneumonia in mice. Compared with WT mice, Trail(-/-) mice demonstrated significantly decreased lung bacterial clearance and survival in response to S. pneumoniae, which was accompanied by significantly reduced apoptosis and caspase 3 cleavage but rather increased necrosis in alveolar macrophages. In WT mice, neutrophils were identified as a major source of intraalveolar released TRAIL, and their depletion led to a shift from apoptosis toward necrosis as the dominant mechanism of alveolar macrophage cell death in pneumococcal pneumonia. Therapeutic application of TRAIL or agonistic anti-DR5 mAb (MD5-1) dramatically improved survival of S. pneumoniae-infected WT mice. Most importantly, neutropenic mice lacking neutrophil-derived TRAIL were protected from lethal pneumonia by MD5-1 therapy. We have identified a previously unrecognized mechanism by which neutrophil-derived TRAIL induces apoptosis of DR5-expressing macrophages, thus promoting early bacterial killing in pneumococcal pneumonia. TRAIL-based therapy in neutropenic hosts may represent a novel antibacterial treatment option.
Collapse
Affiliation(s)
- Kathrin Steinwede
- Department of Experimental Pneumology and 2 Clinic for Pneumology, Hannover School of Medicine, Hannover 30625, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Zhang Y, Zhang R, Zhang H, Liu J, Yang Z, Xu P, Cai W, Lu G, Cui M, Schwendener RA, Shi HZ, Xiong H, Huang B. Microparticles released by Listeria monocytogenes-infected macrophages are required for dendritic cell-elicited protective immunity. Cell Mol Immunol 2012; 9:489-96. [PMID: 23064105 DOI: 10.1038/cmi.2012.33] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Interplay between macrophages and dendritic cells in the processing and presentation of bacterial antigens for T-cell immune responses remains poorly understood. Using a Listeria monocytogenes (Lm) infection model, we demonstrate that dendritic cells (DCs) require the support of macrophages to elicit protective immunity against Lm infection. DCs themselves were inefficient at taking up Lm but capable of taking up microparticles (MPs) released by Lm-infected macrophages. These MPs transferred Lm antigens to DCs, allowing DCs to present Lm antigen to effector T cells. MP-mediated Lm antigen transfer required MHC class I participation, since MHC class I deficiency in macrophages resulted in a significant reduction of T-cell activation. Moreover, the vaccination of mice with MPs from Lm-infected macrophages produced strong protective immunity against Lm infection. We here identify an intrinsic antigen transfer program between macrophages and DCs during Lm infection, and emphasize that macrophages also play an essential role in DC-elicited Lm-specific T-cell responses.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Infection with Listeria monocytogenes shows an early stage of lymphocyte apoptosis. This is an obligatory stage the extent of which depends on infective dose. Lymphocyte apoptosis occurs early and is rapidly superseded, yet it has a strong biological consequence. The immunological effect of lymphocyte apoptosis following infection is increased susceptibility to L. monocytogenes infection due, in part, to upregulation of IL-10 on macrophages and DC. Lymphocyte apoptosis is dependent on bacterial expression of the pore-forming toxin listeriolysin O (LLO). Also, purified LLO can lead to the induction of death pathways similar to infection, demonstrating that it is a killer agent generated by L. monocytogenes. Signaling through the type I interferon receptor potentiates cell death induced by the bacteria or LLO. Infection with L. monocytogenes also causes death of phagocytic cells, the nature and significance of which is not clear at present. Infection with L. monocytogenes is a tractable model to examine pathogen-induced cell death pathways and their possible immunological consequences in multiple cell types following infection.
Collapse
|
25
|
McGrath EE, Marriott HM, Lawrie A, Francis SE, Sabroe I, Renshaw SA, Dockrell DH, Whyte MKB. TNF-related apoptosis-inducing ligand (TRAIL) regulates inflammatory neutrophil apoptosis and enhances resolution of inflammation. J Leukoc Biol 2011; 90:855-65. [PMID: 21562052 PMCID: PMC3644175 DOI: 10.1189/jlb.0211062] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 03/25/2011] [Accepted: 04/08/2011] [Indexed: 01/20/2023] Open
Abstract
Novel therapeutics targeting neutrophilic inflammation are a major unmet clinical need in acute and chronic inflammation. The timely induction of neutrophil apoptosis is critical for inflammation resolution, and it is thought that acceleration of apoptosis may facilitate resolution at inflammatory sites. We previously demonstrated that a death receptor ligand, TRAIL, accelerates neutrophil apoptosis in vitro. We examined the role of TRAIL in neutrophil-dominant inflammation in WT and TRAIL-deficient mice. TRAIL deficiency did not alter constitutive neutrophil apoptosis, whereas exogenous TRAIL accelerated apoptosis of murine peripheral blood neutrophils. We compared TRAIL-deficient and WT mice in two independent models of neutrophilic inflammation: bacterial LPS-induced acute lung injury and zymosan-induced peritonitis. In both models, TRAIL-deficient mice had an enhanced inflammatory response with increased neutrophil numbers and reduced neutrophil apoptosis. Correction of TRAIL deficiency and supraphysiological TRAIL signaling using exogenous protein enhanced neutrophil apoptosis and reduced neutrophil numbers in both inflammatory models with no evidence of effects on other cell types. These data indicate the potential therapeutic benefit of TRAIL in neutrophilic inflammation.
Collapse
Affiliation(s)
| | | | - Allan Lawrie
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| | - Sheila E. Francis
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| | - Ian Sabroe
- Academic Unit of Respiratory Medicine and
| | | | - David H. Dockrell
- Immunology and Infectious Disease, Department of Infection and Immunity, and
| | | |
Collapse
|
26
|
Li H, Li X, Lu Y, Wang X, Zheng SJ. Staphylococcus sciuriexfoliative toxin C is a dimer that modulates macrophage functions. Can J Microbiol 2011; 57:722-9. [PMID: 21854098 DOI: 10.1139/w11-066] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Haihua Li
- State Key Laboratory of Agrobiotechnology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People’s Republic of China
| | - Xiaying Li
- State Key Laboratory of Agrobiotechnology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People’s Republic of China
| | - Ying Lu
- State Key Laboratory of Agrobiotechnology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People’s Republic of China
| | - Xiaojia Wang
- State Key Laboratory of Agrobiotechnology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People’s Republic of China
| | - Shijun J. Zheng
- State Key Laboratory of Agrobiotechnology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People’s Republic of China
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, 2 Yuan-Ming-Yuan West Road, Beijing 100193, People’s Republic of China
| |
Collapse
|
27
|
Borges M, Rosa GT, Appelberg R. The death-promoting molecule tumour necrosis factor-related apoptosis inducing ligand (TRAIL) is not required for the development of peripheral lymphopenia or granuloma necrosis during infection with virulent Mycobacterium avium. Clin Exp Immunol 2011; 164:407-16. [PMID: 21470210 DOI: 10.1111/j.1365-2249.2011.04385.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Disseminated infection with virulent Mycobacterium avium in C57Bl/6 (B6) mice leads to severe lymphocyte depletion in secondary lymphoid organs. In this study, we found an up-regulation of caspase-8 activity in spleen cell extracts from M. avium 25291-infected B6 mice compared to non-infected mice. The activation of this extrinsic apoptotic pathway correlated with an increase in inter-nucleosomal DNA fragmentation in CD4(+) spleen cells, as analysed by the terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assay. These data suggest the involvement of death receptors in the induction of lymphocyte loss in the spleen, but previous work has excluded a role for tumour necrosis factor (TNF) receptors and Fas/CD95 in M. avium-induced lymphopenia. TNF-related apoptosis-inducing ligand (TRAIL) is expressed by different cell types of the immune system and induces apoptosis and killing of tumour cells while sparing normal cells. Here we used TRAIL(-/-) mice to determine if the absence of TRAIL prevented M. avium-induced immune pathology. We found that TRAIL-deficient mice still developed splenic lymphopenia during disseminated infection or granuloma necrosis during low-dose infections while exhibiting slightly increased susceptibility to M. avium 25291 when compared to B6 mice. However, in vivo proliferation of less virulent strains of M. avium was not influenced by TRAIL deficiency despite a decrease in interferon-γ production in infected B6.TRAIL(-/-) mice compared to B6 mice. Our results show that TRAIL does not play a significant role in either M. avium-induced pathology or protective immunity.
Collapse
Affiliation(s)
- M Borges
- Laboratory of Microbiology and Immunology of Infection, Institute for Molecular and Cell Biology, Porto, Portugal
| | | | | |
Collapse
|
28
|
Listeria monocytogenes infection induces prosurvival metabolic signaling in macrophages. Infect Immun 2011; 79:1526-35. [PMID: 21263022 DOI: 10.1128/iai.01195-10] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Host cells use metabolic signaling through the LXRα nuclear receptor to defend against Listeria monocytogenes infection. 25-Hydroxycholesterol is a natural ligand of LXRs that is produced by the enzyme cholesterol 25-hydroxylase (CH25H). We found that expression of Ch25h is upregulated following L. monocytogenes infection in a beta interferon (IFN-β)-dependent fashion. Moreover, increased Ch25h expression promotes survival of L. monocytogenes-infected cells and increases sensitivity of the host to infection. We determined that expression of Cd5l, a prosurvival gene, is controlled by CH25H. In addition, we found that CD5L inhibits activation of caspase-1, promoting survival of infected macrophages. Our results reveal a mechanism by which an intracellular pathogen can prolong survival of infected cells, thus providing itself with a protected environment in which to replicate.
Collapse
|
29
|
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
|
30
|
Unsinger J, Kazama H, McDonough JS, Griffith TS, Hotchkiss RS, Ferguson TA. Sepsis-induced apoptosis leads to active suppression of delayed-type hypersensitivity by CD8+ regulatory T cells through a TRAIL-dependent mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:6766-72. [PMID: 20483771 PMCID: PMC2887093 DOI: 10.4049/jimmunol.0904054] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Patients who survive severe sepsis often display severely compromised immune function. One hallmark of such immune suppression in septic patients is an impaired delayed-type hypersensitivity (DTH) response, manifested by a loss of skin testing to recall Ags. Because sepsis induces significant apoptosis in lymphoid and myeloid cells, and apoptotic cells are themselves tolerogenic, we tested the hypothesis that suppression of DTH is mediated by tolerogenic properties of the apoptotic cells generated during sepsis. Mice subjected to cecal ligation and puncture demonstrated a loss of DTH for the 7 d following cecal ligation and puncture; however, the immune response returned to normal by day 10. Blocking sepsis-induced apoptosis via Bcl-2 overexpression or Bim deficiency prevented the loss of DTH. Importantly, injection of apoptotic cells into Bim-/- mice prevented an effective DTH response, thereby suggesting a causal link between apoptotic cells and immune suppression. Surprisingly, when TRAIL null mice were examined, we found that these animals had significant apoptosis but retained their DTH responses. Further studies revealed that apoptotic cells generated during sepsis induced a CD8+ regulatory T cell that suppressed DTH by TRAIL production. These results establish a link between apoptotic cells and immune suppression during sepsis and suggest TRAIL may be a viable therapeutic target for boosting the adaptive immune response following sepsis.
Collapse
Affiliation(s)
- Jacqueline Unsinger
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Hirotaka Kazama
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110
| | | | - Thomas S. Griffith
- Department of Urology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Richard S. Hotchkiss
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Thomas A. Ferguson
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
31
|
Abstract
Apoptosis ligand 2 tumor necrosis factor (TNF)-related apoptosis-inducing ligand (Apo2L/TRAIL) belongs to a small subset of proapoptotic protein ligands in the TNF superfamily. This subset, which also includes Fas ligand and TNF-alpha, can activate the extrinsic apoptotic cell death pathway on binding to cognate death receptors at the cell surface. Over the past 10 years, Apo2L/TRAIL has emerged as a promising candidate for cancer therapy, on the basis of its unique ability to trigger apoptosis in various types of cancer cells without significant toxicity toward normal cells. Herein, we review key advances in understanding the molecular events that control apoptosis signaling by Apo2L/TRAIL, which may aid in the development of cancer therapies based on the extrinsic apoptotic pathway.
Collapse
Affiliation(s)
- F Gonzalvez
- Department of Molecular Oncology, Genentech Inc., South San Francisco, CA, USA
| | | |
Collapse
|
32
|
Mellier G, Huang S, Shenoy K, Pervaiz S. TRAILing death in cancer. Mol Aspects Med 2009; 31:93-112. [PMID: 19995571 DOI: 10.1016/j.mam.2009.12.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Accepted: 12/02/2009] [Indexed: 12/13/2022]
Abstract
The observation that certain types of cancer express death receptors on their cell surface has triggered heightened interest in exploring the potential of receptor ligation as a novel anti-cancer modality, and since the expression is somewhat restricted to cancer cells the therapeutic implications are very promising. One such death receptor ligand belonging to the tumor necrosis receptor (TNF) superfamily, TNF-related apoptosis-inducing ligand (TRAIL), has been in the limelight as a tumor selective molecule that transmits death signal via ligation to its receptors (TRAIL-R1 and TRAIL-R2 or death receptors 4 and 5; DR4 and DR5). Interestingly, TRAIL-induced apoptosis exhibits hallmarks of extrinsic as well as intrinsic death pathways, and, therefore, is subject to regulation both at the cell surface receptor level as well as more downstream at the post-mitochondrial level. Despite the remarkable selectivity of DR expression on cancer cell surface, development of resistance to TRAIL-induced apoptosis remains a major challenge. Therefore, unraveling the cellular and molecular mechanisms of TRAIL resistance as well as identifying strategies to overcome this problem for an effective therapeutic response remains the cornerstone of many research endeavors. This review aims at presenting an overview of the biology, function and translational relevance of TRAIL with a specific view to discussing the various regulatory mechanisms and the current trends in reverting TRAIL resistance of cancer cells with the obvious implication of an improved clinical outcome.
Collapse
Affiliation(s)
- Gregory Mellier
- Department of Physiology, Yong Loo Lin School of Medicine, Singapore
| | | | | | | |
Collapse
|
33
|
Brost S, Koschny R, Sykora J, Stremmel W, Lasitschka F, Walczak H, Ganten TM. Differential expression of the TRAIL/TRAIL-receptor system in patients with inflammatory bowel disease. Pathol Res Pract 2009; 206:43-50. [PMID: 19954896 DOI: 10.1016/j.prp.2009.09.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 07/16/2009] [Accepted: 09/09/2009] [Indexed: 12/28/2022]
Abstract
TNF-related apoptosis inducing-ligand (TRAIL) is a potent inducer of apoptosis and plays an important role in immune regulation. To explore the role of TRAIL in inflammatory bowel disease (IBD), we examined the expression of the TRAIL/TRAIL-receptor system in colonic resections from patients with ulcerative colitis and Crohn's disease in comparison to normal colon and appendicitis. TRAIL and TRAIL-receptor (TRAIL-R) expression was assessed in resections of normal colon, colon of IBD patients, and appendicitis by immunohistochemistry. TRAIL was downregulated in enterocytes of patients with IBD, but was upregulated in mononuclear cells in areas of active mucosal inflammation. For TRAIL-R1, we detected a strong downregulation in the surface epithelium in IBD but not in appendicitis. TRAIL-R2 and TRAIL-R4 were strongly downregulated in the surface epithelium in any kind of mucosal inflammation. TRAIL and TRAIL-R1 are downregulated in enterocytes, and TRAIL is upregulated in mononuclear cells only in IBD but not in normal colon or appendicitis. This may point to a pathophysiologic role of the TRAIL system in inflammatory bowel disease.
Collapse
Affiliation(s)
- Sylvia Brost
- Department of Gastroenterology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Carrero JA, Calderon B, Vivanco-Cid H, Unanue ER. Recombinant Listeria monocytogenes expressing a cell wall-associated listeriolysin O is weakly virulent but immunogenic. Infect Immun 2009; 77:4371-82. [PMID: 19667043 PMCID: PMC2747929 DOI: 10.1128/iai.00419-09] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 05/25/2009] [Accepted: 08/04/2009] [Indexed: 11/20/2022] Open
Abstract
Listeriolysin O (LLO) is an essential virulence factor for the gram-positive bacterium Listeria monocytogenes. Our goal was to determine if altering the topology of LLO would alter the virulence and toxicity of L. monocytogenes in vivo. A recombinant strain was generated that expressed a surface-associated LLO (sLLO) variant secreted at 40-fold-lower levels than the wild type. In culture, the sLLO strain grew in macrophages, translocated to the cytosol, and induced cell death. However, the sLLO strain showed decreased infectivity, reduced lymphocyte apoptosis, and decreased virulence despite a normal in vitro phenotype. Thus, the topology of LLO in L. monocytogenes was a factor in the pathogenesis of the infection and points to a role of LLO secretion during in vivo infection. The sLLO strain was cleared by severe combined immunodeficient (SCID) mice. Despite the attenuation of virulence, the sLLO strain was immunogenic and capable of eliciting protective T-cell responses.
Collapse
Affiliation(s)
- Javier A Carrero
- Washington University School of Medicine, Department of Pathology and Immunology, 660 South Euclid Ave., St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
35
|
Abstract
The members of the tumour necrosis factor (TNF) superfamily of cytokines play important roles in the regulation of various immune-cell functions. Likewise, induction of cell death by apoptosis is indispensable for the normal functioning of the immune system. There are two major pathways of apoptosis induction. The intrinsic, or mitochondrial, pathway is regulated by the activation and interaction of members of the Bcl-2 family. The extrinsic, or death receptor, pathway is triggered by certain TNF family members when they engage their respective cognate receptors on the surface of the target cell. Hence, cell-to-cell-mediated death signals are induced by activation of these death receptor-ligand systems. Besides TNF itself and the CD95 (Fas/APO-1) ligand (FasL/Apo1L), the TNF-related apoptosis-inducing ligand (TRAIL/Apo2L) belongs to the subfamily of ligands that is responsible for extrinsic induction of cell death. Depending on their status of stimulation, TRAIL can be expressed by various cells of the immune system, amongst them natural killer (NK) cells, T cells, natural killer T cells (NKT cells), dendritic cells and macrophages. TRAIL has been implicated in immunosuppressive, immunoregulatory and immune-effector functions. With respect to pathological challenges, TRAIL and its receptors have been shown to play important roles in the immune response to viral infections and in immune surveillance of tumours and metastases. In this review we summarize the current knowledge on the role of TRAIL and its receptors in the immune system and, based on this, we discuss future directions of research into the diverse functions of this fascinating receptor-ligand system.
Collapse
|
36
|
Abstract
Since the discovery of Tumor Necrosis Factor-Related Apoptosis Inducing Ligand (TRAIL) in 1995, much has been learned about the protein, its receptors and signaling cascade to induce apoptosis and the regulation of its expression. However, the physiologic role or roles that TRAIL may play in vivo are still being explored. The expression of TRAIL on effector T cells and the ability of TRAIL to induce apoptosis in virally infected cells provided early clues that TRAIL may play an active role in the immune defense against viral infections. However, increasing evidence is emerging that TRAIL may have a dual function in the immune system, both as a means to kill virally infected cells and in the regulation of cytokine production. TRAIL has been implicated in the immune response to viral infections (good), and in the pathogenesis of multiple viral infections (bad). Furthermore, several viruses have evolved mechanisms to manipulate TRAIL signaling to increase viral replication (ugly). It is likely that whether TRAIL ultimately has a proviral or antiviral effect will be dependent on the specific virus and the overall cytokine milieu of the host. Knowledge of the factors that determine whether TRAIL is proviral or antiviral is important because the TRAIL system may become a target for development of novel antiviral therapies.
Collapse
Affiliation(s)
- Nathan Cummins
- Division of Infectious Diseases, Mayo Clinic, 2001ST SW, Rochester, MN 55905, USA
| | - Andrew Badley
- Division of Infectious Diseases, Mayo Clinic, 2001ST SW, Rochester, MN 55905, USA
| |
Collapse
|
37
|
Shepard BD, Badley AD. The Biology of TRAIL and the Role of TRAIL-Based Therapeutics in Infectious Diseases. ACTA ACUST UNITED AC 2009; 8:87-101. [PMID: 21857885 DOI: 10.2174/187152109787846060] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
TNF-related apoptosis inducing ligand (TRAIL) is a key mediator of the innate immune response to infection. While TRAIL-mediated apoptosis plays an essential role in the clearance of virus-infected cells, its physiologic role also includes immunosurveilance for cancer cells. Therapeutics that induce TRAIL-mediated apoptosis in cancer cells remain a focus of ongoing investigation in clinical trials, and much has been learned from these studies regarding the efficacy and toxicity of these interventions. These data, combined with data from numerous preclinical studies that detail the important and multifaceted role of TRAIL during infection with human immunodeficiency virus and other viruses, suggest that therapeutic exploitation of TRAIL signaling offers a novel and efficacious strategy for the management of infectious diseases.
Collapse
Affiliation(s)
- Brett D Shepard
- Mayo Clinic College of Medicine, Division of Infectious Diseases, Rochester, MN, 55905, USA
| | | |
Collapse
|
38
|
Semnani RT, Venugopal PG, Mahapatra L, Skinner JA, Meylan F, Chien D, Dorward DW, Chaussabel D, Siegel RM, Nutman TB. Induction of TRAIL- and TNF-alpha-dependent apoptosis in human monocyte-derived dendritic cells by microfilariae of Brugia malayi. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:7081-9. [PMID: 18981128 PMCID: PMC3662363 DOI: 10.4049/jimmunol.181.10.7081] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dysregulation of professional APC has been postulated as a major mechanism underlying Ag-specific T cell hyporesponsiveness in patients with patent filarial infection. To address the nature of this dysregulation, dendritic cells (DC) and macrophages generated from elutriated monocytes were exposed to live microfilariae (mf), the parasite stage that circulates in blood and is responsible for most immune dysregulation in filarial infections. DC exposed to mf for 24-96 h showed a marked increase in cell death and caspase-positive cells compared with unexposed DC, whereas mf exposure did not induce apoptosis in macrophages. Interestingly, 48-h exposure of DC to mf induced mRNA expression of the proapoptotic gene TRAIL and both mRNA and protein expression of TNF-alpha. mAb to TRAIL-R2, TNF-R1, or TNF-alpha partially reversed mf-induced cell death in DC, as did knocking down the receptor for TRAIL-R2 using small interfering RNA. The mf also induced gene expression of BH3-interacting domain death agonist and protein expression of cytochrome c in DC; mf-induced cleavage of BH3-interacting domain death agonist could be shown to induce release of cytochrome c, leading to activation of caspase 9. Our data suggest that mf induce DC apoptosis in a TRAIL- and TNF-alpha-dependent fashion.
Collapse
Affiliation(s)
- Roshanak Tolouei Semnani
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20872, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wang Y, Lu Y, Yu D, Wang Y, Chen F, Yang H, Zheng SJ. Enhanced resistance of restraint-stressed mice to sepsis. THE JOURNAL OF IMMUNOLOGY 2008; 181:3441-8. [PMID: 18714016 DOI: 10.4049/jimmunol.181.5.3441] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Sepsis remains a major health concern across the world. The effects of stress on host resistance to sepsis are still not very clear. To explore the effects of chronic stress on sepsis(') we examined the impact of restraint stress on the resistance of mice to sepsis. Interestingly, it was found that restraint stress enhanced the antisepsis resistance of mice and the concentrations of the proinflammatory cytokines IL-1, IL-6, IL-12, and TNF-alpha in the blood of stressed mice were dramatically reduced post Escherichia coli infection or LPS treatment as compared with that of controls (p < 0.05). In addition, the mRNA expressions of glucocorticoid-induced leucine zipper (GILZ) were up-regulated in the spleen and peritoneal macrophages of mice receiving restraint stress or dexamethasone treatment. These results demonstrate that restraint stress enhances the resistance of mice to sepsis, supporting corticotherapy for sepsis and proposing restraint-stressed mouse as an animal model to elucidate mechanisms of stress-associated, antisepsis resistance.
Collapse
Affiliation(s)
- Yu Wang
- College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
40
|
Carrero JA, Vivanco-Cid H, Unanue ER. Granzymes drive a rapid listeriolysin O-induced T cell apoptosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:1365-74. [PMID: 18606691 PMCID: PMC2562634 DOI: 10.4049/jimmunol.181.2.1365] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The Listeria monocytogenes protein listeriolysin O (LLO) is a pore-forming protein essential for virulence. Although the major role for LLO is to allow L. monocytogenes entry into the cytosol, it also induces apoptosis of activated lymphocytes, an obligatory cellular response that modulates the infection. Induction of apoptosis by LLO proceeds through a fast, caspase-dependent pathway and a slow, caspase-independent pathway. Polyclonal T cell lines were generated from either normal mice or mice deficient in granzyme and perforin proteins, and then treated with apoptogenic doses of LLO. In this study we show that apoptosis of lymphocytes induced by LLO was characterized by activation of caspases as quickly as 30 min that was dependent on the expression of granzymes. In the absence of granzymes, all parameters of apoptosis such as caspase activation, phosphatidylserine exposure, mitochondrial depolarization, and DNA fragmentation were dramatically reduced in magnitude. Removal of perforin inhibited the apoptotic effect of LLO on cells by approximately 50%. Neutralization of intracellular acidification using chloroquine inhibited the rapid apoptotic death. In agreement with these findings granzyme-deficient mice harbored lower bacterial titers and decrease splenic pathology compared with normal mice following L. monocytogenes infection. Thus, LLO exploits apoptotic enzymes of the adaptive immune response to eliminate immune cells and increase its virulence.
Collapse
Affiliation(s)
- Javier A Carrero
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | |
Collapse
|
41
|
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
|
42
|
Russell BH, Vasan R, Keene DR, Koehler TM, Xu Y. Potential dissemination of Bacillus anthracis utilizing human lung epithelial cells. Cell Microbiol 2007; 10:945-57. [PMID: 18067609 DOI: 10.1111/j.1462-5822.2007.01098.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dissemination of Bacillus anthracis spores from the lung is a critical early event in the establishment of inhalational anthrax. We recently reported that B. anthracis could adhere to and be internalized by cultured intestinal epithelial and fibroblast cells. Here, using gentamicin protection assays and/or electron microscopy, we found that Sterne strain 7702 spores were able to adhere to and subsequently be internalized by polarized A549 cells and primary human small airway epithelial cells. We showed for the first time that internalized spores were able to survive and that spores could translocate across an A549 cell barrier from the apical side to the basolateral side without disrupting the barrier integrity, suggesting a transcellular route. In addition, dormant spores of fully virulent Ames and UT500 strains were able to adhere to A549 cells at a frequency similar to that of 7702, whereas the capsule in germinated Ames and UT500 spores prevented adherence. Fluorescence microscopy also revealed that dormant Ames spores were internalized at a frequency similar to that of 7702. These findings highlight the possibility of a novel route of dissemination in which B. anthracis utilizes epithelial cells of the lung. The implications of these results to B. anthracis pathogenesis are discussed.
Collapse
Affiliation(s)
- Brooke H Russell
- Center for Extracellular Matrix Biology, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
43
|
Garifulin O, Qi Z, Shen H, Patnala S, Green MR, Boyartchuk V. Irf3 polymorphism alters induction of interferon beta in response to Listeria monocytogenes infection. PLoS Genet 2007; 3:1587-97. [PMID: 17845078 PMCID: PMC1971118 DOI: 10.1371/journal.pgen.0030152] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Accepted: 07/19/2007] [Indexed: 12/25/2022] Open
Abstract
Genetic makeup of the host plays a significant role in the course and outcome of infection. Inbred strains of mice display a wide range of sensitivities to Listeria monocytogenes infection and thus serve as a good model for analysis of the effect of genetic polymorphism. The outcome of L. monocytogenes infection in mice is influenced by the ability of this bacterium to induce expression of interferon beta mRNA, encoded in mouse by the Ifnb1 (interferon beta 1, fibroblast) gene. Mouse strains that lack components of the IFNβ signaling pathway are substantially more resistant to infection. We found that macrophages from the ByJ substrain of the common C57BL/6 inbred strain of mice are impaired in their ability to induce Ifnb1 expression in response to bacterial and viral infections. We mapped the locus that controls differential expression of Ifnb1 to a region on Chromosome 7 that includes interferon regulatory factor 3 (Irf3), which encodes a transcription factor responsible for early induction of Ifnb1 expression. In C57BL/6ByJ mice, Irf3 mRNA was inefficiently spliced, with a significant proportion of the transcripts retaining intron 5. Analysis of the Irf3 locus identified a single base-pair polymorphism and revealed that intron 5 of Irf3 is spliced by the atypical U12-type spliceosome. We found that the polymorphism disrupts a U12-type branchpoint and has a profound effect on the efficiency of splicing of Irf3. We demonstrate that a naturally occurring change in the splicing control element has a dramatic effect on the resistance to L. monocytogenes infection. Thus, the C57BL/6ByJ mouse strain serves as an example of how a mammalian host can counter bacterial virulence strategies by introducing subtle alteration of noncoding sequences. Specific variances in an individual's DNA, known as genetic polymorphisms, can play a significant role in determining susceptibility to an infectious disease. To identify the genetic polymorphisms that are associated with resistance to the common human bacterial pathogen L. monocytogenes, we have carried out a series of genetic and molecular biology experiments using closely related strains of mice that are differentially susceptible to Listeria infection. Through this analysis, we have identified a spontaneous mutation in an intron of the Irf3 gene, which encodes a key transcription factor involved in innate immunity. This single nucleotide change affects the efficiency with which Irf3 mRNA is spliced, thus limiting the ability of bacteria to induce interferon beta expression in order to suppress innate immune defense. By analyzing this mutation, we found that processing of mouse Irf3 mRNA relies on an atypical U12 splicing mechanism that has been suggested to be a rate-limiting step in gene expression. Our findings not only provide an additional example of an important role of noncoding polymorphisms in control of gene function, but also demonstrate how such polymorphisms can fine tune innate immune response.
Collapse
Affiliation(s)
- Oleg Garifulin
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Zanmei Qi
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Haihong Shen
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Sujatha Patnala
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Michael R Green
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Victor Boyartchuk
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
44
|
Huang B, Zhao J, Shen S, Li H, He KL, Shen GX, Mayer L, Unkeless J, Li D, Yuan Y, Zhang GM, Xiong H, Feng ZH. Listeria monocytogenes promotes tumor growth via tumor cell toll-like receptor 2 signaling. Cancer Res 2007; 67:4346-52. [PMID: 17483348 DOI: 10.1158/0008-5472.can-06-4067] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The contribution of bacterial infection to tumorigenesis is usually ascribed to infection-associated inflammation. An alternate view is that direct interaction of bacteria with tumor cells promotes tumor progression. Here, we show that the microenvironment of large tumors favors bacterial survival, which in turn directly accelerates tumor growth by activating tumor cell Toll-like receptors (TLR). Listeria monocytogenes (Lm) survives in the microenvironment of large but not small tumors, resulting in the promotion of tumor growth. Lm did not affect the percentage of regulatory T cells or myeloid suppressor cells in the tumor. Through TLR2 signaling, Lm activated mitogen-activated protein kinases and nuclear factor-kappaB in tumor cells, resulting in the increased production of nitric oxide and interleukin-6 and increased proliferation of tumor cells. All of these effects were abrogated by silencing expression of TLR2, but not TLR4. The interaction of Helicobacter pylori with tumor cells from gastric carcinoma patients resulted in similar effects. These findings provide a new insight into infection-associated tumorigenesis and illustrate the importance of antibiotic therapy to treat tumors with bacterial infiltration.
Collapse
Affiliation(s)
- Bo Huang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, The People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Eidsmo L, Fluur C, Rethi B, Eriksson Ygberg S, Ruffin N, De Milito A, Akuffo H, Chiodi F. FasL and TRAIL induce epidermal apoptosis and skin ulceration upon exposure to Leishmania major. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:227-39. [PMID: 17200196 PMCID: PMC1762690 DOI: 10.2353/ajpath.2007.060068] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Receptor-mediated apoptosis is proposed as an important regulator of keratinocyte homeostasis in human epidermis. We have previously reported that Fas/FasL interactions in epidermis are altered during cutaneous leishmaniasis (CL) and that keratinocyte death through apoptosis may play a pathogenic role for skin ulceration. To further investigate the alterations of apoptosis during CL, a keratinocyte cell line (HaCaT) and primary human epidermal keratinocytes were incubated with supernatants from Leishmania major-infected peripheral blood mononuclear cells. An apoptosis-specific microarray was used to assess mRNA expression in HaCaT cells exposed to supernatants derived from L. major-infected cultures. Fas and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) mRNA and protein expression were significantly up-regulated, and apoptosis was detected in both HaCaT and human epidermal keratinocyte cells. The keratinocyte apoptosis was partly inhibited through blocking of Fas or FasL and even more efficiently through TRAIL neutralization. Up-regulation of Fas on keratinocytes in epidermis and the presence of FasL-expressing macrophages and T cells in dermis were previously reported by us. In this study, keratinocytes expressing TRAIL, as well as the proapoptotic receptor TRAIL-R2, were detected in skin biopsies from CL cases. We propose that activation of Fas and TRAIL apoptosis pathways, in the presence of inflammatory mediators at the site of infection, leads to tissue destruction and ulceration during CL.
Collapse
Affiliation(s)
- Liv Eidsmo
- Department of Microbiology, Karolinska Institutet, MTC, 171 77 Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Griffith TS, Kazama H, VanOosten RL, Earle JK, Herndon JM, Green DR, Ferguson TA. Apoptotic Cells Induce Tolerance by Generating Helpless CD8+ T Cells That Produce TRAIL. THE JOURNAL OF IMMUNOLOGY 2007; 178:2679-87. [PMID: 17312109 DOI: 10.4049/jimmunol.178.5.2679] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The decision to generate a productive immune response or immune tolerance following pathogenic insult often depends on the context in which T cells first encounter Ag. The presence of apoptotic cells favors the induction of tolerance, whereas immune responses generated with necrotic cells promote immunity. We have examined the tolerance induced by injection of apoptotic cells, a system in which cross-presentation of Ag associated with the dead cells induces CD8+ regulatory (or suppressor) T cells. We observed that haptenated apoptotic cells induced CD8+ suppressor T cells without priming CD4+ T cells for immunity. These CD8+ T cells transferred unresponsiveness to naive recipients. In contrast, haptenated necrotic cells stimulated immunity, but induced CD8+ suppressor T cells when CD4+ T cells were absent. We further found that CD8+ T cells induced by these treatments displayed a "helpless CTL" phenotype and suppress the immune response by producing TRAIL. Animals deficient in TRAIL were resistant to tolerance induction by apoptotic cells. Thus, the outcome of an immune response taking place in the presence of cell death can be determined by the presence of CD4+-mediated Th cell function.
Collapse
Affiliation(s)
- Thomas S Griffith
- Department of Urology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Chen S, Wang Y, Chen F, Yang H, Gan M, Zheng SJ. A highly pathogenic strain of Staphylococcus sciuri caused fatal exudative epidermitis in piglets. PLoS One 2007; 2:e147. [PMID: 17215958 PMCID: PMC1764720 DOI: 10.1371/journal.pone.0000147] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2006] [Accepted: 11/23/2006] [Indexed: 11/18/2022] Open
Abstract
Staphylococcus sciuri are important human pathogens responsible for endocarditis, peritonitis, septic shock, urinary tract infection, pelvic inflammatory disease and wound infections. However, little information is known regarding the pathogenicity of S. sciuri to animals. From the pericardial fluid of a diseased piglet with exudative epidermitis (EE), we isolated a strain of Staphylococcus in pure culture. Surprisingly, this isolate was a member of S. sciuri rather than S. hyicus as identified by its biochemical traits and also by analysis of 23S ribosomal DNA using Internal Transcribed Spacer PCR. In addition, inoculation of newborn piglets with 1x10(10) CFU of the isolate by oral feeding or intra-muscular injection successfully reproduced EE in piglets, which suggested that the oral intake of the pathogen by the animals is one of the major routes of exposure. These unexpected findings prioritized S. sciuri as important zoonotic agents, which may have ramifications for human medicine.
Collapse
|
48
|
Scandurra GM, Williams RBH, Triccas JA, Pinto R, Gicquel B, Slobedman B, Cunningham A, Britton WJ. Effect of phthiocerol dimycocerosate deficiency on the transcriptional response of human macrophages to Mycobacterium tuberculosis. Microbes Infect 2006; 9:87-95. [PMID: 17194612 DOI: 10.1016/j.micinf.2006.10.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2006] [Revised: 09/29/2006] [Accepted: 10/20/2006] [Indexed: 01/06/2023]
Abstract
The control of mycobacterial infections is dependent on the finely tuned synergism between the innate and adaptive immune responses. The macrophage is the major host cell for Mycobacterium tuberculosis and the degree of virulence of mycobacteria may influence the initial macrophage response to infection. The cell wall molecule, phthiocerol dimycocerosate (DIM), is an important virulence factor that influences the early growth of M. tuberculosis in the lungs. To explore the basis for this effect we have compared the early gene response of human THP-1 macrophages to infection with virulent M. tuberculosis and the DIM-deficient DeltafadD26 M. tuberculosis strain using microarrays. Detailed analysis revealed a common core of macrophage genes, which were rapidly induced following infection with both strains, and deficiency of DIM had no significant effect on this initial macrophage transcriptional responses. In addition to chemokines and pro-inflammatory cytokines, the early response genes included components of the Toll-like receptor signalling, antigen presentation and apoptotic pathways, interferon response genes, cell surface receptors and their ligands, including TNF-related apoptosis inducing ligand (TRAIL) and CD40, and other novel genes. Therefore, although fadD26 deficiency is responsible for the early attenuation of the growth of M. tuberculosis in vivo, this effect is not associated with differences in the initial macrophage transcriptional response.
Collapse
Affiliation(s)
- G M Scandurra
- Mycobacterial Research Group, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW 2042, Australia
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Jabbari A, Legge KL, Harty JT. T cell conditioning explains early disappearance of the memory CD8 T cell response to infection. THE JOURNAL OF IMMUNOLOGY 2006; 177:3012-8. [PMID: 16920937 DOI: 10.4049/jimmunol.177.5.3012] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Memory CD8 T cells respond more rapidly to acute intracellular infections than naive CD8 T cells. An understanding of the biological processes involved in memory CD8 T cell recognition of Ag and up-regulation of effector mechanism necessitates analyzing memory CD8 T cells at early time points after infection. In the current study, we show that memory CD8 T cells ostensibly disappear from the spleens, blood, and peripheral organs of mice early after infection with Listeria monocytogenes. This disappearance is critically dependent on Ag, and cell-associated Ag alone can mediate this phenomenon. Further investigations, however, suggest that this disappearance is secondary to T cell-APC interactions, also known as T cell conditioning, and disruption of these putative interactions during splenic processing improves recovery of Ag-specific memory CD8 T cell populations after immunization. Conventional analyses of memory CD8 T cell populations early after infection and possibly in the presence of low levels of Ag (as during chronic infections) may exclude significant numbers of the responding CD8 T cell population.
Collapse
Affiliation(s)
- Ali Jabbari
- Interdisciplinary Graduate Program in Immunology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
50
|
Badovinac VP, Messingham KAN, Griffith TS, Harty JT. TRAIL Deficiency Delays, but Does Not Prevent, Erosion in the Quality of “Helpless” Memory CD8 T Cells. THE JOURNAL OF IMMUNOLOGY 2006; 177:999-1006. [DOI: 10.4049/jimmunol.177.2.999] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|