251
|
Cooperativity of adaptive and innate immunity: implications for cancer therapy. Cancer Immunol Immunother 2011; 60:1061-74. [PMID: 21656157 DOI: 10.1007/s00262-011-1053-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Accepted: 05/26/2011] [Indexed: 02/07/2023]
Abstract
The dichotomy of immunology into innate and adaptive immunity has created conceptual barriers in appreciating the intrinsic two-way interaction between immune cells. An emerging body of evidence in various models of immune rejection, including cancer, indicates an indispensable regulation of innate effector functions by adaptive immune cells. This bidirectional cooperativity in innate and adaptive immune functions has broad implications for immune responses in general and for regulating the tumor-associated inflammation that overrides the protective antitumor immunity. Mechanistic understanding of this two-way immune cross-talk could provide insights into novel strategies for designing better immunotherapy approaches against cancer and other diseases that normally defy immune control.
Collapse
|
252
|
Psarras S, Mavroidis M, Sanoudou D, Davos CH, Xanthou G, Varela AE, Panoutsakopoulou V, Capetanaki Y. Regulation of adverse remodelling by osteopontin in a genetic heart failure model. Eur Heart J 2011; 33:1954-63. [DOI: 10.1093/eurheartj/ehr119] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
253
|
Bertoletti A, Maini MK, Ferrari C. The host-pathogen interaction during HBV infection: immunological controversies. Antivir Ther 2011; 15 Suppl 3:15-24. [PMID: 21041900 DOI: 10.3851/imp1620] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
HBV is a hepatotropic and non-cytopathic virus that causes more than one million deaths annually from liver cirrhosis and hepatocellular carcinoma. As the virus itself is non-cytopathic, it is widely accepted that both viral control and liver pathology are mediated by the host immune system. Until recently, the focus has been on the crucial role of adaptive immune responses in controlling HBV infection, but the potential contribution of the innate system is now an important area of controversy. Unanswered questions include whether and when HBV can trigger components of innate immunity, and whether HBV can actively suppress the induction of innate immunity. We discuss the data available from animal models and human HBV infection addressing the role of innate immunity in the first part of this review. In the second part, we address the immunopathogenesis of the inflammatory events that characterize chronic hepatitis B. The mechanisms thought to be responsible for liver inflammation, namely the intrahepatic recruitment of inflammatory cells, which is orchestrated by chemokines, have been described; however, the underlying immunological triggers are much less clear. The prevailing idea is that liver inflammation results from a recovery of HBV-specific T-cells directly causing liver injury, but this scenario is supported by scanty experimental data. By contrast, recent findings raise the possibility of a contribution from innate components, such as natural killer cells.
Collapse
Affiliation(s)
- Antonio Bertoletti
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore.
| | | | | |
Collapse
|
254
|
Strutt TM, McKinstry KK, Swain SL. Control of innate immunity by memory CD4 T cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 780:57-68. [PMID: 21842365 DOI: 10.1007/978-1-4419-5632-3_6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
How memory CD4 T cells contribute to protection upon pathogen -challenge is not fully understood. Beyond traditional helper functions for CD8 T cell and B cell responses, memory CD4 T cells can have a potent impact on the character and a magnitude of inflammatory responses. Here we discuss how memory CD4 T cell control of innate immunity at early time points after pathogen encounters can influence protective responses. We also discuss important aspects of the mechanism whereby memory CD4 T cells directly and indirectly impact the activation status of antigen-presenting cells and production of inflammatory cytokines and chemokines from multiple cell types. We suggest that control of innate immune responses by the adaptive immune system is a powerful protective mechanism associated with the memory state and represents an important fail-safe in the face of pathogens that fail to trigger robust inflammatory responses through conserved pattern recognition receptors.
Collapse
|
255
|
|
256
|
Chen XH, Yin YJ, Zhang JX. Sepsis and immune response. World J Emerg Med 2011; 2:88-92. [PMID: 25214990 PMCID: PMC4129694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 03/17/2011] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND Sepsis and secondary multiple organ failure in critically ill patients are the major cause of death, but the pathogenesis of sepsis is not clear, especially the dysfunction of the immune system. In this paper, we review the response and regulation of the immune system and the functions of a variety of inflammatory mediators in sepsis. DATA SOURCES Studies were identified by searching MEDLINE and PubMed for articles using the keywords "sepsis", "immune response", and "inflammatory mediator" up to October 2010. Additional papers were identified by a manual search of the references from the key articles. RESULTS THIS SYSTEMATIC REVIEW WAS CONDUCTED OF: 1) the immune response; 2) immune regulation; 3) inflammatory mediators; 4) high-mobility group box 1 protein; 5) the complement system; and 6) the autonomic nervous system. There are no therapeutic approaches available for sepsis that target inflammatory response; the mortality of sepsis has not been significantly reduced. CONCLUSIONS Sepsis is complex and dynamic, and it has a group of heterogeneous syndromes. Since different patients with sepsis have different etiology, susceptibility, and responses, treatment should be prescribed individually.
Collapse
Affiliation(s)
- Xing-hai Chen
- Department of Emergency Medicine, Second Hospital of Jilin University, Changchun 130041, China
| | - Yong-jie Yin
- Department of Emergency Medicine, Second Hospital of Jilin University, Changchun 130041, China
| | - Jing-xiao Zhang
- Department of Emergency Medicine, Second Hospital of Jilin University, Changchun 130041, China
| |
Collapse
|
257
|
Veninga H, de Groot DM, McCloskey N, Owens BM, Dessing MC, Verbeek JS, Nourshargh S, van Eenennaam H, Boots AM, Hamann J. CD97 antibody depletes granulocytes in mice under conditions of acute inflammation via a Fc receptor-dependent mechanism. J Leukoc Biol 2010; 89:413-21. [DOI: 10.1189/jlb.0510280] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
258
|
Suppression of innate immune pathology by regulatory T cells during Influenza A virus infection of immunodeficient mice. J Virol 2010; 84:12564-75. [PMID: 20943986 DOI: 10.1128/jvi.01559-10] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The viral infection of higher vertebrates elicits potent innate and adaptive host immunity. However, an excessive or inappropriate immune response also may lead to host pathology that often is more severe than the direct effects of viral replication. Therefore, several mechanisms exist that regulate the magnitude and class of the immune response. Here, we have examined the potential involvement of regulatory T (Treg) cells in limiting pathology induced by influenza A virus (IAV) infection. Using lymphocyte-deficient mice as hosts, we showed that Treg cell reconstitution resulted in a significant delay in weight loss and prolonged survival following infection. The adoptively transferred Treg cells did not affect the high rate of IAV replication in the lungs of lymphocyte-deficient hosts, and therefore their disease-ameliorating effect was mediated through the suppression of innate immune pathology. Mechanistically, Treg cells reduced the accumulation and altered the distribution of monocytes/macrophages in the lungs of IAV-infected hosts. This reduction in lung monocytosis was associated with a specific delay in monocyte chemotactic protein-2 (MCP-2) induction in the infected lungs. Nevertheless, Treg cells failed to prevent the eventual development of severe disease in lymphocyte-deficient hosts, which likely was caused by the ongoing IAV replication. Indeed, using T-cell-deficient mice, which mounted a T-cell-independent B cell response to IAV, we further showed that the combination of virus-neutralizing antibodies and transferred Treg cells led to the complete prevention of clinical disease following IAV infection. Taken together, these results suggested that innate immune pathology and virus-induced pathology are the two main contributors to pathogenesis during IAV infection.
Collapse
|
259
|
Squamous carcinoma cells influence monocyte phenotype and suppress lipopolysaccharide-induced TNF-alpha in monocytes. Inflammation 2010; 33:207-23. [PMID: 20084448 DOI: 10.1007/s10753-009-9175-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bacteria and chronic inflammation are present in squamous cell carcinoma of the head and neck (HNSCC), but their roles in the pathogenesis of HNSCC are unclear. Our studies described here revealed that human monocytes co-cultured short term with HNSCC cells were more likely to express CD16, and CD16(+) small mononuclear cells were common in HNSCC specimens. In addition, we identified monocytes as the primary source of LPS-induced IL-6 and TNF-alpha in the monocyte-HNSCC co-cultures. Remarkably, relative to LPS-stimulated monocytes cultured alone, HNSCC cells profoundly suppressed LPS-induced TNF-alpha in monocytes, without compromising IL-6 production. High levels of cytoprotective factors like IL-6 and low levels of TNF-alpha are important for the tumor microenvironment that enables tumor cell survival, affects monocyte differentiation and may contribute to tumor colonization by bacteria. This study provides novel observations that HNSCC cells affect monocyte phenotype and function, which are relevant to the regulation of the HNSCC microenvironment.
Collapse
|
260
|
Skewed ratios between CD3(+) T cells and monocytes are associated with poor prognosis in patients with HBV-related acute-on-chronic liver failure. Biochem Biophys Res Commun 2010; 402:30-6. [PMID: 20883666 DOI: 10.1016/j.bbrc.2010.09.096] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 09/23/2010] [Indexed: 12/15/2022]
Abstract
Tempering of the innate immune response by T lymphocytes has been demonstrated to play a critical role in protecting animals from inflammation-induced death; however, its role in humans remains unknown. Patients with HBV-related acute-on-chronic liver failure (ACLF) share a striking similarity to the inflammatory response in septic shock where a hyperactive innate response is observed. The present study attempted to characterize the features of CD3(+) T cells and monocytes and evaluate their clinical implications in 55 patients with HBV-related ACLF, 30 patients with chronic hepatitis B (CHB) and 30 healthy controls (HC). We found that the ratio between circulating CD3(+) T cells and monocytes (T/M) was decreased in ACLF patients, due to decreased CD3 counts and increased monocyte counts compared with CHB and HC subjects. We also found that the T/M ratios were decreased from the early to the intermediate stage and reached the lowest value at the late stage in ACLF patients. Analyses with clinical parameters revealed that T/M ratios were negatively correlated with the Model for End-Stage Liver Disease Score and direct bilirubin, and positively correlated with prothrombin activity. Moreover, increased T/M ratios were observed in patients with good prognosis, but not in patients with a poor outcome; and ACLF patients who received liver transplantation exhibited an increased T/M ratio. Importantly, we found that programmed death-1 receptor (PD-1) was drastically upregulated on both CD4(+) T and CD8(+) T cells in ACLF, which at least in part contributed to the T-cell loss in these patients. Mechanically, the in vitro co-culture assay revealed that both CD4(+) T and CD8(+) T cells, as well as regulatory T cells, could inhibit TNF-α secretion by monocytes. In addition, the TNF-α levels in ACLF serum were negatively correlated with T/M ratios. In conclusion, our study identified the novel potential role of T/M ratio in predicting disease progression and provided novel evidences for further studies of the immunopathogenesis in ACLF.
Collapse
|
261
|
Kusunoki Y, Yamaoka M, Kubo Y, Hayashi T, Kasagi F, Douple EB, Nakachi K. T-cell immunosenescence and inflammatory response in atomic bomb survivors. Radiat Res 2010; 174:870-6. [PMID: 21128811 DOI: 10.1667/rr1847.1] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
In this paper we summarize the long-term effects of A-bomb radiation on the T-cell system and discuss the possible involvement of attenuated T-cell immunity in the disease development observed in A-bomb survivors. Our previous observations on such effects include impaired mitogen-dependent proliferation and IL-2 production, decreases in naive T-cell populations, and increased proportions of anergic and functionally weak memory CD4 T-cell subsets. In addition, we recently found a radiation dose-dependent increase in the percentages of CD25(+)/CD127(-) regulatory T cells in the CD4 T-cell population of the survivors. All these effects of radiation on T-cell immunity resemble effects of aging on the immune system, suggesting that ionizing radiation might direct the T-cell system toward a compromised phenotype and thereby might contribute to an enhanced immunosenescence. Furthermore, there are inverse, significant associations between plasma levels of inflammatory cytokines and the relative number of naïve CD4 T cells, also suggesting that the elevated levels of inflammatory markers found in A-bomb survivors can be ascribed in part to T-cell immunosenescence. We suggest that radiation-induced T-cell immunosenescence may result in activation of inflammatory responses and may be partly involved in the development of aging-associated and inflammation-related diseases frequently observed in A-bomb survivors.
Collapse
Affiliation(s)
- Yoichiro Kusunoki
- Department of Radiobiology, Radiation Effects Research Foundation, Hiroshima, Japan.
| | | | | | | | | | | | | |
Collapse
|
262
|
B-1 cells temper endotoxemic inflammatory responses. Immunobiology 2010; 216:302-8. [PMID: 20817308 DOI: 10.1016/j.imbio.2010.08.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 08/02/2010] [Indexed: 12/12/2022]
Abstract
Sepsis syndrome is caused by inappropriate immune activation due to bacteria and bacterial components released during infection. This syndrome is the leading cause of death in intensive care units. Specialized B-lymphocytes located in the peritoneal and pleural cavities are known as B-1 cells. These cells produce IgM and IL-10, both of which are potent regulators of cell-mediated immunity. It has been suggested that B-1 cells modulate the systemic inflammatory response in sepsis. In this study, we conducted in vitro and in vivo experiments in order to investigate a putative role of B-1 cells in a murine model of LPS-induced sepsis. Macrophages and B-1 cells were studied in monocultures and in co-cultures. The B-1 cells produced the anti-inflammatory cytokine IL-10 in response to LPS. In the B-1 cell-macrophage co-cultures, production of proinflammatory mediators (TNF-α, IL-6 and nitrite) was lower than in the macrophage monocultures, whereas that of IL-10 was higher in the co-cultures. Co-culture of B-1 IL-10(-/-) cells and macrophages did not reduce the production of the proinflammatory mediators (TNF-α, IL-6 and nitrite). After LPS injection, the mortality rate was higher among Balb/Xid mice, which are B-1 cell deficient, than among wild-type mice (65.0% vs. 0.0%). The Balb/Xid mice also presented a proinflammatory profile of TNF-α, IL-6 and nitrite, as well as lower levels of IL-10. In the early phase of LPS stimulation, B-1 cells modulate the macrophage inflammatory response, and the main molecular pathway of that modulation is based on IL-10-mediated intracellular signaling.
Collapse
|
263
|
Park S, Jiang Z, Mortenson ED, Deng L, Radkevich-Brown O, Yang X, Sattar H, Wang Y, Brown NK, Greene M, Liu Y, Tang J, Wang S, Fu YX. The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell 2010; 18:160-70. [PMID: 20708157 PMCID: PMC2923645 DOI: 10.1016/j.ccr.2010.06.014] [Citation(s) in RCA: 426] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Revised: 02/03/2010] [Accepted: 06/22/2010] [Indexed: 01/20/2023]
Abstract
Anti-HER2/neu antibody therapy is reported to mediate tumor regression by interrupting oncogenic signals and/or inducing FcR-mediated cytotoxicity. Here, we demonstrate that the mechanisms of tumor regression by this therapy also require the adaptive immune response. Activation of innate immunity and T cells, initiated by antibody treatment, was necessary. Intriguingly, the addition of chemotherapeutic drugs, although capable of enhancing the reduction of tumor burden, could abrogate antibody-initiated immunity leading to decreased resistance to rechallenge or earlier relapse. Increased influx of both innate and adaptive immune cells into the tumor microenvironment by a selected immunotherapy further enhanced subsequent antibody-induced immunity, leading to increased tumor eradication and resistance to rechallenge. This study proposes a model and strategy for anti-HER2/neu antibody-mediated tumor clearance.
Collapse
Affiliation(s)
- SaeGwang Park
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, Illinois 60637, USA
- Department of Microbiology, College of Medicine, University of INJE, 633-165, Gaegum-Dong, Jin-Gu, Busan, 614-735, Korea
| | - Zhujun Jiang
- Center for Infection and Immunity and National key laboratory of macromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Rd, Chaoyang District, Beijing 100101, China
| | - Eric D. Mortenson
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, Illinois 60637, USA
| | - Liufu Deng
- Center for Infection and Immunity and National key laboratory of macromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Rd, Chaoyang District, Beijing 100101, China
| | - Olga Radkevich-Brown
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, Illinois 60637, USA
| | - Xuanming Yang
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, Illinois 60637, USA
| | - Husain Sattar
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, Illinois 60637, USA
| | - Yang Wang
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, Illinois 60637, USA
| | - Nicholas K. Brown
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, Illinois 60637, USA
| | - Mark Greene
- Department of Pathology, University of Pennsylvania, 252 John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104
| | - Yang Liu
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Jie Tang
- Center for Infection and Immunity and National key laboratory of macromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Rd, Chaoyang District, Beijing 100101, China
| | - Shengdian Wang
- Center for Infection and Immunity and National key laboratory of macromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Rd, Chaoyang District, Beijing 100101, China
| | - Yang-Xin Fu
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, Illinois 60637, USA
- Center for Infection and Immunity and National key laboratory of macromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Rd, Chaoyang District, Beijing 100101, China
| |
Collapse
|
264
|
Shen X, Reng F, Gao F, Uchida Y, Busuttil RW, Kupiec-Weglinski JW, Zhai Y. Alloimmune activation enhances innate tissue inflammation/injury in a mouse model of liver ischemia/reperfusion injury. Am J Transplant 2010; 10:1729-37. [PMID: 20659085 PMCID: PMC3655759 DOI: 10.1111/j.1600-6143.2010.03205.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The deleterious sensitization to donor MHC Ags represents one of the most challenging problems in clinical organ transplantation. Although the role of effector/memory T cells in the rejection cascade has been extensively studied, it remains unknown whether and how these 'Ag-specific' cells influence host innate immunity, such as tissue inflammation associated with ischemia and reperfusion injury (IRI). In this study, we analyzed how allogeneic skin transplant (Tx) affected the sequel of host's own liver damage induced by partial warm ischemia and reperfusion. Our data clearly showed that allo-Tx recipients had increased inflammatory response against IR insult in their native livers, as evidenced by significantly more severe hepatocelluar damage, compared with syngeneic Tx recipient controls, and determined by serum ALT levels, liver histology (Suzuki's score) and intrahepatic proinflammatory gene inductions (TNF-alpha, IL-1beta and CXCL10). The CD4 T cells, but neither CD8 nor NK cells, mediated the detrimental effect of allo-Ag sensitization in liver IRI. Furthermore, CD154, but not IFN-gamma, was the key mechanism in allo-Tx recipients to facilitate IR-triggered liver damage. These results provide new evidence that alloreactive CD4 T cells are capable of enhancing innate tissue inflammation and organ injury via an Ag-nonspecific CD154-dependent but IFN-gamma independent mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuan Zhai
- Address correspondences to: Yuan Zhai, MD, PhD., The Dumont-UCLA Transplant Center 77-120 CHS, BOX: 957054, 10833 Le Conte Ave, Los Angeles, CA 90095-7054. Phone: (310) 825 9426; Fax: (310) 267 2367;
| |
Collapse
|
265
|
Genome-wide expression profiling deciphers host responses altered during dengue shock syndrome and reveals the role of innate immunity in severe dengue. PLoS One 2010; 5:e11671. [PMID: 20652028 PMCID: PMC2907396 DOI: 10.1371/journal.pone.0011671] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 06/22/2010] [Indexed: 12/14/2022] Open
Abstract
Background Deciphering host responses contributing to dengue shock syndrome (DSS), the life-threatening form of acute viral dengue infections, is required to improve both the differential prognosis and the treatments provided to DSS patients, a challenge for clinicians. Methodology/Principal Findings Based on a prospective study, we analyzed the genome-wide expression profiles of whole blood cells from 48 matched Cambodian children: 19 progressed to DSS while 16 and 13 presented respectively classical dengue fever (DF) or dengue hemorrhagic fever grades I/II (DHF). Using multi-way analysis of variance (ANOVA) and adjustment of p-values to control the False Discovery Rate (FDR<10%), we identified a signature of 2959 genes differentiating DSS patients from both DF and DHF, and showed a strong association of this DSS-gene signature with the dengue disease phenotype. Using a combined approach to analyse the molecular patterns associated with the DSS-gene signature, we provide an integrative overview of the transcriptional responses altered in DSS children. In particular, we show that the transcriptome of DSS children blood cells is characterized by a decreased abundance of transcripts related to T and NK lymphocyte responses and by an increased abundance of anti-inflammatory and repair/remodeling transcripts. We also show that unexpected pro-inflammatory gene patterns at the interface between innate immunity, inflammation and host lipid metabolism, known to play pathogenic roles in acute and chronic inflammatory diseases associated with systemic vascular dysfunction, are transcriptionnally active in the blood cells of DSS children. Conclusions/Significance We provide a global while non exhaustive overview of the molecular mechanisms altered in of DSS children and suggest how they may interact to lead to final vascular homeostasis breakdown. We suggest that some mechanisms identified should be considered putative therapeutic targets or biomarkers of progression to DSS.
Collapse
|
266
|
Macroparasites, innate immunity and immunoregulation: developing natural models. Trends Parasitol 2010; 26:540-9. [PMID: 20634138 DOI: 10.1016/j.pt.2010.06.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2009] [Revised: 06/03/2010] [Accepted: 06/07/2010] [Indexed: 12/25/2022]
Abstract
Innate immune receptors carry out surveillance for infection threats and are a proximal controller of the threshold and intensity at which inflammatory responses occur. As such, they are a natural focus for understanding how inflammatory immune reactivity is regulated. This review highlights how little data there are relating to the effect of macroparasites on systemic innate receptor responses. The idea is developed that studies on innate immune function in wild animals exposed to a natural profile of infections, including macroparasites, might be a valuable model in which to test hypotheses about the ultimate cause of aberrant inflammation in modern human populations.
Collapse
|
267
|
Sander LE, Sackett SD, Dierssen U, Beraza N, Linke RP, Müller M, Blander JM, Tacke F, Trautwein C. Hepatic acute-phase proteins control innate immune responses during infection by promoting myeloid-derived suppressor cell function. J Exp Med 2010; 207:1453-64. [PMID: 20530204 PMCID: PMC2901069 DOI: 10.1084/jem.20091474] [Citation(s) in RCA: 265] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 04/29/2010] [Indexed: 12/14/2022] Open
Abstract
Acute-phase proteins (APPs) are an evolutionarily conserved family of proteins produced mainly in the liver in response to infection and inflammation. Despite vast pro- and antiinflammatory properties ascribed to individual APPs, their collective function during infections remains poorly defined. Using a mouse model of polymicrobial sepsis, we show that abrogation of APP production by hepatocyte-specific gp130 deletion, the signaling receptor shared by IL-6 family cytokines, strongly increased mortality despite normal bacterial clearance. Hepatic gp130 signaling through STAT3 was required to control systemic inflammation. Notably, hepatic gp130-STAT3 activation was also essential for mobilization and tissue accumulation of myeloid-derived suppressor cells (MDSCs), a cell population mainly known for antiinflammatory properties in cancer. MDSCs were critical to regulate innate inflammation, and their adoptive transfer efficiently protected gp130-deficient mice from sepsis-associated mortality. The hepatic APPs serum amyloid A and Cxcl1/KC cooperatively promoted MDSC mobilization, accumulation, and survival, and reversed dysregulated inflammation and restored survival of gp130-deficient mice. Thus, gp130-dependent communication between the liver and MDSCs through APPs controls inflammatory responses during infection.
Collapse
Affiliation(s)
- Leif E. Sander
- Department of Medicine III, RWTH University Hospital, 52074 Aachen, Germany
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | | | - Uta Dierssen
- Department of Medicine III, RWTH University Hospital, 52074 Aachen, Germany
| | - Naiara Beraza
- Department of Medicine III, RWTH University Hospital, 52074 Aachen, Germany
| | | | - Michael Müller
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition, Wageningen University, 6700 EV Wageningen, Netherlands
| | | | - Frank Tacke
- Department of Medicine III, RWTH University Hospital, 52074 Aachen, Germany
| | | |
Collapse
|
268
|
Host response to influenza virus: protection versus immunopathology. Curr Opin Immunol 2010; 22:475-81. [PMID: 20594815 DOI: 10.1016/j.coi.2010.06.003] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 06/01/2010] [Indexed: 11/22/2022]
Abstract
Host responses play crucial roles in defense against influenza but sometimes these may contribute to immunopathology. Potentially, this may be more important in disease caused by viruses such as avian influenza A H5N1 or the 1918 H1N1 influenza virus rather than with seasonal influenza or pandemic H1N1 2009 (pdmH1N1). Understanding pathogenesis will help develop novel therapeutic options that minimize immunopathology without impairing beneficial host defenses.
Collapse
|
269
|
Feng H, Guo L, Song Z, Gao H, Wang D, Fu W, Han J, Li Z, Huang B, Li XA. Caveolin-1 protects against sepsis by modulating inflammatory response, alleviating bacterial burden, and suppressing thymocyte apoptosis. J Biol Chem 2010; 285:25154-60. [PMID: 20534584 DOI: 10.1074/jbc.m110.116897] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sepsis is a leading cause of death, which is characterized by uncontrolled inflammatory response. In this study, we report that caveolin-1, a major component of caveolae, is a critical survival factor of sepsis. We induced sepsis using a well established sepsis animal model, cecal ligation and puncture (CLP). CLP induced 67% fatality in caveolin-1 null mice, but only 27% fatality in wild type littermates (p = 0.015). Further studies revealed that mice deficient in caveolin-1 exhibited marked increase in tumor necrosis factor-alpha and interleukin-6 production 20 h following CLP treatment, indicating uncontrolled inflammatory responses in the absence of caveolin-1. Caveolin-1 null mice also had a significant increase in bacteria number recovered from liver and spleen, indicating elevated bacterial burdens. In addition, caveolin-1 null mice had a 2-fold increase in thymocyte apoptosis compared with wild type littermates, indicating caveolin-1 as a critical modulator of thymocyte apoptosis during sepsis. In conclusion, our findings demonstrate that caveolin-1 is a critical protective modulator of sepsis in mice. Caveolin-1 exerts its protective function likely through its roles in modulating inflammatory response, alleviating bacterial burdens, and suppressing thymocyte apoptosis.
Collapse
Affiliation(s)
- Hong Feng
- Department of Pediatrics, University of Kentucky Medical School, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
270
|
Melo ES, Goloubkova T, Barbeiro DF, Gorjão R, Vasconcelos D, Szabo C, Curi R, de Lima Salgado TM, Velasco IT, Soriano FG. Endotoxin tolerance: Selective alterations in gene expression and protection against lymphocyte death. Immunobiology 2010; 215:435-42. [DOI: 10.1016/j.imbio.2009.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 09/02/2009] [Accepted: 09/03/2009] [Indexed: 10/20/2022]
|
271
|
Strutt TM, McKinstry KK, Dibble JP, Winchell C, Kuang Y, Curtis JD, Huston G, Dutton RW, Swain SL. Memory CD4+ T cells induce innate responses independently of pathogen. Nat Med 2010; 16:558-64, 1p following 564. [PMID: 20436484 DOI: 10.1038/nm.2142] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 03/19/2010] [Indexed: 12/12/2022]
Abstract
Inflammation induced by recognition of pathogen-associated molecular patterns markedly affects subsequent adaptive responses. We asked whether the adaptive immune system can also affect the character and magnitude of innate inflammatory responses. We found that the response of memory, but not naive, CD4(+) T cells enhances production of multiple innate inflammatory cytokines and chemokines (IICs) in the lung and that, during influenza infection, this leads to early control of virus. Memory CD4(+) T cell-induced IICs and viral control require cognate antigen recognition and are optimal when memory cells are either T helper type 1 (T(H)1) or T(H)17 polarized but are independent of interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha) production and do not require activation of conserved pathogen recognition pathways. This represents a previously undescribed mechanism by which memory CD4(+) T cells induce an early innate response that enhances immune protection against pathogens.
Collapse
|
272
|
Miethke AG, Saxena V, Shivakumar P, Sabla GE, Simmons J, Chougnet CA. Post-natal paucity of regulatory T cells and control of NK cell activation in experimental biliary atresia. J Hepatol 2010; 52:718-26. [PMID: 20347178 PMCID: PMC2864543 DOI: 10.1016/j.jhep.2009.12.027] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 11/25/2009] [Accepted: 12/09/2009] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Although recent studies have identified important roles for T and NK cells in the pathogenesis of biliary atresia (BA), the mechanisms by which susceptibility to bile duct injury is restricted to the neonatal period are unknown. METHODS We characterised hepatic regulatory T cells (Tregs) by flow cytometry in two groups of neonatal mice challenged with rhesus rotavirus (RRV) at day 7 (no ductal injury) or day 1 of life (resulting in BA), determined the functional interaction with effector cells in co-culture assays, and examined the effect of adoptive transfer of CD4+ cells on the BA phenotype. RESULTS While day 7 RRV infection increased hepatic Tregs (Foxp3+ CD4+ CD25+) by 10-fold within 3 days, no increase in Tregs occurred at this time point following infection on day 1. In vitro, Tregs effectively suppressed NK cell activation by hepatic dendritic cells and decreased the production of pro-inflammatory cytokines, including TNFalpha and IL-15, following RRV infection. In vivo, adoptive transfer of CD4+ cells prior to RRV inoculation led to increased survival, improved weight gain, decreased population of hepatic NK cells, and persistence of donor Tregs in the liver. CONCLUSIONS (1) The liver is devoid of Tregs early after perinatal RRV infection; (2) Tregs suppress DC-dependent activation of naive NK cells in vitro, and Treg-containing CD4+ cells inhibit hepatic NK cell expansion in vivo. Thus, the post-natal absence of Tregs may be a key factor that allows hepatic DCs to act unopposed in NK cell activation during the initiation of neonatal bile duct injury.
Collapse
Affiliation(s)
- Alexander G. Miethke
- Corresponding author: Alexander G. Miethke, Cincinnati Children’s Hospital Medical Center, Division of Pediatric, Gastroenterology, Hepatology and Nutrition, MLC 2010, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA, Tel.: +1 513 636 9078; fax: +1 513 636 5581, (A.G. Miethke)
| | | | | | | | | | | |
Collapse
|
273
|
Adaptive control of innate immunity. Immunol Lett 2010; 131:107-12. [PMID: 20394777 DOI: 10.1016/j.imlet.2010.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 03/30/2010] [Accepted: 04/03/2010] [Indexed: 02/07/2023]
Abstract
The mechanisms by which the immune system responds to an infection or disease depend on a complex interplay between the elements of innate and adaptive immunity. While most of the focus so far has been on the innate instruction of the adaptive immune responses, considerable evidence now suggests an equally important adaptive control of the innate immunity. Several studies yield new insights into how the adaptive immunity by initiating an antigen-specific response can compensate, suppress and activate innate responses at the site of tissue antigen. Here we discuss recent advances in our understanding of the adaptive control of immune effector functions in various pathological and physiological conditions.
Collapse
|
274
|
Taylor JD. COPD and the response of the lung to tobacco smoke exposure. Pulm Pharmacol Ther 2010; 23:376-83. [PMID: 20382250 DOI: 10.1016/j.pupt.2010.04.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 03/18/2010] [Accepted: 04/03/2010] [Indexed: 11/28/2022]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a major cause of death in the western world and increasing in prevalence in developing countries. COPD is characterised by irreversible airflow obstruction, loss of lung tissue, reduced quality of life and high rates of mortality. The major cause of COPD is tobacco smoke. The changes in the innate immune system directed by tobacco smoke exposure lead to a pronounced and chronic inflammation in the lung. This in turn leads to other pathological changes including remodelling and destruction of lung tissue. Tobacco smoke exposure also leads to infection of the lung by bacteria and viruses. These, bacteria, viruses and co-infection are key triggers of acute worsening's of COPD termed exacerbations. COPD exacerbations are an additional major factor in the morbidity and mortality within COPD and are also the major healthcare costs associated with the disease. Within this review we discuss the response of the immune system to cigarette smoke exposure and inappropriate harmful responses. Successful treatment strategies will need to balance the positive effects of reducing inflammatory aspects of the disease whilst retaining some of the needed immune responses triggered by tobacco smoke exposure.
Collapse
Affiliation(s)
- John D Taylor
- Integrative Pharmacology, Biosciences R&D Lung, Respiratory & Inflammation Research Area, AstraZeneca, Lund, Sweden.
| |
Collapse
|
275
|
Maione F, Paschalidis N, Iqbal AJ, Crompton T, Perretti M, D'Acquisto F. Analysis of the inflammatory response in HY-TCR transgenic mice highlights the pathogenic potential of CD4- CD8- T cells. Autoimmunity 2010; 43:672-81. [PMID: 20370575 DOI: 10.3109/08916931003678296] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Transgenic mice expressing a rearranged T cell receptor (TCR)-αβ prematurely at the double-negative stage develop an abnormal population of peripheral T cells that lack CD4 and CD8 expression and are hyper-reactive to anti-TCR antibody stimulation. One such example is the HY-TCR transgenic mice. These mice express a TCR transgenic specific for the HY antigen that is expressed in male but not in female mice. As a result, male mice have an abnormal population of HY(+)/CD4(-)8(-) or HY(+)/CD4(-)CD8(low) T cells that are much lower in female mice. In this study, we investigated the potential patho/physiological function of these cells in vivo using a model of delayed-type hypersensitivity (DTH) reaction: the λ-carrageenan-induced paw edema. Interestingly, while both male and female HY-TCR mice develop a classical biphasic inflammatory response to λ-carrageenan, the degree of inflammation in the former was much higher than that in the latter. This was accompanied by a selective expansion of HY(+)/CD4(-)8(-) and HY(+)/CD4(-)CD8(low) T cells in male mice and by a markedly increased production of typical DTH cytokines compared with cells from female mice. These results were specific since analysis of the inflammatory response of HY-TCR transgenic mice subjected to zymosan-induced peritonitis showed no differences between male and female mice. Together, these findings provide novel evidence for the pathological role of self-reactive CD4(-)CD8(-) T cells, previously described in several autoimmune strains and recently identified in patients suffering from autoimmune diseases such as systemic lupus erythematosus.
Collapse
Affiliation(s)
- Francesco Maione
- William Harvey Research Institute, Barts and the London School of Medicine, Charterhouse Square, London, EC1M 6BQ, UK
| | | | | | | | | | | |
Collapse
|
276
|
Usharauli D. Chronic infection and the origin of adaptive immune system. Med Hypotheses 2010; 75:241-3. [PMID: 20299160 DOI: 10.1016/j.mehy.2010.02.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Accepted: 02/23/2010] [Indexed: 11/28/2022]
Abstract
It has been speculated that the rise of the adaptive immune system in jawed vertebrates some 400 million years ago gave them a superior protection to detect and defend against pathogens that became more elusive and/or virulent to the host that had only innate immune system. First, this line of thought implies that adaptive immune system was a new, more sophisticated layer of host defense that operated independently of the innate immune system. Second, the natural consequence of this scenario would be that pathogens would have exercised so strong an evolutionary pressure that eventually no host could have afforded not to have an adaptive immune system. Neither of these arguments is supported by the facts. First, new experimental evidence has firmly established that operation of adaptive immune system is critically dependent on the ability of the innate immune system to detect invader-pathogens and second, the absolute majority of animal kingdom survives just fine with only an innate immune system. Thus, these data raise the dilemma: If innate immune system was sufficient to detect and protect against pathogens, why then did adaptive immune system develop in the first place? In contrast to the innate immune system, the adaptive immune system has one important advantage, precision. By precision I mean the ability of the defense system to detect and remove the target, for example, infected cells, without causing unwanted bystander damage of surrounding tissue. While the target precision per se is not important for short-term immune response, it becomes a critical factor when the immune response is long-lasting, as during chronic infection. In this paper I would like to propose new, "toxic index" hypothesis where I argue that the need to reduce the collateral damage to the tissue during chronic infection(s) was the evolutionary pressure that led to the development of the adaptive immune system.
Collapse
Affiliation(s)
- David Usharauli
- T Cell Tolerance and Memory Section (Ghost Lab), Laboratory of Cellular and Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bldg. 4, Room 111, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| |
Collapse
|
277
|
Tang H, Fu Y. A new role for T cells in dampening innate inflammatory responses. SCIENCE CHINA-LIFE SCIENCES 2010; 53:190-4. [DOI: 10.1007/s11427-010-0040-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 01/15/2010] [Indexed: 12/13/2022]
|
278
|
Thangavelu G, Smolarchuk C, Anderson CC. Co-inhibitory molecules: Controlling the effectors or controlling the controllers? SELF NONSELF 2010; 1:77-88. [PMID: 21487510 DOI: 10.4161/self.1.2.11548] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 02/15/2010] [Accepted: 02/16/2010] [Indexed: 12/31/2022]
Abstract
Nearly forty years ago the concept was proposed that lymphocytes are negatively regulated by what are now called co-inhibitory signals. Nevertheless, it is only the more recent identification of numerous co-inhibitors and their critical functions that has brought co-inhibition to the forefront of immunologic research. Although co-inhibitory signals have been considered to directly regulate conventional T cells, more recent data has indicated a convergence between co-inhibitory signals and the other major negative control mechanism in the periphery that is mediated by regulatory T cells. Furthermore, it is now clear that lymphocytes are not the sole domain of co-inhibitory signals, as cells of the innate immune system, themselves controllers of immunity, are regulated by co-inhibitors they express. Thus, in order to better understand negative regulation in the periphery and apply this knowledge to the treatment of disease, a major focus for the future should be the definition of the conditions where co-inhibition controls effector cells intrinsically versus extrinsically (via regulatory or innate cells).
Collapse
Affiliation(s)
- Govindarajan Thangavelu
- Department of Surgery; Alberta Diabetes Institute; University of Alberta; Edmonton, Alberta Canada
| | | | | |
Collapse
|
279
|
Zavitz CCJ, Bauer CMT, Gaschler GJ, Fraser KM, Strieter RM, Hogaboam CM, Stampfli MR. Dysregulated macrophage-inflammatory protein-2 expression drives illness in bacterial superinfection of influenza. THE JOURNAL OF IMMUNOLOGY 2010; 184:2001-13. [PMID: 20065113 DOI: 10.4049/jimmunol.0903304] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Influenza virus infection is a leading cause of death and disability throughout the world. Influenza-infected hosts are vulnerable to secondary bacterial infection, however, and an ensuing bacterial pneumonia is actually the predominant cause of influenza-attributed deaths during pandemics. A number of mechanisms have been proposed by which influenza may predispose to superinfection with an unrelated or heterologous pathogen, but the subsequent interaction between the host, virus, and bacteria remains an understudied area. In this study, we develop and examine a novel model of heterologous pulmonary infection in which an otherwise subclinical Bordetella parapertussis infection synergizes with an influenza virus infection to yield a life-threatening secondary pneumonia. Despite a profound pulmonary inflammatory response and unaltered viral clearance, bacterial clearance was significantly impaired in heterologously infected mice. No deficits were observed in pulmonary or systemic adaptive immune responses or the viability or function of infiltrating inflammatory cells to explain this phenomenon, and we provide evidence that the onset of severe pulmonary inflammation actually precedes the increased bacterial burden, suggesting that exacerbated inflammation is independent of bacterial burden. To that end, neutralization of the ELR(+) inflammatory chemokine MIP-2 (CXCL2/GRO-beta) attenuated the inflammation, weight loss, and clinical presentation of heterologously infected mice without impacting bacterial burden. These data suggest that pulmonary inflammation, rather than pathogen burden, is the key threat during bacterial superinfection of influenza and that selective chemokine antagonists may be a novel therapeutic intervention in cases of bacterial superinfection of influenza.
Collapse
Affiliation(s)
- Caleb C J Zavitz
- Medical Sciences Program, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
280
|
Perreault C. The Origin and Role of MHC Class I-Associated Self-Peptides. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 92:41-60. [DOI: 10.1016/s1877-1173(10)92003-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
281
|
Melo E, Barbeiro H, Ariga S, Goloubkova T, Curi R, Velasco I, Vasconcelos D, Soriano F. Immune cells and oxidative stress in the endotoxin tolerance mouse model. Braz J Med Biol Res 2010; 43:57-67. [DOI: 10.1590/s0100-879x2009007500027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Accepted: 11/26/2009] [Indexed: 08/30/2023] Open
|
282
|
Rubin B, Gairin JE. Concepts and ways to amplify the antitumor immune response. Curr Top Microbiol Immunol 2010; 344:97-128. [PMID: 20680804 DOI: 10.1007/82_2010_89] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this chapter, a detailed description of how the innate and adaptive immune responses interact with malignant cells is presented. In addition, we discuss how developing tumors establish themselves, and how they benefit on one hand and organize their defense against the immune system on the other hand. New data from three tumor model systems in mice are discussed; in particular, the intricate interactions between the immune cells and the tumor cells are highlighted. With the present data and knowledge, we conclude that a first prerequisite for the combat against tumors is the activation of the innate immune system via external danger signals or damage signals and internal danger signals. The second prerequisite for efficient tumor cell eradication is combined therapeutic approaches of physical, chemical, pharmacological, and immunological origin. Finally, we propose new ways for further investigation of the relationship linking tumor cells and our defense system. It appears mandatory to understand how the malignant cells render the adaptive immune cells tolerant instead of turning them into aggressive effectors and memory cells. Perhaps, the most important thing, for immunologists and clinicians, to understand is that tumor cells must not be viewed just as antigens but much more.
Collapse
Affiliation(s)
- Bent Rubin
- UMR 2587 CNRS-Pierre Fabre, Institut de Sciences et Technologies du Médicament de Toulouse, 3, rue des Satellites, 31400, Toulouse, France
| | | |
Collapse
|
283
|
Mucosal perspectives in pneumococcal vaccine development: a meeting summary: a one-day international workshop focusing on stimulating research and collaborations on this topic. Vaccine 2009; 28:2-6. [PMID: 19786131 DOI: 10.1016/j.vaccine.2009.09.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 09/11/2009] [Indexed: 11/22/2022]
|
284
|
Cuff S, Dolton G, Matthews RJ, Gallimore A. Antigen specificity determines the pro- or antitumoral nature of CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2009; 184:607-14. [PMID: 20007540 DOI: 10.4049/jimmunol.0804089] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Although CD8+ T cells are usually considered antitumoral, several recent studies report that the cells can also promote tumor progression. Using the melanoma cell line B16 as a murine model of pulmonary metastasis, we examined whether the pro- versus antitumoral effects of CD8+ T cells relate to their Ag specificity. Results of the study indicate that although CD8+ T cells specific for tumor Ags promote tumor rejection, CD8+ T cells specific for unrelated Ags promote tumor progression. We found the effect to be partly attributable to CD8+ T cells dampening effective antitumor NK cell responses. Notably, activation of CD8+ T cell responses by an unrelated stimulus, in this case infection with influenza virus, increased the number of pulmonary tumor nodules. These data provide a rationale for previously unexplained data identifying contrasting roles for CD8+ T cells in tumor progression.
Collapse
Affiliation(s)
- Simone Cuff
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff, United Kingdom.
| | | | | | | |
Collapse
|
285
|
D'Alessio FR, Tsushima K, Aggarwal NR, West EE, Willett MH, Britos MF, Pipeling MR, Brower RG, Tuder RM, McDyer JF, King LS. CD4+CD25+Foxp3+ Tregs resolve experimental lung injury in mice and are present in humans with acute lung injury. J Clin Invest 2009; 119:2898-913. [PMID: 19770521 DOI: 10.1172/jci36498] [Citation(s) in RCA: 420] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Accepted: 07/15/2009] [Indexed: 01/11/2023] Open
Abstract
Acute lung injury (ALI) is characterized by rapid alveolar injury, inflammation, cytokine induction, and neutrophil accumulation. Although early events in the pathogenesis of ALI have been defined, the mechanisms underlying resolution are unknown. As a model of ALI, we administered intratracheal (i.t.) LPS to mice and observed peak lung injury 4 days after the challenge, with resolution by day 10. Numbers of alveolar lymphocytes increased as injury resolved. To examine the role of lymphocytes in this response, lymphocyte-deficient Rag-1-/- and C57BL/6 WT mice were exposed to i.t. LPS. The extent of injury was similar between the groups of mice through day 4, but recovery was markedly impaired in the Rag-1-/- mice. Adoptive transfer studies revealed that infusion of CD4+CD25+Foxp3+ Tregs as late as 24 hours after i.t. LPS normalized resolution in Rag-1-/- mice. Similarly, Treg depletion in WT mice delayed recovery. Treg transfer into i.t. LPS-exposed Rag-1-/- mice also corrected the elevated levels of alveolar proinflammatory cytokines and increased the diminished levels of alveolar TGF-beta and neutrophil apoptosis. Mechanistically, Treg-mediated resolution of lung injury was abrogated by TGF-beta inhibition. Moreover, BAL of patients with ALI revealed dynamic changes in CD3+CD4+CD25hiCD127loFoxp3+ cells. These results indicate that Tregs modify innate immune responses during resolution of lung injury and suggest potential targets for treating ALI, for which there are no specific therapies currently available.
Collapse
Affiliation(s)
- Franco R D'Alessio
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, Maryland 21224, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
286
|
Shivakumar P, Sabla GE, Whitington P, Chougnet CA, Bezerra JA. Neonatal NK cells target the mouse duct epithelium via Nkg2d and drive tissue-specific injury in experimental biliary atresia. J Clin Invest 2009; 119:2281-90. [PMID: 19662681 DOI: 10.1172/jci38879] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Biliary atresia is a neonatal obstructive cholangiopathy that progresses to end-stage liver disease. Although the etiology is unknown, a neonatal adaptive immune signature has been mechanistically linked to obstruction of the extrahepatic bile ducts. Here, we investigated the role of the innate immune response in the pathogenesis of biliary atresia. Analysis of livers of infants at diagnosis revealed that NK cells populate the vicinity of intrahepatic bile ducts and overexpress several genes involved in cytotoxicity. Using a model of rotavirus-induced biliary atresia in newborn mice, we found that activated NK cells also populated murine livers and were the most abundant cells in extrahepatic bile ducts at the time of obstruction. Rotavirus-primed hepatic NK cells lysed cholangiocytes in a contact- and Nkg2d-dependent fashion. Depletion of NK cells and blockade of Nkg2d each prevented injury of the duct epithelium after rotavirus infection, maintained continuity of duct lumen between the liver and duodenum, and enabled bile flow, despite the presence of virus in the tissue and the overexpression of proinflammatory cytokines. These findings identify NK cells as key initiators of cholangiocyte injury via Nkg2d and demonstrate that injury to the duct epithelium drives the phenotype of experimental biliary atresia.
Collapse
Affiliation(s)
- Pranavkumar Shivakumar
- Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229-3039, USA
| | | | | | | | | |
Collapse
|
287
|
Tanriver Y, Martín-Fontecha A, Ratnasothy K, Lombardi G, Lechler R. Superantigen-Activated Regulatory T Cells Inhibit the Migration of Innate Immune Cells and the Differentiation of Naive T Cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:2946-56. [DOI: 10.4049/jimmunol.0803953] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
288
|
Asp L, Beraki S, Kristensson K, Ogren SO, Karlsson H. Neonatal infection with neurotropic influenza A virus affects working memory and expression of type III Nrg1 in adult mice. Brain Behav Immun 2009; 23:733-41. [PMID: 19362585 DOI: 10.1016/j.bbi.2009.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Accepted: 04/05/2009] [Indexed: 12/28/2022] Open
Abstract
Epidemiological studies suggest that early life infections may contribute to the development of psychiatric disorders characterized by cognitive deficits. Here, we studied the effects of a neonatal influenza A/WSN/33 virus infection on locomotor activity, working memory and emotional behavior in adult mice. In addition to wild type mice, immunodeficient (Tap1(-/-)) mice lacking functional CD8(+) T cells, were included in the study to model the potential influence of a genetic deficit relating to virus clearance. Three to four months after the infection, infected Tap1(-/-) mice, but not wild type mice, exhibited deficits in working memory as well as increased rearing activity and anxiety. In the medial prefrontal cortices of these infected Tap1(-/-) mice reduced levels of type III Nrg1 transcripts were observed supporting a role for neuregulin 1 signaling in neuronal circuits involved in working memory. Virus replication, distribution or clearance did not differ between the two genotypes. The lack of CD8(+) T cells, however, appeared to contribute to a more pronounced glia response in Tap1(-/-) than in wild type mice. Thus, the present study suggest that the risk of developing deficits in cognitive and emotional behavior following a CNS infection during brain development is influenced by genetic variation in genes involved in the immune response.
Collapse
Affiliation(s)
- Linnéa Asp
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
289
|
Crozat K, Vivier E, Dalod M. Crosstalk between components of the innate immune system: promoting anti-microbial defenses and avoiding immunopathologies. Immunol Rev 2009; 227:129-49. [PMID: 19120481 DOI: 10.1111/j.1600-065x.2008.00736.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Because it reaches full functional efficacy rapidly upon encounter with a pathogen, the innate immune system is considered as the first line of defense against infections. The sensing of microbes or of transformed or infected cells, through innate immune recognition receptors (referred to as activating I2R2), initiates pro-inflammatory responses and innate immune effector functions. Other I2R2 with inhibitory properties bind self-ligands constitutively expressed in host. However, this dichotomy in the recognition of foreign or induced self versus constitutive self by I2R2 is not always respected in certain non-infectious conditions reminiscent of immunopathologies. In this review, we discuss that immune mechanisms have evolved to avoid inappropriate inflammatory disorders in individuals. Molecular crossregulation exists between components of I2R2 signaling pathways, and intricate interactions between cells from both innate and adaptive immune systems set the bases of controlled immune responses. We also pinpoint that, like T or B cells, some cells of the innate immune system must go through education processes to prevent autoreactivity. In addition, we illustrate how gene expression profiling of immune cell types is a useful tool to find functional homologies between cell subsets of different species and to speculate about unidentified functions of these cells in the responses to pathogen infections.
Collapse
Affiliation(s)
- Karine Crozat
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerranée, Marseille, France
| | | | | |
Collapse
|
290
|
Vaccination with an attenuated strain of Francisella novicida prevents T-cell depletion and protects mice infected with the wild-type strain from severe sepsis. Infect Immun 2009; 77:4314-26. [PMID: 19635830 DOI: 10.1128/iai.00654-09] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensis is the causative agent of zoonotic tularemia, a severe pneumonia in humans, and Francisella novicida causes a similarly severe tularemia in mice upon inhalation. The correlates of protective immunity, as well as the virulence mechanisms of this deadly pathogen, are not well understood. In the present study, we compared the host immune responses of lethally infected and vaccinated mice to highlight the host determinants of protection from this disease. Intranasal infection with an attenuated mutant (Mut) of F. novicida lacking a 58-kDa hypothetical protein protected C57BL/6 mice from a subsequent challenge with the fully virulent wild-type strain U112 via the same route. The protection conferred by Mut vaccination was associated with reduced bacterial burdens in systemic organs, as well as the absence of bacteremia. Also, there was reduced lung pathology and associated cell death in the lungs of vaccinated mice. Both vaccinated and nonvaccinated mice displayed an initial 2-day delay in upregulation of signature inflammatory mediators after challenge. Whereas the nonvaccinated mice developed severe sepsis characterized by hypercytokinemia and T-cell depletion, the vaccinated mice displayed moderated cytokine induction and contained increased numbers of alphabeta T cells. The recall response in vaccinated mice consisted of a characteristic Th1-type response in terms of cytokines, as well as antibody isotypes. Our results show that a regulated Th1 type of cell-mediated and humoral immunity in the absence of severe sepsis is associated with protection from respiratory tularemia, whereas a deregulated host response leading to severe sepsis contributes to mortality.
Collapse
|
291
|
Sun Y, Brown NK, Ruddy MJ, Miller ML, Lee Y, Wang Y, Murphy KM, Pfeffer K, Chen L, Kaye J, Fu YX. B and T lymphocyte attenuator tempers early infection immunity. THE JOURNAL OF IMMUNOLOGY 2009; 183:1946-51. [PMID: 19587015 DOI: 10.4049/jimmunol.0801866] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Coinhibitory pathways are thought to act in later stages of an adaptive immune response, but whether coinhibition contributes to early innate immunity is unclear. We show that engagement of the newly discovered coinhibitory receptor B and T lymphocyte attenuator (BTLA) by herpesvirus entry mediator (HVEM) is critical for negatively regulating early host immunity against intracellular bacteria. Both HVEM(-/-) and BTLA(-/-), but not LIGHT(-/-), mice are more resistant to listeriosis compared with wild-type mice, and blockade of the BTLA pathway promotes, while engagement inhibits, early bacterial clearance. Differences in bacterial clearance were seen as early as 1 day postinfection, implicating the initial innate response. Therefore, innate cell function in BTLA(-/-) mice was studied. We show that innate cells from BTLA(-/-) mice secrete significantly more proinflammatory cytokines upon stimulation with heat-killed Listeria. These results provide the first evidence that a coinhibitory pathway plays a critical role in regulating early host innate immunity against infection.
Collapse
Affiliation(s)
- Yonglian Sun
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
292
|
Hotson AN, Hardy JW, Hale MB, Contag CH, Nolan GP. The T cell STAT signaling network is reprogrammed within hours of bacteremia via secondary signals. THE JOURNAL OF IMMUNOLOGY 2009; 182:7558-68. [PMID: 19494279 DOI: 10.4049/jimmunol.0803666] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The delicate balance between protective immunity and inflammatory disease is challenged during sepsis, a pathologic state characterized by aspects of both a hyperactive immune response and immunosuppression. The events driven by systemic infection by bacterial pathogens on the T cell signaling network that likely control these responses have not been illustrated in great detail. We characterized how intracellular signaling within the immune compartment is reprogrammed at the single cell level when the host is challenged with a high level of pathogen. To accomplish this, we applied flow cytometry to measure the phosphorylation potential of key signal transduction proteins during acute bacterial challenge. We modeled the onset of sepsis by i.v. administration of avirulent strains of Listeria monocytogenes and Escherichia coli to mice. Within 6 h of bacterial challenge, T cells were globally restricted in their ability to respond to specific cytokine stimulations as determined by assessing the extent of STAT protein phosphorylation. Mechanisms by which this negative feedback response occurred included SOCS1 and SOCS3 gene up-regulation and IL-6-induced endocystosis of the IL-6 receptor. Additionally, macrophages were partially tolerized in their ability to respond to TLR agonists. Thus, in contrast to the view that there is a wholesale immune activation during sepsis, one immediate host response to blood-borne bacteria was induction of a refractory period during which leukocyte activation by specific stimulations was attenuated.
Collapse
Affiliation(s)
- Andrew N Hotson
- Department of Microbiology and Immunology, The Baxter Laboratory of Genetic Pharmacology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
293
|
Rittirsch D, Flierl MA, Day DE, Nadeau BA, Zetoune FS, Sarma JV, Werner CM, Wanner GA, Simmen HP, Huber-Lang MS, Ward PA. Cross-talk between TLR4 and FcgammaReceptorIII (CD16) pathways. PLoS Pathog 2009; 5:e1000464. [PMID: 19503602 PMCID: PMC2685003 DOI: 10.1371/journal.ppat.1000464] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Accepted: 05/04/2009] [Indexed: 01/10/2023] Open
Abstract
Pathogen-pattern-recognition by Toll-like receptors (TLRs) and pathogen clearance after immune complex formation via engagement with Fc receptors (FcRs) represent central mechanisms that trigger the immune and inflammatory responses. In the present study, a linkage between TLR4 and FcgammaR was evaluated in vitro and in vivo. Most strikingly, in vitro activation of phagocytes by IgG immune complexes (IgGIC) resulted in an association of TLR4 with FcgammaRIII (CD16) based on co-immunoprecipitation analyses. Neutrophils and macrophages from TLR4 mutant (mut) mice were unresponsive to either lipopolysaccharide (LPS) or IgGIC in vitro, as determined by cytokine production. This phenomenon was accompanied by the inability to phosphorylate tyrosine residues within immunoreceptor tyrosine-based activation motifs (ITAMs) of the FcRgamma-subunit. To transfer these findings in vivo, two different models of acute lung injury (ALI) induced by intratracheal administration of either LPS or IgGIC were employed. As expected, LPS-induced ALI was abolished in TLR4 mut and TLR4(-/-) mice. Unexpectedly, TLR4 mut and TLR4(-/-) mice were also resistant to development of ALI following IgGIC deposition in the lungs. In conclusion, our findings suggest that TLR4 and FcgammaRIII pathways are structurally and functionally connected at the receptor level and that TLR4 is indispensable for FcgammaRIII signaling via FcRgamma-subunit activation.
Collapse
MESH Headings
- Acute Lung Injury/immunology
- Acute Lung Injury/metabolism
- Analysis of Variance
- Animals
- Antigen-Antibody Complex/immunology
- Cells, Cultured
- Cytokines/metabolism
- Immunoglobulin G/immunology
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Lipopolysaccharides/immunology
- Lung/immunology
- Lung/metabolism
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Male
- Mice
- Mice, Knockout
- Phosphorylation
- Receptor Cross-Talk
- Receptor, Anaphylatoxin C5a
- Receptors, Complement/genetics
- Receptors, Complement/metabolism
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
- Signal Transduction
- Toll-Like Receptor 4/genetics
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
- Daniel Rittirsch
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Traumatology, University Hospital Zurich, Zurich, Switzerland
| | - Michael A. Flierl
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Danielle E. Day
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Brian A. Nadeau
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Firas S. Zetoune
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - J. Vidya Sarma
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Clement M. Werner
- Department of Traumatology, University Hospital Zurich, Zurich, Switzerland
| | - Guido A. Wanner
- Department of Traumatology, University Hospital Zurich, Zurich, Switzerland
| | - Hans-Peter Simmen
- Department of Traumatology, University Hospital Zurich, Zurich, Switzerland
| | - Markus S. Huber-Lang
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Peter A. Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
294
|
Kinsey GR, Sharma R, Huang L, Li L, Vergis AL, Ye H, Ju ST, Okusa MD. Regulatory T cells suppress innate immunity in kidney ischemia-reperfusion injury. J Am Soc Nephrol 2009; 20:1744-53. [PMID: 19497969 DOI: 10.1681/asn.2008111160] [Citation(s) in RCA: 284] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Both innate and adaptive mechanisms participate in the pathogenesis of kidney ischemia-reperfusion injury (IRI), but the role of regulatory immune mechanisms is unknown. We hypothesized that the anti-inflammatory effects of CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs) protect against renal IRI. Partial depletion of Tregs with an anti-CD25 mAb potentiated kidney damage induced by IRI. Reducing the number of Tregs resulted in more neutrophils, macrophages, and innate cytokine transcription in the kidney after IRI but did not affect CD4(+) T cells or B cells. We performed adoptive transfer of lymph node cells from wild-type mice or FoxP3-deficient Scurfy mice into T cell- and B cell-deficient RAG-1 knockout mice to generate mice with and without FoxP3(+) Tregs, respectively. FoxP3(+) Treg-deficient mice accumulated a greater number of inflammatory leukocytes after renal IRI than mice containing Tregs. To confirm that a lack of Tregs potentiated renal injury, we co-transferred isolated Tregs and Scurfy lymph node cells; Treg repletion significantly attenuated IRI-induced renal injury and leukocyte accumulation. Furthermore, although adoptive transfer of wild-type Tregs into RAG-1 knockout mice was sufficient to prevent kidney IRI, transfer of IL-10-deficient Tregs was not. Taken together, these results demonstrate that Tregs modulate injury after kidney IRI through IL-10-mediated suppression of the innate immune system.
Collapse
Affiliation(s)
- Gilbert R Kinsey
- Division of Nephrology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
295
|
Guarda G, Dostert C, Staehli F, Cabalzar K, Castillo R, Tardivel A, Schneider P, Tschopp J. T cells dampen innate immune responses through inhibition of NLRP1 and NLRP3 inflammasomes. Nature 2009; 460:269-73. [PMID: 19494813 DOI: 10.1038/nature08100] [Citation(s) in RCA: 201] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Accepted: 04/16/2009] [Indexed: 12/20/2022]
Abstract
Inflammation is a protective attempt by the host to remove injurious stimuli and initiate the tissue healing process. The inflammatory response must be actively terminated, however, because failure to do so can result in 'bystander' damage to tissues and diseases such as arthritis or type-2 diabetes. Yet the mechanisms controlling excessive inflammatory responses are still poorly understood. Here we show that mouse effector and memory CD4(+) T cells abolish macrophage inflammasome-mediated caspase-1 activation and subsequent interleukin 1beta release in a cognate manner. Inflammasome inhibition is observed for all tested NLRP1 (commonly called NALP1) and NLRP3 (NALP3 or cryopyrin) activators, whereas NLRC4 (IPAF) inflammasome function and release of other inflammatory mediators such as CXCL2, interleukin 6 and tumour necrosis factor are not affected. Suppression of the NLRP3 inflammasome requires cell-to-cell contact and can be mimicked by macrophage stimulation with selected ligands of the tumour necrosis factor family, such as CD40L (also known as CD40LG). In a NLRP3-dependent peritonitis model, effector CD4(+) T cells are responsible for decreasing neutrophil recruitment in an antigen-dependent manner. Our findings reveal an unexpected mechanism of inflammasome inhibition, whereby effector and memory T cells suppress potentially damaging inflammation, yet leave the primary inflammatory response, crucial for the onset of immunity, intact.
Collapse
Affiliation(s)
- Greta Guarda
- Department of Biochemistry, University of Lausanne, Chemin des Boveresses 155, CH-1066 Epalinges, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
296
|
Duffaut C, Galitzky J, Lafontan M, Bouloumié A. Unexpected trafficking of immune cells within the adipose tissue during the onset of obesity. Biochem Biophys Res Commun 2009; 384:482-5. [PMID: 19422792 DOI: 10.1016/j.bbrc.2009.05.002] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Accepted: 05/01/2009] [Indexed: 11/20/2022]
Abstract
The primary inflammatory events occurring in the adipose tissue (AT) during high fat diet (HFD)-induced obesity are poorly defined. The present study was undertaken to characterize, in wild-type(+/+) and lymphocyte deficient RAG2(-/-) mice under HFD, the changes in AT immune cells by flow cytometry analyses. In (+/+) mice, early accumulation of AT B-cells was observed, followed by increased AT T-cell numbers and finally by the appearance of insulin resistance and AT macrophage accumulation. Lack of lymphocytes in the RAG2(-/-) mice did not affect the onset of obesity and the state of insulin resistance. However, a striking accumulation of AT NK cells and activated macrophages was detected. The present study demonstrates that AT is the site of an unexpected dynamic in innate and adaptive cells during diet-induced obesity and insulin resistance. Moreover it appears that early AT lymphocyte infiltration could be considered a protective process to temper adipose tissue inflammation.
Collapse
Affiliation(s)
- Carine Duffaut
- Equipe AVENIR/Institut National de la Santé et de la Recherche Médicale (INSERM), U858, Hôpital Rangueil Bat L4, Toulouse, France.
| | | | | | | |
Collapse
|
297
|
Kuhla A, Eipel C, Abshagen K, Siebert N, Menger MD, Vollmar B. Role of the perforin/granzyme cell death pathway in D-Gal/LPS-induced inflammatory liver injury. Am J Physiol Gastrointest Liver Physiol 2009; 296:G1069-76. [PMID: 19264954 DOI: 10.1152/ajpgi.90689.2008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cytotoxic T lymphocytes and their granule components, such as perforin and granzyme, play an important role in the defense of hepatic infections caused by different pathogens. Moreover, it has been shown in vitro that hepatocytes can initiate cell death via a perforin-dependent mechanism. Although it is well known that hepatocellular apoptosis in D-galactosamine/lipopolysaccharide (D-Gal/LPS)-associated liver failure is mediated by TNF-alpha-dependent Fas/FasL cytotoxicity, there is no information on the role of perforin-mediated mechanisms in vivo. Therefore, we studied whether the cytolytic perforin/granzyme pathway contributes to the D-Gal/LPS-associated hepatotoxicity. Perforin knockout (Pko) mice showed significantly higher hepatic TNF-alpha and IL-6 mRNA expression as well as plasma TNF-alpha and IL-6 concentrations within the first hour upon D-Gal/LPS challenge compared with perforin wild-type (Pwt) mice. At 6 h upon D-Gal/LPS challenge, Pko mice further presented with higher transaminase release and onconecrotic tissue damage, whereas hepatocellular apoptosis and caspase-3 cleavage remained unaffected by the perforin deficiency. Pretreatment with a recombinant human TNF-alpha receptor fusion protein attenuated necrotic and apoptotic tissue damage and reduced plasma transaminase activities as well as cytokine release, thereby preventing acute liver failure in Pko mice as effectively as in Pwt mice. These data do not only confirm the significance of TNF-alpha as distal mediator of hepatic injury in this model but simultaneously reveal a contribution of a perforin-dependent immunoregulation, limiting the D-Gal/LPS-induced overwhelming cytokine release and onconecrotic tissue injury.
Collapse
Affiliation(s)
- Angela Kuhla
- Institute for Experimental Surgery, University of Rostock, Rostock, Germany
| | | | | | | | | | | |
Collapse
|
298
|
Jackson JA, Friberg IM, Bolch L, Lowe A, Ralli C, Harris PD, Behnke JM, Bradley JE. Immunomodulatory parasites and toll-like receptor-mediated tumour necrosis factor alpha responsiveness in wild mammals. BMC Biol 2009; 7:16. [PMID: 19386086 PMCID: PMC2685781 DOI: 10.1186/1741-7007-7-16] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Accepted: 04/22/2009] [Indexed: 01/07/2023] Open
Abstract
Background Immunological analyses of wild populations can increase our understanding of how vertebrate immune systems respond to 'natural' levels of exposure to diverse infections. A major recent advance in immunology has been the recognition of the central role of phylogenetically conserved toll-like receptors in triggering innate immunity and the subsequent recruitment of adaptive response programmes. We studied the cross-sectional associations between individual levels of systemic toll-like receptor-mediated tumour necrosis factor alpha responsiveness and macro- and microparasite infections in a natural wood mouse (Apodemus sylvaticus) population. Results Amongst a diverse group of macroparasites, only levels of the nematode Heligmosomoides polygyrus and the louse Polyplax serrata were correlated (negatively) with innate immune responsiveness (measured by splenocyte tumour necrosis factor alpha responses to a panel of toll-like receptor agonists). Polyplax serrata infection explained a strikingly high proportion of the total variation in innate responses. Contrastingly, faecal oocyst count in microparasitic Eimeria spp. was positively associated with innate immune responsiveness, most significantly for the endosomal receptors TLR7 and TLR9. Conclusion Analogy with relevant laboratory models suggests the underlying causality for the observed patterns may be parasite-driven immunomodulatory effects on the host. A subset of immunomodulatory parasite species could thus have a key role in structuring other infections in natural vertebrate populations by affecting the 'upstream' innate mediators, like toll-like receptors, that are important in initiating immunity. Furthermore, the magnitude of the present result suggests that populations free from immunosuppressive parasites may exist at 'unnaturally' elevated levels of innate immune activation, perhaps leading to an increased risk of immunopathology.
Collapse
Affiliation(s)
- Joseph A Jackson
- School of Biology, University of Nottingham, University Park, Nottingham, UK.
| | | | | | | | | | | | | | | |
Collapse
|
299
|
Abstract
The immune system defends the host against pathogenic attacks by micro-organisms and their products. It does not react against self-components due to the relatively efficient negative selection of developing T lymphocytes in the thymus. This process does permit T cells with low avidity against self to be present in the T cell repertoire. Such cells play an important physiological role as the host needs so-called autoimmune reactions in order to eliminate dying cells or transformed tumour cells. One of the mysteries in immunology is how the host maintains beneficial autoimmune reactions and avoids pathogenic autoimmune reactions. Activation of the adaptive T lymphocytes is mediated by the low avidity interaction between T-cell antigen receptors and antigenic peptides associated with major histocompatibility complex class I or class II molecules. This interaction is strengthened by T-cell co-receptors such as CD2, CD4, CD8, CD28 and CD154, which react with ligands expressed by cells of the innate immune system. In recent years, the importance of pre-activation of the innate immune system for initiation of adaptive T-cell immune responses has been appreciated. In the present review, I will summarize our work on how natural immunity plays an important role in determining the level of beneficial autoimmune reactions against cancer.
Collapse
Affiliation(s)
- B Rubin
- Institut de Science et Technologies du Médicament de Toulouse, UMR 2587 CNRS-Pierre Fabre, Rue des Satellites, Toulouse, France.
| |
Collapse
|
300
|
Sharma J, Li Q, Mishra BB, Teale JM. Lethal pulmonary infection with Francisella novicida causes depletion of alphabeta T cells from lungs. Cell Immunol 2009; 257:1-4. [PMID: 19356746 DOI: 10.1016/j.cellimm.2009.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 03/11/2009] [Accepted: 03/12/2009] [Indexed: 11/25/2022]
Abstract
Respiratory Francisella infections induce a delayed innate immune response followed by a severe sepsis like condition. In this study, mice infected intranasally with Francisella novicida showed a depletion of alphabeta T cells in lungs while exhibiting large accumulations of other leukocytes correlating with disease severity. The depleted T cells were predominantly CD4(+). The alphabeta T cells in infected mice showed significantly higher levels of Annexin V binding than those in mock control mice suggesting increased apoptosis of T cells. These results suggest that lack of transition from an innate to adaptive host response is associated with lethality of respiratory tularemia.
Collapse
Affiliation(s)
- Jyotika Sharma
- Department of Biology, South Texas Center for Emerging Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249-1644, USA
| | | | | | | |
Collapse
|