551
|
Ibrahim J, Nguyen AH, Rehman A, Ochi A, Jamal M, Graffeo CS, Henning JR, Zambirinis CP, Fallon N, Barilla R, Badar S, Mitchell A, Rao R, Acehan D, Frey AB, Miller G. Dendritic cell populations with different concentrations of lipid regulate tolerance and immunity in mouse and human liver. Gastroenterology 2012; 143:1061-72. [PMID: 22705178 PMCID: PMC3459067 DOI: 10.1053/j.gastro.2012.06.003] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 05/07/2012] [Accepted: 06/08/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Immune cells of the liver must be able to recognize and react to pathogens yet remain tolerant to food molecules and other nonpathogens. Dendritic cells (DCs) are believed to contribute to hepatic tolerance. Lipids have been implicated in dysfunction of DCs in cancer. Therefore, we investigated whether high lipid content in liver DCs affects induction of tolerance. METHODS Mouse and human hepatic nonparenchymal cells were isolated by mechanical and enzymatic digestion. DCs were purified by fluorescence-activated cell sorting or with immunomagnetic beads. DC lipid content was assessed by flow cytometry, immune fluorescence, and electron microscopy and by measuring intracellular component lipids. DC activation was determined from surface phenotype and cytokine profile. DC function was assessed in T-cell, natural killer (NK) cell, and NKT cell coculture assays as well as in vivo. RESULTS We observed 2 distinct populations of hepatic DCs in mice and humans based on their lipid content and expression of markers associated with adipogenesis and lipid metabolism. This lipid-based dichotomy in DCs was unique to the liver and specific to DCs compared with other hepatic immune cells. However, rather than mediate tolerance, the liver DC population with high concentrations of lipid was immunogenic in multiple models; they activated T cells, NK cells, and NKT cells. Conversely, liver DCs with low levels of lipid induced regulatory T cells, anergy to cancer, and oral tolerance. The immunogenicity of lipid-rich liver DCs required their secretion of tumor necrosis factor α and was directly related to their high lipid content; blocking DC synthesis of fatty acids or inhibiting adipogenesis (by reducing endoplasmic reticular stress) reduced DC immunogenicity. CONCLUSIONS Human and mouse hepatic DCs are composed of distinct populations that contain different concentrations of lipid, which regulates immunogenic versus tolerogenic responses in the liver.
Collapse
Affiliation(s)
| | | | - Adeel Rehman
- S. Arthur Localio Laboratory, Department of Surgery
| | - Atsuo Ochi
- S. Arthur Localio Laboratory, Department of Surgery
| | - Mohsin Jamal
- S. Arthur Localio Laboratory, Department of Surgery
| | | | | | | | - Nina Fallon
- S. Arthur Localio Laboratory, Department of Surgery
| | | | - Sana Badar
- S. Arthur Localio Laboratory, Department of Surgery
| | - Aaron Mitchell
- S. Arthur Localio Laboratory, Department of Cell Biology, NYU School of Medicine
| | | | - Devrim Acehan
- S. Arthur Localio Laboratory, Department of Cell Biology, NYU School of Medicine
| | - Alan B. Frey
- S. Arthur Localio Laboratory, Department of Cell Biology, NYU School of Medicine
| | - George Miller
- S. Arthur Localio Laboratory, Department of Surgery
- S. Arthur Localio Laboratory, Department of Cell Biology, NYU School of Medicine
| |
Collapse
|
552
|
Stauffer JK, Scarzello AJ, Jiang Q, Wiltrout RH. Chronic inflammation, immune escape, and oncogenesis in the liver: a unique neighborhood for novel intersections. Hepatology 2012; 56:1567-74. [PMID: 22378061 PMCID: PMC3381981 DOI: 10.1002/hep.25674] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 02/13/2012] [Indexed: 12/11/2022]
Abstract
Sustained hepatic inflammation, driven by alcohol consumption, nonalcoholic fatty liver disease, and/or chronic viral hepatitis (hepatitis B and C), results in damage to parenchyma, oxidative stress, and compensatory regeneration/proliferation. There is substantial evidence linking these inflammation-associated events with the increased incidence of hepatocellular carcinogenesis. Although acute liver inflammation can play a vital and beneficial role in response to liver damage or acute infection, the effects of chronic liver inflammation, including liver fibrosis and cirrhosis, are sufficient in a fraction of individuals to initiate the process of transformation and the development of hepatocellular carcinoma. This review highlights immune-dependent mechanisms that may be associated with hepatocellular oncogenesis, including critical transformative events/pathways in the context of chronic inflammation and subverted tolerogenesis.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/pathology
- Fatty Liver/immunology
- Fatty Liver/pathology
- Hepatitis B, Chronic/immunology
- Hepatitis B, Chronic/pathology
- Hepatitis C, Chronic/immunology
- Hepatitis C, Chronic/pathology
- Hepatitis, Chronic/immunology
- Hepatitis, Chronic/pathology
- Hepatitis, Viral, Human/immunology
- Hepatitis, Viral, Human/pathology
- Humans
- Liver Cirrhosis/immunology
- Liver Cirrhosis/pathology
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Non-alcoholic Fatty Liver Disease
- Oxidative Stress/immunology
- Oxidative Stress/physiology
- Precancerous Conditions/pathology
- Prognosis
Collapse
Affiliation(s)
| | | | | | - Robert H. Wiltrout
- Correspondence: Robert H. Wiltrout, NCI-Frederick, Bldg 428, Rm 48A, Frederick, MD 21702, Telephone:301-496-4345, Fax:301-496-0775,
| |
Collapse
|
553
|
The liver's imprint on CD8⁺ T cell priming. J Hepatol 2012; 57:718-9. [PMID: 22814455 DOI: 10.1016/j.jhep.2012.07.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 07/09/2012] [Accepted: 07/11/2012] [Indexed: 12/04/2022]
|
554
|
Murray PJ, Smale ST. Restraint of inflammatory signaling by interdependent strata of negative regulatory pathways. Nat Immunol 2012; 13:916-24. [PMID: 22990889 DOI: 10.1038/ni.2391] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Activation of Toll-like receptor (TLR) signaling and related pathways by microbial products drives inflammatory responses, host-defense pathways and adaptive immunity. The cost of excessive inflammation is cell and tissue damage, an underlying cause of many acute and chronic diseases. Coincident with activation of TLR signaling, a plethora of anti-inflammatory pathways and mechanisms begin to modulate inflammation until tissue repair is complete. Whereas most studies have focused on the signaling components immediately downstream of the TLRs, this Review summarizes the different levels of anti-inflammatory pathways that have evolved to abate TLR signaling and how they are integrated to prevent cell and tissue destruction.
Collapse
Affiliation(s)
- Peter J Murray
- Department of Infectious Diseases and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
| | | |
Collapse
|
555
|
Wilson RA. Virulence factors of schistosomes. Microbes Infect 2012; 14:1442-50. [PMID: 22982150 DOI: 10.1016/j.micinf.2012.09.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 07/18/2012] [Accepted: 09/03/2012] [Indexed: 12/25/2022]
Abstract
This review considers whether the products of schistosomes in the mammalian host can be considered as virulence factors. These include: the cercarial secretions used in infection, those of the migrating schistosomulum, surface-exposed proteins of adult worms in the portal system and their gut vomitus in the context of immune evasion, secretions of the egg facilitating its escape from gut tissues and micro-exon gene products.
Collapse
Affiliation(s)
- R Alan Wilson
- Centre for Immunology & Infection, Department of Biology, University of York, Heslington, York YO10 5DD, UK.
| |
Collapse
|
556
|
Braza F, Soulillou JP, Brouard S. Gene expression signature in transplantation tolerance. Clin Chim Acta 2012; 413:1414-8. [DOI: 10.1016/j.cca.2012.04.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 03/09/2012] [Accepted: 04/18/2012] [Indexed: 01/21/2023]
|
557
|
Stross L, Günther J, Gasteiger G, Asen T, Graf S, Aichler M, Esposito I, Busch DH, Knolle P, Sparwasser T, Protzer U. Foxp3+ regulatory T cells protect the liver from immune damage and compromise virus control during acute experimental hepatitis B virus infection in mice. Hepatology 2012; 56:873-83. [PMID: 22487943 DOI: 10.1002/hep.25765] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Accepted: 03/22/2012] [Indexed: 12/15/2022]
Abstract
UNLABELLED The strength of antiviral T cell responses correlates with clearance of hepatitis B virus (HBV) infection, but the immunological mechanisms mitigating or suppressing HBV-specific T cells are still poorly understood. In this study, we examined the role of CD4(+) Foxp3(+) regulatory T cells (Tregs) in a mouse model of acute HBV infection. We initiated HBV infection via an adenoviral vector transferring a 1.3-fold overlength HBV genome (AdHBV) into transgenic DEREG mice, where Tregs can be transiently but selectively depleted by injection of diphtheria toxin. The effect of Treg depletion on the outcome of HBV infection was characterized by detailed virological, immunological, and histopathological analysis. Numbers of Tregs increase in the liver rapidly after initiation of HBV replication. Initial depletion of Tregs revealed their complex regulatory function during acute infection. Tregs mitigated immunomediated liver damage by down-regulating the antiviral activity of effector T cells by limiting cytokine production and cytotoxicity, but did not influence development of HBV-specific CD8 T cells or development of memory T cells. Furthermore, Tregs controlled the recruitment of innate immune cells such as macrophages and dendritic cells to the infected liver. As a consequence, Tregs significantly delayed clearance of HBV from blood and infected hepatocytes. CONCLUSION Tregs limit immunomediated liver damage early after an acute infection of the liver, thereby contributing to conservation of tissue integrity and organ function at the cost of prolonging virus clearance.
Collapse
Affiliation(s)
- Leonhard Stross
- Institutes of Virology, Immunology, and Hygiene, Technische Universität München / Helmholtz Zentrum München, München, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
558
|
Gottrand G, Taleb K, Ragon I, Bergot AS, Goldstein JD, Marodon G. Intrathymic injection of lentiviral vector curtails the immune response in the periphery of normal mice. J Gene Med 2012; 14:90-9. [PMID: 22228582 DOI: 10.1002/jgm.1650] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Gene transfer in the thymus, based on HIV-derived lentiviral vectors, is a promising avenue for modulation of T cell selection and autoimmunity. However, the impact of intrathymic (IT) injections on an antigen-specific immune response elicited in the periphery of normal mice has not been investigated yet. METHODS Highly concentrated stocks of lentiviral vectors expressing the soluble form of hemaglutinin of the influenza virus (LvHA) were injected in the thymus of normal BALB/c mice. The CD4 and CD8-mediated immune responses to HA after peripheral immunization were measured by various parameters. RESULTS We first show that a lentiviral vector expressing the luciferase was detected for at least 2 months after IT-injections. We then show that the LvHA vector could elicit a functional CD4- and CD8-T cell-mediated immune responses in the peripheral lymphoid organs of BALB/c mice. IT-injection of the LvHA vector significantly curbed this response: lower numbers of transferred HA-specific CD4(+) T cells were found in LvHA-injected compared to control animals. Furthermore, lower frequencies of HA-specific CD8(+) T cells, interferon γ-producing cells and cytotoxic cells were detected from 3 weeks to 3 months in LvHA-injected mice compared to controls. However, these reduced CD8-mediated responses were not increased after depletion of CD25(+) cells in vitro or in vivo. CONCLUSIONS The results obtained in the present study show that injection of the LvHA lentiviral vector significantly curtailed the immune response to the same antigen in the periphery. Increased selection of HA-specific regulatory T cells and negative selection of HA-specific CD8(+) T cell precursors may explain the results. Our work establish the feasibility of IT-injections of lentiviral vectors to manipulate T cell tolerance in the thymus of normal mice, for basic and pre-clinical research.
Collapse
Affiliation(s)
- Gaëlle Gottrand
- Université Pierre et Marie Curie, UPMC University of Paris 06, Paris, France
| | | | | | | | | | | |
Collapse
|
559
|
Oo YH, Adams DH. Regulatory T cells and autoimmune hepatitis: defective cells or a hostile environment? J Hepatol 2012; 57:6-8. [PMID: 22522382 DOI: 10.1016/j.jhep.2012.04.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 04/12/2012] [Accepted: 04/13/2012] [Indexed: 12/04/2022]
|
560
|
Tian Z, Gershwin ME, Zhang C. Regulatory NK cells in autoimmune disease. J Autoimmun 2012; 39:206-15. [PMID: 22704425 DOI: 10.1016/j.jaut.2012.05.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 05/20/2012] [Indexed: 12/26/2022]
Abstract
As major components of innate immunity, NK cells not only exert cell-mediated cytotoxicity against tumor cells or infected cells, but also act to regulate the function of other immune cells by secretion of cytokines and chemokines, thus providing surveillance in early defense against viruses, intracellular bacteria and cancer cells. However, the effector function of NK cells must be exquisitely controlled in order to prevent inadvertent attack against self normal cells. The activity of NK cells is defined by integration of signals coming from inhibitory and activation receptors. Inhibitory receptors not only distinguish healthy from diseased cells by recognize self-MHC class I molecules on cell surfaces with "missing-self" model, but also provide an educational signal that generates functional NK cells. NK cells enrich in immunotolerance organ and recent findings of different regulatory NK cell subsets have indicated the unique role of NK cells in maintenance of homeostasis. Once the self-tolerance is broken, autoimmune response may occur. Although data has demonstrated that NK cells play important role in autoimmune disorders, NK cells seemed to act as a two edged weapon and play opposite roles with both regulatory and inducer activity even in the same disease. The precise role and regulatory mechanisms need to be further determined. In this review, we focus on recent research on the association of NK cells and antoimmune diseases, particularly the genetic correlation, the immune tolerance and misrecognition of NK cells, the regulatory function of NK cells, and their potential role in autoimmunity.
Collapse
Affiliation(s)
- Zhigang Tian
- School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.
| | | | | |
Collapse
|
561
|
van Golen RF, van Gulik TM, Heger M. The sterile immune response during hepatic ischemia/reperfusion. Cytokine Growth Factor Rev 2012; 23:69-84. [PMID: 22609105 DOI: 10.1016/j.cytogfr.2012.04.006] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 04/16/2012] [Indexed: 12/14/2022]
Abstract
Hepatic ischemia and reperfusion elicits an immune response that lacks a microbial constituent yet poses a potentially lethal threat to the host. In this sterile setting, the immune system is alarmed by endogenous danger signals that are release by stressed and dying liver cells. The detection of these immunogenic messengers by sentinel leukocyte populations constitutes the proximal trigger for a self-perpetuating cycle of inflammation, in which consecutive waves of cytokines and chemokines orchestrate the influx of various leukocyte subsets that ultimately confer tissue destruction. This review focuses on the temporal organization of sterile hepatic inflammation, using surgery-induced trauma as a template disease state.
Collapse
Affiliation(s)
- Rowan F van Golen
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
562
|
Luan X, Liao W, Lai X, He Y, Liu Y, Gong J, Li J. Dynamic Changes of Indoleamine 2,3-Dioxygenase of Kupffer Cells in Rat Liver Transplant Rejection and Tolerance. Transplant Proc 2012; 44:1045-7. [DOI: 10.1016/j.transproceed.2012.01.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
563
|
Abstract
Oral tolerance is the state of local and systemic immune unresponsiveness that is induced by oral administration of innocuous antigen such as food proteins. An analogous but more local process also regulates responses to commensal bacteria in the large intestine and, together, mucosally induced tolerance appears to prevent intestinal disorders such as food allergy, celiac disease, and inflammatory bowel diseases. Here we discuss the anatomical basis of antigen uptake and recognition in oral tolerance and highlight possible mechanisms underlying the immunosuppression. We propose a model of stepwise induction of oral tolerance in which specialized populations of mucosal dendritic cells and the unique microenvironment of draining mesenteric lymph nodes combine to generate regulatory T cells that undergo subsequent expansion in the small intestinal lamina propria. The local and systemic effects of these regulatory T cells prevent potentially dangerous hypersensitivity reactions to harmless antigens derived from the intestine and hence are crucial players in immune homeostasis.
Collapse
Affiliation(s)
- O Pabst
- Institute of Immunology, Hannover Medical School, Hannover, Germany,() or AM Mowat ()
| | - A M Mowat
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland,() or AM Mowat ()
| |
Collapse
|
564
|
Szabolcs P, Burlingham WJ, Thomson AW. Tolerance after solid organ and hematopoietic cell transplantation. Biol Blood Marrow Transplant 2012; 18:S193-200. [PMID: 22226107 DOI: 10.1016/j.bbmt.2011.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Paul Szabolcs
- Division of Blood and Marrow Transplantation and Cellular Therapies, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224, USA.
| | | | | |
Collapse
|
565
|
Matta BM, Raimondi G, Rosborough BR, Sumpter TL, Thomson AW. IL-27 production and STAT3-dependent upregulation of B7-H1 mediate immune regulatory functions of liver plasmacytoid dendritic cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:5227-37. [PMID: 22508931 DOI: 10.4049/jimmunol.1103382] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Plasmacytoid dendritic cells (pDCs) are highly specialized APCs that, in addition to their well-recognized role in anti-viral immunity, also regulate immune responses. Liver-resident pDCs are considerably less immunostimulatory than those from secondary lymphoid tissues and are equipped to promote immune tolerance/regulation through various mechanisms. IL-27 is an IL-12 family cytokine that regulates the function of both APCs and T cells, although little is known about its role in pDC immunobiology. In this study, we show that mouse liver pDCs express higher levels of IL-27p28 and EBV-induced protein 3 (Ebi3) compared with those of splenic pDCs. Both populations of pDCs express the IL-27Rα/WSX-1; however, only liver pDCs significantly upregulate expression of the coregulatory molecule B7 homolog-1 (B7-H1) in response to IL-27. Inhibition of STAT3 activation completely abrogates IL-27-induced upregulation of B7-H1 expression on liver pDCs. Liver pDCs treated with IL-27 increase the percentage of CD4(+)Foxp3(+) T cells in MLR, which is dependent upon expression of B7-H1. pDCs from Ebi3-deficient mice lacking functional IL-27 show increased capacity to stimulate allogeneic T cell proliferation and IFN-γ production in MLR. Liver but not spleen pDCs suppress delayed-type hypersensitivity responses to OVA, an effect that is lost with Ebi3(-/-) and B7-H1(-/-) liver pDCs compared with wild-type liver pDCs. These data suggest that IL-27 signaling in pDCs promotes their immunoregulatory function and that IL-27 produced by pDCs contributes to their capacity to regulate immune responses in vitro and in vivo.
Collapse
Affiliation(s)
- Benjamin M Matta
- Starzl Transplantation Institute and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
566
|
Dangi A, Sumpter TL, Kimura S, Stolz DB, Murase N, Raimondi G, Vodovotz Y, Huang C, Thomson AW, Gandhi CR. Selective expansion of allogeneic regulatory T cells by hepatic stellate cells: role of endotoxin and implications for allograft tolerance. THE JOURNAL OF IMMUNOLOGY 2012; 188:3667-77. [PMID: 22427640 DOI: 10.4049/jimmunol.1102460] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatic stellate cells (HSCs) may play an important role in hepatic immune regulation by producing numerous cytokines/chemokines and expressing Ag-presenting and T cell coregulatory molecules. Due to disruption of the endothelial barrier during cold-ischemic storage and reperfusion of liver grafts, HSCs can interact directly with cells of the immune system. Endotoxin (LPS), levels of which increase in liver diseases and transplantation, stimulates the synthesis of many mediators by HSCs. We hypothesized that LPS-stimulated HSCs might promote hepatic tolerogenicity by influencing naturally occurring immunosuppressive CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs). Following their portal venous infusion, allogeneic CD4(+) T cells, including Tregs, were found closely associated with HSCs, and this association increased in LPS-treated livers. In vitro, both unstimulated and LPS-stimulated HSCs upregulated Fas (CD95) expression on conventional CD4(+) T cells and induced their apoptosis in a Fas/Fas ligand-dependent manner. By contrast, HSCs induced Treg proliferation, which required cell-cell contact and was MHC class II-dependent. This effect was augmented when HSCs were pretreated with LPS. LPS increased the expression of MHC class II, CD80, and CD86 and stimulated the production of IL-1α, IL-1β, IL-6, IL-10 and TNF-α by HSCs. Interestingly, production of IL-1α, IL-1β, IL-6, and TNF-α was strongly inhibited, but that of IL-10 enhanced in LPS-pretreated HSC/Treg cocultures. Adoptively transferred allogeneic HSCs migrated to the secondary lymphoid tissues and induced Treg expansion in lymph nodes. These data implicate endotoxin-stimulated HSCs as important immune regulators in liver transplantation by inducing selective expansion of tolerance-promoting Tregs and reducing inflammation and alloimmunity.
Collapse
Affiliation(s)
- Anil Dangi
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
567
|
Cotugno G, Annunziata P, Barone MV, Karali M, Banfi S, Auricchio A. Impact of age at administration, lysosomal storage, and transgene regulatory elements on AAV2/8-mediated rat liver transduction. PLoS One 2012; 7:e33286. [PMID: 22428010 PMCID: PMC3302848 DOI: 10.1371/journal.pone.0033286] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 02/08/2012] [Indexed: 12/24/2022] Open
Abstract
Liver-directed gene transfer is being investigated for the treatment of systemic or liver-specific diseases. Recombinant vectors based on adeno-associated virus serotype 8 (AAV2/8) efficiently transduce liver cells allowing long term transgene expression after a single administration in animal models and in patients.We evaluated the impact on AAV2/8-mediated rat liver transduction of the following variables: i) age at vector administration, ii) presence of lysosomal storage in liver cells, and iii) regulatory elements included in the transgene expression cassette. We found that systemic administration of AAV2/8 to newborn rats results in vector genome dilution and reduced transduction efficacy when compared to adult injected animals, presumably due to hepatocyte proliferation. Accumulation of glycosaminoglycans in lysosomes does not impact on levels and distribution of AAV2/8-mediated liver transduction. Transgene expression occurs in hepatocytes but not in Kupffer or liver endothelial cells when the liver-specific thyroxine-binding-globulin promoter is used. However, extra-hepatic transduction is observed in the spleen and kidney of animals injected at birth. The use of target sequences for the hematopoietic-specific microRNA miR142-3p does not improve liver transduction efficacy neither reduce immune responses to the lysosomal enzyme arylsulfatase B. The inclusion of a variant of the Woodchuck hepatitis virus post-transcriptional regulatory element (WPRE-m) decreases AAV2/8-mediated liver transduction levels.As AAV2/8-mediated liver gene transfer is entering in the clinical arena, these data will provide relevant information to the design of efficient AAV2/8-based therapeutic strategies.
Collapse
Affiliation(s)
- Gabriella Cotugno
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Medical Genetics, Dept. of Pediatrics, “Federico II” University, Naples, Italy
| | - Patrizia Annunziata
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Medical Genetics, Dept. of Pediatrics, “Federico II” University, Naples, Italy
| | - Maria Vittoria Barone
- Department of Pediatrics and European Laboratory for the Investigation of Food-Induced Diseases, “Federico II” University, Naples, Italy
| | | | - Sandro Banfi
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Medical Genetics, Dept. of General Pathology, Second University of Naples, Naples, Italy
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Medical Genetics, Dept. of Pediatrics, “Federico II” University, Naples, Italy
| |
Collapse
|
568
|
Kutscher S, Bauer T, Dembek C, Sprinzl M, Protzer U. Design of therapeutic vaccines: hepatitis B as an example. Microb Biotechnol 2012; 5:270-82. [PMID: 21958338 PMCID: PMC3815787 DOI: 10.1111/j.1751-7915.2011.00303.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 08/22/2011] [Accepted: 08/22/2011] [Indexed: 12/18/2022] Open
Abstract
Therapeutic vaccines are currently developed for chronic viral infections, such as human papillomavirus (HPV), human immunodeficiency virus (HIV), herpesvirus and hepatitis B (HBV) and C (HCV) virus infections. As an alternative to antiviral treatment or to support only partially effective therapy a therapeutic vaccine shall activate the patient's immune system to fight and finally control or ideally even eliminate the virus. Whereas the success of prophylactic vaccination is based on rapid neutralization of the invading pathogen by antibodies, virus control and elimination of infected cells require T cells. Therefore, induction of a multi-specific and multifunctional T-cell response against key viral antigens is a paradigm of therapeutic vaccination--besides activation of a humoral immune response to limit virus spread. In this review, we describe options to develop a therapeutic vaccine for chronic viral infections using HBV as a promising example.
Collapse
Affiliation(s)
| | | | | | | | - Ulrike Protzer
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Trogerstr. 30, 81675 München, Germany
| |
Collapse
|
569
|
Razakandrainibe R, Pelleau S, Grau GE, Jambou R. Antigen presentation by endothelial cells: what role in the pathophysiology of malaria? Trends Parasitol 2012; 28:151-60. [PMID: 22365903 DOI: 10.1016/j.pt.2012.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 01/20/2012] [Accepted: 01/20/2012] [Indexed: 11/19/2022]
Abstract
Disruption of the endothelial cell (EC) barrier leads to pathology via edema and inflammation. During infections, pathogens are known to invade the EC barrier and modulate vascular permeability. However, ECs are semi-professional antigen-presenting cells, triggering T-cell costimulation and specific immune-cell activation. This in turn leads to the release of inflammatory mediators and the destruction of infected cells by effectors such as CD8(+) T-cells. During malaria, transfer of parasite antigens to the EC surface is now established. At the same time, CD8 activation seems to play a major role in cerebral malaria. We summarize here some of the pathways leading to antigen presentation by ECs and address the involvement of these mechanisms in the pathophysiology of cerebral malaria.
Collapse
|
570
|
Abstract
The liver has vital metabolic and clearance functions that involve the uptake of nutrients, waste products and pathogens from the blood. In addition, its unique immunoregulatory functions mediated by local expression of co-inhibitory receptors and immunosuppressive mediators help to prevent inadvertent organ damage. However, these tolerogenic properties render the liver an attractive target site for pathogens. Although most pathogens that reach the liver via the blood are eliminated or controlled by local innate and adaptive immune responses, some pathogens (such as hepatitis viruses) can escape immune control and persist in hepatocytes, causing substantial morbidity and mortality worldwide. Here, we review our current knowledge of the mechanisms of liver targeting by pathogens and describe the interplay between pathogens and host factors that promote pathogen elimination and maintain organ integrity or that allow pathogen persistence.
Collapse
|
571
|
Defining early human NK cell developmental stages in primary and secondary lymphoid tissues. PLoS One 2012; 7:e30930. [PMID: 22319595 PMCID: PMC3272048 DOI: 10.1371/journal.pone.0030930] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 12/26/2011] [Indexed: 11/19/2022] Open
Abstract
A better understanding of human NK cell development in vivo is crucial to exploit NK cells for immunotherapy. Here, we identified seven distinctive NK cell developmental stages in bone marrow of single donors using 10-color flow cytometry and found that NK cell development is accompanied by early expression of stimulatory co-receptor CD244 in vivo. Further analysis of cord blood (CB), peripheral blood (PB), inguinal lymph node (inLN), liver lymph node (liLN) and spleen (SPL) samples showed diverse distributions of the NK cell developmental stages. In addition, distinctive expression profiles of early development marker CD33 and C-type lectin receptor NKG2A between the tissues, suggest that differential NK cell differentiation may take place at different anatomical locations. Differential expression of NKG2A and stimulatory receptors (e.g. NCR, NKG2D) within the different subsets of committed NK cells demonstrated the heterogeneity of the CD56(bright)CD16⁺/⁻ and CD56(dim)CD16⁺ subsets within the different compartments and suggests that microenvironment may play a role in differential in situ development of the NK cell receptor repertoire of committed NK cells. Overall, differential in situ NK cell development and trafficking towards multiple tissues may give rise to a broad spectrum of mature NK cell subsets found within the human body.
Collapse
|
572
|
Nakamoto N, Ebinuma H, Kanai T, Chu PS, Ono Y, Mikami Y, Ojiro K, Lipp M, Love PE, Saito H, Hibi T. CCR9+ macrophages are required for acute liver inflammation in mouse models of hepatitis. Gastroenterology 2012; 142:366-76. [PMID: 22079594 DOI: 10.1053/j.gastro.2011.10.039] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 10/25/2011] [Accepted: 10/26/2011] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Antigen-presenting cells (APCs) are involved in the induction of liver inflammation. We investigated the roles of specific APCs in the pathogenesis of acute liver injury in mice. METHODS We used concanavalin A (con A) or carbon tetrachloride to induce acute liver inflammation in mice and studied the roles of macrophages that express CCR9. RESULTS After injection of con A, we detected CCR9(+)CD11b(+)CD11c(-) macrophages that express tumor necrosis factor (TNF)-α in livers of mice, whereas CCR9(+)Siglec-H(+)CD11b(-)CD11c(low) plasmacytoid DCs (pDCs), which are abundant in normal livers, disappeared. The CCR9(+) macrophages were also detected in the livers of RAG-2(-/-) mice, which lack lymphocytes and natural killer T cells, after injection of con A. Under inflammatory conditions, CCR9(+) macrophages induced naive CD4(+) T cells to become interferon gamma-producing Th1 cells in vivo and in vitro. CCR9(-/-) mice injected with con A did not develop hepatitis unless they also received CCR9(+) macrophages from mice that received con A; more CCR9(+) macrophages accumulated in their inflamed livers than CCR9(+) pDCs, CCR9(-) pDCs, or CCR9(-) macrophages isolated from mice that had received injections of con A. Levels of CCL25 messenger RNA increased in livers after injection of con A; neutralizing antibodies against CCL25 reduced the induction of hepatitis by con A by blocking the migration of CCR9(+) macrophages and their production of TNF-α. Peripheral blood samples from patients with acute hepatitis had greater numbers of TNF-α-producing CCR9(+)CD14(+)CD16(high) monocytes than controls. CONCLUSIONS CCR9(+) macrophages contribute to the induction of acute liver inflammation in mouse models of hepatitis.
Collapse
Affiliation(s)
- Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
573
|
Chinnadurai R, Velazquez V, Grakoui A. Hepatic transplant and HCV: a new playground for an old virus. Am J Transplant 2012; 12:298-305. [PMID: 22044693 DOI: 10.1111/j.1600-6143.2011.03812.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Hepatitis C virus (HCV) infection is a major global health problem affecting 170 million people worldwide. The majority of infected individuals fail to resolve their infection, with a significant number developing chronic, progressive HCV-related liver disease. HCV infection is the leading indication for liver transplantation and unfortunately, all patients with detectable viral load before transplantation will have rapid, recurrent infection. What remain to be determined are factors contributing to the severity of HCV recurrence. Such factors are unique to the posttransplant setting and include: viral genetic diversity and composition, immunosuppression, donor/recipient age and sex, genetic factors and the liver microenvironment. Importantly, the possibility that the severity of HCV recurrence might be also influenced by factors related to the primary course of disease (i.e. viral set point, previously acquired adaptations of the virus) must be further evaluated. In this sense, recurrent HCV infection should not be regarded merely as another acute infection, but rather, it should be cautioned that problems first arising during the primary course of disease may be accentuated during recurrence. Development of novel therapeutic approaches will require a thorough understanding of viral and host determinants of infection resolution and how these factors may change in the posttransplant setting.
Collapse
Affiliation(s)
- R Chinnadurai
- Department of Medicine, Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | | | | |
Collapse
|
574
|
Hösel M, Broxtermann M, Janicki H, Esser K, Arzberger S, Hartmann P, Gillen S, Kleeff J, Stabenow D, Odenthal M, Knolle P, Hallek M, Protzer U, Büning H. Toll-like receptor 2-mediated innate immune response in human nonparenchymal liver cells toward adeno-associated viral vectors. Hepatology 2012; 55:287-97. [PMID: 21898480 DOI: 10.1002/hep.24625] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED Adeno-associated viral vectors (rAAV) are frequently used in gene therapy trials. Although rAAV vectors are of low immunogenicity, humoral as well as T cell responses may be induced. While the former limits vector reapplication, the expansion of cytotoxic T cells correlates with liver inflammation and loss of transduced hepatocytes. Because adaptive immune responses are a consequence of recognition by the innate immune system, we aimed to characterize cell autonomous immune responses elicited by rAAV in primary human hepatocytes and nonparenchymal liver cells. Surprisingly, Kupffer cells, but also liver sinusoidal endothelial cells, mounted responses to rAAV, whereas neither rAAV2 nor rAAV8 were recognized by hepatocytes. Viral capsids were sensed at the cell surface as pathogen-associated molecular patterns by Toll-like receptor 2. In contrast to the Toll-like receptor 9-mediated recognition observed in plasmacytoid dendritic cells, immune recognition of rAAV in primary human liver cells did not induce a type I interferon response, but up-regulated inflammatory cytokines through activation of nuclear factor κB. CONCLUSION Using primary human liver cells, we identified a novel mechanism of rAAV recognition in the liver, demonstrating that alternative means of sensing rAAV particles have evolved. Minimizing this recognition will be key to improving rAAV-mediated gene transfer and reducing side effects in clinical trials due to immune responses against rAAV.
Collapse
Affiliation(s)
- Marianna Hösel
- Department I of Internal Medicine, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
575
|
Bohne F, Martínez-Llordella M, Lozano JJ, Miquel R, Benítez C, Londoño MC, Manzia TM, Angelico R, Swinkels DW, Tjalsma H, López M, Abraldes JG, Bonaccorsi-Riani E, Jaeckel E, Taubert R, Pirenne J, Rimola A, Tisone G, Sánchez-Fueyo A. Intra-graft expression of genes involved in iron homeostasis predicts the development of operational tolerance in human liver transplantation. J Clin Invest 2012; 122:368-382. [PMID: 22156196 PMCID: PMC3248302 DOI: 10.1172/jci59411] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 10/26/2011] [Indexed: 12/13/2022] Open
Abstract
Following organ transplantation, lifelong immunosuppressive therapy is required to prevent the host immune system from destroying the allograft. This can cause severe side effects and increased recipient morbidity and mortality. Complete cessation of immunosuppressive drugs has been successfully accomplished in selected transplant recipients, providing proof of principle that operational allograft tolerance is attainable in clinical transplantation. The intra-graft molecular pathways associated with successful drug withdrawal, however, are not well defined. In this study, we analyzed sequential blood and liver tissue samples collected from liver transplant recipients enrolled in a prospective multicenter immunosuppressive drug withdrawal clinical trial. Before initiation of drug withdrawal, operationally tolerant and non-tolerant recipients differed in the intra-graft expression of genes involved in the regulation of iron homeostasis. Furthermore, as compared with non-tolerant recipients, operationally tolerant patients exhibited higher serum levels of hepcidin and ferritin and increased hepatocyte iron deposition. Finally, liver tissue gene expression measurements accurately predicted the outcome of immunosuppressive withdrawal in an independent set of patients. These results point to a critical role for iron metabolism in the regulation of intra-graft alloimmune responses in humans and provide a set of biomarkers to conduct drug-weaning trials in liver transplantation.
Collapse
Affiliation(s)
- Felix Bohne
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Marc Martínez-Llordella
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Juan-José Lozano
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Rosa Miquel
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Carlos Benítez
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - María-Carlota Londoño
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Tommaso-María Manzia
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Roberta Angelico
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Dorine W. Swinkels
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Harold Tjalsma
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Marta López
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Juan G. Abraldes
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Eliano Bonaccorsi-Riani
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Elmar Jaeckel
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Richard Taubert
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Antoni Rimola
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Giuseppe Tisone
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Alberto Sánchez-Fueyo
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
576
|
Song MG, Kang B, Jeon JY, Chang J, Lee S, Min CK, Youn H, Choi EY. In vivo imaging of differences in early donor cell proliferation in graft-versus-host disease hosts with different pre-conditioning doses. Mol Cells 2012; 33:79-86. [PMID: 22228184 PMCID: PMC3887749 DOI: 10.1007/s10059-012-2228-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 10/18/2011] [Accepted: 10/19/2011] [Indexed: 01/28/2023] Open
Abstract
Graft-versus-host disease (GVHD) results from immunemediated attacks on recipient tissues by donor-originated cells through the recognition of incompatible antigens expressed on host cells. The pre-conditioning irradiation dose is a risk factor influencing GVHD severity. In this study, using newly generated luciferase transgenic mice on a B6 background (B6.Luc(Tg)) as bone marrow and splenocyte donors, we explored the effects of irradiation doses on donor cell dynamics in major histocompatibility complex (MHC)-matched allogeneic GVHD hosts via bioluminescence imaging (BLI). Results from BLI of GVHD hosts showed higher emission intensities of luminescence signals from hosts irradiated with 900 cGy as compared with those irradiated with 400 cGy. In particular, BLI signals from target organs, such as the spleen, liver, and lung, and several different lymph nodes fluctuated with similar time kinetics soon after transplantation, reflecting the synchronous proliferation of donor cells in the different organs in hosts irradiated with 900 cGy. The kinetic curves of the BLI signals were not synchronized between the target organs and the secondary organs in hosts irradiated with 400 cGy. These results demonstrate that pre-conditioning doses influence the kinetics and degree of proliferation in the target organs soon after transplantation. The results from this study are the first describing donor cell dynamics in MHC-matched allogeneic GVHD hosts and the influence of irradiation doses on proliferation dynamics, and will provide spatiotemporal information to help understand GVHD pathophysiology.
Collapse
Affiliation(s)
- Myung Geun Song
- Department of Tumor Biology, Seoul National University College of Medicine, Seoul 110-799,
Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul 110-799,
Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799,
Korea
| | - Bora Kang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799,
Korea
| | - Ji Yeong Jeon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799,
Korea
| | - Jun Chang
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750,
Korea
| | - Seungbok Lee
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul 110-749,
Korea
| | - Chang-Ki Min
- Department of Internal Medicine, The Catholic University of Korea College of Medicine, St. Mary’s Hospital, Seoul 137-701,
Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul 110-799,
Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799,
Korea
- Cancer Imaging Center, Seoul National University Cancer Hospital, Seoul 110-799,
Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799,
Korea
| |
Collapse
|
577
|
Bode JG, Albrecht U, Häussinger D, Heinrich PC, Schaper F. Hepatic acute phase proteins--regulation by IL-6- and IL-1-type cytokines involving STAT3 and its crosstalk with NF-κB-dependent signaling. Eur J Cell Biol 2011; 91:496-505. [PMID: 22093287 DOI: 10.1016/j.ejcb.2011.09.008] [Citation(s) in RCA: 294] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 09/15/2011] [Accepted: 09/15/2011] [Indexed: 12/16/2022] Open
Abstract
The function of the liver as an important constituent of the immune system involved in innate as well as adaptive immunity is warranted by different highly specialized cell populations. As the major source of acute phase proteins, including secreted pathogen recognition receptors (PRRs), short pentraxins, components of the complement system or regulators of iron metabolism, hepatocytes are essential constituents of innate immunity and largely contribute to the control of a systemic inflammatory response. The production of acute phase proteins in hepatocytes is controlled by a variety of different cytokines released during the inflammatory process with IL-1- and IL-6-type cytokines as the leading regulators operating both as a cascade and as a network having additive, inhibitory, or synergistic regulatory effects on acute phase protein expression. Hence, IL-1β substantially modifies IL-6-induced acute phase protein production as it almost completely abrogates production of acute phase proteins such as γ-fibrinogen, α(2)-macroglobulin or α(1)-antichymotrypsin, whereas production of for example hepcidin, C-reactive protein and serum amyloid A is strongly up-regulated. This switch-like regulation of IL-6-induced acute phase protein production by IL-1β is due to a complex processing of the intracellular signaling events activated in response to IL-6 and/or IL-1β, with the crosstalk between STAT3- and NF-κB-mediated signal transduction being of particular importance. Recent data suggest that in this context complex formation between STAT3 and the p65 subunit of NF-κB might be of key importance. The present review summarizes the regulation of acute phase protein production focusing on the role of the crosstalk of STAT3- and NF-κB-driven pathways for transcriptional control of acute phase gene expression.
Collapse
Affiliation(s)
- Johannes G Bode
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital, Medical Faculty, Heinrich-Heine University, Moorenstraße 5, D-40225 Düsseldorf, Germany.
| | | | | | | | | |
Collapse
|
578
|
Nace G, Evankovich J, Eid R, Tsung A. Dendritic cells and damage-associated molecular patterns: endogenous danger signals linking innate and adaptive immunity. J Innate Immun 2011; 4:6-15. [PMID: 22086146 DOI: 10.1159/000334245] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2011] [Accepted: 10/10/2011] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells critical in regulating the adaptive immune response. The role of DCs is dichotomous; they may both present antigens and the appropriate stimulatory molecules to initiate an adaptive immune response, or they may induce tolerance and release anti-inflammatory signals. The activation of immature DCs, required for the expression of the necessary costimulatory T cell molecules, is dependent on pattern recognition receptors. In addition to the pathogen-derived ligands of pattern recognition receptors, several damage-associated molecular patterns (DAMPs) have recently been shown to interact with DCs and dramatically affect their ultimate function. The complex interplay of DAMPs on DCs is clinically important, with implications for transplantation, tumor immunity, autoimmunity, chronic inflammation and other conditions of sterile inflammation such as ischemia reperfusion injury. In this review, we will focus on the role of DAMPs in DC function.
Collapse
Affiliation(s)
- Gary Nace
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
579
|
Abstract
In the steady state, hepatic antigen (Ag)-presenting cells (APC) generally dampen systemic inflammatory responses to gut-derived Ags. Our studies focus on the role of specific liver APC populations, both non-parenchymal cells (dendritic cells [DC], Kupffer cells, and hepatic stellate cells [HSC]) and parenchymal cells, in the molecular regulation of tissue damage (ischemia and reperfusion [I/R] injury) and immunity following liver transplantation. We focus on factors that either promote or overwhelm the natural tendency of the liver to suppress inflammatory/immune responses. We are also examining molecular mechanisms that regulate liver DC maturation and function and that determine their role in the control of allogeneic T-cell function and the fate of the transplanted liver. Our studies are also aimed at elucidating mechanisms by which HSC regulate DC and T-cell function. These investigations may provide new targets for therapeutic intervention in liver inflammation.
Collapse
|
580
|
Meneses-Ruiz DM, Laclette JP, Aguilar-Díaz H, Hernández-Ruiz J, Luz-Madrigal A, Sampieri A, Vaca L, Carrero JC. Mucosal delivery of ACNPV baculovirus driving expression of the Gal-lectin LC3 fragment confers protection against amoebic liver abscess in hamster. Int J Biol Sci 2011; 7:1345-56. [PMID: 22110386 PMCID: PMC3221370 DOI: 10.7150/ijbs.7.1345] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/01/2011] [Indexed: 12/23/2022] Open
Abstract
Mucosal vaccination against amoebiasis using the Gal-lectin of E. histolytica has been proposed as one of the leading strategies for controlling this human disease. However, most mucosal adjuvants used are toxic and the identification of safe delivery systems is necessary. Here, we evaluate the potential of a recombinant Autographa californica baculovirus driving the expression of the LC3 fragment of the Gal-lectin to confer protection against amoebic liver abscess (ALA) in hamsters following oral or nasal immunization. Hamsters immunized by oral route showed complete absence (57.9%) or partial development (21%) of ALA, resulting in some protection in 78.9% of animals when compared with the wild type baculovirus and sham control groups. In contrast, nasal immunization conferred only 21% of protection efficacy. Levels of ALA protection showed lineal correlation with the development of an anti-amoebic cellular immune response evaluated in spleens, but not with the induction of seric IgG anti-amoeba antibodies. These results suggest that baculovirus driving the expression of E. histolytica vaccine candidate antigens is useful for inducing protective cellular and humoral immune responses following oral immunization, and therefore it could be used as a system for mucosal delivery of an anti-amoebic vaccine.
Collapse
Affiliation(s)
- D M Meneses-Ruiz
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México. A.P. 70228, México D.F., México
| | | | | | | | | | | | | | | |
Collapse
|
581
|
Abstract
UNLABELLED Antigen cross-presentation is a principal function of specialized antigen-presenting cells of bone marrow origin such as dendritic cells. Although these cells are sometimes known as "professional" antigen-presenting cells, nonbone marrow-derived cells may also act as antigen-presenting cells. Here, using four-way liver cell isolation and parallel comparison of candidate antigen-presenting cells, we show that, depending on the abundance of antigen-donor cells, different subsets of liver cells could cross-present a hepatocyte-associated antigen. This function was observed in both liver sinusoidal endothelial cells and Kupffer cells even at very low antigen concentration, as well as when using soluble protein. Antigen cross-presentation by liver cells induced efficient CD8+ T-cell proliferation in a similar manner to classical dendritic cells from spleen. However, proliferated cells expressed a lower level of T-cell activation markers and intracellular interferon-gamma levels. In contrast to classical spleen dendritic cells, cross-presentation by liver antigen-presenting cells was predominantly dependent on intercellular adhesion molecule-1. CONCLUSION Hepatic sinusoids are an environment rich in antigen cross-presenting activity. However, the liver's resident antigen-presenting cells cause partial T-cell activation. These results clarify how the liver can act as a primary site of CD8+ T-cell activation, and why immunity against hepatocyte pathogens is sometimes ineffective.
Collapse
Affiliation(s)
- Mohammad R Ebrahimkhani
- Malaria Program, Seattle Biomedical Research Institute, 307 Westlake Avenue N., Seattle, WA 98109, USA
- MIT Center for Environmental Health Sciences, Department of Biological Engineering, Department of Biology, Massachusetts Institute of Technology (MIT), 77 Massachusetts Avenue, 56-255, Cambridge, MA 02139, USA
| | - Isaac Mohar
- Malaria Program, Seattle Biomedical Research Institute, 307 Westlake Avenue N., Seattle, WA 98109, USA
| | - Ian N Crispe
- Malaria Program, Seattle Biomedical Research Institute, 307 Westlake Avenue N., Seattle, WA 98109, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| |
Collapse
|
582
|
Pessayre D, Fromenty B, Berson A, Robin MA, Lettéron P, Moreau R, Mansouri A. Central role of mitochondria in drug-induced liver injury. Drug Metab Rev 2011; 44:34-87. [PMID: 21892896 DOI: 10.3109/03602532.2011.604086] [Citation(s) in RCA: 191] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A frequent mechanism for drug-induced liver injury (DILI) is the formation of reactive metabolites that trigger hepatitis through direct toxicity or immune reactions. Both events cause mitochondrial membrane disruption. Genetic or acquired factors predispose to metabolite-mediated hepatitis by increasing the formation of the reactive metabolite, decreasing its detoxification, or by the presence of critical human leukocyte antigen molecule(s). In other instances, the parent drug itself triggers mitochondrial membrane disruption or inhibits mitochondrial function through different mechanisms. Drugs can sequester coenzyme A or can inhibit mitochondrial β-oxidation enzymes, the transfer of electrons along the respiratory chain, or adenosine triphosphate (ATP) synthase. Drugs can also destroy mitochondrial DNA, inhibit its replication, decrease mitochondrial transcripts, or hamper mitochondrial protein synthesis. Quite often, a single drug has many different effects on mitochondrial function. A severe impairment of oxidative phosphorylation decreases hepatic ATP, leading to cell dysfunction or necrosis; it can also secondarily inhibit ß-oxidation, thus causing steatosis, and can also inhibit pyruvate catabolism, leading to lactic acidosis. A severe impairment of β-oxidation can cause a fatty liver; further, decreased gluconeogenesis and increased utilization of glucose to compensate for the inability to oxidize fatty acids, together with the mitochondrial toxicity of accumulated free fatty acids and lipid peroxidation products, may impair energy production, possibly leading to coma and death. Susceptibility to parent drug-mediated mitochondrial dysfunction can be increased by factors impairing the removal of the toxic parent compound or by the presence of other medical condition(s) impairing mitochondrial function. New drug molecules should be screened for possible mitochondrial effects.
Collapse
Affiliation(s)
- Dominique Pessayre
- INSERM, U, Centre de Recherche Bichat Beaujon CRB, Faculté de Médecine Xavier-Bichat, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
583
|
Ryan EJ, O'Farrelly C. The affect of chronic hepatitis C infection on dendritic cell function: a summary of the experimental evidence. J Viral Hepat 2011; 18:601-7. [PMID: 21794024 DOI: 10.1111/j.1365-2893.2011.01453.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chronic hepatitis C virus (HCV) infection occurs in patients who fail to mount an effective T-cell response against the virus. One hypothesis for poor anti-viral immunity in these patients is that the virus impedes the immune response by disabling dendritic cells (DCs), cells that play a key role in pathogen recognition and initiation of adaptive immunity. Initial studies in the 1990s supported this hypothesis, as they clearly demonstrated that monocyte-derived DCs obtained from patients with chronic HCV infection displayed a reduced ability to stimulate lymphocyte proliferation. However, over the last 20 years, the situation has become more ambiguous. Many studies support the initial observation of a DC defect, while others using different patient cohorts or technologies have clearly demonstrated intact DC function in patients with chronic HCV. It is likely that the true situation lies somewhere in between. Just as there is a spectrum of disease in patients with chronic HCV, DCs obtained from different patients may display different properties. It is important to reconcile these divergent findings, as a clearer understanding of how the virus affects DC function will facilitate the development of immunotherapy and therapeutic vaccination strategies for patients with chronic HCV infection.
Collapse
Affiliation(s)
- E J Ryan
- School of Biochemistry and Immunology, Trinity College, Dublin 2, Ireland.
| | | |
Collapse
|
584
|
Abstract
One of the most fundamental problems in immunology is the seemingly schizophrenic ability of the immune system to launch robust immunity against pathogens, while acquiring and maintaining a state of tolerance to the body's own tissues and the trillions of commensal microorganisms and food antigens that confront it every day. A fundamental role for the innate immune system, particularly dendritic cells (DCs), in orchestrating immunological tolerance has been appreciated, but emerging studies have highlighted the nature of the innate receptors and the signaling pathways that program DCs to a tolerogenic state. Furthermore, several studies have emphasized the major role played by cellular interactions and the microenvironment in programming tolerogenic DCs. Here, we review these studies and suggest that the innate control of tolerogenic responses can be viewed as different hierarchies of organization, in which DCs, their innate receptors and signaling networks, and their interactions with other cells and local microenvironments represent different levels of the hierarchy.
Collapse
Affiliation(s)
- Santhakumar Manicassamy
- Emory Vaccine Center, Yerkes National Primate Research Center, Department of Pathology, Emory University School of Medicine, Atlanta, GA 30329, USA
| | | |
Collapse
|
585
|
Zhao Q, Xiao X, Wu Y, Wei Y, Zhu LY, Zhou J, Kuang DM. Interleukin-17-educated monocytes suppress cytotoxic T-cell function through B7-H1 in hepatocellular carcinoma patients. Eur J Immunol 2011; 41:2314-22. [PMID: 21674477 DOI: 10.1002/eji.201041282] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Revised: 05/11/2011] [Accepted: 05/13/2011] [Indexed: 01/01/2023]
Abstract
Substantial evidence indicates that inflammation is a critical component of tumor progression. The proinflammatory IL-17-producing cells have recently been detected in tumors, but the effect of IL-17 on antigen-presenting cells in tumors is presently unknown. We recently found that B7-H1(+) macrophages (Mφs) were enriched predominantly in the peritumoral stroma of hepatocellular carcinomas (HCCs). Here, we found a positive correlation between IL-17-producing cells and B7-H1-expressing Mφs in the same area. The B7-H1(+) monocytes/Mφs from HCC tissues expressed significantly more HLA-DR, CD80, and CD86 than B7-H1(-) cells. Accordingly, IL-17 could activate monocytes to express B7-H1 in a dose-dependent manner. Although culture supernatants derived from hepatoma cells also induced B7-H1 expression on monocytes, IL-17 additionally increased hepatoma-mediated B7-H1 expression. Autocrine inflammatory cytokines released from IL-17-activated monocytes stimulated B7-H1 expression. Moreover, these IL-17-exposed monocytes effectively suppressed cytotoxic T-cell immunity in vitro; the effect could be reversed by blocking B7-H1 on those monocytes. Consistent with this, cytotoxic T cells from HCC tissues expressed significant B7-H1 receptor programmed death 1 (PD-1) and exhibited an exhausted phenotype. These data reveal a fine-tuned collaborative action between different stromal cells to counteract T-cell responses in tumors. Such IL-17-mediated immune tolerance should be considered for the rational design of effective immune-based anti-cancer therapies.
Collapse
Affiliation(s)
- Qiyi Zhao
- School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
586
|
Cardenas PA, Huang Y, Ildstad ST. The role of pDC, recipient T(reg) and donor T(reg) in HSC engraftment: Mechanisms of facilitation. CHIMERISM 2011; 2:65-70. [PMID: 22163063 DOI: 10.4161/chim.2.3.17588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 06/20/2011] [Accepted: 08/01/2011] [Indexed: 11/19/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) has been utilized for treatment of many hematologic malignancies, genetic and metabolic disorders, and hemoglobinopathies such as sickle cell disease and thalassemia. It also induces donor-specific tolerance to organ and tissue transplants. The widespread success of HSCT is hampered by the toxicities of immunosuppression and development of graft-versus-host disease (GVHD). The mechanism of induction of transplantation tolerance (reciprocal donor/host) is still an elusive challenge in allogeneic HSCT. An understanding of the mechanisms for induction of tolerance and the critical cells involved in this process has resulted in novel cell-based therapies poised to be translated to clinical application. The focus of this review is those cells of interest.Bone marrow-derived plasmacytoid dendritic cells induce naïve T cells to differentiate to become antigen-specific regulatory T cells (T(reg)), creating a milieu for the induction of transplantation tolerance. Recently, CD8(+)/TCR(-) facilitating cells (FC), a novel cell population in mouse bone marrow, have been shown to potently enhance engraftment of allogeneic HSC without causing GVHD. The predominant subpopulation of FC resembles plasmacytoid precursor dendritic cells. FC induce antigen-specific T(reg) in vivo. Notably, FC address one major concern that has prevented the implementation of T(reg) cell therapy in the clinic: to expand T(reg) and have them remain tolerogenic in vivo. FC are novel in that they induce an antigen-specific regulatory milieu in vivo. The discovery of FC has opened new alternatives to expanded criteria in bone marrow transplantation that were previously restricted to human leukocyte antigen-matched recipients. The focus of this review is to cover what is currently known about the mechanism of FC action in inducing tolerance and preventing GVHD and hostversus-graft reactivity.
Collapse
Affiliation(s)
- Paul A Cardenas
- Institute for Cellular Therapeutics; University of Louisville; Louisville, KY USA
| | | | | |
Collapse
|
587
|
Schildberg FA, Wojtalla A, Siegmund SV, Endl E, Diehl L, Abdullah Z, Kurts C, Knolle PA. Murine hepatic stellate cells veto CD8 T cell activation by a CD54-dependent mechanism. Hepatology 2011; 54:262-72. [PMID: 21488077 DOI: 10.1002/hep.24352] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
UNLABELLED The liver has a role in T cell tolerance induction, which is mainly achieved through the functions of tolerogenic hepatic antigen-presenting cells (APCs) and regulatory T cells. Hepatic stellate cells (HSCs) are known to have various immune functions, which range from immunogenic antigen presentation to the induction of T cell apoptosis. Here we report a novel role for stellate cells in vetoing the priming of naive CD8 T cells. Murine and human HSCs and stromal cells (but not hepatocytes) prevented the activation of naive T cells by dendritic cells, artificial APCs, and phorbol 12-myristate 13-acetate/ionomycin by a cell contact-dependent mechanism. The veto function for inhibiting T cell activation was directly correlated with the activation state of HSCs and was most pronounced in HSCs from fibrotic livers. Mechanistically, high expression levels of CD54 simultaneously restricted the expression of interleukin-2 (IL-2) receptor and IL-2 in T cells, and this was responsible for the inhibitory effect because exogenous IL-2 overcame the HSC veto function. CONCLUSION Our results demonstrate a novel function of HSCs in the local skewing of immune responses in the liver through the prevention of local stimulation of naive T cells. These results not only indicate a beneficial role in hepatic fibrosis, for which increased CD54 expression on HSCs could attenuate further T cell activation, but also identify IL-2 as a key cytokine in mediating local T cell immunity to overcome hepatic tolerance.
Collapse
Affiliation(s)
- Frank A Schildberg
- Institutes of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | |
Collapse
|
588
|
Self-antigen presentation by mouse B cells results in regulatory T-cell induction rather than anergy or clonal deletion. Blood 2011; 118:984-91. [PMID: 21652680 DOI: 10.1182/blood-2011-02-336115] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multiple mechanisms operate to ensure T-cell tolerance toward self-antigens. Three main processes have been described: clonal deletion, anergy, and deviation to CD4(+) regulatory T cells (Tregs) that suppress autoreactive T cells that have escaped the first 2 mechanisms. Although it is accepted that dendritic cells (DCs) and B cells contribute in maintaining T-cell tolerance to self-antigens, their relative contribution and the processes involved under physiologic conditions remain only partially characterized. In this study, we used different transgenic mouse models to obtain chimeras where a neo self-antigen is expressed by thymic epithelium and/or by DCs or B cells. We found that expression of cognate ligand in the thymus enhances antigen-specific FoxP3(+) cells independently of whether the self-antigen is expressed on thymic epithelium or only on DCs, but not on B cells. On the contrary, self-antigen expression by B cells was very efficient in inducing FoxP3(+) cells in the periphery, whereas self-antigen expression by DC led mainly to deletion and anergy of antigen-specific FoxP3(-) cells. The results presented in this study underline the role of B cells in Treg induction and may have important implications in clinical protocols aimed at the peripheral expansion of Tregs in patients.
Collapse
|
589
|
Schurich A, Khanna P, Lopes AR, Han KJ, Peppa D, Micco L, Nebbia G, Kennedy PTF, Geretti AM, Dusheiko G, Maini MK. Role of the coinhibitory receptor cytotoxic T lymphocyte antigen-4 on apoptosis-Prone CD8 T cells in persistent hepatitis B virus infection. Hepatology 2011; 53:1494-503. [PMID: 21360567 DOI: 10.1002/hep.24249] [Citation(s) in RCA: 250] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED An excess of coinhibitory signals has been proposed to drive the T-cell exhaustion characteristic of persistent viral infections. In this study we examined the contribution of the coinhibitory receptor cytotoxic T lymphocyte antigen-4 (CTLA-4) to CD8 T cell tolerance in chronic hepatitis B virus (HBV) infection (CHB). CD8 T cells in patients with CHB have an increased propensity to express the coinhibitory receptor CTLA-4 and this correlates with viral load. CTLA-4 is up-regulated on those HBV-specific CD8 T cells with the highest levels of the proapoptotic protein Bim, which we have previously shown mediates their premature attrition; abrogation of CTLA-4-mediated coinhibition can reduce Bim expression. Longitudinal study of CHB patients beginning antiviral therapy reveals that HBV DNA suppression induces transient reconstitution of HBV-specific CD8 T cells but does not reprogram their CTLA-4(hi) Bim(hi) tolerogenic phenotype. Blocking CTLA-4 is able to increase the expansion of interferon gamma (IFN-γ)-producing HBV-specific CD8 T cells in both the peripheral and intrahepatic compartments. The rescue of anti-HBV responses by either CTLA-4 or PD-L1 blockade is nonredundant. CONCLUSION CTLA-4 is expressed by HBV-specific CD8 T cells with high levels of Bim and helps to drive this proapoptotic phenotype. CTLA-4 blockade could form one arm of a therapeutic approach to modulate the diverse patterns of coregulation of T-cell exhaustion in this heterogeneous disease.
Collapse
|
590
|
Abstract
Expression by DCs of co-inhibitory molecules such as programmed death ligand-1 (PD-L1/B7-H1/CD274), a member of the B7 superfamily, is crucial for the downregulation of T-cell responses and the maintenance of immune homeostasis. Exposure of immature DCs to danger-associated molecular patterns (DAMPS) or pathogen-associated molecular patterns (PAMPs) generally results in their maturation and acquisition of immunostimulatory function. However, exposure of DCs to TLR ligands early during their differentiation can inhibit further differentiation and confer tolerogenic properties on these APCs. A report in this issue of The European Journal of Immunology reveals that early inhibition of human DC differentiation from blood monocytes by TLR agonists is associated with a tolerogenic phenotype and Treg generation. The tolerogenic function of these APCs is dependent on MAPK-induced IL-6 and IL-10 production, which drives STAT-3-mediated PD-L1 expression. These observations link IL-10 and IL-6 to PD-L1 expression, providing a new dimension to the anti-inflammatory properties of these cytokines. These findings also have implications for understanding the inherent function of DCs in non-lymphoid tissues such as the liver and lung, where they are exposed to PAMPs that are found constitutively in the local microenvironment.
Collapse
Affiliation(s)
- Tina L Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
591
|
Ichikawa S, Mucida D, Tyznik AJ, Kronenberg M, Cheroutre H. Hepatic stellate cells function as regulatory bystanders. THE JOURNAL OF IMMUNOLOGY 2011; 186:5549-55. [PMID: 21460203 DOI: 10.4049/jimmunol.1003917] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Regulatory T cells (Tregs) contribute significantly to the tolerogenic nature of the liver. The mechanisms, however, underlying liver-associated Treg induction are still elusive. We recently identified the vitamin A metabolite, retinoic acid (RA), as a key controller that promotes TGF-β-dependent Foxp3(+) Treg induction but inhibits TGF-β-driven Th17 differentiation. To investigate whether the RA producing hepatic stellate cells (HSC) are part of the liver tolerance mechanism, we investigated the ability of HSC to function as regulatory APC. Different from previous reports, we found that highly purified HSC did not express costimulatory molecules and only upregulated MHC class II after in vitro culture in the presence of exogenous IFN-γ. Consistent with an insufficient APC function, HSC failed to stimulate naive OT-II TCR transgenic CD4(+) T cells and only moderately stimulated α-galactosylceramide-primed invariant NKT cells. In contrast, HSC functioned as regulatory bystanders and promoted enhanced Foxp3 induction by OT-II TCR transgenic T cells primed by spleen dendritic cells, whereas they greatly inhibited the Th17 differentiation. Furthermore, the regulatory bystander capacity of the HSC was completely dependent on their ability to produce RA. Our data thus suggest that HSC can function as regulatory bystanders, and therefore, by promoting Tregs and suppressing Th17 differentiation, they might represent key players in the mechanism that drives liver-induced tolerance.
Collapse
Affiliation(s)
- Shintaro Ichikawa
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
592
|
Ota T, Ota M, Duong BH, Gavin AL, Nemazee D. Liver-expressed Igkappa superantigen induces tolerance of polyclonal B cells by clonal deletion not kappa to lambda receptor editing. ACTA ACUST UNITED AC 2011; 208:617-29. [PMID: 21357741 PMCID: PMC3058582 DOI: 10.1084/jem.20102265] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Analysis of tolerance in a polyclonal wild-type B cell population demonstrates apoptosis of cells reactive to antigen expressed on liver membrane. Little is know about the nature of peripheral B cell tolerance or how it may vary in distinct lineages. Although autoantibody transgenic studies indicate that anergy and apoptosis are involved, some studies claim that receptor editing occurs. To model peripheral B cell tolerance in a normal, polyclonal immune system, we generated transgenic mice expressing an Igκ–light chain–reactive superantigen targeted to the plasma membrane of hepatocytes (pAlb mice). In contrast to mice expressing κ superantigen ubiquitously, in which κ cells edit efficiently to λ, in pAlb mice, κ B cells underwent clonal deletion. Their κ cells failed to populate lymph nodes, and the remaining splenic κ cells were anergic, arrested at a semi-mature stage without undergoing receptor editing. In the liver, κ cells recognized superantigen, down-regulated surface Ig, and expressed active caspase 3, suggesting ongoing apoptosis at the site of B cell receptor ligand expression. Some, apparently mature, κ B1 and follicular B cells persisted in the peritoneum. BAFF (B cell–activating factor belonging to the tumor necrosis factor family) overexpression rescued splenic κ B cell maturation and allowed κ cells to populate lymph nodes. Our model facilitates analysis of tissue-specific autoimmunity, tolerance, and apoptosis in a polyclonal B cell population. The results suggest that deletion, not editing, is the major irreversible pathway of tolerance induction among peripheral B cells.
Collapse
Affiliation(s)
- Takayuki Ota
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
593
|
|