Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Hepatol Commun ; 3(10): 1296-1310, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31592075

ABSTRACT

Current therapeutics for chronic infection with hepatitis B virus (HBV) rarely induce functional cure due to the immunotolerant status of patients. Small molecule agonists targeting toll-like receptor 7 (TLR7) have been shown to elicit a functional cure in animal models of HBV but sometimes with poor tolerability due to immune-related toxicities. In an effort to increase the therapeutic window of TLR7 agonists to treat chronic hepatitis B (CHB), we developed an oral TLR7 agonist, APR002, designed to act locally in the gastrointestinal tract and liver, thus minimizing systemic exposure and improving tolerability. Here, we describe the pharmacokinetic/pharmacodynamic (PK/PD) profile of APR002 in mice and uninfected woodchucks as well as the safety and antiviral efficacy in combination with entecavir (ETV) in woodchucks with CHB. Treatment of woodchucks chronically infected with woodchuck hepatitis virus (WHV) with weekly oral doses of APR002 was well-tolerated. While APR002 and ETV single agents did not elicit sustained viral control, combination therapy resulted in durable immune-mediated suppression of the chronic infection. These woodchucks also had detectable antibodies to viral antigens, enhanced interferon-stimulated gene expression, and loss of WHV covalently closed circular DNA. Conclusion: APR002 is a novel TLR7 agonist exhibiting a distinct PK/PD profile that in combination with ETV can safely attain a functional cure in woodchucks with chronic WHV infection. Our results support further investigation of liver-targeted TLR7 agonists in human CHB.

2.
Malar J ; 12: 316, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24025732

ABSTRACT

BACKGROUND: One of the main causes of mortality from severe malaria in Plasmodium falciparum infections is cerebral malaria (CM). An important host genetic component determines the susceptibility of an individual to develop CM or to clear the infection and become semi-immune. As such, the identification of genetic loci associated with susceptibility or resistance may serve to modulate disease severity. METHODOLOGY: The Plasmodium berghei mouse model for experimental cerebral malaria (ECM) reproduces several disease symptoms seen in human CM, and two different phenotypes, a susceptible (FVB/NJ) and a resistant mouse strain (DBA/2J), were examined. RESULTS: FVB/NJ mice died from infection within ten days, whereas DBA/2J mice showed a gender bias: males survived on average nineteen days and females either died early with signs of ECM or survived for up to three weeks. A comparison of brain pathology between FVB/NJ and DBA/2J showed no major differences with regard to brain haemorrhages or the number of parasites and CD3+ cells in the microvasculature. However, significant differences were found in the peripheral blood of infected mice: For example resistant DBA/2J mice had significantly higher numbers of circulating basophils than did FVB/NJ mice on day seven. Analysis of the F2 offspring from a cross of DBA/2J and FVB/NJ mice mapped the genetic locus of the underlying survival trait to chromosome 9 with a Lod score of 4.9. This locus overlaps with two previously identified resistance loci (char1 and pymr) from a blood stage malaria model. CONCLUSIONS: Survival best distinguishes malaria infections between FVB/NJ and DBA/2J mice. The importance of char1 and pymr on chromosome 9 in malaria resistance to P. berghei was confirmed. In addition there was an association of basophil numbers with survival.


Subject(s)
Chromosomes, Human, Pair 9 , Disease Resistance , Genetic Loci , Malaria, Cerebral/genetics , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Animals , Basophils/immunology , Disease Models, Animal , Female , Humans , Male , Mice , Survival Analysis
3.
J Immunol ; 182(4): 2213-20, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19201875

ABSTRACT

Exosomes are small membrane vesicles of endocytic origin that are secreted by most cells in culture, but are also present in serum. They contain a wide array of protein ligands on their surface, which has led to the hypothesis that they might mediate intercellular communication. Indeed, data support that exosomes can transfer Ags to dendritic cells (DC), and, interestingly, that these DC can subsequently induce T cell priming or tolerance. We have investigated whether this concept can be expanded to antiviral immunity. We isolated exosomes from supernatant of cultured bone marrow-derived DC (BMDC) that were infected with lymphocytic choriomeningitis virus (LCMV) or loaded with an immunodominant LCMV peptide, and characterized them by flow cytometry upon binding to beads. We then incubated the exosome preparations with BMDC and looked at their potential to activate LCMV gp33-specific naive and memory CD8 T cells. We found that exosomes do not significantly contribute to CD8 T cell cross-priming in vitro. Additionally, exosomes derived from in vitro-infected BMDC did not exhibit significant in vivo priming activity, as evidenced by the lack of protection following exosome vaccination. Thus, DC-derived exosomes do not appear to contribute significantly to CTL priming during acute LCMV infection.


Subject(s)
Cross-Priming/immunology , Dendritic Cells/immunology , Exosomes/immunology , Lymphocytic Choriomeningitis/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Bone Marrow Cells/immunology , Dendritic Cells/virology , Exosomes/virology , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Cytotoxic/virology
4.
J Exp Med ; 205(6): 1409-22, 2008 Jun 09.
Article in English | MEDLINE | ID: mdl-18474629

ABSTRACT

Autoimmune liver diseases, such as autoimmune hepatitis (AIH) and primary biliary cirrhosis, often have severe consequences for the patient. Because of a lack of appropriate animal models, not much is known about their potential viral etiology. Infection by liver-tropic viruses is one possibility for the breakdown of self-tolerance. Therefore, we infected mice with adenovirus Ad5 expressing human cytochrome P450 2D6 (Ad-2D6). Ad-2D6-infected mice developed persistent autoimmune liver disease, apparent by cellular infiltration, hepatic fibrosis, "fused" liver lobules, and necrosis. Similar to type 2 AIH patients, Ad-2D6-infected mice generated type 1 liver kidney microsomal-like antibodies recognizing the immunodominant epitope WDPAQPPRD of cytochrome P450 2D6 (CYP2D6). Interestingly, Ad-2D6-infected wild-type FVB/N mice displayed exacerbated liver damage when compared with transgenic mice expressing the identical human CYP2D6 protein in the liver, indicating the presence of a stronger immunological tolerance in CYP2D6 mice. We demonstrate for the first time that infection with a virus expressing a natural human autoantigen breaks tolerance, resulting in a chronic form of severe, autoimmune liver damage. Our novel model system should be instrumental for studying mechanisms involved in the initiation, propagation, and precipitation of virus-induced autoimmune liver diseases.


Subject(s)
Adenoviridae Infections/enzymology , Adenoviridae Infections/immunology , Autoantigens/immunology , Cytochrome P-450 CYP2D6/immunology , Immune Tolerance , Liver/enzymology , Liver/immunology , Adenoviridae/genetics , Animals , Genes, Reporter , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Inbred Strains , Mice, Transgenic
5.
J Autoimmun ; 27(3): 166-73, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17045460

ABSTRACT

CD8(+) cytotoxic T lymphocytes (CTL) can rapidly kill beta-cells and therefore contribute to the development of type 1 diabetes (T1D). CTL-mediated beta-cell killing can occur via perforin-mediated lysis, Fas-Fas-L interaction, and the secretion of TNF-alpha or IFN-gamma. The secretion of IFN-gamma can contribute to beta-cell death directly by eliciting nitric oxide production, and indirectly by upregulating MHC class I and 'unmasking' beta-cells for recognition by CTL. Earlier studies in the RIP-LCMV mouse model of diabetes showed that disruption of beta-cell IFN-gamma signaling alone abolished the direct detrimental effects of IFN-gamma, but not MHC class I upregulation. Suppressor of cytokine signaling-1 (SOCS-1) represses several crucial cytokine signaling pathways simultaneously, among them IFN-gamma and IL-1-beta. We therefore evaluated the protective capacity of islet cell SOCS-1 expression in the CD8(+) mediated RIP-LCMV diabetes model. Clinical disease was prevented in over 90% of the mice. Not only absence of MHC-I and Fas upregulation, but also resistance to cytokine-induced killing of beta-cells and a complete lack of CXCL-10 (IP10) production in islets led to a lack of islet infiltration and impaired activation of autoaggressive CD4(+) and CD8(+) T-cells in these mice. Thus, SOCS expression renders beta-cells resistant to CTL attack in a mouse model of T1D.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/genetics , Insulin-Secreting Cells/metabolism , Suppressor of Cytokine Signaling Proteins/biosynthesis , Animals , Cytotoxicity, Immunologic , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/virology , Disease Models, Animal , Flow Cytometry , Histocompatibility Antigens Class I/biosynthesis , Immunohistochemistry , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymphocytic choriomeningitis virus , Mice , Mice, Transgenic , Microscopy, Confocal , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Cytokine/biosynthesis , Signal Transduction/physiology , Suppressor of Cytokine Signaling 1 Protein , fas Receptor/biosynthesis
6.
J Clin Invest ; 116(5): 1371-81, 2006 May.
Article in English | MEDLINE | ID: mdl-16628253

ABSTRACT

Safe induction of autoantigen-specific long-term tolerance is the "holy grail" for the treatment of autoimmune diseases. In animal models of type 1 diabetes, oral or i.n. immunization with islet antigens induces Tregs that are capable of bystander suppression. However, such interventions are only effective early in the prediabetic phase. Here, we demonstrate that a novel combination treatment with anti-CD3epsilon-specific antibody and i.n. proinsulin peptide can reverse recent-onset diabetes in 2 murine diabetes models with much higher efficacy than with monotherapy with anti-CD3 or antigen alone. In vivo, expansion of CD25(+)Foxp3(+) and insulin-specific Tregs producing IL-10, TGF-beta, and IL-4 was strongly enhanced. These cells could transfer dominant tolerance to immunocompetent recent-onset diabetic recipients and suppressed heterologous autoaggressive CD8 responses. Thus, combining a systemic immune modulator with antigen-specific Treg induction is more efficacious in reverting diabetes. Since Tregs act site-specifically, this strategy should also be expected to reduce the potential for systemic side effects.


Subject(s)
Autoantigens/therapeutic use , CD3 Complex/chemistry , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/therapy , Proinsulin/therapeutic use , T-Lymphocytes, Regulatory/metabolism , Administration, Intranasal , Animals , Autoantigens/administration & dosage , CD3 Complex/immunology , Interleukin-10/biosynthesis , Interleukin-4/biosynthesis , Mice , Mice, Transgenic , Proinsulin/administration & dosage , Remission Induction , Transforming Growth Factor beta/biosynthesis
7.
J Virol ; 80(1): 270-80, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16352552

ABSTRACT

CTLA-4 is considered one of the most potent negative regulators of T-cell activation. To circumvent experimental limitations due to fatal lymphoproliferative disease associated with genetic ablation of CTLA-4, we have used radiation chimeras reconstituted with a mixture of CTLA-4+/+ and CTLA-4-/- bone marrow that retain a normal phenotype and allow the evaluation of long-term T-cell immunity under conditions of intrinsic CTLA-4 deficiency. Following virus infection, we profiled primary, memory, and secondary CD8+ and CD4+ T-cell responses directed against eight different viral epitopes. Our data demonstrate unaltered antigen-driven proliferation, acquisition of effector functions, distribution of epitope hierarchies, T-cell receptor repertoire selection, functional avidities, and long-term memory maintenance in the absence of CTLA-4. Moreover, regulation of memory T-cell survival and homeostatic proliferation, as well as secondary responses, was equivalent in virus-specific CTLA4+/+ and CTL-A-4-/- T-cell populations. Thus, lack of CTLA-4 expression by antigen-specific T cells can be compensated for by extrinsic factors in the presence of CTLA-4 expression by other cells. These findings have implications for the physiologic, pathological, and therapeutic regulation of costimulation.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , CTLA-4 Antigen , Gene Expression Regulation/immunology , Gene Silencing , Lymphocyte Activation/immunology , Mice , T-Lymphocytes, Regulatory/immunology
8.
J Immunol ; 175(6): 3516-24, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16148094

ABSTRACT

During inflammation, chemokines are conductors of lymphocyte trafficking. The chemokine CXCL10 is expressed early after virus infection. In a virus-induced mouse model for type 1 diabetes, CXCL10 blockade abrogated disease by interfering with trafficking of autoaggressive lymphocytes to the pancreas. We have generated transgenic rat insulin promotor (RIP)-CXCL10 mice expressing CXCL10 in the beta cells of the islets of Langerhans to evaluate how bystander inflammation influences autoimmunity. RIP-CXCL10 mice have islet infiltrations by mononuclear cells and limited impairment of beta cell function, but not spontaneous diabetes. RIP-CXCL10 mice crossed to RIP-nucleoprotein (NP) mice expressing the NP of the lymphocytic choriomeningitis virus in the beta cells had massively accelerated type 1 diabetes after lymphocytic choriomeningitis virus infection. Mechanistically, we found a drastic increase in NP-specific, autoaggressive CD8 T cells in the pancreas after infection. In situ staining with H-2D(b)(NP(396)) tetramers revealed islet infiltration by NP-specific CD8 T cells in RIP-NP-CXCL10 mice early after infection. Our results indicate that CXCL10 expression accelerates the autoimmune process by enhancing the migration of Ag-specific lymphocytes to their target site.


Subject(s)
Chemokines, CXC/genetics , Chemotaxis, Leukocyte , Diabetes Mellitus, Type 1/etiology , Islets of Langerhans/metabolism , Animals , Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL10 , Chemokines, CXC/analysis , Chemokines, CXC/physiology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/virology , Inflammation/immunology , Islets of Langerhans/chemistry , Lymphocytic choriomeningitis virus , Mice , Mice, Transgenic , T-Cell Antigen Receptor Specificity , Viral Proteins/immunology
9.
J Exp Med ; 199(8): 1153-62, 2004 Apr 19.
Article in English | MEDLINE | ID: mdl-15096540

ABSTRACT

The T-box transcription factor T-bet is known to control lineage commitment and interferon-gamma production by T helper 1 (Th1) CD4 lymphocytes. We report here that T-bet is essential for development of CD8 lymphocyte-dependent autoimmune diabetes (type 1 diabetes [T1D]) in the rat insulin promoter-lymphocytic choriomeningitis virus (LCMV) transgenic model for virally induced T1D. In the absence of T-bet, autoaggressive (anti-LCMV) CD8 lymphocytes were reduced in number and produced less IFN-gamma, but increased IL-2 compared with controls. Further analysis showed that T-bet intrinsically controls the generation, but not apoptosis, maintenance, or secondary expansion of antiviral effector/memory CD8 lymphocytes. This observation points toward a therapeutic opportunity for the treatment of T1D and other autoimmune disorders.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Transcription Factors/immunology , Animals , Autoimmunity , Chimera , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Immunologic Memory , Interferon-gamma/biosynthesis , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Rats , T-Box Domain Proteins , Transcription Factors/deficiency , Transcription Factors/genetics
10.
Diabetes ; 53(3): 591-6, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14988242

ABSTRACT

Tissue-specific expression of Fas-ligand (Fas-L) can provide immune privilege by inducing apoptosis of "invading" lymphocytes expressing Fas. However, accelerated diabetes has been reported in transgenic mice expressing Fas-L in islets (RIP-Fas-L) as a result of Fas-dependent fratricide of beta-cells after transfer of diabetogenic clones. Here we studied whether Fas-L could protect islets from autoaggressive CD8 lymphocytes in a transgenic model of virally induced diabetes (RIP-LCMV-NP transgenic mice), in which the autoaggressive response is directed to a viral nucleoprotein (NP) expressed as a transgene in beta-cells. Indeed, disease incidence after viral (lymphocytic choriomeningitis virus [LCMV]) infection was reduced by approximately 30%, which was associated with a decrease of autoaggressive CD8 NP-specific lymphocytes in islets and pancreatic draining lymph nodes. However, surprisingly, a high degree (50%) of diabetes was seen in mice that expressed only Fas-L but not the viral transgene (NP) in beta-cells after infection with LCMV. This was due to induction of Fas on beta-cells after LCMV infection of the pancreas, resulting in Fas/Fas-L-mediated fratricide. Thus, although Fas-L can lend some immune privilege to islet cells, local virus-induced inflammation will induce Fas on beta-cells, leading to their mutual destruction if Fas-L is present. Expression of Fas-L therefore might not be protective in situations in which viral inflammation can be expected, resulting in Fas induction on the targeted cell itself.


Subject(s)
Islets of Langerhans/pathology , Lymphocytic Choriomeningitis/physiopathology , Membrane Glycoproteins/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Crosses, Genetic , Fas Ligand Protein , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Islets of Langerhans/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus , Mice , Mice, Transgenic , Spleen/immunology
11.
J Clin Invest ; 113(1): 74-84, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14702111

ABSTRACT

Viruses can cause but can also prevent autoimmune disease. This dualism has certainly hampered attempts to establish a causal relationship between viral infections and type 1 diabetes (T1D). To develop a better mechanistic understanding of how viruses can influence the development of autoimmune disease, we exposed prediabetic mice to various viral infections. We used the well-established NOD and transgenic RIP-LCMV models of autoimmune diabetes. In both cases, infection with the lymphocytic choriomeningitis virus (LCMV) completely abrogated the diabetic process. Interestingly, such therapeutic viral infections resulted in a rapid recruitment of T lymphocytes from the islet infiltrate to the pancreatic draining lymph node, where increased apoptosis was occurring. In both models this was associated with a selective and extensive expression of the chemokine IP-10 (CXCL10), which predominantly attracts activated T lymphocytes, in the pancreatic draining lymph node, and in RIP-LCMV mice it depended on the viral antigenic load. In RIP-LCMV mice, blockade of TNF-alpha or IFN-gamma in vivo abolished the prevention of T1D. Thus, virally induced proinflammatory cytokines and chemokines can influence the ongoing autoaggressive process beneficially at the preclinical stage, if produced at the correct location, time, and levels.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic choriomeningitis virus/physiology , Prediabetic State/prevention & control , Aging , Animals , Diabetes Mellitus, Type 1/virology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred NOD , Mice, Transgenic , Polymerase Chain Reaction , Prediabetic State/virology , Time Factors
12.
Eur J Immunol ; 32(1): 113-21, 2002 01.
Article in English | MEDLINE | ID: mdl-11754351

ABSTRACT

Administration of autoantigens through DNA immunizations or via the oral route can prevent progression of islet destruction and lower the incidence of type 1 diabetes in animal models. This beneficial effect is mediated by autoreactive regulatory CD4 lymphocytes, and it is known that their induction depends on the precise dose and route of antigen administration. However, it is not clear which endogenous factors determine when such immunizations lead to activation of regulatory versus aggressive autoreactive lymphocytes and how a deleterious outcome can be avoided. Here we describe novel observations made in an animal model for virally induced type 1 diabetes, showing that the endogenous expression levels of the islet antigens and glutamic acid decarboxylase determine whether immunization with these antigens is beneficial or detrimental. Lower expression levels in beta-cells support immune regulation resulting in induction of autoreactive, regulatory cells characterized by increased IL-4 production (Th2-like), whereas higher levels favor Th1-like autoaggressive responses characterized by augmented IFN-gamma generation. Co-immunization with an IL-4-expressing plasmid reduces the risk of augmenting autoaggression and in this way increases the safety margin of this immune-based therapy. Our findings will be of importance for designing safe antigen-specific interventions for human type 1 diabetes.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Type 1/prevention & control , Glutamate Decarboxylase/immunology , Interleukin-4/immunology , Isoenzymes/immunology , Vaccines, DNA/immunology , Animals , Antigens, Viral/genetics , Autoantibodies/biosynthesis , Autoantigens/genetics , Cell Line , Chlorocebus aethiops , Consumer Product Safety , Cricetinae , Diabetes Mellitus, Type 1/physiopathology , Female , Gene Expression , Glutamate Decarboxylase/genetics , Glycoproteins/genetics , Humans , Insulin/metabolism , Interleukin-4/genetics , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Isoenzymes/genetics , Lymphocytic choriomeningitis virus , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Plasmids , Th1 Cells/immunology , Th2 Cells/immunology , Treatment Outcome , Vaccination , Vaccines, DNA/genetics , Vero Cells , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...