Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Hum Gene Ther ; 32(7-8): 341-348, 2021 04.
Article in English | MEDLINE | ID: mdl-33213214

ABSTRACT

Tight control of transgene expression is key to ensure the efficacy of a wide range of gene therapy interventions, in which the magnitude and duration of gene expression have to be adjusted to therapeutic needs, thereby limiting secondary effects. The development of upgraded strategies to link transgene expression to pathological stress episodes is an unmet need in gene therapy. Here, we propose an expression strategy that associates transgene expression to an intracellular stress coping mechanism, the unfolded protein response. Specifically, we harnessed the cis elements required to sustain the noncanonical splicing of X-box binding protein 1 (XBP1) messenger RNA (mRNA) in response to the dysfunction of the endoplasmic reticulum (ER), a situation commonly known as ER stress, to drive the expression of heterologous genes. Since ER stress features a wide variety of pathological conditions, including viral infections, cancer, or metabolic disorders, this new expression module stimulates the synthesis of therapeutic genes as a response to cellular damage, and ensures their expression only when necessary. Validation of this inducible expression system was performed in vitro and in vivo, and its potential to limit/inhibit viral infections has been shown in proof-of principle experiments.


Subject(s)
Hepatitis B virus , Signal Transduction , Endoplasmic Reticulum Stress/genetics , Genetic Therapy , Unfolded Protein Response/genetics
2.
Methods Enzymol ; 636: 129-172, 2020.
Article in English | MEDLINE | ID: mdl-32178817

ABSTRACT

Transforming growth factor beta (TGF-ß) is a potent pleiotropic polypeptide cytokine, with a complex and context dependent control of its activation, signaling and effector functions. This cytokine is pivotal in the regulation of immunological responses, tumor initiation and development, stromal homeostasis and all their intricate related interactions. Last decade advances in cancer immunotherapy have reactivated the clinical interest on potential drug with TGF-ß inhibition effect, combined with immunomodulating enhancer drugs. The correct quantification of the in vitro and in vivo biological activity of this cytokine is essential to understand the intrinsic underlying biological mechanisms and TGF-ß role in the immune system, tumor and stromal codevelopment, modulation and interactions. There is a wide variety of available procedures to quantify TGF-ß activity, which includes different methodological approximations like ELISA, Bioassays including reporter gene assays, Flow cytometry (FC), Western blotting (WB), immunochemical/fluorescence microscopy, among others. Here, we detail available methods for TGF-ß biological activity analysis, together with their applicability and suitability for each experimental setting, in order to get a complete analytical perspective and more comprehensive information along the development and design of combined antitumor immunotherapies, which include the inhibition of TGF-ß biological activity.


Subject(s)
Neoplasms , Transforming Growth Factor beta , Cytokines , Humans , Neoplasms/drug therapy , Signal Transduction
3.
J Tissue Eng Regen Med ; 14(1): 123-134, 2020 01.
Article in English | MEDLINE | ID: mdl-31677236

ABSTRACT

Adeno-associated viruses (AAV) have become one of the most promising tools for gene transfer in clinics. Among all the serotypes, AAV9 has been described as the most efficient for cardiac transduction. In order to achieve optimal therapeutic delivery in heart disease, we have explored AAV9 transduction efficiency in an infarcted heart using different routes of administration and promoters, including a cardiac-specific one. AAV9 vectors carrying luciferase or green fluorescence protein under the control of the ubiquitous elongation-factor-1-alpha or the cardiac-specific troponin-T (TnT) promoters were administered by intramyocardial or intravenous injection, either in healthy or myocardial-infarcted mice. The transduction efficacy and specificity, the time-course expression, and the safety of each vector were tested. High transgene expression levels were found in the heart, but not in the liver, of mice receiving AAV-TnT, which was significantly higher after intramyocardial injection regardless of ischemia-induction. On the contrary, high hepatic transgene expression levels were detected with the elongation-factor-1-alpha-promoter, independently of the administration route and heart damage. Moreover, tissue-specific green fluorescence protein expression was found in cardiomyocytes with the TnT vector, whereas minimal cardiac expression was detected with the ubiquitous one. Interestingly, we found that myocardial infarction greatly increased the transcriptional activity of AAV genomes. Our findings show that the use of cardiac promoters allows for specific and stable cardiac gene expression, which is optimal and robust when intramyocardially injected. Furthermore, our data indicate that the pathological status of the tissue can alter the transcriptional activity of AAV genomes, an aspect that should be carefully evaluated for clinical applications.


Subject(s)
Dependovirus/genetics , Myocardial Ischemia/pathology , Animals , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Genome, Viral , Green Fluorescent Proteins/metabolism , HEK293 Cells , Heart/physiology , Humans , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/cytology , Peptide Elongation Factor 1/metabolism , Promoter Regions, Genetic , Tissue Distribution , Transduction, Genetic , Transgenes , Troponin T/metabolism
6.
J Clin Invest ; 127(10): 3624-3639, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28846069

ABSTRACT

Hematopoietic stem cells (HSCs) remain mostly quiescent under steady-state conditions but switch to a proliferative state following hematopoietic stress, e.g., bone marrow (BM) injury, transplantation, or systemic infection and inflammation. The homeostatic balance between quiescence, self-renewal, and differentiation of HSCs is strongly dependent on their interactions with cells that constitute a specialized microanatomical environment in the BM known as the HSC niche. Here, we identified the secreted extracellular matrix protein Del-1 as a component and regulator of the HSC niche. Specifically, we found that Del-1 was expressed by several cellular components of the HSC niche, including arteriolar endothelial cells, CXCL12-abundant reticular (CAR) cells, and cells of the osteoblastic lineage. Del-1 promoted critical functions of the HSC niche, as it regulated long-term HSC (LT-HSC) proliferation and differentiation toward the myeloid lineage. Del-1 deficiency in mice resulted in reduced LT-HSC proliferation and infringed preferentially upon myelopoiesis under both steady-state and stressful conditions, such as hematopoietic cell transplantation and G-CSF- or inflammation-induced stress myelopoiesis. Del-1-induced HSC proliferation and myeloid lineage commitment were mediated by ß3 integrin on hematopoietic progenitors. This hitherto unknown Del-1 function in the HSC niche represents a juxtacrine homeostatic adaptation of the hematopoietic system in stress myelopoiesis.


Subject(s)
Carrier Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Myelopoiesis , Stem Cell Niche , Stress, Physiological , Animals , Calcium-Binding Proteins , Carrier Proteins/genetics , Cell Adhesion Molecules , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Endothelial Cells/metabolism , Humans , Integrin beta3/genetics , Integrin beta3/metabolism , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout
7.
J Hepatol ; 67(4): 669-679, 2017 10.
Article in English | MEDLINE | ID: mdl-28527664

ABSTRACT

BACKGROUND & AIMS: Studying hepatitis delta virus (HDV) and developing new treatments is hampered by the limited availability of small animal models. Herein, a description of a robust mouse model of HDV infection that mimics several important characteristics of the human disease is presented. METHODS: HDV and hepatitis B virus (HBV) replication competent genomes were delivered to the mouse liver using adeno-associated viruses (AAV; AAV-HDV and AAV-HBV). Viral load, antigen expression and genomes were quantified at different time points after AAV injection. Furthermore, liver pathology, genome editing, and the activation of the innate immune response were evaluated. RESULTS: AAV-HDV infection initiated HDV replication in mouse hepatocytes. Genome editing was confirmed by the presence of small and large HDV antigens and sequencing. Viral replication was detected for 45days, even after the AAV-HDV vector had almost disappeared. In the presence of HBV, HDV infectious particles were detected in serum. Furthermore, as observed in patients, co-infection was associated with the reduction of HBV antigen expression and the onset of liver damage that included the alteration of genes involved in the development of liver pathologies. HDV replication induced a sustained type I interferon response, which was significantly reduced in immunodeficient mice and almost absent in mitochondrial antiviral signaling protein (MAVS)-deficient mice. CONCLUSION: The animal model described here reproduces important characteristics of human HDV infection and provides a valuable tool for characterizing the viral infection and for developing new treatments. Furthermore, MAVS was identified as a main player in HDV detection and adaptive immunity was found to be involved in the amplification of the innate immune response. Lay summary: Co-infection with hepatitis B and D virus (HBV and HDV, respectively) often causes a more severe disease condition than HBV alone. Gaining more insight into HDV and developing new treatments is hampered by limited availability of adequate immune competent small animal models and new ones are needed. Here, a mouse model of HDV infection is described, which mimics several important characteristics of the human disease, such as the initiation and maintenance of replication in murine hepatocytes, genome editing and, in the presence of HBV, generation of infectious particles. Lastly, the involvement of an adaptive immunity and the intracellular signaling molecule MAVS in mounting a strong and lasting innate response was shown. Thus, our model serves as a useful tool for the investigation of HDV biology and new treatments.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Hepatitis D/immunology , Interferon-beta/biosynthesis , Adaptive Immunity , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Coinfection/immunology , Coinfection/pathology , Coinfection/virology , Dependovirus/genetics , Disease Models, Animal , Genome, Viral , Hepatitis B/complications , Hepatitis B/immunology , Hepatitis B/virology , Hepatitis B Antigens/metabolism , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Hepatitis D/complications , Hepatitis D/virology , Hepatitis Delta Virus/genetics , Hepatitis Delta Virus/immunology , Hepatitis Delta Virus/physiology , Hepatitis delta Antigens/metabolism , Humans , Immunity, Innate , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological , Signal Transduction/immunology , Virus Replication
8.
Article in English | MEDLINE | ID: mdl-27895015

ABSTRACT

Sepsis still constitutes an unmet clinical need, which could benefit from novel adjunctive strategies to conventional antibiotic therapy. The soluble form of the scavenger-like human CD6 lymphocyte receptor (shCD6) binds to key pathogenic components from Gram-positive and -negative bacteria and shows time- and dose-dependent efficacy in mouse models of monobacterial sepsis. The objective of the present work was to demonstrate the effectiveness of infusing mouse and human sCD6 by different systemic routes, either alone or as adjunctive therapy to gold standard antibiotics, in a lethal model of polymicrobial sepsis. To this end, C57BL/6 mice undergoing high-grade septic shock induced by cecal ligation and puncture (CLP; ≥90% lethality) were infused via the intraperitoneal (i.p.) or intravenous (i.v.) route with shCD6 at different doses and time points, either alone or in combination with imipenem/cilastatin (I/C) at a dose of 33 mg/kg of body weight every 8 h. Significantly reduced mortality and proinflammatory cytokine levels were observed by i.p. infusion of a single shCD6 dose (1.25 mg/kg) 1 h pre- or post-CLP. When using the i.v. route, mice survival was significantly extended by starting shCD6 infusion at later time points post-CLP (up to 6 h after CLP). Significant adjunctive effects on mouse survival were observed by i.p. or i.v. infusion of shCD6 in combination with i.p. I/C post-CLP. Similar results were obtained in mice expressing high sustained levels (5 to 10 µg/ml) of mouse sCD6 in serum by means of transduction with hepatotropic adeno-associated virus (AAV). Taken together, the data support the conserved antibacterial effects of human and mouse sCD6 and their use as adjunctive therapy in experimental models of complex and severe polymicrobial sepsis.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cilastatin/therapeutic use , Imipenem/therapeutic use , Animals , Cecum/injuries , Cilastatin, Imipenem Drug Combination , Cytokines/metabolism , Dependovirus/genetics , Disease Models, Animal , Drug Combinations , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Humans , Ligation , Male , Mice , Mice, Inbred C57BL , Receptors, Scavenger/metabolism , Sepsis
9.
Cell Stem Cell ; 19(3): 284-6, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27588743

ABSTRACT

Successful hematopoietic regeneration demands preservation of stemness while enabling expansion and differentiation into blood lineages. Now, Silberstein et al. (2016) and Goncalves et al. (2016) identify a ribonuclease secreted by proximal niche cells that simultaneously drives quiescence of HSCs and proliferation of myeloid progenitors and dramatically enhances hematopoietic recovery after HSC transplantation.


Subject(s)
Hematopoietic Stem Cells , Stem Cell Niche , Cell Differentiation , Humans , Regeneration
10.
Diabetologia ; 59(12): 2702-2710, 2016 12.
Article in English | MEDLINE | ID: mdl-27600278

ABSTRACT

AIMS/HYPOTHESIS: In the postprandial state, the liver regulates glucose homeostasis by glucose uptake and conversion to glycogen and lipids. Glucose and insulin signalling finely regulate glycogen synthesis through several mechanisms. Glucose uptake in hepatocytes is favoured by the insulin receptor isoform A (IRA), rather than isoform B (IRB). Thus, we hypothesised that, in hepatocytes, IRA would increase glycogen synthesis by promoting glucose uptake and glycogen storage. METHODS: We addressed the role of insulin receptor isoforms on glycogen metabolism in vitro in immortalised neonatal hepatocytes. In vivo, IRA or IRB were specifically expressed in the liver using adeno-associated virus vectors in inducible liver insulin receptor knockout (iLIRKO) mice, a model of type 2 diabetes. The role of IR isoforms in glycogen synthesis and storage in iLIRKO was subsequently investigated. RESULTS: In immortalised hepatocytes, IRA, but not IRB expression induced an increase in insulin signalling that was associated with elevated glycogen synthesis, glycogen synthase activity and glycogen storage. Similarly, elevated IRA, but not IRB expression in the livers of iLIRKO mice induced an increase in glycogen content. CONCLUSIONS/INTERPRETATION: We provide new insight into the role of IRA in the regulation of glycogen metabolism in cultured hepatocytes and in the livers of a mouse model of type 2 diabetes. Our data strongly suggest that IRA is more efficient than IRB at promoting glycogen synthesis and storage. Therefore, we suggest that IRA expression in the liver could provide an interesting therapeutic approach for the regulation of hepatic glucose content and glycogen storage.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glycogen Phosphorylase/metabolism , Glycogen Synthase/metabolism , Glycogen/metabolism , Liver Glycogen/metabolism , Liver/metabolism , Protein Isoforms/metabolism , Receptor, Insulin/metabolism , Animals , Blotting, Western , Cell Line , Diabetes Mellitus, Type 2/genetics , Glucose/metabolism , Glycogen Phosphorylase/genetics , Glycogen Synthase/genetics , Glycogenolysis , Hepatocytes , Mice , Mice, Knockout , Protein Isoforms/genetics , Receptor, Insulin/genetics
11.
J Immunol ; 197(6): 2145-56, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27511737

ABSTRACT

The etiopathogenesis of autoimmune hepatitis (AIH) remains poorly understood. In this study, we sought to develop an animal model of human AIH to gain insight into the immunological mechanisms driving this condition. C57BL/6 mice were i.v. injected with adeno-associated viral vectors encoding murine IL-12 or luciferase under the control of a liver-specific promoter. Organ histology, response to immunosuppressive therapy, and biochemical and immunological parameters, including Ag-specific humoral and cellular response, were analyzed. Mechanistic studies were carried out using genetically modified mice and depletion of lymphocyte subpopulations. Adeno-associated virus IL-12-treated mice developed histological, biochemical, and immunological changes resembling type 1 AIH, including marked and persistent liver mononuclear cell infiltration, hepatic fibrosis, hypergammaglobulinemia, anti-nuclear and anti-smooth muscle actin Abs, and disease remission with immunosuppressive drugs. Interestingly, transgenic IL-12 was short-lived, but endogenous IL-12 expression was induced, and both IL-12 and IFN-γ remained elevated during the entire study period. IFN-γ was identified as an essential mediator of liver damage, and CD4 and CD8 T cells but not NK, NKT, or B cells were essential executors of hepatic injury. Furthermore, both MHC class I and MHC class II expression was upregulated at the hepatocellular membrane, and induction of autoreactive liver-specific T cells was detected. Remarkably, although immunoregulatory mechanisms were activated, they only partially mitigated liver damage. Thus, low and transient expression of transgenic IL-12 in hepatocytes causes loss of tolerance to hepatocellular Ags, leading to chronic hepatitis resembling human AIH type 1. This model provides a practical tool to explore AIH pathogenesis and novel therapies.


Subject(s)
Hepatitis, Autoimmune/etiology , Interleukin-12/genetics , Liver/metabolism , Animals , Dependovirus/genetics , Female , Hepatitis, Autoimmune/drug therapy , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Hypergammaglobulinemia/etiology , Immune Tolerance , Immunosuppressive Agents/therapeutic use , Interferon-gamma/biosynthesis , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Receptor/physiology , T-Lymphocytes/immunology
12.
Dis Model Mech ; 9(11): 1271-1281, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27562101

ABSTRACT

Type 2 diabetes mellitus is a complex metabolic disease and its pathogenesis involves abnormalities in both peripheral insulin action and insulin secretion. Previous in vitro data showed that insulin receptor isoform A, but not B, favours basal glucose uptake through its specific association with endogenous GLUT1/2 in murine hepatocytes and beta cells. With this background, we hypothesized that hepatic expression of insulin receptor isoform A in a mouse model of type 2 diabetes could potentially increase the glucose uptake of these cells, decreasing the hyperglycaemia and therefore ameliorating the diabetic phenotype. To assure this hypothesis, we have developed recombinant adeno-associated viral vectors expressing insulin receptor isoform A (IRA) or isoform B (IRB) under the control of a hepatocyte--specific promoter. Our results demonstrate that in the long term, hepatic expression of IRA in diabetic mice is more efficient than IRB in ameliorating glucose intolerance. Consequently, it impairs the induction of compensatory mechanisms through beta cell hyperplasia and/or hypertrophy that finally lead to beta cell failure, reverting the diabetic phenotype in about 8 weeks. Our data suggest that long-term hepatic expression of IRA could be a promising therapeutic approach for the treatment of type 2 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose Intolerance/metabolism , Receptor, Insulin/metabolism , Animals , Cell Proliferation , Dependovirus/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Glucose/metabolism , Glucose Intolerance/pathology , Green Fluorescent Proteins/metabolism , Homeostasis , Hyperplasia , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Liver/metabolism , Mice, Knockout , Protein Isoforms/metabolism
13.
J Virol ; 90(19): 8563-74, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27440883

ABSTRACT

UNLABELLED: In chronic hepatitis B (CHB), failure to control hepatitis B virus (HBV) is associated with T cell dysfunction. HBV transgenic mice mirror many features of the human disease, including T cell unresponsiveness, and thus represent an appropriate model in which to test novel therapeutic strategies. To date, the tolerant state of CD8(+) T cells in these animals could be altered only by strong immunogens or by immunization with HBV antigen-pulsed dendritic cells; however, the effectors induced were unable to suppress viral gene expression or replication. Because of the known stimulatory properties of alpha interferon (IFN-α) and interleukin-15 (IL-15), this study explored the therapeutic potential of liver-directed gene transfer of these cytokines in a murine model of CHB using adeno-associated virus (AAV) delivery. This combination not only resulted in a reduction in the viral load in the liver and the induction of an antibody response but also gave rise to functional and specific CD8(+) immunity. Furthermore, when splenic and intrahepatic lymphocytes from IFN-α- and IL-15-treated animals were transferred to new HBV carriers, partial antiviral immunity was achieved. In contrast to previous observations made using either cytokine alone, markedly attenuated PD-L1 induction in hepatic tissue was observed upon coadministration. An initial study with CHB patient samples also gave promising results. Hence, we demonstrated synergy between two stimulating cytokines, IL-15 and IFN-α, which, given together, constitute a potent approach to significantly enhance the CD8(+) T cell response in a state of immune hyporesponsiveness. Such an approach may be useful for treating chronic viral infections and neoplastic conditions. IMPORTANCE: With 350 million people affected worldwide and 600,000 annual deaths due to HBV-induced liver cirrhosis and/or hepatocellular carcinoma, chronic hepatitis B (CHB) is a major health problem. However, current treatment options are costly and not very effective and/or need to be administered for life. The unprecedented efficacy of the strategy described in our paper may offer an alternative and is relevant for a broad spectrum of readers because of its clear translational importance to other chronic viral infections in which a hyporesponsive antigen-specific T cell repertoire prevents clearance of the pathogen.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Interferon-alpha/administration & dosage , Interleukin-15/administration & dosage , Adenoviridae/genetics , Animals , Disease Models, Animal , Drug Carriers , Genetic Therapy , Hepatitis B Antibodies/blood , Interferon-alpha/genetics , Interleukin-15/genetics , Liver/virology , Mice, Transgenic , Treatment Outcome , Viral Load
14.
Oncotarget ; 7(31): 49008-49026, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27356750

ABSTRACT

Interleukin-15 (IL-15) is a cell growth-factor that regulates lymphocyte function and homeostasis. Its strong immunostimulatory activity coupled with an apparent lack of toxicity makes IL-15 an exciting candidate for cancer therapy, somehow limited by its short half-life in circulation. To increase IL-15 bioavailability we constructed a recombinant adeno-associated vector expressing murine IL-15 (AAV-mIL15) in the liver. Mice injected with AAV-mIL15 showed sustained and vector dose-dependent levels of IL-15/IL-15Rα complexes in serum, production of IFN-γ and activation of CD8+ T-cells and macrophages. The antitumoral efficacy of AAV-mIL15 was tested in a mouse model of metastatic colorectal cancer established by injection of MC38 cells. AAV-mIL15 treatment slightly inhibits MC38 tumor-growth and significantly increases the survival of mice. However, mIL-15 sustained expression was associated with development of side effects like hepatosplenomegaly, liver damage and the development of haematological stress, which results in the expansion of hematopoietic precursors in the bone marrow. To elucidate the mechanism, we treated IFN-γ receptor-, RAG1-, CD1d- and µMT-deficient mice and performed adoptive transfer of bone marrow cells from WT mice to RAG1-defcient mice. We demonstrated that the side effects of murine IL-15 administration were mainly mediated by IFN-γ-producing T-cells. CONCLUSIONS: IL-15 induces the activation and survival of effector immune cells that are necessary for its antitumoral activity; but, long-term exposure to IL-15 is associated with the development of important side effects mainly mediated by IFN-γ-producing T-cells. Strategies to modulate T-cell activation should be combined with IL-15 administration to reduce secondary adverse events while maintaining its antitumoral effect.


Subject(s)
Interferon-gamma/metabolism , Interleukin-15/pharmacology , T-Lymphocytes/metabolism , Adoptive Transfer , Animals , Antigens, CD1d/metabolism , Bone Marrow Cells/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Colorectal Neoplasms/therapy , Dependovirus , Disease Progression , Female , Gene Expression Regulation , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors , Homeodomain Proteins/metabolism , Humans , Killer Cells, Natural/metabolism , Liver/metabolism , Lymphocyte Activation , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
J Innate Immun ; 7(5): 466-81, 2015.
Article in English | MEDLINE | ID: mdl-25966783

ABSTRACT

RIG-I-like receptors (RLRs) are cellular sensor proteins that detect certain RNA species produced during viral infections. RLRs activate a signaling cascade that results in the production of IFN-ß as well as several other cytokines with antiviral and proinflammatory activities. We explored the potential of different constructs based on RLRs to induce the IFN-ß pathway and create an antiviral state in type I IFN-unresponsive models. A chimeric construct composed of RIG-I 2CARD and the first 200 amino acids of MAVS (2CARD-MAVS200) showed an enhanced ability to induce IFN-ß when compared to other stimulatory constructs. Furthermore, this human chimeric construct showed a superior ability to activate IFN-ß expression in cells from various species. This construct was found to overcome the restrictions of blocking IFN-ß induction or signaling by a number of viral IFN-antagonist proteins. Additionally, the antiviral activity of this chimera was demonstrated in influenza virus and HBV infection mouse models using adeno-associated virus (AAV) vectors as a delivery vehicle. We propose that AAV vectors expressing 2CARD-MAVS200 chimeric protein can reconstitute IFN-ß induction and recover a partial antiviral state in different models that do not respond to recombinant IFN-ß treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD-box RNA Helicases/metabolism , Hepatitis B virus/immunology , Hepatitis B, Chronic/therapy , Influenza A virus/immunology , Interferon-beta/metabolism , Orthomyxoviridae Infections/therapy , Adaptor Proteins, Signal Transducing/genetics , Animals , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Dependovirus/genetics , Disease Models, Animal , Female , HEK293 Cells , Hepatitis B, Chronic/immunology , Humans , Immunity, Innate , Interferon-beta/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Peptide Fragments/genetics , Receptors, Immunologic , Recombinant Fusion Proteins/genetics , Signal Transduction/genetics
16.
J Hepatol ; 63(2): 329-36, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25772035

ABSTRACT

BACKGROUND & AIMS: Current hepatitis B virus (HBV) management is challenging as treatment with nucleos(t)ide analogues needs to be maintained indefinitely and because interferon (IFN)-α therapy is associated with considerable toxicity. Previously, we showed that linking IFNα to apolipoprotein A-I generates a molecule (IA) with distinct antiviral and immunostimulatory activities which lacks the hematological toxicity of IFNα. METHODS: Here, we analyse the antiviral potential of an adeno-associated vector encoding IFNα fused to apolipoprotein A-I (AAV-IA) in comparison to a vector encoding only IFNα (AAV-IFN) in two animal models of chronic hepadnavirus infection. RESULTS: In HBV transgenic mice, we found that both vectors induced marked reductions in serum and liver HBV DNA and in hepatic HBV RNA but AAV-IFN caused lethal pancytopenia. Woodchucks with chronic hepatitis virus (WHV) infection that were treated by intrahepatic injection of vectors encoding the woodchuck sequences (AAV-wIFN or AAV-wIA), experienced only a slight reduction of viremia which was associated with hematological toxicity and high mortality when using AAV-wIFN, while AAV-wIA was well tolerated. However, when we tested AAV-wIA or a control vector encoding woodchuck apolipoprotein A-I (AAV-wApo) in combination with entecavir, we found that AAV-wApo-treated animals exhibited an immediate rebound of viral load upon entecavir withdrawal while, in AAV-wIA-treated woodchucks, viremia and antigenemia remained at low levels for several weeks following entecavir interruption. CONCLUSIONS: Treatment with AAV-IA is safe and elicits antiviral effects in animal models with difficult to treat chronic hepadnavirus infection. AAV-IA in combination with nucleos(t)ide analogues represents a promising approach for the treatment of HBV infection in highly viremic patients.


Subject(s)
Apolipoprotein A-I/metabolism , DNA, Viral/genetics , Genetic Therapy/methods , Hepadnaviridae/genetics , Hepatitis B, Chronic/therapy , Interferon-alpha/therapeutic use , Liver/drug effects , Animals , Antiviral Agents/therapeutic use , Disease Models, Animal , Female , Genetic Vectors , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/virology , Liver/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic
17.
Haematologica ; 100(8): 1014-22, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25715405

ABSTRACT

Interferon-α is a potent antiviral agent and a vigorous adjuvant in the induction of T-cell responses but its use is limited by hematologic toxicity. Interferon-α alters hematopoietic stem cell dormancy and impairs myelocytic and erythrocytic/megakaryocytic differentiation from hematopoietic progenitors. However, the effect of chronic interferon-α exposure on hematopoietic precursors has still not been well characterized. Here, we transduced the liver of mice with an adenoassociated vector encoding interferon-α to achieve sustained high serum levels of the cytokine. The bone marrow of these animals showed diminished long-term and short-term hematopoietic stem cells, reduction of multipotent progenitor cells, and marked decrease of B cells, but significant increase in the proportion of CD8(+) and CD4(+)CD8(+) T cells. Upon adoptive transfer to RAG(-/-) mice, bone marrow cells from interferon-α-treated animals generated CD4(+) and CD8(+) T cells while CD19(+), CD11b(+) and NK1.1(+) lineages failed to develop. These effects are associated with the transcriptional downregulation of transcription factors involved in B-cell differentiation and modulation of key factors for T-cell development. Thus, sustained interferon-α exposure causes hematopoietic stem cells exhaustion and drives common lymphoid progenitors towards T-cell generation.


Subject(s)
Cell Differentiation/drug effects , Interferon-alpha/pharmacology , Lymphopoiesis/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Gene Expression , Gene Expression Regulation/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunophenotyping , Interferon-alpha/genetics , Leukocyte Count , Leukocytes/cytology , Leukocytes/metabolism , Lymphopoiesis/genetics , Male , Mice , Mice, Knockout , Transcription Factors/genetics , Transcription Factors/metabolism
18.
EMBO Mol Med ; 6(10): 1246-62, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25100745

ABSTRACT

Insulin-like growth factor 2 (IGF2) was recently found to play a critical role in memory consolidation in rats and mice, and hippocampal or systemic administration of recombinant IGF2 enhances memory. Here, using a gene therapy-based approach with adeno-associated virus (AAV), we show that IGF2 overexpression in the hippocampus of aged wild-type mice enhances memory and promotes dendritic spine formation. Furthermore, we report that IGF2 expression decreases in the hippocampus of patients with Alzheimer's disease, and this leads us to hypothesize that increased IGF2 levels may be beneficial for treating the disease. Thus, we used the AAV system to deliver IGF2 or IGF1 into the hippocampus of the APP mouse model Tg2576 and demonstrate that IGF2 and insulin-like growth factor 1 (IGF1) rescue behavioural deficits, promote dendritic spine formation and restore normal hippocampal excitatory synaptic transmission. The brains of Tg2576 mice that overexpress IGF2 but not IGF1 also show a significant reduction in amyloid levels. This reduction probably occurs through an interaction with the IGF2 receptor (IGF2R). Hence, IGF2 and, to a lesser extent, IGF1 may be effective treatments for Alzheimer's disease.


Subject(s)
Alzheimer Disease/physiopathology , Insulin-Like Growth Factor II/metabolism , Memory Disorders/physiopathology , Synaptic Transmission/physiology , Aged , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cells, Cultured , Dendritic Spines/genetics , Dendritic Spines/physiology , Dependovirus/genetics , Disease Models, Animal , Female , Genetic Vectors/genetics , HEK293 Cells , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/genetics , Male , Memory Disorders/genetics , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , Synaptic Transmission/genetics
19.
PLoS One ; 8(7): e67748, 2013.
Article in English | MEDLINE | ID: mdl-23844082

ABSTRACT

Recombinant adenoassociated viral vectors (rAAV) have proven to be excellent candidates for gene therapy clinical applications. Recent results showed that cellular immunity to AAV represents a major challenge facing the clinical use of systemic administration of these vectors. Interestingly, no preclinical animal model has previously fully reproduced the clinical findings. The aim of the present work was to enhance the T cell immune response against AAV capsid in mice by the administration of a rAAV expressing the immunostimulatory cytokine IL-12. Our results indicate that although IL-12 expression enhanced the AAV capsid-specific immune response it failed to eliminate transduced hepatocytes and long-term expression was achieved. We found that AAV-mediated transgene expression is altered by IL-12-induced liver inflammation. However, IL-12 expression has no effect over preexisting AAV-mediated transgene expression. IL-12 down-regulates AAV mediated transgene expression via induction of IFN-γ production by NK and T cells, but without altering the transduction efficiency measured by viral genomes. Our results indicate that liver inflammation affects the formation of transcriptionally active AAV vector genomes through an unknown mechanism that can be avoided by the use of DNA-demethylating or anti-inflammatory agents.


Subject(s)
Dependovirus/immunology , Gene Expression/immunology , Hepatocytes/immunology , Interleukin-12/immunology , Liver/immunology , Transcription, Genetic/immunology , Animals , Capsid/immunology , Dependovirus/genetics , Female , Genetic Therapy/methods , Genetic Vectors/immunology , Hepatocytes/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Liver/pathology , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transgenes/immunology
20.
Gastroenterology ; 144(4): 818-828.e4, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23295442

ABSTRACT

BACKGROUND & AIMS: α1-Antichymotrypsin (α1-ACT), a member of the serpin family (SERPINA3), is an acute-phase protein secreted by hepatocytes in response to cytokines such as oncostatin M. α1-ACT is a protease inhibitor thought to limit tissue damage produced by excessive inflammation-associated proteolysis. However, α1-ACT also is detected in the nuclei of cells, where its activities are unknown. Expression of α1-ACT is down-regulated in human hepatocellular carcinoma (HCC) tissues and cells; we examined its roles in liver regeneration and HCC proliferation. METHODS: We measured levels of α1-ACT messenger RNA in human HCC samples and healthy liver tissue. We reduced levels of α1-ACT using targeted RNA interference in human HCC (HepG2) and mouse hepatocyte (AML12) cell lines, and overexpressed α1-ACT from lentiviral vectors in Huh7 (HCC) cells and adeno-associated viral vectors in livers of mice. We assessed proliferation, differentiation, and chromatin compaction in cultured cells, and liver regeneration and tumor formation in mice. RESULTS: Reducing levels of α1-ACT promoted proliferation of HCC cells in vitro. Oncostatin M up-regulated α1-ACT expression and nuclear translocation, which inhibited HCC cell proliferation and activated differentiation of mouse hepatocytes. We identified amino acids required for α1-ACT nuclear localization, and found that α1-ACT inhibits cell-cycle progression and anchorage-independent proliferation of HCC cells. HCC cells that overexpressed α1-ACT formed smaller tumors in mice than HCC cells that did not express the protein. α1-ACT was observed to self-associate and polymerize in the nuclei of cells; nuclear α1-ACT strongly bound chromatin to promote a condensed state that could prevent cell proliferation. CONCLUSIONS: α1-ACT localizes to the nuclei of hepatic cells to control chromatin condensation and proliferation. Overexpression of α1-ACT slows the growth of HCC xenograft tumors in nude mice.


Subject(s)
Carcinoma, Hepatocellular/pathology , Heterochromatin/metabolism , Liver Neoplasms/pathology , Liver Regeneration/physiology , alpha 1-Antichymotrypsin/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms, Experimental , Liver Regeneration/genetics , Mice , Mice, Nude , RNA, Messenger/analysis , Sensitivity and Specificity , Transplantation, Heterologous , alpha 1-Antichymotrypsin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...