Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Mol Ther ; 25(8): 1790-1804, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28550974

ABSTRACT

Lentiviral vector genomic RNA requires sequences that partially overlap wild-type HIV-1 gag and env genes for packaging into vector particles. These HIV-1 packaging sequences constitute 19.6% of the wild-type HIV-1 genome and contain functional cis elements that potentially compromise clinical safety. Here, we describe the development of a novel lentiviral vector (LTR1) with a unique genomic structure designed to prevent transfer of HIV-1 packaging sequences to patient cells, thus reducing the total HIV-1 content to just 4.8% of the wild-type genome. This has been achieved by reconfiguring the vector to mediate reverse-transcription with a single strand transfer, instead of the usual two, and in which HIV-1 packaging sequences are not copied. We show that LTR1 vectors offer improved safety in their resistance to remobilization in HIV-1 particles and reduced frequency of splicing into human genes. Following intravenous luciferase vector administration to neonatal mice, LTR1 sustained a higher level of liver transgene expression than an equivalent dose of a standard lentivirus. LTR1 vectors produce reverse-transcription products earlier and start to express transgenes significantly quicker than standard lentiviruses after transduction. Finally, we show that LTR1 is an effective lentiviral gene therapy vector as demonstrated by correction of a mouse hemophilia B model.


Subject(s)
Gene Transfer Techniques , Genetic Vectors/genetics , HIV-1/genetics , RNA, Viral , Regulatory Sequences, Ribonucleic Acid , Transduction, Genetic , Animals , Cell Line , Disease Models, Animal , Factor IX/genetics , Gene Expression , Gene Order , Genes, Reporter , Genetic Therapy , Genome, Viral , HIV Long Terminal Repeat , Hemophilia B/blood , Hemophilia B/genetics , Hemophilia B/therapy , Humans , Mice , Proviruses/genetics , Recombination, Genetic , Transgenes , Virus Replication/genetics
2.
Mediators Inflamm ; 2013: 245804, 2013.
Article in English | MEDLINE | ID: mdl-24453413

ABSTRACT

Macrophages are an integral part of the innate immune system and key players in pathogen clearance and tissue remodelling. Both functions are accomplished by a pivotal network of different macrophage subtypes, including proinflammatory M1 and anti-inflammatory M2 macrophages. Previously, our laboratory identified the transcription factor interferon regulatory factor 5 (IRF5) as the master regulator of the M1 macrophage polarisation. IRF5 was found to be highly expressed in human M1 compared to M2 macrophages. Furthermore, IRF5 dictates the expression of proinflammatory genes such as IL12b and IL23a whilst repressing anti-inflammatory genes like IL10. Here we show that murine bone marrow derived macrophages differentiated in vitro with GM-CSF are also characterised by high levels of IRF5 mRNA and protein and express proinflammatory cytokines upon LPS stimulation. These macrophages display characteristic expression of M1-marker MHC II but lack the M2-marker CD206. Significantly, we develop intracellular staining of IRF5- expressing macrophages and utilise it to recapitulate the in vitro results in an in vivo model of antigen-induced arthritis, emphasising their physiological relevance. Thus, we establish the species-invariant role of IRF5 in controlling the inflammatory macrophage phenotype both in vitro and in in vivo.


Subject(s)
Inflammation/etiology , Interferon Regulatory Factors/physiology , Macrophages/physiology , Animals , Arthritis, Experimental/etiology , Biomarkers , Cytokines/genetics , Disease Models, Animal , Gene Expression Profiling , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interferon Regulatory Factors/analysis , Interferon Regulatory Factors/genetics , Lectins, C-Type/analysis , Lipopolysaccharides/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Mannose Receptor , Mannose-Binding Lectins/analysis , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , Receptors, Cell Surface/analysis
3.
Hum Gene Ther ; 34(7-8): 273-288, 2023 04.
Article in English | MEDLINE | ID: mdl-36927149

ABSTRACT

The liver is a prime target for in vivo gene therapies using recombinant adeno-associated viral vectors. Multiple clinical trials have been undertaken for this target in the past 15 years; however, we are still to see market approval of the first liver-targeted adeno-associated virus (AAV)-based gene therapy. Inefficient expression of the therapeutic transgene, vector-induced liver toxicity and capsid, and/or transgene-mediated immune responses reported at high vector doses are the main challenges to date. One of the contributing factors to the insufficient clinical outcomes, despite highly encouraging preclinical data, is the lack of robust, biologically and clinically predictive preclinical models. To this end, this study reports findings of a functional evaluation of 6 AAV vectors in 12 preclinical models of the human liver, with the aim to uncover which combination of models is the most relevant for the identification of AAV capsid variant for safe and efficient transgene delivery to primary human hepatocytes. The results, generated by studies in models ranging from immortalized cells, iPSC-derived and primary hepatocytes, and primary human hepatic organoids to in vivo models, increased our understanding of the strengths and weaknesses of each system. This should allow the development of novel gene therapies targeting the human liver.


Subject(s)
Dependovirus , Liver , Humans , Dependovirus/genetics , Liver/metabolism , Genetic Therapy/methods , Hepatocytes/metabolism , Capsid Proteins/metabolism , Tropism , Genetic Vectors/genetics
4.
Mol Ther Methods Clin Dev ; 23: 135-146, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34703837

ABSTRACT

X-linked inherited ornithine transcarbamylase deficiency (OTCD) is the most common disorder affecting the liver-based urea cycle, a pathway enabling detoxification of nitrogen waste and endogenous arginine biosynthesis. Patients develop acute hyperammonemia leading to neurological sequelae or death despite the best-accepted therapy based on ammonia scavengers and protein-restricted diet. Liver transplantation is curative but associated with procedure-related complications and lifelong immunosuppression. Adeno-associated viral (AAV) vectors have demonstrated safety and clinical benefits in a rapidly growing number of clinical trials for inherited metabolic liver diseases. Engineered AAV capsids have shown promising enhanced liver tropism. Here, we conducted a good-laboratory practice-compliant investigational new drug-enabling study to assess the safety of intravenous liver-tropic AAVLK03 gene transfer of a human codon-optimized OTC gene. Juvenile cynomolgus monkeys received vehicle and a low and high dose of vector (2 × 1012 and 2 × 1013 vector genome (vg)/kg, respectively) and were monitored for 26 weeks for in-life safety with sequential liver biopsies at 1 and 13 weeks post-vector administration. Upon completion of monitoring, animals were euthanized to study vector biodistribution, immune responses, and histopathology. The product was well tolerated with no adverse clinical events, predominant hepatic biodistribution, and sustained supra-physiological OTC overexpression. This study supports the clinical deployment of intravenous AAVLK03 for severe OTCD.

5.
Mol Ther Methods Clin Dev ; 20: 357-365, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33553484

ABSTRACT

Lentiviral (LV) vectors based on human immunodeficiency virus type I (HIV-1) package two copies of their single-stranded RNA into vector particles. Normally, this RNA genome is reverse transcribed into a double-stranded DNA provirus that integrates into the cell genome, providing permanent gene transfer and long-term expression. Integration-deficient LV vectors have been developed to reduce the frequency of genomic integration and thereby limit their persistence in dividing cells. Here, we describe optimization of a reverse-transcriptase-deficient LV vector, which enables direct translation of LV RNA genomes upon cell entry, for transient expression of vector payloads as mRNA without a DNA intermediate. We have engineered a novel LV genome arrangement in which HIV-1 sequences are removed from the 5' end, to enable ribosomal entry from the 5' 7-methylguanylate cap for efficient translation of the vector payload. We have shown that this LV-mediated mRNA delivery platform provides transient transgene expression in vitro and in vivo. This has a potential application in gene and cell therapy scenarios requiring temporary payload expression in cells and tissues that can be targeted with pseudotyped LV vectors.

6.
EMBO Mol Med ; 13(2): e13158, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33369168

ABSTRACT

Urea cycle disorders (UCD) are inherited defects in clearance of waste nitrogen with high morbidity and mortality. Novel and more effective therapies for UCD are needed. Studies in mice with constitutive activation of autophagy unravelled Beclin-1 as druggable candidate for therapy of hyperammonemia. Next, we investigated efficacy of cell-penetrating autophagy-inducing Tat-Beclin-1 (TB-1) peptide for therapy of the two most common UCD, namely ornithine transcarbamylase (OTC) and argininosuccinate lyase (ASL) deficiencies. TB-1 reduced urinary orotic acid and improved survival under protein-rich diet in spf-ash mice, a model of OTC deficiency (proximal UCD). In AslNeo/Neo mice, a model of ASL deficiency (distal UCD), TB-1 increased ureagenesis, reduced argininosuccinate, and improved survival. Moreover, it alleviated hepatocellular injury and decreased both cytoplasmic and nuclear glycogen accumulation in AslNeo/Neo mice. In conclusion, Beclin-1-dependent activation of autophagy improved biochemical and clinical phenotypes of proximal and distal defects of the urea cycle.


Subject(s)
Argininosuccinic Aciduria , Ornithine Carbamoyltransferase Deficiency Disease , Urea Cycle Disorders, Inborn , Animals , Autophagy , Beclin-1/genetics , Mice
7.
iScience ; 23(12): 101808, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33305175

ABSTRACT

Explosion of gene therapy approaches for treating rare monogenic and common liver disorders created an urgent need for disease models able to replicate human liver cellular environment. Available models lack 3D liver structure or are unable to survive in long-term culture. We aimed to generate and test a 3D culture system that allows long-term maintenance of human liver cell characteristics. The in vitro whole-organ "Bioreactor grown Artificial Liver Model" (BALM) employs a custom-designed bioreactor for long-term 3D culture of human induced pluripotent stem cells-derived hepatocyte-like cells (hiHEPs) in a mouse decellularized liver scaffold. Adeno-associated viral (AAV) and lentiviral (LV) vectors were introduced by intravascular injection. Substantial AAV and LV transgene expression in the BALM-grown hiHEPs was detected. Measurement of secreted proteins in the media allowed non-invasive monitoring of the system. We demonstrated that humanized whole-organ BALM is a valuable tool to generate pre-clinical data for investigational medicinal products.

8.
Sci Rep ; 10(1): 2121, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034258

ABSTRACT

We have previously designed a library of lentiviral vectors to generate somatic-transgenic rodents to monitor signalling pathways in diseased organs using whole-body bioluminescence imaging, in conscious, freely moving rodents. We have now expanded this technology to adeno-associated viral vectors. We first explored bio-distribution by assessing GFP expression after neonatal intravenous delivery of AAV8. We observed widespread gene expression in, central and peripheral nervous system, liver, kidney and skeletal muscle. Next, we selected a constitutive SFFV promoter and NFκB binding sequence for bioluminescence and biosensor evaluation. An intravenous injection of AAV8 containing firefly luciferase and eGFP under transcriptional control of either element resulted in strong and persistent widespread luciferase expression. A single dose of LPS-induced a 10-fold increase in luciferase expression in AAV8-NFκB mice and immunohistochemistry revealed GFP expression in cells of astrocytic and neuronal morphology. Importantly, whole-body bioluminescence persisted up to 240 days. We have validated a novel biosensor technology in an AAV system by using an NFκB response element and revealed its potential to monitor signalling pathway in a non-invasive manner in a model of LPS-induced inflammation. This technology complements existing germline-transgenic models and may be applicable to other rodent disease models.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Mice, Transgenic/genetics , Animals , Biosensing Techniques/methods , Gene Expression/genetics , Green Fluorescent Proteins/genetics , Inflammation/genetics , Luciferases, Firefly/genetics , Mice , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Spleen Focus-Forming Viruses/genetics , Transcription, Genetic/genetics
9.
Hum Gene Ther ; 30(1): 79-87, 2019 01.
Article in English | MEDLINE | ID: mdl-30027761

ABSTRACT

Recombinant adeno-associated virus (rAAV) vectors are a promising platform for in vivo gene therapy. The presence of neutralizing antibodies (Nab) against AAV capsids decreases cell transduction efficiency and is a common exclusion criterion for participation in clinical trials. Novel engineered capsids are being generated to improve gene delivery to the target cells and facilitate success of clinical trials; however, the prevalence of antibodies against such capsids remains largely unknown. We therefore assessed the seroprevalence of antibodies against a novel synthetic liver-tropic capsid AAV-LK03. We measured seroprevalence of immunoglobulin (Ig)G (i.e., neutralizing and nonneutralizing) antibodies and Nab to AAV-LK03 in a cohort of 323 UK patients (including 260 pediatric) and 52 juvenile rhesus macaques. We also performed comparative analysis of seroprevalence of Nab against wild-type AAV8 and AAV3B capsids. Overall IgG seroprevalence for AAV-LK03 was 39% in human samples. The titer increased with age. Prevalence of Nab was 23%, 35%, and 18% for AAV-LK03, AAV3B, and AAV8, respectively, with the lowest seroprevalence between 3 and 17 years of age for all serotypes. Presence of Nab against AAV-LK03 decreased from 36% in the youngest cohort (birth to 6 months) to 7% in older primary school-age children (9-11 years) and then progressively increased to 54% in late adulthood. Cross-reactivity between serotypes was >60%. Nab seroprevalence in macaques was 62%, 85%, and 40% for AAV-LK03, AAV3B, and AAV8, respectively. When planning for AAV gene therapy clinical trials, knowing the seropositivity of the target population is critical. In the population studied, AAV seroprevalence for AAV serotypes tested was low. However, high cross-reactivity between AAV serotypes remains a barrier for re-injection. Shifts in Nab seroprevalence during the first decade need to be confirmed by longitudinal studies. This possibility suggests that pediatric patients could respond differently to AAV therapy according to age. If late childhood is an ideal age window, intervention at an early age when maternal Nab levels are high may be challenging. Nab-positive children excluded from trials could be rescreened for eligibility at regular intervals because this status may change.


Subject(s)
Antibodies, Viral/immunology , Dependovirus/immunology , Genetic Vectors/adverse effects , Seroepidemiologic Studies , Adolescent , Antibodies, Neutralizing , Antibodies, Viral/blood , Capsid/immunology , Child , Child, Preschool , Cross Reactions , Dependovirus/classification , Dependovirus/genetics , Female , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Infant , Infant, Newborn , Male , Population Surveillance , Sex Factors , Transduction, Genetic , United Kingdom/epidemiology
10.
Nat Med ; 24(9): 1317-1323, 2018 09.
Article in English | MEDLINE | ID: mdl-30013199

ABSTRACT

For inherited genetic diseases, fetal gene therapy offers the potential of prophylaxis against early, irreversible and lethal pathological change. To explore this, we studied neuronopathic Gaucher disease (nGD), caused by mutations in GBA. In adult patients, the milder form presents with hepatomegaly, splenomegaly and occasional lung and bone disease; this is managed, symptomatically, by enzyme replacement therapy. The acute childhood lethal form of nGD is untreatable since enzyme cannot cross the blood-brain barrier. Patients with nGD exhibit signs consistent with hindbrain neurodegeneration, including neck hyperextension, strabismus and, often, fatal apnea1. We selected a mouse model of nGD carrying a loxP-flanked neomycin disruption of Gba plus Cre recombinase regulated by the keratinocyte-specific K14 promoter. Exclusive skin expression of Gba prevents fatal neonatal dehydration. Instead, mice develop fatal neurodegeneration within 15 days2. Using this model, fetal intracranial injection of adeno-associated virus (AAV) vector reconstituted neuronal glucocerebrosidase expression. Mice lived for up to at least 18 weeks, were fertile and fully mobile. Neurodegeneration was abolished and neuroinflammation ameliorated. Neonatal intervention also rescued mice but less effectively. As the next step to clinical translation, we also demonstrated the feasibility of ultrasound-guided global AAV gene transfer to fetal macaque brains.


Subject(s)
Fetus/metabolism , Genetic Therapy , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/therapy , Animals , Gaucher Disease/genetics , Gaucher Disease/therapy , Humans , Infant , Injections, Intravenous , Injections, Intraventricular , Mice, Inbred C57BL
11.
Nat Commun ; 9(1): 3505, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30158522

ABSTRACT

Argininosuccinate lyase (ASL) belongs to the hepatic urea cycle detoxifying ammonia, and the citrulline-nitric oxide (NO) cycle producing NO. ASL-deficient patients present argininosuccinic aciduria characterised by hyperammonaemia, multiorgan disease and neurocognitive impairment despite treatment aiming to normalise ammonaemia without considering NO imbalance. Here we show that cerebral disease in argininosuccinic aciduria involves neuronal oxidative/nitrosative stress independent of hyperammonaemia. Intravenous injection of AAV8 vector into adult or neonatal ASL-deficient mice demonstrates long-term correction of the hepatic urea cycle and the cerebral citrulline-NO cycle, respectively. Cerebral disease persists if ammonaemia only is normalised but is dramatically reduced after correction of both ammonaemia and neuronal ASL activity. This correlates with behavioural improvement and reduced cortical cell death. Thus, neuronal oxidative/nitrosative stress is a distinct pathophysiological mechanism from hyperammonaemia. Disease amelioration by simultaneous brain and liver gene transfer with one vector, to treat both metabolic pathways, provides new hope for hepatocerebral metabolic diseases.


Subject(s)
Argininosuccinate Lyase/metabolism , Argininosuccinic Aciduria/metabolism , Argininosuccinic Aciduria/therapy , Animals , Argininosuccinate Lyase/genetics , Argininosuccinic Aciduria/genetics , Brain Diseases/genetics , Brain Diseases/metabolism , Brain Diseases/therapy , Citrulline/metabolism , Genetic Therapy , Hyperammonemia/genetics , Hyperammonemia/metabolism , Hyperammonemia/therapy , Liver/cytology , Mice , Neurons/metabolism , Nitric Oxide/metabolism , Nitrosative Stress/genetics , Nitrosative Stress/physiology
12.
Sci Rep ; 7: 41874, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28157201

ABSTRACT

Molecular mechanisms regulating liver repair following cholestatic injury remain largely unknown. We have combined a mouse model of acute cholestatic liver injury, partial bile duct ligation (pBDL), with a novel longitudinal bioimaging methodology to quantify transcription factor activity during hepatic injury and repair. We administered lentiviral transcription factor activated luciferase/eGFP reporter (TFAR) cassettes to neonatal mice enabling longitudinal TFAR profiling by continued bioimaging throughout the lives of the animals and following pBDL in adulthood. Neonatal intravascular injection of VSV-G pseudotyped lentivirus resulted in almost exclusive transduction of hepatocytes allowing analysis of hepatocyte-specific transcription factor activity. We recorded acute but transient responses with NF-κB and Smad2/3 TFAR whilst our Notch reporter was repressed over the 40 days of evaluation post-pBDL. The bipotent hepatic progenitor cell line, HepaRG, can be directed to differentiate into hepatocytes and biliary epithelia. We found that forced expression of the Notch inhibitor NUMB in HepaRG resulted in enhanced hepatocyte differentiation and proliferation whereas over-expressing the Notch agonist JAG1 resulted in biliary epithelial differentiation. In conclusion, our data demonstrates that hepatocytes rapidly upregulate NF-κB and Smad2/3 activity, whilst repressing Notch signalling. This transcriptional response to cholestatic liver injury likely promotes partial de-differentiation to allow pro-regenerative proliferation of hepatocytes.


Subject(s)
Cholestasis/metabolism , Hepatocytes/metabolism , Liver Diseases/metabolism , Signal Transduction , 3T3 Cells , Animals , Cell Differentiation , Cell Proliferation , Cholestasis/complications , Cholestasis/diagnostic imaging , Epithelial Cells/cytology , Epithelial Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Hepatocytes/cytology , Hepatocytes/physiology , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Lentivirus/genetics , Liver Diseases/diagnostic imaging , Liver Diseases/etiology , Luciferases/genetics , Luciferases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism
13.
Sci Rep ; 7(1): 6374, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743959

ABSTRACT

Luciferase bioimaging in living animals is increasingly being applied in many fields of biomedical research. Rodent imaging usually involves anaesthetising the animal during data capture, however, the biological consequences of anaesthesia have been largely overlooked. We have evaluated luciferase bioimaging in conscious, unrestrained mice after neonatal intracranial or intravascular administration of lentiviral, luciferase reporter cassettes (biosensors); we present real-time analyses from the first day of life to adulthood. Anaesthetics have been shown to exert both neurotoxic and neuroprotective effects during development and in models of brain injury. Mice subjected to bioimaging after neonatal intracranial or intravascular administration of biosensors, targeting the brain and liver retrospectively showed no significant difference in luciferase expression when conscious or unconscious throughout development. We applied conscious bioimaging to the assessment of NFκB and STAT3 transcription factor activated reporters during the earliest stages of development in living, unrestrained pups. Our data showed unique longitudinal activities for NFκB and STAT3 in the brain of conscious mice. Conscious bioimaging was applied to a neonatal mouse model of cerebral palsy (Hypoxic-Ischaemic Encephalopathy). Imaging of NFκB reporter before and after surgery showed a significant increase in luciferase expression, coinciding with secondary energy failure, in lesioned mice compared to controls.


Subject(s)
Brain/metabolism , Cerebral Palsy/metabolism , Liver/metabolism , Luciferases/metabolism , Molecular Imaging/methods , Animals , Animals, Newborn , Biosensing Techniques/methods , Cerebral Palsy/surgery , Consciousness , Disease Models, Animal , Genetic Vectors/administration & dosage , Injections, Intra-Arterial , Lentivirus/genetics , Luciferases/genetics , Mice , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Recombinant Proteins/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
14.
Sci Rep ; 5: 11842, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26138224

ABSTRACT

The application of transcription factor activated luciferase reporter cassettes in vitro is widespread but potential for in vivo application has not yet been realized. Bioluminescence imaging enables non-invasive tracking of gene expression in transfected tissues of living rodents. However the mature immune response limits luciferase expression when delivered in adulthood. We present a novel approach of tissue-targeted delivery of transcription factor activated luciferase reporter lentiviruses to neonatal rodents as an alternative to the existing technology of generating germline transgenic light producing rodents. At this age, neonates acquire immune tolerance to the conditionally responsive luciferase reporter. This simple and transferrable procedure permits surrogate quantitation of transcription factor activity over the lifetime of the animal. We show principal efficacy by temporally quantifying NFκB activity in the brain, liver and lungs of somatotransgenic reporter mice subjected to lipopolysaccharide (LPS)-induced inflammation. This response is ablated in Tlr4(-/-) mice or when co-administered with the anti-inflammatory glucocorticoid analogue dexamethasone. Furthermore, we show the malleability of this technology by quantifying NFκB-mediated luciferase expression in outbred rats. Finally, we use somatotransgenic bioimaging to longitudinally quantify LPS- and ActivinA-induced upregulation of liver specific glucocorticoid receptor and Smad2/3 reporter constructs in somatotransgenic mice, respectively.


Subject(s)
Luciferases, Firefly/genetics , Transcription Factors/physiology , Transcriptional Activation/immunology , Animals , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Lentivirus/genetics , Lipopolysaccharides/pharmacology , Luciferases, Firefly/biosynthesis , Mice , NIH 3T3 Cells , Organ Specificity , Rats, Sprague-Dawley , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics
15.
Arthritis Rheumatol ; 66(10): 2728-38, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24965881

ABSTRACT

OBJECTIVE: Tumor necrosis factor (TNF) signals via 2 receptors, TNFR type I (TNFRI) and TNFRII, with distinct cellular distribution and signaling functions. In rheumatoid arthritis (RA), the net effect of TNFR signaling favors inflammatory responses while inhibiting the activity of regulatory T cells. TNFRII signaling has been shown to promote Treg cell function. To assess the relative contributions of TNFRI and TNFRII signaling to inflammatory and regulatory responses in vivo, we compared the effect of TNF blockade, hence TNFRI/II, versus TNFRI alone in collagen-induced arthritis (CIA) as a model of RA. METHODS: Mice with established arthritis were treated for 10 days with anti-mouse TNFRI domain antibody (dAb; DMS5540), an isotype control dAb (DMS5538), or murine TNFRII genetically fused with mouse IgG1 Fc domain (mTNFRII-Fc) beginning on the day of arthritis onset, and disease progression was monitored. Systemic cytokine concentrations and numbers of T cell subsets in lymph nodes and spleens were measured, and intrinsic Treg cell function was determined by ex vivo suppression assays. RESULTS: Progression of CIA was suppressed similarly by TNFRI (DMS5540) and TNFRI/II (mTNFRII-Fc) blockade. However, blockade of TNFRI/II led to increased effector T cell activity, which was not observed after selective TNFRI blockade, suggesting an immunoregulatory role of TNFRII. In support of this, TNFRI blockade, but not TNFRI/II blockade, expanded and activated Treg cells. Furthermore, a dramatic increase in expression of the Treg cell signature genes FoxP3 and TNFRII was observed in joints undergoing remission, which supports the notion that these molecules have a physiologic role in the resolution of inflammation. CONCLUSION: We propose that a therapeutic strategy that targets TNFRI while sparing TNFRII has the potential to both inhibit inflammation and promote Treg cell activity, which might be superior to TNF blockade.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Single-Domain Antibodies/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/immunology , Cell Proliferation/drug effects , Forkhead Transcription Factors/metabolism , Inflammation/drug therapy , Inflammation/immunology , Male , Mice , Mice, Inbred DBA , Recombinant Fusion Proteins/pharmacology , Single-Domain Antibodies/pharmacology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/immunology
16.
Arthritis Res Ther ; 12(3): R107, 2010.
Article in English | MEDLINE | ID: mdl-20525198

ABSTRACT

INTRODUCTION: Type 4 phosphodiesterases (PDE4) play an important role in immune cells through the hydrolysis of the second messenger, cAMP. Inhibition of PDE4 has previously been shown to suppress immune and inflammatory responses, demonstrating PDE4 to be a valid therapeutic target for immune-mediated pathologies. We assessed the anti-inflammatory effects of a novel PDE4 inhibitor, apremilast, in human synovial cells from rheumatoid arthritis (RA) patients, as well as two murine models of arthritis. METHODS: Cells liberated from tissue excised from arthritic joints of RA patients were cultured in the presence of increasing concentrations of apremilast for 48 hours and spontaneous tumour necrosis factor-alpha (TNFalpha) production was analysed in culture supernatants by ELISA. In addition, arthritis was induced in BALB/c and DBA/1 mice by passive transfer of anti-type II collagen mAb and immunisation with type II collagen, respectively. Mice with established arthritis received 5 or 25 mg/kg apremilast and disease severity was monitored relative to mice receiving vehicle alone. At the end of the study, paws were removed and processed for histopathological assessment. Behavioural effects of apremilast, relative to rolipram, were assessed in naïve DBA/1 mice using an automated activity monitor (LABORAS). RESULTS: Apremilast dose dependently inhibited spontaneous release of TNFalpha from human rheumatoid synovial membrane cultures. Furthermore, apremilast significantly reduced clinical score in both murine models of arthritis over a ten day treatment period and maintained a healthy joint architecture in a dose-dependent manner. Importantly, unlike rolipram, apremilast demonstrated no adverse behavioural effects in naïve mice. CONCLUSIONS: Apremilast is an orally available PDE4 inhibitor that reduces TNFalpha production from human synovial cells and significantly suppresses experimental arthritis. Apremilast appears to be a potential new agent for the treatment of rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/metabolism , Phosphodiesterase 4 Inhibitors , Synovial Membrane/metabolism , Thalidomide/analogs & derivatives , Tumor Necrosis Factor-alpha/metabolism , Animals , Arthritis, Rheumatoid/pathology , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Phosphodiesterase Inhibitors/adverse effects , Phosphodiesterase Inhibitors/therapeutic use , Rolipram/adverse effects , Rolipram/therapeutic use , Severity of Illness Index , Synovial Membrane/drug effects , Thalidomide/pharmacology , Thalidomide/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL