Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
J Neuroinflammation ; 19(1): 155, 2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35715845

ABSTRACT

BACKGROUND: Vagus nerve stimulation has been suggested to affect immune responses, partly through a neuronal circuit requiring sympathetic innervation of the splenic nerve bundle and norepinephrine (NE) release. Molecular and cellular mechanisms of action remain elusive. Here, we investigated the therapeutic value of this neuromodulation in inflammatory bowel disease (IBD) by applying electrical splenic nerve bundle stimulation (SpNS) in mice with dextran sulfate sodium (DSS)-induced colitis. METHODS: Cuff electrodes were implanted around the splenic nerve bundle in mice, whereupon mice received SpNS or sham stimulation. Stimulation was applied 6 times daily for 12 days during DSS-induced colitis. Colonic and splenic tissues were collected for transcriptional analyses by qPCR and RNA-sequencing (RNA-seq). In addition, murine and human splenocytes were stimulated with lipopolysaccharide (LPS) in the absence or presence of NE. Single-cell RNA-seq data from publicly available data sets were analyzed for expression of ß-adrenergic receptors (ß-ARs). RESULTS: Colitic mice undergoing SpNS displayed reduced colon weight/length ratios and showed improved Disease Activity Index scores with reduced Tumor Necrosis Factor α mRNA expression in the colon compared with sham stimulated mice. Analyses of splenocytes from SpNS mice using RNA-seq demonstrated specific immune metabolism transcriptome profile changes in myeloid cells. Splenocytes showed expression of ß-ARs in myeloid and T cells. Cytokine production was reduced by NE in mouse and human LPS-stimulated splenocytes. CONCLUSIONS: Together, our results demonstrate that SpNS reduces clinical features of colonic inflammation in mice with DSS-induced colitis possibly by inhibiting splenic myeloid cell activation. Our data further support exploration of the clinical use of SpNS for patients with IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Colitis/chemically induced , Colitis/therapy , Colon/metabolism , Colon/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Electric Stimulation , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/therapy , Lipopolysaccharides/adverse effects , Mice , Mice, Inbred C57BL
2.
Ann Rheum Dis ; 80(6): 714-726, 2021 06.
Article in English | MEDLINE | ID: mdl-33602797

ABSTRACT

Animal models for inflammatory arthritides such as rheumatoid arthritis (RA) and psoriatic arthritis are widely accepted and frequently used to identify pathological mechanisms and validate novel therapeutic strategies. Unfortunately, many publications reporting on these animal studies lack detailed description and appropriate assessment of the distinct histopathological features of arthritis: joint inflammation, cartilage damage and bone erosion. Therefore, the European consortium BeTheCure, consisting of 38 academic and industrial partners from 15 countries, set as goal to standardise the histological evaluation of joint sections from animal models of inflammatory arthritis. The consensual approach of a task force including 16 academic and industrial scientists as well as laboratory technicians has resulted in the development of the Standardised Microscopic Arthritis Scoring of Histological sections ('SMASH') recommendations for a standardised processing and microscopic scoring of the characteristic histopathological features of arthritis, exemplified by four different rodent models for arthritis: murine collagen-induced arthritis, collagen-antibody-induced arthritis, human tumour necrosis factor transgenic Tg197 mice and rat pristane-induced arthritis, applicable to any other inflammatory arthritis model. Through standardisation, the SMASH recommendations are designed to improve and maximise the information derived from in vivo arthritis experiments and to promote reproducibility and transparent reporting on such studies. In this manuscript, we will discuss and provide recommendations for analysis of histological joint sections: identification of the regions of interest, sample preparation, staining procedures and quantitative scoring methods. In conclusion, awareness of the different features of the arthritis pathology in animal models of inflammatory arthritis is of utmost importance for reliable research outcome, and the standardised histological processing and scoring methods in these SMASH recommendations will help increase uniformity and reproducibility in preclinical research on inflammatory arthritis.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Rats , Reproducibility of Results
3.
Surg Radiol Anat ; 43(3): 377-384, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33104863

ABSTRACT

PURPOSE: To evaluate the morphology and course of the splenic artery, which might impact the surgical implantation of systems that stimulate the nerves surrounding the splenic artery. Experimental studies indicate that these nerves play an important part in immune modulation, and might be a potential target in the treatment of autoimmune diseases. METHODS: This retrospective cohort study made use of contrast-enhanced CT images from 40 male and 40 female patients (age 30-69) that underwent a CT examination of the aorta, kidneys or pancreas. Anatomic features were described including total splenic artery length, calibers, tortuosity, the presence of arterial loops and the branching pattern of the splenic artery. RESULTS: No age-gender-related differences could be found related to tortuosity or branching pattern. The length of splenic artery in contact with pancreatic tissue decreased with increasing age, but was not different between genders. Artery diameters were wider in male compared to female subjects. Loops of variable directions, that represent a part of the artery that curls out of the pancreatic tissue, were identified in each age-gender category and were present in nearly all subjects (86%). CONCLUSION: This study suggests that although some anatomic features of the splenic artery are subject to factors as age and gender, the tortuosity of the splenic artery is not age dependent. Most subjects had one or multiple loops, which can serve as a target for neuromodulatory devices. Future studies should investigate whether splenic nerve stimulation is safe and feasible.


Subject(s)
Splenic Artery/anatomy & histology , Adult , Age Factors , Aged , Female , Humans , Male , Middle Aged , Pancreas/blood supply , Pancreas/diagnostic imaging , Retrospective Studies , Sex Factors , Splenic Artery/diagnostic imaging , Tomography, X-Ray Computed
4.
Mol Med ; 25(1): 1, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30616543

ABSTRACT

BACKGROUND: Both the parasympathetic and sympathetic nervous system exert control over innate immune responses. In inflammatory bowel disease, sympathetic innervation in intestinal mucosa is reduced. Our aim was to investigate the role of sympathetic innervation to the intestine on regulation of the innate immune responses. METHODS: In lipopolysaccharide (LPS)-stimulated macrophages, we evaluated the effect of adrenergic receptor activation on cytokine production and metabolic profile. In vivo, the effect of sympathetic denervation on mucosal innate immune responses using 6-hydroxydopamine (6-OHDA), or using surgical transection of the superior mesenteric nerve (sympathectomy) was tested in Rag1-/- mice that lack T- and B-lymphocytes. RESULTS: In murine macrophages, adrenergic ß2 receptor activation elicited a dose-dependent reduction of LPS-induced cytokines, reduced LPS-induced glycolysis and increased maximum respiration. Sympathectomy led to a significantly decreased norepinephrine concentration in intestinal tissue. Within 14 days after sympathectomy, mice developed clinical signs of colitis, colon oedema and excess colonic cytokine production. Both 6-OHDA and sympathectomy led to prominent goblet cell depletion and histological damage of colonic mucosa. CONCLUSIONS: We conclude that the sympathetic nervous system plays a regulatory role in constraining innate immune cell reactivity towards microbial challenges, likely via the adrenergic ß2 receptor.


Subject(s)
Colitis/immunology , Immunity, Innate , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/innervation , Sympathetic Nervous System/immunology , Adrenergic beta-2 Receptor Agonists/pharmacology , Albuterol/pharmacology , Animals , Cells, Cultured , Colitis/pathology , Colon/drug effects , Colon/pathology , Cytokines/genetics , Cytokines/immunology , Female , Humans , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Oxidopamine/pharmacology
5.
Brain Behav Immun ; 80: 238-246, 2019 08.
Article in English | MEDLINE | ID: mdl-30885844

ABSTRACT

The autonomic nervous system innervates all lymphoid tissues including the spleen therefore providing a link between the central nervous system and the immune system. The only known mechanism of neural inhibition of inflammation in the spleen relies on the production of norepinephrine by splenic catecholaminergic fibers which binds to ß2-adrenergic receptors (ß 2-ARs) of CD4+ T cells. These CD4+ T cells trigger the release of acetylcholine that inhibits the secretion of inflammatory cytokines by macrophages through α7 nicotinic acetylcholine receptor (α7nAchRs) signaling. While the vagal anti-inflammatory pathway has been extensively studied in rodents, it remains to be determined whether it coexists with other neural pathways. Here, we have found that three nerve branches project to the spleen in mice. While two of these nerves are associated with an artery and contain catecholaminergic fibers, the third is located at the apex of the spleen and contain both catecholaminergic and cholinergic fibers. We found that electrical stimulation of the apical nerve, but not the arterial nerves, inhibited inflammation independently of lymphocytes. In striking contrast to the anti-inflammatory pathway mechanism described so far, we also found that the inhibition of inflammation by apical nerve electrical stimulation relied on signaling by both ß 2-ARs and α7nAchRs in myeloid cells, with these two signaling pathways acting in parallel. Most importantly, apical splenic nerve electrical stimulation mitigated clinical symptoms in a mouse model of rheumatoid arthritis further providing the proof-of-concept that such an approach could be beneficial in patients with Immune-mediated inflammatory diseases.


Subject(s)
Myeloid Cells/immunology , Receptors, Adrenergic/immunology , Receptors, Nicotinic/immunology , Spleen/immunology , Spleen/innervation , Acetylcholine/metabolism , Animals , Electric Stimulation , Female , Mice, Inbred C57BL , Mice, Transgenic , Norepinephrine/metabolism , Spleen/physiopathology , Tumor Necrosis Factor-alpha/immunology , Vagus Nerve/immunology , Vagus Nerve Stimulation
6.
Arthritis Rheum ; 65(5): 1203-12, 2013 May.
Article in English | MEDLINE | ID: mdl-23335223

ABSTRACT

OBJECTIVE: Glucocorticoid-induced leucine zipper (GILZ) has effects on inflammatory pathways that suggest it to be a key inhibitory regulator of the immune system, and its expression is exquisitely sensitive to induction by glucocorticoids. We undertook this study to test our hypothesis that GILZ deficiency would exacerbate experimental immune-mediated inflammation and impair the effects of glucocorticoids on inflammation and, correspondingly, that exogenous GILZ would inhibit these events. METHODS: GILZ(-/-) mice were generated using the Cre/loxP system, and responses were studied in delayed-type hypersensitivity (DTH), antigen-induced arthritis (AIA), K/BxN serum-transfer arthritis, and lipopolysaccharide (LPS)-induced cytokinemia. Therapeutic expression of GILZ via administration of recombinant adeno-associated virus expressing the GILZ gene (GILZ-rAAV) was compared to the effects of glucocorticoid in collagen-induced arthritis (CIA). RESULTS: Increased T cell proliferation and DTH were observed in GILZ(-/-) mice, but neither AIA nor K/BxN serum-transfer arthritis was affected, and GILZ deficiency did not affect LPS-induced cytokinemia. Deletion of GILZ did not impair the effects of exogenous glucocorticoids on CIA or cytokinemia. In contrast, overexpression of GILZ in joints significantly inhibited CIA, with an effect similar to that of dexamethasone. CONCLUSION: Despite effects on T cell activation, GILZ deficiency had no effect on effector pathways of arthritis and was unexpectedly redundant with effects of glucocorticoids. These findings do not support the hypothesis that GILZ is central to the actions of glucocorticoids, but the efficacy of exogenous GILZ in CIA suggests that further evaluation of GILZ in inflammatory disease is required.


Subject(s)
Arthritis, Experimental/therapy , Hypersensitivity, Delayed/therapy , Transcription Factors/genetics , Adenoviridae/genetics , Animals , Arthritis, Experimental/genetics , Cell Proliferation , Dexamethasone/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Gene Targeting , Genetic Therapy/methods , Glucocorticoids/pharmacology , Hypersensitivity, Delayed/genetics , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , Transcription Factors/deficiency , Transduction, Genetic
7.
Front Neurosci ; 17: 1125492, 2023.
Article in English | MEDLINE | ID: mdl-37123375

ABSTRACT

The magnitude of innate inflammatory immune responses is dependent on interactions between peripheral neural and immune cells. In particular, a cholinergic anti-inflammatory pathway (CAP) has been identified in the spleen whereby noradrenaline (NA) released by splenic nerves binds to ß2-adrenergic receptors (ß2-AR) on CD4+ T cells which, in turn, release acetylcholine (ACh). The binding of ACh to α7 acetylcholine receptors (α7-AChR) expressed by splenic macrophages inhibits the production of inflammatory cytokines, including tumor necrosis factor (TNF). However, the role of ACh-secreting CD4+ T-cells in the CAP is still controversial and largely based on the absence of this anti-inflammatory pathway in mice lacking T-cells (nude, FoxN1-/-). Using four conscious, non-lymphopenic transgenic mouse models, we found that, rather than acting on CD4+ T-cells, NA released by splenic nerve terminals acts directly onto ß2-AR on splenic myeloid cells to exert this anti-inflammatory effect. We also show that, while larger doses of LPS are needed to trigger CAP in nude mouse strain compared to other strains, TNF production can be inhibited in these animals lacking CD4+ T-cell by stimulating either the vagus or the splenic nerve. We demonstrate that CD4+ T-cells are dispensable for the CAP after antibody-mediated CD4+ T-cell depletion in wild type mice. Furthermore, we found that NA-mediated inhibition of in vitro LPS-induced TNF secretion by human or porcine splenocytes does not require α7-AChR signaling. Altogether our data demonstrate that activation of the CAP by stimulation of vagus or splenic nerves in mice is mainly mediated by direct binding of NA to ß2-AR on splenic macrophages, and suggest that the same mechanism is at play in larger species.

8.
Ann Rheum Dis ; 71(8): 1411-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22523426

ABSTRACT

OBJECTIVE: The mechanisms contributing to the persistent activation of macrophages in rheumatoid arthritis (RA) are not fully understood. Some studies suggest that endogenous toll-like receptor (TLR) ligands promote the chronic inflammation observed in RA. The objective of this study was to identify endogenous TLR ligands expressed in RA synovial tissue (ST) based on their ability to bind the extracellular domains of TLR2 or TLR4. METHODS: A yeast two-hybrid cDNA library was constructed from ST obtained by arthroscopy from patients with RA and screened using the extracellular domains of TLR2 and TLR4 as the bait. Interactions between TLRs and Snapin were demonstrated by reciprocal co-immunoprecipitation. ST was examined by histology, and single- and two-colour immunohistochemistry and quantitative reverse transcriptase PCR. Snapin (SNAP - associated protein) expression in macrophages was examined by Western Blot analysis and confocal microscopy. The ability of Snapin to activate through TLR2 was examined. RESULTS: Employing a yeast two-hybrid system, Snapin was the most frequently identified molecule that interacted with TLR2. These results were confirmed by pull-down of in vitro-expressed Snapin together with TLR2. By immunohistochemistry and quantitative reverse transcriptase PCR, Snapin was highly expressed in RA ST, and it was readily detected in macrophages, where it co-localised in the late endosomes. ST Snapin expression correlated with inflammation and was not disease specific. Finally, Snapin was capable of activating through TLR2. CONCLUSION: These observations identify Snapin as a novel endogenous TLR2 ligand in RA, and thus support a role for persistent TLR2 signalling in the pathogenesis of RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Synovial Membrane/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Vesicular Transport Proteins/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/surgery , Blotting, Western , Endosomes/metabolism , Endosomes/pathology , Gene Expression , Gene Library , Humans , Ligands , Macrophage Activation , Macrophages/metabolism , Protein Binding , Saccharomyces cerevisiae/physiology , Synovial Membrane/pathology , Two-Hybrid System Techniques , Vesicular Transport Proteins/genetics
9.
Front Neurosci ; 16: 1088628, 2022.
Article in English | MEDLINE | ID: mdl-36620453

ABSTRACT

Introduction: The autonomic nervous system is a key regulator of inflammation. Electrical stimulation of the vagus nerve has been shown to have some preclinical efficacy. However, only a few clinical studies have been reported to treat inflammatory diseases. The present study evaluates, for the first time, neuromodulation of the splenic arterial neurovascular bundle (SpA NVB) in patients undergoing minimally invasive esophagectomy (MIE), in which the SpA NVB is exposed as part of the procedure. Methods: This single-center, single-arm study enrolled 13 patients undergoing MIE. During the abdominal phase of the MIE, a novel cuff was placed around the SpA NVB, and stimulation was applied. The primary endpoint was the feasibility and safety of cuff application and removal. A secondary endpoint included the impact of stimulation on SpA blood flow changes during the stimulation, and an exploratory point was C-reactive protein (CRP) levels on postoperative day (POD) 2 and 3. Results: All patients successfully underwent placement, stimulation, and removal of the cuff on the SpA NVB with no adverse events related to the investigational procedure. Stimulation was associated with an overall reduction in splenic arterial blood flow but not with changes in blood pressure or heart rate. When compared to historic Propensity Score Matched (PSM) controls, CRP levels on POD2 (124 vs. 197 mg/ml, p = 0.032) and POD3 (151 vs. 221 mg/ml, p = 0.033) were lower in patients receiving stimulation. Conclusion: This first-in-human study demonstrated for the first time that applying a cuff around the SpA NVB and subsequent stimulation is safe, feasible, and may have an effect on the postoperative inflammatory response following MIE. These findings suggest that SpA NVB stimulation may offer a new method for immunomodulatory therapy in acute or chronic inflammatory conditions.

10.
Mol Med ; 17(9-10): 937-48, 2011.
Article in English | MEDLINE | ID: mdl-21607292

ABSTRACT

The immunomodulatory effect of the autonomic nervous system has raised considerable interest over the last decades. Studying the influence on the immune system and the role in inflammation of the sympathetic as well as the parasympathetic nervous system not only will increase our understanding of the mechanism of disease, but also could lead to the identification of potential new therapeutic targets for chronic immune-mediated inflammatory diseases, such as rheumatoid arthritis (RA). An imbalanced autonomic nervous system, with a reduced parasympathetic and increased sympathetic tone, has been a consistent finding in RA patients. Studies in animal models of arthritis have shown that influencing the sympathetic (via α- and ß-adrenergic receptors) and the parasympathetic (via the nicotinic acetylcholine receptor α7nAChR or by electrically stimulating the vagus nerve) nervous system can have a beneficial effect on inflammation markers and arthritis. The immunosuppressive effect of the parasympathetic nervous system appears less ambiguous than the immunomodulatory effect of the sympathetic nervous system, where activation can lead to increased or decreased inflammation depending on timing, doses and kind of adrenergic agent used. In this review we will discuss the current knowledge of the role of both the sympathetic (SNS) and parasympathetic nervous system (PNS) in inflammation with a special focus on the role in RA. In addition, potential antirheumatic strategies that could be developed by targeting these autonomic pathways are discussed.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Parasympathetic Nervous System/physiology , Sympathetic Nervous System/physiology , Animals , Humans , Inflammation/physiopathology , Models, Biological , Receptors, Adrenergic, beta/physiology , Receptors, Nicotinic/physiology , alpha7 Nicotinic Acetylcholine Receptor
11.
Ann Rheum Dis ; 70(8): 1357-62, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21628308

ABSTRACT

The development of novel treatments for rheumatoid arthritis (RA) requires the interplay between clinical observations and studies in animal models. Given the complex molecular pathogenesis and highly heterogeneous clinical picture of RA, there is an urgent need to dissect its multifactorial nature and to propose new strategies for preventive, early and curative treatments. Research on animal models has generated new knowledge on RA pathophysiology and aetiology and has provided highly successful paradigms for innovative drug development. Recent focus has shifted towards the discovery of novel biomarkers, with emphasis on presymptomatic and emerging stages of human RA, and towards addressing the pathophysiological mechanisms and subsequent efficacy of interventions that underlie different disease variants. Shifts in the current paradigms underlying RA pathogenesis have also led to increased demand for new (including humanised) animal models. There is therefore an urgent need to integrate the knowledge on human and animal models with the ultimate goal of creating a comprehensive 'pathogenesis map' that will guide alignment of existing and new animal models to the subset of disease they mimic. This requires full and standardised characterisation of all models at the genotypic, phenotypic and biomarker level, exploiting recent technological developments in 'omics' profiling and computational biology as well as state of the art bioimaging. Efficient integration and dissemination of information and resources as well as outreach to the public will be necessary to manage the plethora of data accumulated and to increase community awareness and support for innovative animal model research in rheumatology.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Animals , Antirheumatic Agents/therapeutic use , Arthritis, Experimental/physiopathology , Arthritis, Experimental/prevention & control , Arthritis, Rheumatoid/physiopathology , Arthritis, Rheumatoid/prevention & control , Drug Discovery/methods , Therapies, Investigational/methods
12.
Am J Pathol ; 177(6): 3010-24, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20971740

ABSTRACT

Changes in the expression and activation status of Ras proteins are thought to contribute to the pathological phenotype of stromal fibroblast-like synoviocytes (FLS) in rheumatoid arthritis, a prototypical immune-mediated inflammatory disease. Broad inhibition of Ras and related proteins has shown protective effects in animal models of arthritis, but each of the Ras family homologues (ie, H-, K-, and N-Ras) makes distinct contributions to cellular activation. We examined the expression of each Ras protein in synovial tissue and FLS obtained from patients with rheumatoid arthritis and other forms of inflammatory arthritis. Each Ras protein was expressed in synovial tissue and cultured FLS. Each homolog was also activated following FLS stimulation with tumor necrosis factor-α or interleukin (IL)-1ß. Constitutively active mutants of each Ras protein enhanced IL-1ß-induced FLS matrix metalloproteinase-3 production, while only active H-Ras enhanced IL-8 production. Gene silencing demonstrated that each Ras protein contributed to IL-1ß-dependent IL-6 production, while H-Ras and N-Ras supported IL-1ß-dependent matrix metalloproteinase-3 and IL-8 production, respectively. The overlap in contributions of Ras homologues to FLS activation suggests that broad targeting of Ras GTPases in vivo suppresses global inflammation and joint destruction in arthritis. Consistent with this, simultaneous silencing of H-Ras, K-Ras, and N-Ras expression significantly reduces inflammation and joint destruction in murine collagen-induced arthritis, while specific targeting of N-Ras alone is less effective in providing clinical benefits.


Subject(s)
Arthritis, Experimental/genetics , Genes, ras/genetics , Inflammation/genetics , Joints/pathology , RNA Interference/physiology , Adult , Aged , Animals , Arthritis, Experimental/pathology , Cells, Cultured , Cohort Studies , Female , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gene Expression/physiology , Genes, ras/physiology , Humans , Inflammation/pathology , Jurkat Cells , Male , Mice , Mice, Inbred C57BL , Middle Aged , Multigene Family , Sequence Homology
13.
Arthritis Rheum ; 62(3): 765-70, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20187132

ABSTRACT

OBJECTIVE: RNA interference (RNAi) is a powerful tool for sequence-specific gene silencing, and interest in its application in human diseases is growing. Given the success of recent strategies for administering gene therapy in rheumatoid arthritis using recombinant vectors such as adeno-associated virus type 5 (rAAV5) for optimized intraarticular gene transfer, we undertook the present study to determine the feasibility of using rAAV5-mediated RNAi-based therapy in arthritis. METHODS: We developed rAAV5 vectors expressing short hairpin small interfering RNA (shRNA) against tumor necrosis factor alpha (TNFalpha) under H1 promoter, and carrying the enhanced green fluorescent protein (eGFP) reporter gene under cytomegalovirus promoter (rAAV5-shTNF). TNFalpha gene silencing was validated in vitro with mouse macrophages. Mice with collagen-induced arthritis were injected in the ankle and knee joints, at disease onset, with either rAAV5-shTNF or control rAAV5-eGFP vectors (5 x 10(9) particles). Arthritis severity was assessed clinically and histologically, and immunologic response was examined. Local and systemic transgene expression was monitored using quantitative reverse transcriptase-polymerase chain reaction, immunohistochemical analysis, and enzyme-linked immunosorbent assay. RESULTS: After a single injection of rAAV5-shTNF into inflamed joints, local TNFalpha gene silencing provided rapid and long-term suppression of arthritis progression and reduced joint damage compared with that observed in control groups. Treatment with rAAV5-shTNF was associated with decreased proliferation and interferon-gamma production by antigen-stimulated T cells from draining lymph nodes, and the potency of this treatment was similar to that observed with other treatment strategies targeting TNFalpha at the protein level, either locally or systemically. CONCLUSION: Our data present the first proof-of-concept for the application of rAAV5-mediated RNAi-based gene therapy for local blockade of inflammation in experimental arthritis.


Subject(s)
Arthritis, Experimental/therapy , RNA Interference/physiology , RNA, Small Interfering/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Animals , Dependovirus , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Gene Silencing/physiology , Genetic Vectors , Green Fluorescent Proteins/genetics , Immunohistochemistry , Injections, Intra-Articular , Mice , Tumor Necrosis Factor-alpha/metabolism
14.
Ann Rheum Dis ; 69(9): 1717-23, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20511609

ABSTRACT

BACKGROUND: The alpha7 subunit of nicotinic acetylcholine receptors (alpha7nAChR) can negatively regulate the synthesis and release of proinflammatory cytokines by macrophages and fibroblast-like synoviocytes in vitro. In addition, stimulation of the alpha7nAChR can reduce the severity of arthritis in murine collagen-induced arthritis (CIA). OBJECTIVE: To provide more insight into the role of the alpha7nAChR in the pathogenesis of arthritis by investigating the effect of the absence of alpha7nAChR in CIA in alpha7-deficient (alpha7nAChR(-/-)) compared with wild-type (WT) mice. METHODS: CIA was induced in alpha7nAChR(-/-) and WT littermate mice at day 0 by immunisation with chicken collagen type II (cCII) followed by a booster injection with cCII on day 20. Mice were killed on day 44 or day 63 and arthritis activity as well as radiological and histological damage were scored. The effects on the immune response were evaluated by measurement of antigen-specific antibodies and cytokines, and evaluation of the effects on antigen-specific stimulated spleen cells. RESULTS: In alpha7nAChR(-/-) mice a significant increase in the incidence and severity of arthritis as well as increased synovial inflammation and joint destruction were seen. Exacerbation of CIA was associated with elevated systemic proinflammatory cytokines and enhanced T-helper cell 1 (Th1)-cytokine and tumour necrosis factor alpha production by spleen cells. Moreover, a specific decrease in the collagen-specific 'Th1-associated' IgG2a response was seen, whereas IgG1 titres were unaffected. CONCLUSIONS: The results presented here indicate that immune cell function in a model of rheumatoid arthritis is regulated by the cholinergic system and, at least in part, mediated by the alpha7nAChR.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Receptors, Nicotinic/physiology , Acute Disease , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/pathology , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/immunology , Bone Diseases, Metabolic/pathology , Cartilage Diseases/etiology , Cartilage Diseases/immunology , Cartilage Diseases/pathology , Chemokine CCL2/blood , Disease Progression , Female , Male , Mice , Mice, Knockout , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/blood , alpha7 Nicotinic Acetylcholine Receptor
15.
Curr Gene Ther ; 9(3): 160-70, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19519361

ABSTRACT

Nuclear factor (NF)-kappaB is regarded as one of the most important transcription factors and plays an essential role in the transcriptional activation of pro-inflammatory cytokines, cell proliferation and survival. NF-kappaB can be activated via two distinct NF-kappaB signal transduction pathways, the so-called canonical and non-canonical pathways, and has been demonstrated to play a key role in a wide range of inflammatory diseases and various types of cancer. Much effort has been put in strategies to inhibit NF-kappaB activation, for example by the development of pharmacological compounds that selectively inhibit NF-kappaB activity and therefore would be beneficial for immunotherapy of transplantation, autoimmune and allergic diseases, as well as an adjuvant approach in patients treated with chemotherapy for cancer. Gene therapy targeting NF-kappaB is a promising new strategy with the potential of long-term effects and has been explored in a wide variety of diseases, ranging from cancer to transplantation medicine and autoimmune diseases. In this review we discuss recent progress made in the development of NF-kappaB targeted gene therapy and the evolution towards clinical application.


Subject(s)
Genetic Therapy/trends , NF-kappa B/genetics , Neoplasms/therapy , Animals , Genetic Therapy/methods , Humans , Inflammation/therapy , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Transcriptional Activation/drug effects
16.
Cells ; 8(7)2019 07 02.
Article in English | MEDLINE | ID: mdl-31269754

ABSTRACT

Inflammatory bowel diseases (IBD) have a complex, multifactorial pathophysiology with an unmet need for effective treatment. This calls for novel strategies to improve disease outcome and quality of life for patients. Increasing evidence suggests that autonomic nerves and neurotransmitters, as well as neuropeptides, modulate the intestinal immune system, and thereby regulate the intestinal inflammatory processes. Although the autonomic nervous system is classically divided in a sympathetic and parasympathetic branch, both play a pivotal role in the crosstalk with the immune system, with the enteric nervous system acting as a potential interface. Pilot clinical trials that employ vagus nerve stimulation to reduce inflammation are met with promising results. In this paper, we review current knowledge on the innervation of the gut, the potential of cholinergic and adrenergic systems to modulate intestinal immunity, and comment on ongoing developments in clinical trials.


Subject(s)
Enteric Nervous System/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/innervation , Neuroimmunomodulation/immunology , Vagus Nerve Stimulation , Clinical Trials as Topic , Colon/immunology , Colon/innervation , Humans , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/immunology , Treatment Outcome
17.
Hum Gene Ther ; 28(2): 168-178, 2017 02.
Article in English | MEDLINE | ID: mdl-27806641

ABSTRACT

Gene therapy has potential to treat rheumatic diseases; however, the presence of macrophages in the joint might hamper adeno-associated viral vector-mediated gene delivery. Here we demonstrate that in arthritic, but also in healthy, mice administration of agents that influence macrophage activity/number and/or addition of empty decoy capsids substantially improve the efficacy of recombinant adeno-associated viral vector 5 transgene expression in the joint. Pretreatment with triamcinolone or clodronate liposomes improved luciferase expression over a period of 4 weeks. Similar results were seen when empty decoy capsids were added to full genome containing capsids in a 5:1 ratio. In a study to assess the duration of expression as well as to investigate the combination of these two approaches, we observed a synergistic enhancement of gene expression, sustained for at least 12 weeks. The enhancement of gene expression was independent of the route of administration of triamcinolone (intra-articular or intramuscular). In healthy mice it was demonstrated that the combination improved expression of the transgene significantly, in a serotype independent manner. These data have implications for future applications of gene therapy to the joint and for other tissues with an abundance of macrophages.


Subject(s)
Arthritis/therapy , Capsid/metabolism , Dependovirus/genetics , Genetic Vectors/administration & dosage , Joints/metabolism , Macrophages/metabolism , Transgenes/physiology , Animals , Arthritis/genetics , Joints/pathology , Macrophages/cytology , Mice
18.
Hum Gene Ther ; 17(8): 821-32, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16942442

ABSTRACT

Nuclear factor (NF)-kappaB is highly activated in the synovium of rheumatoid arthritis (RA) patients, and can induce transcription of many proinflammatory molecules. Phosphorylation of inhibitor of kappaB (IkappaB) proteins is an important step in NF-kappaB activation and under inflammatory conditions is regulated predominantly by IkappaB kinase (IKK)beta. Consequently, specific targeting of IKK beta in the joint, using gene therapy, presents a sophisticated treatment option for arthritis. In the present study we investigated the effect of inhibiting IKK beta in adjuvant arthritis (AA) in rats, using recombinant adeno-associated virus (rAAV)-mediated intraarticular gene therapy. For this purpose rAAV5 carrying the dominant negative IKK beta gene (AAV5.IKK beta dn) or control AAV5.eGFP was injected into the right ankle joint. Rats treated with AAV5.IKK beta dn in early arthritis exhibited significantly reduced paw swelling (p < 0.05). Immunohistochemical analysis of synovial tissue revealed reduced levels of interleukin (IL)-6 (p = 0.005) and tumor necrosis factor-alpha (TNF-alpha) (p = 0.03), whereas IL-10 levels were not affected. No significant effect was found on cartilage and bone destruction, or on matrix metalloproteinase-3 and tissue inhibitor of matrix metalloproteinase-1 expression. Injection of AAV5.IKK beta dn in the preclinical phase showed only a marginal effect on arthritis. Importantly, in this study we also demonstrate for the first time that our vector is capable of transducing human RA whole synovial tissue biopsies ex vivo, resulting in reduced IL-6 production after TNF-alpha stimulation (p = 0.03). In conclusion, we are the first to demonstrate that rAAV5 can be used to successfully deliver a therapeutic gene (IKK beta dn) to the synovium, resulting in reduced severity of inflammation in AA in vivo and proinflammatory cytokine production in human RA synovial tissue ex vivo. This translational research represents a crucial next step toward the development of gene therapy for application in humans.


Subject(s)
Arthritis, Rheumatoid/therapy , Dependovirus , Genes, Dominant , Genetic Therapy , I-kappa B Kinase/genetics , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cytokines/genetics , Cytokines/metabolism , Genetic Therapy/methods , Humans , I-kappa B Kinase/metabolism , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Rats , Rats, Inbred Lew , Synovial Membrane/metabolism , Synovial Membrane/pathology
20.
J Rheumatol ; 42(9): 1672-6, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26178282

ABSTRACT

OBJECTIVE: To investigate responsiveness, discrimination, and construct validity of a composite change index (CCI) for the assessment of single-joint involvement in inflammatory arthritis. METHODS: Evaluation of standardized response means (SRM), Guyatt effect size, and Spearman rank correlation coefficient in a randomized controlled trial investigating the effect of an intraarticular etanercept injection. RESULTS: The CCI showed a high SRM (1.68) and high Guyatt effect size (2.72). Both visual analog scale of pain and functionality had a moderate Guyatt effect size (2.06, 2.44) and high SRM (0.81, 0.97). CONCLUSION: This study supports the use of the CCI as a single-joint assessment after single-joint intervention. CLINICAL TRIAL REGISTRATION: NTR-1210.


Subject(s)
Antirheumatic Agents/administration & dosage , Arthritis, Psoriatic/drug therapy , Arthritis, Rheumatoid/drug therapy , Etanercept/administration & dosage , Knee Joint/pathology , Antirheumatic Agents/therapeutic use , Arthritis, Psoriatic/pathology , Arthritis, Rheumatoid/pathology , Disability Evaluation , Double-Blind Method , Etanercept/therapeutic use , Humans , Injections, Intra-Articular , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL