Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Autoimmun ; 146: 103219, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38696927

ABSTRACT

Tissue repair is disturbed in fibrotic diseases like systemic sclerosis (SSc), where the deposition of large amounts of extracellular matrix components such as collagen interferes with organ function. LAIR-1 is an inhibitory collagen receptor highly expressed on tissue immune cells. We questioned whether in SSc, impaired LAIR-1-collagen interaction is contributing to the ongoing inflammation and fibrosis. We found that SSc patients do not have an intrinsic defect in LAIR-1 expression or function. Instead, fibroblasts from healthy controls and SSc patients stimulated by soluble factors that drive inflammation and fibrosis in SSc deposit disorganized collagen products in vitro, which are dysfunctional LAIR-1 ligands. This is dependent of matrix metalloproteinases and platelet-derived growth factor receptor signaling. In support of a non-redundant role of LAIR-1 in the control of fibrosis, we found that LAIR-1-deficient mice have increased skin fibrosis in response to repeated injury and in the bleomycin mouse model for SSc. Thus, LAIR-1 represents an essential control mechanism for tissue repair. In fibrotic disease, excessive collagen degradation may lead to a disturbed feedback loop. The presence of functional LAIR-1 in patients provides a therapeutic opportunity to reactivate this intrinsic negative feedback mechanism in fibrotic diseases.

2.
J Clin Invest ; 131(5)2021 03 01.
Article in English | MEDLINE | ID: mdl-33645549

ABSTRACT

IgE induced by type 2 immune responses in atopic dermatitis is implicated in the progression of atopic dermatitis to other allergic diseases, including food allergies, allergic rhinitis, and asthma. However, the keratinocyte-derived signals that promote IgE and ensuing allergic diseases remain unclear. Herein, in a mouse model of atopic dermatitis-like skin inflammation induced by epicutaneous Staphylococcus aureus exposure, keratinocyte release of IL­36α along with IL-4 triggered B cell IgE class-switching, plasma cell differentiation, and increased serum IgE levels-all of which were abrogated in IL-36R-deficient mice or anti-IL­36R-blocking antibody-treated mice. Moreover, skin allergen sensitization during S. aureus epicutaneous exposure-induced IL-36 responses was required for the development of allergen-specific lung inflammation. In translating these findings, elevated IL­36 cytokines in human atopic dermatitis skin and in IL­36 receptor antagonist-deficiency patients coincided with increased serum IgE levels. Collectively, keratinocyte-initiated IL­36 responses represent a key mechanism and potential therapeutic target against allergic diseases.


Subject(s)
Dermatitis, Atopic/immunology , Immunoglobulin E/immunology , Interleukin-1/immunology , Keratinocytes/immunology , Plasma Cells/immunology , Staphylococcus aureus/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Dermatitis, Atopic/genetics , Dermatitis, Atopic/microbiology , Humans , Immunoglobulin Class Switching , Immunoglobulin E/genetics , Interleukin-1/genetics , Interleukin-4/genetics , Interleukin-4/immunology , Keratinocytes/microbiology , Mice , Mice, Knockout , Plasma Cells/pathology
3.
Commun Biol ; 4(1): 172, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33558616

ABSTRACT

IL-36, which belongs to the IL-1 superfamily, is increasingly linked to neutrophilic inflammation. Here, we combined in vivo and in vitro approaches using primary mouse and human cells, as well as, acute and chronic mouse models of lung inflammation to provide mechanistic insight into the intercellular signaling pathways and mechanisms through which IL-36 promotes lung inflammation. IL-36 receptor deficient mice exposed to cigarette smoke or cigarette smoke and H1N1 influenza virus had attenuated lung inflammation compared with wild-type controls. We identified neutrophils as a source of IL-36 and show that IL-36 is a key upstream amplifier of lung inflammation by promoting activation of neutrophils, macrophages and fibroblasts through cooperation with GM-CSF and the viral mimic poly(I:C). Our data implicate IL-36, independent of other IL-1 family members, as a key upstream amplifier of neutrophilic lung inflammation, providing a rationale for targeting IL-36 to improve treatment of a variety of neutrophilic lung diseases.


Subject(s)
Interleukin-1/metabolism , Lung/metabolism , Neutrophil Activation , Neutrophils/metabolism , Orthomyxoviridae Infections/metabolism , Pneumonia, Viral/metabolism , Receptors, Interleukin-1/metabolism , Animals , Cells, Cultured , Cigarette Smoking , Disease Models, Animal , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Interleukin-1/genetics , Lung/immunology , Lung/virology , Macrophage Activation , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/virology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Pneumonia, Viral/genetics , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Receptors, Interleukin-1/genetics , Signal Transduction
4.
Br J Clin Pharmacol ; 87(4): 1824-1838, 2021 04.
Article in English | MEDLINE | ID: mdl-32986868

ABSTRACT

AIMS: To evaluate the safety, pharmacokinetics and pharmacodynamics of single- and multiple-rising doses (MRDs) of BI 705564 and establish proof of mechanism. METHODS: BI 705564 was studied in 2 placebo-controlled, Phase I clinical trials testing single-rising doses (1-160 mg) and MRDs (1-80 mg) of BI 705564 over 14 days in healthy male volunteers. Blood samples were analysed for BI 705564 plasma concentration, Bruton's tyrosine kinase (BTK) target occupancy (TO) and CD69 expression in B cells stimulated ex vivo. A substudy was conducted in allergic, otherwise healthy, MRD participants. Safety was assessed in both studies. RESULTS: All doses of BI 705564 were well tolerated. Geometric mean BI 705564 plasma terminal half-life ranged from 10.1 to 16.9 hours across tested doses, with no relevant accumulation after multiple dosing. Doses ≥20 mg resulted in ≥85% average TO that was maintained for ≥48 hours after single-dose administration. Functional effects of BTK signalling were demonstrated by dose-dependent inhibition of CD69 expression. In allergic participants, BI 705564 treatment showed a trend in wheal size reduction in a skin prick test and complete inhibition of basophil activation. Mild bleeding-related adverse events were observed with BI 705564; bleeding time increased in 1/12 participants (8.3%) who received placebo vs 26/48 (54.2%) treated with BI 705564. CONCLUSION: BI 705564 showed efficient target engagement through durable TO and inhibition of ex vivo B-cell activation, and proof of mechanism through effects on allergic skin responses. Mild bleeding-related adverse events were probably related to inhibition of platelet aggregation by BTK inhibition.


Subject(s)
B-Lymphocytes , Platelet Aggregation , Agammaglobulinaemia Tyrosine Kinase , Healthy Volunteers , Humans , Male , Protein Kinase Inhibitors/adverse effects , Signal Transduction
5.
Clin Immunol ; 223: 108640, 2021 02.
Article in English | MEDLINE | ID: mdl-33296718

ABSTRACT

Lupus nephritis (LN) is a serious end organ complication of systemic lupus erythematosus. Nephrotoxic serum nephritis (NTN) is an inducible model of LN, which utilizes passive transfer of pre-formed nephrotoxic antibodies to initiate disease. In previous studies, we demonstrated that the Bruton's tyrosine kinase inhibitor, BI-BTK-1, prevents the development of nephritis in NTN when treatment was started prior to nephrotoxic serum transfer, and reverses established proteinuria as well. We manipulated the initiation and duration of BI-BTK-1 therapy in NTN to study its delayed therapeutic effects when treatment is given later in the disease course, as well as to further understand what effect BI-BTK-1 is having to prevent initiation of nephritis with early treatment. Early treatment and remission induction each correlated with decreased inflammatory macrophages, CD4+ and CD8+ T cells, and decreased B220+ B cells. Additionally, an increased proportion of resident macrophages within the CD45+ population favored a delay of disease onset and remission induction. We also studied the cellular processes involved in reactivation of nephritis by withdrawing BI-BTK-1 treatment at different time points. Treatment cessation led to either early or later onset of renal flares inversely dependent on the initial duration of BTK inhibition, as assessed by increased proteinuria and BUN levels and worse renal pathology. These flares were associated with an increase in kidney CD45+ infiltrates, including myeloid cell populations. IL-6, CD14, and CCL2 were also increased in mice developing late flares. These analyses point to the role of macrophages as an important contributor to the pathogenesis of immune mediated nephritis, and further support the therapeutic potential of BTK inhibition in this disease and related conditions.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Kidney/pathology , Lupus Nephritis/drug therapy , Macrophages/immunology , Protein Kinase Inhibitors/therapeutic use , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Animals , Disease Models, Animal , Humans , Leukocyte Common Antigens/metabolism , Mice , Mice, 129 Strain , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Proteinuria
6.
PLoS One ; 15(11): e0242863, 2020.
Article in English | MEDLINE | ID: mdl-33253326

ABSTRACT

Pathophysiology of systemic sclerosis (SSc, Scleroderma), an autoimmune rheumatic disease, comprises of mechanisms that drive vasculopathy, inflammation and fibrosis. Understanding of the disease and associated clinical heterogeneity has advanced considerably in the past decade, highlighting the necessity of more specific targeted therapy. While many of the recent trials in SSc failed to meet the primary end points that predominantly relied on changes in modified Rodnan skin scores (MRSS), sub-group analysis, especially those focused on the basal skin transcriptomic data have provided insights into patient subsets that respond to therapies. These findings suggest that deeper understanding of the molecular changes in pathways is very important to define disease drivers in various patient subgroups. In view of these challenges, we performed meta-analysis on 9 public available SSc microarray studies using a novel pathway pivoted approach combining consensus clustering and machine learning assisted feature selection. Selected pathway modules were further explored through cluster specific topological network analysis in search of novel therapeutic concepts. In addition, we went beyond previously described SSc class divisions of 3 clusters (e.g. inflammation, fibro-proliferative, normal-like) and expanded into a much finer stratification in order to profile SSc patients more accurately. Our analysis unveiled an important 80 pathway signatures that differentiated SSc patients into 8 unique subtypes. The 5 pathway modules derived from such signature successfully defined the 8 SSc subsets and were validated by in-silico cellular deconvolution analysis. Myeloid cells and fibroblasts involvement in different clusters were confirmed and linked to corresponding pathway activities. Collectively, our findings revealed more complex disease subtypes in SSc; Key gene mediators such as IL6, FGFR1, TLR7, PLCG2, IRK2 identified by network analysis underscored the scientific rationale for exploring additional targets in treatment of SSc.


Subject(s)
Scleroderma, Systemic/genetics , Transcription, Genetic , Transcriptome/genetics , Computational Biology , Fibrosis/genetics , Fibrosis/pathology , Humans , Interleukin-6/genetics , Machine Learning , Phospholipase C gamma/genetics , Potassium Channels, Inwardly Rectifying/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/genetics , Skin/metabolism , Skin/pathology , Toll-Like Receptor 7/genetics
7.
Autoimmun Rev ; 19(11): 102668, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32942031

ABSTRACT

Lupus nephritis (LN) is a significant complication of systemic lupus erythematosus (SLE), increasing its morbidity and mortality. Although the current standard of care helps suppress disease activity, it is associated with toxicity and ultimately does not cure SLE. At present, there are no therapies specifically indicated for the treatment of LN and there is an unmet need in this disease where treatment remains a challenge. The CD40-CD40L pathway is central to SLE pathogenesis and the generation of autoantibodies and their deposition in the kidneys, resulting in renal injury in patients with LN. CD40 is expressed on immune cells (including B cells, monocytes and dendritic cells) and also non-haematopoietic cells. Interactions between CD40L on T cells and CD40 on B cells in the renal interstitium are critical for the local expansion of naive B cells and autoantibody-producing B cells in LN. CD40L-mediated activation of myeloid cells and resident kidney cells, including endothelial cells, proximal tubular epithelial cells, podocytes and mesangial cells, further amplifies the inflammatory milieu in the interstitium and the glomeruli. Several studies have highlighted the upregulated expression of CD40 in LN kidney biopsies, and preclinical data have demonstrated the importance of the CD40-CD40L pathway in murine SLE and LN. Blocking this pathway is expected to ameliorate inflammation driven by infiltrating immune cells and resident kidney cells. Initial experimental therapeutic interventions targeting the CD40-CD40L pathway, based on CD40L antibodies, were associated with an increased incidence of thrombosis. However, this safety issue has not been observed with second-generation CD40/CD40L antibodies that have been engineered to prevent platelet activation. With these advancements, together with recent preclinical and clinical findings, it is anticipated that selective blockade of the CD40-CD40L pathway may address the unmet treatment needs in SLE, LN and other autoimmune diseases.


Subject(s)
CD40 Antigens/immunology , CD40 Ligand/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , Animals , Humans , Kidney/immunology , Kidney/physiopathology
8.
Nat Rev Rheumatol ; 15(12): 705-730, 2019 12.
Article in English | MEDLINE | ID: mdl-31712723

ABSTRACT

Fibrosis is defined as an excessive deposition of connective tissue components and can affect virtually every organ system, including the skin, lungs, liver and kidney. Fibrotic tissue remodelling often leads to organ malfunction and is commonly associated with high morbidity and mortality. The medical need for effective antifibrotic therapies is thus very high. However, the extraordinarily high costs of drug development and the rare incidence of many fibrotic disorders hinder the development of targeted therapies for individual fibrotic diseases. A potential strategy to overcome this challenge is to target common mechanisms and core pathways that are of central pathophysiological relevance across different fibrotic diseases. The factors influencing susceptibility to and initiation of these diseases are often distinct, with disease-specific and organ-specific risk factors, triggers and sites of first injury. Fibrotic remodelling programmes with shared fibrotic signalling responses such as transforming growth factor-ß (TGFß), platelet-derived growth factor (PDGF), WNT and hedgehog signalling drive disease progression in later stages of fibrotic diseases. The convergence towards shared responses has consequences for drug development as it might enable the development of general antifibrotic compounds that are effective across different disease entities and organs. Technological advances, including new models, single-cell technologies and gene editing, could provide new insights into the pathogenesis of fibrotic diseases and the development of drugs for their treatment.


Subject(s)
Connective Tissue Diseases , Disease Management , Extracellular Matrix Proteins/metabolism , Genetic Predisposition to Disease , Immunity, Cellular , Immunologic Factors/therapeutic use , Animals , Connective Tissue Diseases/drug therapy , Connective Tissue Diseases/etiology , Connective Tissue Diseases/metabolism , Fibrosis , Humans
9.
Ann Rheum Dis ; 78(6): 754-760, 2019 06.
Article in English | MEDLINE | ID: mdl-30902820

ABSTRACT

OBJECTIVE: To evaluate the safety, efficacy and therapeutic mechanism of BI 655064, an antagonistic anti-CD40 monoclonal antibody, in patients with rheumatoid arthritis (RA) and an inadequate response to methotrexate (MTX-IR). METHODS: In total, 67 patients were randomised to receive weekly subcutaneous doses of 120 mg BI 655064 (n=44) or placebo (n=23) for 12 weeks. The primary endpoint was the proportion of patients who achieved 20% improvement in American College of Rheumatology criteria (ACR20) at week 12. Safety was assessed in patients who received at least one dose of study drug. RESULTS: At week 12, the primary endpoint was not met, with 68.2% of patients treated with BI 655064 achieving an ACR20 vs 45.5% with placebo (p=0.064); using Bayesian analysis, the posterior probability of seeing a difference greater than 35% was 42.9%. BI 655064 was associated with greater changes in CD40-CD40L pathway-related markers, including reductions in inflammatory and bone resorption markers (interleukin-6, matrix metalloproteinase-3, receptor activator of nuclear factor-κB ligand), concentration of autoantibodies (immunoglobulin [Ig]G rheumatoid factor [RF], IgM RF, IgA RF) and CD95+ activated B-cell subsets. No serious adverse events (AEs) related to BI 655064 treatment or thromboembolic events occurred; reported AEs were mainly of mild intensity. CONCLUSION: Although blockade of the CD40-CD40L pathway with BI 655064 in MTX-IR patients with RA resulted in marked changes in clinical and biological parameters, including reductions in activated B-cells, autoantibody production and inflammatory and bone resorption markers, with a favourable safety profile, clinical efficacy was not demonstrated in this small phase IIa study. TRIAL REGISTRATION NUMBER: NCT01751776.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/physiopathology , Autoantibodies/blood , B-Lymphocyte Subsets/drug effects , Biomarkers/blood , Bone Remodeling/drug effects , CD40 Ligand/antagonists & inhibitors , Double-Blind Method , Female , Humans , Inflammation Mediators/metabolism , Injections, Subcutaneous , Male , Methotrexate/therapeutic use , Middle Aged , Severity of Illness Index , Young Adult
10.
Clin Immunol ; 197: 205-218, 2018 12.
Article in English | MEDLINE | ID: mdl-30339790

ABSTRACT

Lupus nephritis is a common disease manifestation of SLE, in which immune complex deposition and macrophage activation are important contributors to disease pathogenesis. Bruton's tyrosine kinase (BTK) plays an important role in both B cell and FcgammaR mediated myeloid cell activation. In the current study, we examined the efficacy of BI-BTK-1, a recently described irreversible BTK inhibitor, in the classical NZB × NZW F1 (NZB/W) and MRL/lpr spontaneous mouse models of SLE. NZB/W mice were randomly assigned to a treatment (0.3 mg/kg, 1 mg/kg, 3 mg/kg and 10 mg/kg) or control group and began treatment at 22 weeks of age. The experimental setup was similar in MRL/lpr mice, but with a single treated (10 mg/kg, beginning at 8-9 weeks of age) and control group. A separate experiment was performed in the MRL/lpr strain to assess the ability of BI-BTK-1 to reverse established kidney disease. Early treatment with BI-BTK-1 significantly protected NZB/W and MRL/lpr mice from the development of proteinuria, correlating with significant renal histological protection, decreased anti-DNA titers, and increased survival in both strains. BI-BTK-1 treated mice displayed a significant decrease in nephritis-associated inflammatory mediators (e.g. LCN2 and IL-6) in the kidney, combined with a significant inhibition of immune cell infiltration and accumulation. Importantly, BI-BTK-1 treatment resulted in the reversal of established kidney disease. BTK inhibition significantly reduced total B cell numbers and all B cell subsets (immature, transitional, follicular, marginal zone, and class switched) in the spleen of NZB/W mice. Overall, the significant efficacy of BI-BTK-1 in ameliorating multiple pathological endpoints associated with kidney disease in two distinct murine models of spontaneous lupus nephritis provides a strong rationale for BTK inhibition as a promising treatment approach for lupus nephritis.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Kidney/drug effects , Lupus Nephritis/pathology , Protein Kinase Inhibitors/pharmacology , Animals , Antibodies, Antinuclear/drug effects , Antibodies, Antinuclear/immunology , B-Lymphocyte Subsets/drug effects , B-Lymphocyte Subsets/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , DNA/immunology , Disease Models, Animal , Interleukin-6/immunology , Interleukin-6/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Lipocalin-2/drug effects , Lipocalin-2/immunology , Lipocalin-2/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/immunology , Mice , Mice, Inbred MRL lpr , Mice, Inbred NZB , Proteinuria/immunology , Random Allocation , Spleen/cytology , Spleen/drug effects
11.
Arthritis Res Ther ; 20(1): 10, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29370834

ABSTRACT

BACKGROUND: Systemic lupus erythematosus (SLE) is a systemic autoimmune disease that affects different end organs, including skin and brain. We and others have previously shown the importance of macrophages in the pathogenesis of cutaneous and neuropsychiatric lupus. Additionally, autoantibodies produced by autoreactive B cells are thought to play a role in both the skin and central nervous system pathologies associated with SLE. METHODS: We used a novel inhibitor of Bruton's tyrosine kinase (BTK), BI-BTK-1, to target both macrophage and B cell function in the MRL-lpr/lpr murine model of SLE, and examined the effect of treatment on skin and brain disease. RESULTS: We found that treatment with BI-BTK-1 significantly attenuated the lupus associated cutaneous and neuropsychiatric disease phenotypes in MRL/lpr mice. Specifically, BI-BTK-1 treated mice had fewer macroscopic and microscopic skin lesions, reduced cutaneous cellular infiltration, and diminished inflammatory cytokine expression compared to control mice. BTK inhibition also significantly improved cognitive function, and decreased accumulation of T cells, B cells, and macrophages within the central nervous system, specifically the choroid plexus. CONCLUSIONS: Directed therapies may improve the response rate in lupus-driven target organ involvement, and decrease the dangerous side effects associated with global immunosuppression. Overall, our results suggest that inhibition of BTK may be a promising therapeutic option for cutaneous and neuropsychiatric disease associated with SLE.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Brain Diseases/prevention & control , Enzyme Inhibitors/pharmacology , Lupus Erythematosus, Systemic/complications , Skin Diseases/prevention & control , Agammaglobulinaemia Tyrosine Kinase/immunology , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Brain Diseases/etiology , Brain Diseases/immunology , Cognition/drug effects , Cognition/physiology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression/drug effects , Humans , Lupus Erythematosus, Systemic/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred MRL lpr , Skin Diseases/etiology , Skin Diseases/immunology
12.
Immunity ; 47(1): 118-134.e8, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28709802

ABSTRACT

Secretory immunoglobulin A (SIgA) enhances host-microbiota symbiosis, whereas SIgM remains poorly understood. We found that gut IgM+ plasma cells (PCs) were more abundant in humans than mice and clonally related to a large repertoire of memory IgM+ B cells disseminated throughout the intestine but rare in systemic lymphoid organs. In addition to sharing a gut-specific gene signature with memory IgA+ B cells, memory IgM+ B cells were related to some IgA+ clonotypes and switched to IgA in response to T cell-independent or T cell-dependent signals. These signals induced abundant IgM which, together with SIgM from clonally affiliated PCs, recognized mucus-embedded commensals. Bacteria recognized by human SIgM were dually coated by SIgA and showed increased richness and diversity compared to IgA-only-coated or uncoated bacteria. Thus, SIgM may emerge from pre-existing memory rather than newly activated naive IgM+ B cells and could help SIgA to anchor highly diverse commensal communities to mucus.


Subject(s)
Angiodysplasia/immunology , B-Lymphocytes/immunology , Colonic Neoplasms/immunology , Colonic Polyps/immunology , Immunoglobulin M/metabolism , Intestines/immunology , Plasma Cells/immunology , Adult , Aged , Aged, 80 and over , Animals , Clone Cells , Female , Gastrointestinal Microbiome/immunology , Humans , Immunity, Mucosal , Immunoglobulin A/metabolism , Immunoglobulin Class Switching , Immunologic Memory , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Symbiosis
13.
Sci Rep ; 6: 26164, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27192942

ABSTRACT

Lupus nephritis (LN) is a potentially dangerous end organ pathology that affects upwards of 60% of lupus patients. Bruton's tyrosine kinase (BTK) is important for B cell development, Fc receptor signaling, and macrophage polarization. In this study, we investigated the effects of a novel, highly selective and potent BTK inhibitor, BI-BTK-1, in an inducible model of LN in which mice receive nephrotoxic serum (NTS) containing anti-glomerular antibodies. Mice were treated once daily with vehicle alone or BI-BTK-1, either prophylactically or therapeutically. When compared with control treated mice, NTS-challenged mice treated prophylactically with BI-BTK-1 exhibited significantly attenuated kidney disease, which was dose dependent. BI-BTK-1 treatment resulted in decreased infiltrating IBA-1+ cells, as well as C3 deposition within the kidney. RT-PCR on whole kidney RNA and serum profiling indicated that BTK inhibition significantly decreased levels of LN-relevant inflammatory cytokines and chemokines. Renal RNA expression profiling by RNA-seq revealed that BI-BTK-1 dramatically modulated pathways related to inflammation and glomerular injury. Importantly, when administered therapeutically, BI-BTK-1 reversed established proteinuria and improved renal histopathology. Our results highlight the important role for BTK in the pathogenesis of immune complex-mediated nephritis, and BTK inhibition as a promising therapeutic target for LN.


Subject(s)
Antigen-Antibody Complex/metabolism , Enzyme Inhibitors/administration & dosage , Lupus Nephritis/drug therapy , Lupus Nephritis/prevention & control , Protein-Tyrosine Kinases/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase , Animals , Blood Chemical Analysis , Complement C3/analysis , Cytokines/analysis , Disease Models, Animal , Gene Expression Profiling , Kidney/pathology , Lupus Nephritis/chemically induced , Lupus Nephritis/pathology , Mice , Sequence Analysis, RNA , Treatment Outcome
14.
Discov Med ; 20(108): 43-9, 2015.
Article in English | MEDLINE | ID: mdl-26321086

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease which results in multiple different end organ pathologies, including the kidney. Lupus nephritis (LN) is one of the most serious complications of SLE, and a leading cause of morbidity and mortality. Current treatment options are suboptimal, involving non-specific immunosuppression which exposes patients to potentially serious side effects with no guarantee of remission. More targeted therapeutic approaches may improve treatment results. Many studies have implicated macrophages as actively contributing to LN pathogenesis in both human and murine disease. Indeed, various studies have shown that depletion of macrophage populations, inhibition of macrophage recruitment, and disruption of inflammatory macrophage activation and polarization have significantly ameliorated nephritis in several different murine LN models. The current literature explores targeting macrophages by several different means, including the CSF-1/CSF-1R signaling axis, the CX3CL1/CX3CR1 signaling axis, the CCL2/CCR2 signaling axis, and Bruton's Tyrosine Kinase (BTK), all of which hold promise as targets for future LN treatments. These studies highlight the potential benefit of targeting macrophages in LN, and emphasize the need for future investigations to discern the ideal mean(s) for targeting macrophages in LN.


Subject(s)
Lupus Nephritis/immunology , Lupus Nephritis/therapy , Macrophages/immunology , Animals , Chemokines/immunology , Disease Models, Animal , Humans , Lupus Nephritis/pathology , Macrophages/pathology , Mice , Receptors, Chemokine/immunology , Signal Transduction/immunology
15.
Oxid Med Cell Longev ; 2013: 853210, 2013.
Article in English | MEDLINE | ID: mdl-23819015

ABSTRACT

Neutrophil infiltration plays a major role in the pathogenesis of myocardial injury. Oxidative injury is suggested to be a central mechanism of the cellular damage after acute myocardial infarction. This study is pertained to the prognostic role of a tetrapeptide derivative PEP1261 (BOC-Lys(BOC)-Arg-Asp-Ser(tBu)-OtBU), a peptide sequence (39-42) of lactoferrin, studied in the modulation of neutrophil functions in vitro by measuring the reactive oxygen species (ROS) generation, lysosomal enzymes release, and enhanced expression of C proteins. The groundwork experimentation was concerned with the isolation of neutrophils from the normal and acute myocardial infarct rats to find out the efficacy of PEP1261 in the presence of a powerful neutrophil stimulant, phorbol 12-myristate 13 acetate (PMA). Stimulation of neutrophils with PMA resulted in an oxidative burst of superoxide anion and enhanced release of lysosomal enzymes and expression of complement proteins. The present study further demonstrated that the free radicals increase the complement factors in the neutrophils confirming the role of ROS. PEP1261 treatment significantly reduced the levels of superoxide anion and inhibited the release of lysosomal enzymes in the stimulated control and infarct rat neutrophils. This study demonstrated that PEP1261 significantly inhibited the effect on the ROS generation as well as the mRNA synthesis and expression of the complement factors in neutrophils isolated from infarct heart.


Subject(s)
Lysosomes/enzymology , Neutrophils/metabolism , Oligopeptides/pharmacology , Reactive Oxygen Species/metabolism , Animals , Complement System Proteins/metabolism , Densitometry , Female , Lysosomes/drug effects , Myocardial Infarction/pathology , Neutrophils/drug effects , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Rats , Rats, Wistar
17.
Arthritis Rheum ; 65(9): 2380-91, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23754328

ABSTRACT

OBJECTIVE: Bruton's tyrosine kinase (BTK) plays a critical role in B cell development and function. We recently described a selective BTK inhibitor, RN486, that blocks B cell receptor (BCR) and Fcγ receptor signaling and is efficacious in animal models of arthritis. The aim of this study was to examine the potential efficacy of BTK in systemic lupus erythematosus (SLE), using an NZB × NZW mouse model of spontaneous SLE. METHODS: Mice received RN486 or its vehicle (administered in chow) at a final concentration of 30 mg/kg for 8 weeks, starting at 32 weeks of age. RESULTS: The administration of RN486 completely stopped disease progression, as determined by histologic and functional analyses of glomerular nephritis. The efficacy was associated with striking inhibition of B cell activation, as demonstrated by a significant reduction in CD69 expression in response to BCR crosslinking. RN486 markedly reduced the secretion of IgG anti-double-stranded DNA (anti-dsDNA) secretion, as determined by enzyme-linked immunosorbent and enzyme-linked immunospot assays. Flow cytometric analysis demonstrated depletion of CD138(high) B220(low) plasma cells in the spleen. RN486 inhibited secretion of IgG anti-dsDNA but not IgM anti-dsDNA, suggesting that pharmacologic blockade of BTK resembles the reported transgenic expression of low levels of endogenous BTK in B cells. In addition, RN486 may also impact the effector function of autoantibodies, as evidenced by a significant reduction in immune complex-mediated activation of human monocytes in vitro and down-regulation of the expression of macrophage-related and interferon-inducible genes in both the kidneys and spleens of treated mice. CONCLUSION: Collectively, our data suggest that BTK inhibitors may simultaneously target autoantibody-producing and effector cells in SLE, thus constituting a promising therapeutic alternative for this disease.


Subject(s)
B-Lymphocytes/pathology , Glomerulonephritis/drug therapy , Kidney Glomerulus/pathology , Lupus Erythematosus, Systemic/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase , Animals , Antigen-Antibody Complex/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Disease Models, Animal , Disease Progression , Down-Regulation , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Lectins, C-Type/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred NZB , Receptors, IgG/metabolism
18.
Arthritis Rheum ; 64(10): 3399-408, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22674120

ABSTRACT

OBJECTIVE: To analyze the mechanism for the therapeutic effects of tumor necrosis factor α (TNFα) inhibition in a murine model of systemic lupus erythematosus. METHODS: We used the (NZB × NZW)F(1) (NZB/NZW) mouse model of interferon-α-induced lupus nephritis and treated mice with TNF receptor type II (TNFRII) Ig after TNFα expression was detected in the kidneys. Autoantibodies were measured by enzyme-linked immunosorbent assay (ELISA), and autoantibody- forming cells were determined using an enzyme-linked immunospot assay. Activation of splenocytes was analyzed by flow cytometry. Kidneys were harvested and analyzed using flow cytometry, immunohistochemistry, ELISA, Western blotting, and real-time polymerase chain reaction. RESULTS: TNFRII Ig treatment stabilized nephritis and markedly prolonged survival. Autoantibody production and systemic immune activation were not inhibited, but the renal response to glomerular immune complex deposition was attenuated. This was associated with decreases in renal production of chemokines, renal endothelial cell activation, interstitial F4/80(high) macrophage accumulation, tubular damage, and oxidative stress. In contrast, perivascular lymphoid aggregates containing B cells, T cells, and dendritic cells accumulated unabated. CONCLUSION: Our data suggest that TNFα is a critical cytokine that amplifies the response of the nephron to immune complex deposition, but that it has less influence on the response of the systemic vasculature to inflammation.


Subject(s)
Antigen-Antibody Complex/drug effects , Kidney/drug effects , Lupus Nephritis/drug therapy , Macrophages/drug effects , Tumor Necrosis Factor-alpha/immunology , Animals , Antigen-Antibody Complex/immunology , Autoantibodies/blood , Autoantibodies/immunology , Disease Models, Animal , Interferon-alpha , Kidney/immunology , Kidney/metabolism , Kidney Glomerulus/drug effects , Kidney Glomerulus/immunology , Kidney Glomerulus/metabolism , Lupus Nephritis/chemically induced , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Spleen/drug effects , Spleen/immunology , Spleen/metabolism
19.
J Immunol ; 189(2): 988-1001, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22723521

ABSTRACT

Lupus nephritis (LN) is a serious manifestation of systemic lupus erythematosus. Therapeutic studies in mouse LN models do not always predict outcomes of human therapeutic trials, raising concerns about the human relevance of these preclinical models. In this study, we used an unbiased transcriptional network approach to define, in molecular terms, similarities and differences among three lupus models and human LN. Genome-wide gene-expression networks were generated using natural language processing and automated promoter analysis and compared across species via suboptimal graph matching. The three murine models and human LN share both common and unique features. The 20 commonly shared network nodes reflect the key pathologic processes of immune cell infiltration/activation, endothelial cell activation/injury, and tissue remodeling/fibrosis, with macrophage/dendritic cell activation as a dominant cross-species shared transcriptional pathway. The unique nodes reflect differences in numbers and types of infiltrating cells and degree of remodeling among the three mouse strains. To define mononuclear phagocyte-derived pathways in human LN, gene sets activated in isolated NZB/W renal mononuclear cells were compared with human LN kidney profiles. A tissue compartment-specific macrophage-activation pattern was seen, with NF-κB1 and PPARγ as major regulatory nodes in the tubulointerstitial and glomerular networks, respectively. Our study defines which pathologic processes in murine models of LN recapitulate the key transcriptional processes active in human LN and suggests that there are functional differences between mononuclear phagocytes infiltrating different renal microenvironments.


Subject(s)
Crosses, Genetic , Disease Models, Animal , Gene Regulatory Networks/immunology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Transcription, Genetic/immunology , Animals , Gene Expression Profiling , Glomerulonephritis, IGA/genetics , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/pathology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lupus Nephritis/genetics , Male , Mice , Mice, Inbred NZB , Nephritis, Interstitial/genetics , Nephritis, Interstitial/immunology , Nephritis, Interstitial/pathology , Proteinuria/genetics , Proteinuria/immunology , Proteinuria/pathology
20.
J Immunol ; 187(3): 1506-13, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21705616

ABSTRACT

The critical role of IFN-α in the pathogenesis of human systemic lupus erythematosus has been highlighted in recent years. Exposure of young lupus-prone NZB/W F1 mice to IFN-α in vivo leads to an accelerated lupus phenotype that is dependent on T cells and is associated with elevated serum levels of BAFF, IL-6, and TNF-α, increased splenic expression of IL-6 and IL-21, formation of large germinal centers, and the generation of large numbers of short-lived plasma cells that produce IgG2a and IgG3 autoantibodies. In this study, we show that both IgG2a and IgG3 autoantibodies are pathogenic in IFN-α-accelerated lupus, and their production can be dissociated by using low-dose CTLA4-Ig. Only high-dose CTLA4-Ig attenuates both IgG2a and IgG3 autoantibody production and significantly delays death from lupus nephritis. In contrast, BAFF/APRIL blockade has no effect on germinal centers or the production of IgG anti-dsDNA Abs but, if given at the time of IFN-α challenge, delays the progression of lupus by attenuating systemic and renal inflammation. Temporary remission of nephritis induced by combination therapy with cyclophosphamide, anti-CD40L Ab, and CTLA4-Ig is associated with the abrogation of germinal centers and depletion of short-lived plasma cells, but relapse occurs more rapidly than in conventional NZB/W F1 mice. This study demonstrates that IFN-α renders NZB/W F1 relatively resistant to therapeutic intervention and suggests that the IFN signature should be considered when randomizing patients into groups and analyzing the results of human clinical trials in systemic lupus erythematosus.


Subject(s)
Crosses, Genetic , Immunity, Innate/immunology , Interferon-alpha/adverse effects , Lupus Nephritis/drug therapy , Lupus Nephritis/immunology , Abatacept , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Autoantibodies/biosynthesis , B-Cell Activating Factor/antagonists & inhibitors , B-Cell Activating Factor/biosynthesis , B-Cell Activating Factor/blood , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Immunity, Innate/genetics , Immunoconjugates/therapeutic use , Interferon-alpha/administration & dosage , Lac Operon/immunology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/pathology , Mice , Mice, Inbred NZB , Recombinant Fusion Proteins/therapeutic use , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL
...