Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Front Immunol ; 15: 1393814, 2024.
Article in English | MEDLINE | ID: mdl-38895113

ABSTRACT

Systemic lupus erythematosus (SLE) is classified by instinctual classification criteria. A valid proclamation is that these formally accepted SLE classification criteria legitimate the syndrome as being difficult to explain and therefore enigmatic. SLE involves scientific problems linked to etiological factors and criteria. Our insufficient understanding of the clinical condition uniformly denoted SLE depends on the still open question of whether SLE is, according to classification criteria, a well-defined one disease entity or represents a variety of overlapping indistinct syndromes. Without rational hypotheses, these problems harm clear definition(s) of the syndrome. Why SLE is not anchored in logic, consequent, downstream interdependent and interactive inflammatory networks may rely on ignored predictive causality principles. Authoritative classification criteria do not reflect consequent causality criteria and do not unify characterization principles such as diagnostic criteria. We need now to reconcile legendary scientific achievements to concretize the delimitation of what SLE really is. Not all classified SLE syndromes are "genuine SLE"; many are theoretically "SLE-like non-SLE" syndromes. In this study, progressive theories imply imperative challenges to reconsider the fundamental impact of "the causality principle". This may offer us logic classification and diagnostic criteria aimed at identifying concise SLE syndromes as research objects. Can a systems science approach solve this problem?


Subject(s)
Lupus Erythematosus, Systemic , Humans , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/immunology , DNA , Causality
2.
Front Immunol ; 15: 1346619, 2024.
Article in English | MEDLINE | ID: mdl-38361929

ABSTRACT

The basic initiative related to this study is derived from the fact that systemic lupus erythematosus (SLE) is a unique and fertile system science subject. We are, however, still far from understanding its nature. It may be fair to indicate that we are spending more time and resources on studying the complexity of classified SLE than studying the validity of classification criteria. This study represents a theoretical analysis of current instinctual SLE classification criteria based on "the causality principle." The discussion has its basis on the radical scientific traditions introduced by Robert Koch and Louis Pasteur. They announced significant changes in our thinking of disease etiology through the implementation of the modern version of "the causality principle." They influenced all aspects of today's medical concepts and research: the transformation of medical science from studies of symptoms to study their causes, relevant for monosymptomatic diseases as for syndromes. Their studies focused on bacteria as causes of infectious diseases and on how the immune system adapts to control and prevent contagious spreading. This is the most significant paradigm shift in the modern history of medicine and resulted in radical changes in our view of the immune system. They described acquired post-infection immunity and active immunization by antigen-specific vaccines. The paradigm "transformation" has a great theoretical impact also on current studies of autoimmune diseases like SLE: symptoms and their cause(s). In this study, the evolution of SLE classification and diagnostic criteria is discussed from "the causality principle" perspective, and if contemporary SLE classification criteria are as useful as believed today for SLE research. This skepticism is based on the fact that classification criteria are not selected based on cogent causal strategies. The SLE classification criteria do not harmonize with Koch's and Pasteur's causality principle paradigms and not with Witebsky's Koch-derived postulates for autoimmune and infectious diseases. It is not established whether the classification criteria can separate SLE as a "one disease entity" from "SLE-like non-SLE disorders"-the latter in terms of SLE imitations. This is discussed here in terms of weight, rank, and impact of the classification criteria: Do they all originate from "one basic causal etiology"? Probably not.


Subject(s)
Autoimmune Diseases , Communicable Diseases , Lupus Erythematosus, Systemic , Humans , Antibodies, Antinuclear , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/etiology
3.
Autoimmun Rev ; 22(12): 103470, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37884202

ABSTRACT

Molecular and cellular aspects of the autoimmune pathophysiology in SLE is linked to the "The causality principle". SLE Classification Criteria identify per definition disease measures (here: synonymous with classification criteria), but not diagnostic criteria within a classical framework. These two mostly theoretical criteria collections represent a salient conflict between phenomenology and the causality principle - between disease measures and molecular interactions that promote such measures, in other words their cause(s). Essentially, each criterion evolves from immunogenic and inflammatory signals - some are interconnected, some are not. Disparate signals instigated by disparate causes. These may promote clinically heterogenous SLE cohorts with respect to organ affection, autoimmunity, and disease course. There is today no concise measures or arguments that settle whether SLE cohorts evolve from one decisive etiological factor (homogenous cohorts), or if disparate patho-biological factors promote SLE (heterogenous cohorts). Current SLE cohorts are not ideal substrates to serve as study objects if the research aims are to describe etiology, and molecular interactions that cause - and link - primary and secondary pathophysiological events together - events that account for early and progressive SLE. We have to develop SLE criteria allowing us to identify definable categories of SLE in order to describe etiology, pathophysiology and diagnostic criteria of delimitated SLE versions. In this regard, the causality principle is central to define dominant etiologies of individual SLE categories, and subsequent and consequent down-stream diagnostic disease measures. In this sense, we may whether we like it or not identify different SLE categories like "genuine SLE" and "SLE-like non-SLE" syndromes. Many aspects of this problem are thoroughly discussed in this study.


Subject(s)
Autoimmunity , Lupus Erythematosus, Systemic , Humans , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/genetics
4.
Front Immunol ; 13: 1011591, 2022.
Article in English | MEDLINE | ID: mdl-36248792

ABSTRACT

It is, so to say, not a prerogative authority assigned to SLE classification criteria that allow them to declare something definitively important about SLE. This is particularly true as criteria-based classification processes overrule the highly needed evolution of concise diagnostic criteria. It is classification criteria that allocate SLE patients into cohorts intended to describe the nature of their disease. Therefore, all major SLE classification criteria since the 1971 preliminary criteria usurp the role of diagnostic criteria. Today´s practice silently accept that the SLE classification process "diagnose" SLE patients despite the fact that classification criteria are not accepted as diagnostic criteria! This is a central paradox in contemporary SLE research strategies. Contemporary SLE cohorts are designed to investigate SLE´s etiological features. However, each cohort that is categorized by classification criteria has one central inherent problem. From theoretical and practical arguments, they embody multiple distinct clinical phenotypes. This raises the critical and principal question if phenotypically heterogenic SLE cohorts are useful to identify basic SLE-specific etiology(ies) and disease process(es). In times to come, we must prioritize development of firm diagnostic criteria for SLE, as the classification criteria have not contributed to reduce the enigmatic character of the syndrome. No radical improvements are visible in the horizon that may lead to concise investigations of SLE in well-defined homogenous SLE cohorts. We must develop new strategies where studies of phenotypically standardized cohorts of SLE must be central elements. Problems related to contemporary SLE classification criteria are contemplated, analyzed, and critically discussed in this study.


Subject(s)
Lupus Erythematosus, Systemic , Cohort Studies , Humans , Lupus Erythematosus, Systemic/diagnosis
5.
Front Immunol ; 12: 808008, 2021.
Article in English | MEDLINE | ID: mdl-35087528

ABSTRACT

Systemic lupus erythematosus (SLE) is diagnosed and classified by criteria, or by experience, intuition and traditions, and not by scientifically well-defined etiology(ies) or pathogenicity(ies). One central criterion and diagnostic factor is founded on theoretical and analytical approaches based on our imperfect definition of the term "The anti-dsDNA antibody". "The anti-dsDNA antibody" holds an archaic position in SLE as a unique classification criterium and pathogenic factor. In a wider sense, antibodies to unique transcriptionally active or silent DNA structures and chromatin components may have individual and profound nephritogenic impact although not considered yet - not in theoretical nor in descriptive or experimental contexts. This hypothesis is contemplated here. In this analysis, our state-of-the-art conception of these antibodies is probed and found too deficient with respect to their origin, structural DNA specificities and clinical/pathogenic impact. Discoveries of DNA structures and functions started with Miescher's Nuclein (1871), via Chargaff, Franklin, Watson and Crick, and continues today. The discoveries have left us with a DNA helix that presents distinct structures expressing unique operations of DNA. All structures are proven immunogenic! Unique autoimmune antibodies are described against e.g. ssDNA, elongated B DNA, bent B DNA, Z DNA, cruciform DNA, or individual components of chromatin. In light of the massive scientific interest in anti-DNA antibodies over decades, it is an unexpected observation that the spectrum of DNA structures has been known for decades without being implemented in clinical immunology. This leads consequently to a critical analysis of historical and contemporary evidence-based data and of ignored and one-dimensional contexts and hypotheses: i.e. "one antibody - one disease". In this study radical viewpoints on the impact of DNA and chromatin immunity/autoimmunity are considered and discussed in context of the pathogenesis of lupus nephritis.


Subject(s)
Antibodies, Antinuclear/immunology , Lupus Erythematosus, Systemic/etiology , Antibody Formation/immunology , Antibody Specificity/immunology , Autoimmunity , Chromatin/chemistry , Chromatin/immunology , Cross Reactions/immunology , DNA/chemistry , DNA/immunology , Disease Susceptibility/immunology , Humans , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/therapy , Nucleic Acid Conformation , Peptides/immunology , Phospholipids/immunology , Proteins
6.
Front Immunol ; 11: 569234, 2020.
Article in English | MEDLINE | ID: mdl-33123142

ABSTRACT

One cannot discuss anti-dsDNA antibodies and lupus nephritis without discussing the nature of Systemic lupus erythematosus (SLE). SLE is insistently described as a prototype autoimmune syndrome, with anti-dsDNA antibodies as a central biomarker and a pathogenic factor. The two entities, "SLE" and "The Anti-dsDNA Antibody," have been linked in previous and contemporary studies although serious criticism to this mutual linkage have been raised: Anti-dsDNA antibodies were first described in bacterial infections and not in SLE; later in SLE, viral and parasitic infections and in malignancies. An increasing number of studies on classification criteria for SLE have been published in the aftermath of the canonical 1982 American College of Rheumatology SLE classification sets of criteria. Considering these studies, it is surprising to observe a nearby complete absence of fundamental critical/theoretical discussions aimed to explain how and why the classification criteria are linked in context of etiology, pathogenicity, or biology. This study is an attempt to prioritize critical comments on the contemporary definition and classification of SLE and of anti-dsDNA antibodies in context of lupus nephritis. Epidemiology, etiology, pathogenesis, and measures of therapy efficacy are implemented as problems in the present discussion. In order to understand whether or not disparate clinical SLE phenotypes are useful to determine its basic biological processes accounting for the syndrome is problematic. A central problem is discussed on whether the clinical role of anti-dsDNA antibodies from principal reasons can be accepted as a biomarker for SLE without clarifying what we define as an anti-dsDNA antibody, and in which biologic contexts the antibodies appear. In sum, this study is an attempt to bring to the forum critical comments on the contemporary definition and classification of SLE, lupus nephritis and anti-dsDNA antibodies. Four concise hypotheses are suggested for future science at the end of this analytical study.


Subject(s)
Autoimmunity , Disease Susceptibility/immunology , Lupus Erythematosus, Systemic/etiology , Animals , Antibodies, Antinuclear/immunology , Antibody Affinity/immunology , Autoantibodies/immunology , Autoantigens/immunology , Biomarkers , DNA/immunology , Evidence-Based Medicine/methods , Genetic Predisposition to Disease , Humans , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/metabolism , Research , Syndrome
7.
Front Immunol ; 10: 1104, 2019.
Article in English | MEDLINE | ID: mdl-31156647

ABSTRACT

This study aims to understand what lupus nephritis is, its origin, clinical context, and its pathogenesis. Truly, we encounter many conceptual and immanent tribulations in our attempts to search for the pathogenesis of this disease-and how to explain its assumed link to SLE. Central in the present landscape stay a short history of the early studies that substantiated the structures of isolated or chromatin-assembled mammalian dsDNA, and its assumed, highly controversial role in induction of anti-dsDNA antibodies. Arguments discussed here may provoke the view that anti-dsDNA antibodies are not what we think they are, as they may be antibodies operational in quite different biological contexts, although they bind dsDNA by chance. This may not mean that these antibodies are not pathogenic but they do not inform how they are so. This theoretical study centers the content around the origin and impact of extra-cellular DNA, and if dsDNA has an effect on the adaptive immune system. The pathogenic potential of chromatin-anti-dsDNA antibody interactions is limited to incite lupus nephritis and dermatitis which may be linked in a common pathogenic process. These are major criteria in SLE classification systems but are not shared with other defined manifestations in SLE, which may mean that they are their own disease entities, and not integrated in SLE. Today, the models thought to explain lupus nephritis are divergent and inconsistent. We miss a comprehensive perspective to try the different models against each other. To do this, we need to take all elements of the syndrome SLE into account. This can only be achieved by concentrating on the interactions between autoimmunity, immunopathology, deviant cell death and necrotic chromatin in context of elements of system science. System science provides a framework where data generated by experts can be compared, and tested against each other. This approach open for consensus on central elements making up "lupus nephritis" to separate what we agree on and how to understand the basis for conflicting models. This has not been done yet in a systematic context.


Subject(s)
Antibodies, Antinuclear/immunology , Autoimmunity , DNA/immunology , Disease Susceptibility/immunology , Lupus Nephritis/etiology , Animals , Antibody Formation/immunology , Autoantigens/immunology , Chromatin/genetics , Chromatin/immunology , Chromatin/metabolism , Cross Reactions/immunology , Humans , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Nephritis/metabolism , Lupus Nephritis/pathology
8.
Front Immunol ; 9: 387, 2018.
Article in English | MEDLINE | ID: mdl-29545801

ABSTRACT

Systemic lupus erythematosus (SLE) is an inadequately defined syndrome. Etiology and pathogenesis remain largely unknown. SLE is on the other hand a seminal syndrome that has challenged immunologists, biologists, genetics, and clinicians to solve its nature. The syndrome is characterized by multiple, etiologically unlinked manifestations. Unexpectedly, they seem to occur in different stochastically linked clusters, although single gene defects may promote a smaller spectrum of symptoms/criteria typical for SLE. There is no known inner coherence of parameters (criteria) making up the disease. These parameters are, nevertheless, implemented in The American College of Rheumatology (ACR) and The Systemic Lupus Collaborating Clinics (SLICC) criteria to classify SLE. Still, SLE is an abstraction since the ACR or SLICC criteria allow us to define hundreds of different clinical SLE phenotypes. This is a major point of the present discussion and uses "The anti-dsDNA antibody" as an example related to the problematic search for biomarkers for SLE. The following discussion will show how problematic this is: the disease is defined through non-coherent classification criteria, its complexity is recognized and accepted, its pathogenesis is plural and poorly understood. Therapy is focused on dominant symptoms or organ manifestations, and not on the syndrome itself. From basic scientific evidences, we can add substantial amount of data that are not sufficiently considered in clinical medicine, which may change the paradigms linked to what "The Anti-DNA antibody" is-and is not-in context of the imperfectly defined syndrome SLE.


Subject(s)
Antibodies, Antinuclear/metabolism , Lupus Erythematosus, Systemic/immunology , Models, Immunological , Rheumatology , Animals , Biomarkers/metabolism , Evidence-Based Medicine , Humans , Interdisciplinary Communication , Lupus Erythematosus, Systemic/diagnosis , Phenotype
9.
PLoS One ; 12(11): e0188863, 2017.
Article in English | MEDLINE | ID: mdl-29190833

ABSTRACT

FcγRIIB-/-yaa mice develop severe lupus glomerulonephritis due to lack of an inhibitory immune cell receptor combined with a Y-chromosome linked autoimmune accelerator mutation. In the present study, we have investigated nephritis development and progression in FcγRIIB-/-yaa mice to find shared features with NZB/NZW F1 lupus prone mice and human disease. We sacrificed 25 male FcγRIIB-/-yaa mice at various disease stages, and grouped them according to activity and chronicity indices for lupus nephritis. Glomerular morphology and localization of electron dense deposits containing IgG were further determined by immune electron microscopy. Renal DNase I and pro-inflammatory cytokine mRNA levels were measured by real-time quantitative PCR. DNase I protein levels was assessed by immunohistochemistry and zymography. Our results demonstrate early development of electron dense deposits containing IgG in FcγRIIB-/-yaa mice, before detectable levels of serum anti-dsDNA antibodies. Similar to NZB/NZW F1, electron dense deposits in FcγRIIB-/-yaa progressed from being confined to the mesangium in the early stage of lupus nephritis to be present also in capillary glomerular basement membranes. In the advanced stage of lupus nephritis, renal DNase I was lost on both transcriptional and protein levels, which has previously been shown in NZB/NZW F1 mice and in human disease. Although lupus nephritis appears on different genetic backgrounds, our findings suggest similar processes when comparing different murine models and human lupus nephritis.


Subject(s)
Deoxyribonuclease I/metabolism , Kidney Glomerulus/pathology , Lupus Nephritis/pathology , Receptors, IgG/genetics , Animals , Disease Progression , Immunoglobulin G/metabolism , Kidney Glomerulus/enzymology , Kidney Glomerulus/metabolism , Kidney Tubules/metabolism , Lupus Nephritis/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Toll-Like Receptor 7/metabolism
10.
Front Immunol ; 8: 362, 2017.
Article in English | MEDLINE | ID: mdl-28424695

ABSTRACT

Deimination, a posttranslational modification of arginine to citrulline carried out by peptidylarginine deiminases, may compromise tolerance of self-antigens. Patients with connective tissue autoimmunity, particularly rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), or Felty's syndrome, present with autoantibodies to deiminated histones (dH), which thus form a category of antibodies to citrullinated protein antigens (ACPA). In general, ACPA are a sensitive diagnostic for RA and may form in response to the release of nuclear chromatin (DNA plus dH) from granulocytes, usually referred to as neutrophil extracellular traps. The aim of this study was to examine spontaneously autoimmune mice for autoantibodies and T cell responses to dH. We compared IgG binding to deiminated and non-deiminated histones (nH) by ELISA and Western blotting in spontaneously autoimmune strains of (NZB × NZW) F1 and NZM2410 together with their derivative congenic strains, C57BL/6.Sle1 and C57BL/6.Sle1.Sle3, which display profound autoreactivity against nuclear self-antigens. The splenocyte proliferation against the two antigens was determined in the spontaneously autoimmune (NZB × NZW) F1 strain from which other autoimmune strains used in the study were derived. Immunizations with dH and nH were attempted in BALB/c mice to assess their splenocyte response. Splenocytes from BALB/c mice and from autoimmune mice at the time of conversion to autoimmunity proliferated strongly in response to dH, yet serum IgG from autoimmune (NZB × NZW) F1, NZM2410, and C57BL/6.Sle1.Sle3 mice displayed a remarkable bias against binding to dH. At the time of seroconversion, the antibodies already exhibited preference for nH, and only nH were recovered from circulating immune complexes. Analysis of histone deimination showed constitutive deimination in thymic extracts from C57BL/6 and C57BL/6.Sle1.Sle2.Sle3 triply congenic mice and in spleens of autoimmune triply congenic mice. Our study demonstrates that tolerance mechanisms against dH are intact in BALB/c and C57BL/6 mice and continue to be effective in mice with overt autoimmunity to nH. We conclude that, in contrast to human RA and SLE patients, where we frequently observe autoantibodies against dH, autoimmune mice maintain strong tolerance mechanisms to prevent the development of autoantibodies to dH.

11.
Ann Rheum Dis ; 76(3): 554-561, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27884822

ABSTRACT

OBJECTIVES: Treat-to-target recommendations have identified 'remission' as a target in systemic lupus erythematosus (SLE), but recognise that there is no universally accepted definition for this. Therefore, we initiated a process to achieve consensus on potential definitions for remission in SLE. METHODS: An international task force of 60 specialists and patient representatives participated in preparatory exercises, a face-to-face meeting and follow-up electronic voting. The level for agreement was set at 90%. RESULTS: The task force agreed on eight key statements regarding remission in SLE and three principles to guide the further development of remission definitions:1. Definitions of remission will be worded as follows: remission in SLE is a durable state characterised by …………………. (reference to symptoms, signs, routine labs).2. For defining remission, a validated index must be used, for example, clinical systemic lupus erythematosus disease activity index (SLEDAI)=0, British Isles lupus assessment group (BILAG) 2004 D/E only, clinical European consensus lupus outcome measure (ECLAM)=0; with routine laboratory assessments included, and supplemented with physician's global assessment.3. Distinction is made between remission off and on therapy: remission off therapy requires the patient to be on no other treatment for SLE than maintenance antimalarials; and remission on therapy allows patients to be on stable maintenance antimalarials, low-dose corticosteroids (prednisone ≤5 mg/day), maintenance immunosuppressives and/or maintenance biologics.The task force also agreed that the most appropriate outcomes (dependent variables) for testing the prognostic value (construct validity) of potential remission definitions are: death, damage, flares and measures of health-related quality of life. CONCLUSIONS: The work of this international task force provides a framework for testing different definitions of remission against long-term outcomes.


Subject(s)
Lupus Erythematosus, Systemic/drug therapy , Adrenal Cortex Hormones/therapeutic use , Antibodies, Antinuclear/blood , Antimalarials/therapeutic use , Complement System Proteins/metabolism , Consensus , DNA/immunology , Humans , Immunosuppressive Agents/therapeutic use , Lupus Erythematosus, Systemic/blood , Maintenance Chemotherapy , Remission Induction , Severity of Illness Index , Symptom Flare Up
12.
Semin Nephrol ; 35(5): 427-38, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26573545

ABSTRACT

Lupus nephritis is one of the most serious manifestations of systemic lupus erythematosus, and represents one of the criteria implemented to classify systemic lupus erythematosus. Although studied for decades, no consensus has been reached related to the basic cellular, molecular, and immunologic mechanism(s) responsible for lupus nephritis. No causal treatments have been developed; therapy is approached mainly with nonspecific immunosuppressive medications. More detailed insight into disease mechanisms therefore is indispensable to develop new therapeutic strategies. In this review, contemporary knowledge on the pathogenic mechanisms of lupus nephritis is discussed based on recent data in murine and human lupus nephritis. Specific focus is given to the effect of anti-double-stranded DNA/antinucleosome antibodies in the kidneys and whether they bind exposed chromatin fragments in glomeruli or whether they bind inherent glomerular structures by cross-recognition. Overall, the data presented here favor the exposed chromatin model because we did not find any indication to substantiate the anti-double-stranded DNA antibody cross-reacting model. At the end of this review we present data on why chromatin fragments are expressed in the glomeruli of patients with lupus nephritis, and discuss how this knowledge can be used to direct the development of future therapies.


Subject(s)
Antibodies, Antinuclear/immunology , Chromatin/immunology , Kidney Glomerulus/immunology , Lupus Nephritis/immunology , Animals , Cross Reactions , Deoxyribonuclease I/genetics , Deoxyribonuclease I/metabolism , Disease Models, Animal , Fibrinolytic Agents/therapeutic use , Heparin/therapeutic use , Humans , Immunoglobulin G/immunology , Immunosuppressive Agents/therapeutic use , Kidney/immunology , Kidney/metabolism , Kidney Glomerulus/metabolism , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/drug therapy , Lupus Nephritis/metabolism , Matrix Metalloproteinases/metabolism , Mice , Molecular Chaperones/therapeutic use , Molecular Targeted Therapy
13.
PLoS One ; 10(6): e0129485, 2015.
Article in English | MEDLINE | ID: mdl-26065428

ABSTRACT

We have demonstrated that the renal endonuclease DNaseI is up-regulated in mesangial nephritis while down-regulated during progression of the disease. To determine the basis for these reciprocal DNaseI expression profiles we analyse processes accounting for an early increase in renal DNaseI expression. Main hypotheses were that i. the mesangial inflammation and secreted pro-inflammatory cytokines directly increase DNaseI protein expression in tubular cells, ii. the anti-apoptotic protein tumor necrosis factor receptor-associated protein 1 (Trap 1) is down-regulated by increased expression of DNaseI due to transcriptional interference, and iii. pro-inflammatory cytokines promote nuclear translocation of a variant of DNaseI. The latter hypothesis emerges from the fact that anti-DNaseI antibodies stained tubular cell nuclei in murine and human lupus nephritis. The present study was performed on human tubular epithelial cells stimulated with pro-inflammatory cytokines. Expression of the DNaseI and Trap 1 genes was determined by qPCR, confocal microscopy, gel zymography, western blot and by immune electron microscopy. Results from in vitro cell culture experiments were analysed for biological relevance in kidneys from (NZBxNZW)F1 mice and human patients with lupus nephritis. Central data indicate that stimulating the tubular cells with TNFα promoted increased DNaseI and reduced Trap 1 expression, while TNFα and IL-1ß stimulation induced nuclear translocation of the DNaseI. TNFα-stimulation resulted in 3 distinct effects; increased DNaseI and IL-1ß gene expression, and nuclear translocation of DNaseI. IL-1ß-stimulation solely induced nuclear DNaseI translocation. Tubular cells stimulated with TNFα and simultaneously transfected with IL-1ß siRNA resulted in increased DNaseI expression but no nuclear translocation. This demonstrates that IL-1ß promotes nuclear translocation of a cytoplasmic variant of DNaseI since translocation clearly was not dependent on DNaseI gene activation. Nuclear translocated DNaseI is shown to be enzymatically inactive, which may point at a new, yet unknown function of renal DNaseI.


Subject(s)
Deoxyribonuclease I/metabolism , Interleukin-1beta/metabolism , Kidney Tubules/metabolism , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Animals , Case-Control Studies , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Cytokines/metabolism , Deoxyribonuclease I/genetics , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , Interleukin-1beta/genetics , Kidney Tubules/cytology , Kidney Tubules/drug effects , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Mice, Inbred BALB C , Mice, Mutant Strains , Molecular Sequence Data , Protein Transport , Tumor Necrosis Factor-alpha/pharmacology
14.
Semin Immunopathol ; 36(3): 301-11, 2014 May.
Article in English | MEDLINE | ID: mdl-24763531

ABSTRACT

Systemic lupus erythematosus (SLE) is a systemic autoimmune disease with various clinical manifestations affecting different tissues. A characteristic feature of SLE is the presence of autoantibodies against double-stranded (ds)DNA, histones and nucleosomes, and other chromatin components. SLE is a prototype type III hypersensitivity reaction. Local deposition of anti-nuclear antibodies in complex with released chromatin induces serious inflammatory conditions by activation of the complement system. The severe renal manifestation, lupus nephritis, is classified based on histological findings in renal biopsies. Apoptotic debris, including chromatin, is present in the extracellular matrix and circulation of patients with SLE. This may be due to an aberrant process of apoptosis and/or insufficient clearance of apoptotic cells/chromatin. The non-cleared apoptotic debris may lead to activation of both the innate and adaptive immune systems. In addition, an aberrant presentation of peptides by antigen-presenting cells, disturbed selection processes for lymphocytes, and deregulated lymphocyte responses may be involved in the development of autoimmunity. In the present review, we briefly will summarize current knowledge on the pathogenesis of SLE. We will also critically discuss and challenge central issues that need to be addressed in order to fully understand the pathogenic mechanisms involved in the development of SLE and in order to have an improved diagnosis for SLE. Disappointingly, in our opinion, there are still more questions than answers for the pathogenesis, diagnosis, and treatment of SLE.


Subject(s)
Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/etiology , Animals , Antibodies, Antinuclear/immunology , Apoptosis/immunology , Autoantibodies/immunology , Chromatin/immunology , Chromatin/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Deoxyribonuclease I/genetics , Disease Progression , Gene Silencing , Humans , Immune Tolerance , Lupus Erythematosus, Systemic/complications , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Transcription, Genetic
16.
Clin Dev Immunol ; 2013: 791262, 2013.
Article in English | MEDLINE | ID: mdl-24151519

ABSTRACT

We have previously demonstrated that continuous infusion of low molecular weight (LMW) heparin delays autoantibody production and development of lupus nephritis in (NZBxNZW)F1 (B/W) mice. In this study we investigated the effect of LMW heparin on renal cytokine and chemokine expression and on nucleosome-mediated activation of nucleosome-specific splenocytes. Total mRNA extracted from kidneys of heparin-treated or -untreated B/W mice was analysed by qPCR for the expression of several cytokines, chemokines, and Toll-like receptors. Splenocytes taken from B/W mice were stimulated with nucleosomes with or without the presence of heparin. Splenocyte cell proliferation as thymidine incorporation and the expression of costimulatory molecules and cell activation markers were measured. Heparin treatment of B/W mice reduced the in vivo expression of CCR2, IL1 ß , and TLR7 compared to untreated B/W mice. Nucleosome-induced cell proliferation of splenocytes was not influenced by heparin. The expression of CD80, CD86, CD69, CD25, CTLA-4, and TLR 2, 7, 8, and 9 was upregulated upon stimulation by nucleosomes, irrespective of whether heparin was added to the cell culture or not. In conclusion, treatment with heparin lowers the kidney expression of proinflammatory mediators in B/W mice but does not affect nucleosomal activation of splenocytes.


Subject(s)
Cytokines/genetics , Gene Expression Regulation/drug effects , Heparin, Low-Molecular-Weight/pharmacology , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Kidney/pathology , Lupus Nephritis/genetics , Lupus Nephritis/pathology , Lupus Nephritis/prevention & control , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Nucleosomes/immunology , Nucleosomes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
17.
Mol Med ; 19: 161-9, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23752208

ABSTRACT

Autoantibodies to components of chromatin, which include double-stranded DNA (dsDNA), histones and nucleosomes, are central in the pathogenesis of lupus nephritis. How anti-chromatin autoantibodies exert their nephritogenic activity, however, is controversial. One model assumes that autoantibodies initiate inflammation when they cross-react with intrinsic glomerular structures such as components of membranes, matrices or exposed nonchromatin ligands released from cells. Another model suggests glomerular deposition of autoantibodies in complex with chromatin, thereby inducing classic immune complex-mediated tissue damage. Recent data suggest acquired error of renal chromatin degradation due to the loss of renal DNaseI enzyme activity is an important contributing factor to the development of lupus nephritis in lupus-prone (NZBxNZW)F1 mice and in patients with lupus nephritis. Down-regulation of DNaseI expression results in reduced chromatin fragmentation and in deposition of extracellular chromatin-IgG complexes in glomerular basement membranes in individuals who produce IgG anti-chromatin autoantibodies. The main focus of the present review is to discuss whether exposed chromatin fragments in glomeruli are targeted by potentially nephritogenic anti-dsDNA autoantibodies or if the nephritogenic activity of these autoantibodies is explained by cross-reaction with intrinsic glomerular constituents or if both models coexist in diseased kidneys. In addition, the role of silencing of the renal DNaseI gene and the biological consequences of reduced chromatin fragmentation in nephritic kidneys are discussed.


Subject(s)
Antibodies, Antinuclear/immunology , Lupus Nephritis/immunology , Animals , Chromatin/immunology , Deoxyribonuclease I/genetics , Humans , Lupus Nephritis/genetics
18.
Autoimmunity ; 45(8): 588-92, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23013317

ABSTRACT

Loss of immunological tolerance results in autoimmunity and may finally end in autoimmune disorders. For an autoimmune response against chromatin, autologous chromatin (nucleosomes) is assumed to activate both chromatin-specific B and T cells with a resulting anti-chromatin antibody response. As only fragmental elements of this process have been described, we do not have the full insight to justify this model in vivo. Early experimental immunization with methylated bovine serum albumin-DNA complexes elicited antibodies to various forms of synthetic ssDNA/dsDNA, but notably not to mammalian dsDNA. Thus, for a long time with intense research, the general result was that all forms of ssDNA and dsDNA, but mammalian B helical DNA, had an immunogenic potential. Summarizing these results, a preliminary conclusion was settled, saying that mammalian dsDNA was not immunogenic while other forms of DNA were really immunogenic in the situation where they were in complex with proteins. Recent studies have focused on nuclease deficiencies as a condition where chromatin may be presented to the immune system in an immunogenic form. However, although such deficiencies may provide information as to how chromatin may be exposed and targeted by relevant antibodies, data demonstrate that nuclease deficiencies is not in general correlated with autoimmunity to components of chromatin. This review discusses these topics, and provides information that may explain processes that account for anti-dsDNA antibody responses in vivo.


Subject(s)
Antibodies, Antinuclear/immunology , Autoimmunity , Chromatin/immunology , DNA-Binding Proteins/immunology , DNA/immunology , Deoxyribonucleases/metabolism , Animals , Deoxyribonucleases/deficiency , Endodeoxyribonucleases/deficiency , Endodeoxyribonucleases/metabolism , Humans , Lupus Nephritis/immunology
19.
PLoS One ; 7(3): e34080, 2012.
Article in English | MEDLINE | ID: mdl-22479529

ABSTRACT

Recent studies demonstrate that transformation of mild lupus nephritis into end-stage disease is imposed by silencing of renal DNaseI gene expression in (NZBxNZW)F1 mice. Down-regulation of DNaseI results in reduced chromatin fragmentation, and in deposition of extracellular chromatin-IgG complexes in glomerular basement membranes in individuals that produce IgG anti-chromatin antibodies. The main focus of the present study is to describe the biological consequences of renal DNaseI shut-down and reduced chromatin fragmentation with a particular focus on whether exposed large chromatin fragments activate Toll like receptors and the necrosis-related Clec4e receptor in murine and human lupus nephritis. Furthermore, analyses where performed to determine if matrix metalloproteases are up-regulated as a consequence of chromatin-mediated Toll like receptors/Clec4e stimulation. Mouse and human mRNA expression levels of DNaseI, Toll like receptors 7-9, Clec4e, pro-inflammatory cytokines and MMP2/MMP9 were determined and compared with in situ protein expression profiles and clinical data. We demonstrate that exposure of chromatin significantly up-regulate Toll like receptors and Clec4e in mice, and also but less pronounced in patients with lupus nephritis treated with immunosuppresants. In conclusion, silencing of renal DNaseI gene expression initiates a cascade of inflammatory signals leading to progression of both murine and human lupus nephritis. Principal component analyses biplot of data from murine and human lupus nephrits demonstrate the importance of DNaseI gene shut down for progression of the organ disease.


Subject(s)
Deoxyribonuclease I/genetics , Kidney/enzymology , Lectins, C-Type/genetics , Lupus Nephritis/genetics , Membrane Proteins/genetics , Receptors, Immunologic/genetics , Animals , Cells, Cultured , Chromatin/chemistry , Cytokines/metabolism , Disease Models, Animal , Female , Gene Silencing , Humans , Inflammation , Lectins, C-Type/metabolism , Lupus Nephritis/enzymology , Membrane Proteins/metabolism , Mice , Principal Component Analysis , RNA, Messenger/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Toll-Like Receptors/metabolism
20.
Med Oncol ; 29(1): 219-26, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21221851

ABSTRACT

Imatinib, a small molecule inhibitor of ABL, PDGFR and C-KIT, has revolutionized treatment of chronic myeloid leukaemia (CML). However, resistance to treatment is of increasing importance and often is due to point mutations in the Abl kinase domain (Abl KD). Here, we analysed clinical outcome and mutation status in two independent Nordic populations (n = 77) of imatinib-resistant CML patients. We detected BCR-ABL transcripts containing point mutations of residues in the P-loop, A-loop and other kinase domain residues in 32 patients (42%). In contrast to previous data, mutations in BCR-ABL were as frequently found in patients with primary resistance (56%) as with secondary resistance (53%). No T315I mutations were found in the study cohort. BCR-ABL splice variants were identified in a significant number of our cases (19%): BCR-ABL transcripts of variable length; a variant fusion transcript joining BCR exon 14 sequences to ABL exon 4; partial, in-frame-deletion of exon 4 due to induction of a cryptic splice site by the L248V and finally, alternative splicing of ABL exon 7 sequences. Though the majority of splice variants observed in this study do not encode functional proteins, alternative splicing appears to represent a common phenomenon in the biology of CML. We conclude that Abl KD point mutations represent a major mechanism of imatinib resistance. Other sequence irregularities were also detected, but their significance in conferring resistance is unclear. Diagnostic strategies looking for imatinib-resistant clones should be designed to detect a broader profile of BCR-ABL variants than just point mutations.


Subject(s)
Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Point Mutation , Adult , Aged , Antineoplastic Agents/therapeutic use , Benzamides , DNA Mutational Analysis , Female , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Male , Middle Aged , Piperazines/therapeutic use , Protein Isoforms/genetics , Protein Structure, Tertiary , Pyrimidines/therapeutic use , Real-Time Polymerase Chain Reaction , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...