Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.935
Filter
Add more filters

Publication year range
1.
Cell ; 166(4): 991-1003, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27477514

ABSTRACT

Small immune complexes cause type III hypersensitivity reactions that frequently result in tissue injury. The responsible mechanisms, however, remain unclear and differ depending on target organs. Here, we identify a kidney-specific anatomical and functional unit, formed by resident macrophages and peritubular capillary endothelial cells, which monitors the transport of proteins and particles ranging from 20 to 700 kDa or 10 to 200 nm into the kidney interstitium. Kidney-resident macrophages detect and scavenge circulating immune complexes "pumped" into the interstitium via trans-endothelial transport and trigger a FcγRIV-dependent inflammatory response and the recruitment of monocytes and neutrophils. In addition, FcγRIV and TLR pathways synergistically "super-activate" kidney macrophages when immune complexes contain a nucleic acid. These data identify a physiological function of tissue-resident kidney macrophages and a basic mechanism by which they initiate the inflammatory response to small immune complexes in the kidney.


Subject(s)
Immune Complex Diseases/immunology , Kidney/cytology , Kidney/immunology , Macrophages/immunology , Animals , Antigen-Antibody Complex , Endothelial Cells , Macrophages/cytology , Mice, Inbred C57BL , Microscopy, Immunoelectron , Monocytes/cytology , Monocytes/immunology , Neutrophils/cytology , Neutrophils/immunology , Receptors, IgG/immunology
2.
Lupus ; 33(5): 502-510, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38470859

ABSTRACT

OBJECTIVES: In addition to various immunosuppressive agents, belimumab and anifrolumab became available in Japan. We aimed to investigate glucocorticoid-free clinical remission in a single-centre retrospective cohort in October 2023. METHODS: Our cohort included patients with SLE who needed to start or increase glucocorticoids for disease activity and were followed up for more than 1 year. We investigated the rate of achievement of clinical remission off corticosteroids (CR off C), defined as no clinical score on the SLEDAI-2K without glucocorticoids, baseline predictors of CR off C, medications used when CR off C was achieved, and flare rates following CR off C. RESULTS: Out of the 60 patients followed for an average of 5.4 (±2.6) years, 17 (28.3%) achieved CR off C in 3.6 (±1.2) years after enrolment. Use of belimumab and anifrolumab accounted for eight (47.1%) of the achievers. Among the baseline data, male sex, recent enrolment, high glucocorticoid dose, and detection of immune complex (IC) significantly predicted CR off C, while lupus nephritis (LN) and a low C3 level tended to predict it. In the multivariate analysis, IC detection was the only predictor of CR off C. Clinical flares were observed in 5.9% of the achievers during a median 1.2 years after achievement of CR off C. CONCLUSION: In the era of biologics, CR off C was achieved in 28.3% of the patient cohort requiring the start or increase of glucocorticoids for disease activity, with a relatively low rate of flares, suggesting that glucocorticoid-free clinical remission is an achievable target in SLE. IC disease, represented by male sex or nephritis, is likely to benefit from currently available medications.


Subject(s)
Biological Products , Immune Complex Diseases , Lupus Erythematosus, Systemic , Humans , Male , Glucocorticoids/therapeutic use , Retrospective Studies , Biological Products/therapeutic use , Severity of Illness Index , Lupus Erythematosus, Systemic/drug therapy , Immunosuppressive Agents/therapeutic use , Immune Complex Diseases/drug therapy , Antigen-Antibody Complex
3.
Am J Dermatopathol ; 45(2): 86-89, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36669070

ABSTRACT

ABSTRACT: Injection site reactions are defined as skin reactions at the injection site to drugs administered subcutaneously. Pathophysiologically, these reactions are based on different immunological mechanisms. We report the case of a 49-year-old patient with type 1 diabetes mellitus (first diagnosis in 1994 at the age of 23 years). Continuous subcutaneous insulin infusion using an insulin pump has been used for many years. The patient presented to the department of dermatology with progressive symptoms in the area of the insulin injection sites on the lower abdomen, accompanied by pain, burning, erythema, tenderness, and the formation of subcutaneous nodules. Previous attempts to use different insulins and to change the injection sites did not improve his symptoms. Furthermore, the symptoms appeared within hours after the insulin pump was attached, so that the injection site has to be changed as soon as every 48 hours. No anaphylactic shock was reported at any time. Multiple histological specimens were obtained from an older lesion on the abdomen as well as from test sites after standard allergological tests (prick and intradermal tests) of various insulins. Histologically, these biopsies showed the image of an extensive deep-reaching small vessel vasculitis with the aspect of an urticarial vasculitis and confirmed the diagnosis of an injection-site reaction that can be characterized as a type III hypersensitivity reaction.


Subject(s)
Diabetes Mellitus, Type 1 , Drug Hypersensitivity , Immune Complex Diseases , Urticaria , Humans , Young Adult , Adult , Middle Aged , Diabetes Mellitus, Type 1/drug therapy , Injection Site Reaction/etiology , Insulin/adverse effects , Urticaria/chemically induced
4.
Proteomics ; 22(19-20): e2100242, 2022 10.
Article in English | MEDLINE | ID: mdl-35964289

ABSTRACT

Systemic lupus erythematosus is a common autoimmune inflammatory disease which is associated with increases in autoantibodies and immune complexes that deposit in the kidney. The MRL-lpr mouse is a common mouse model used for the study of lupus and immune complex glomerulonephritis but very little is known about the plasma proteome changes in this model. We performed in-depth quantitative proteome profiling on MRL-lpr and control (strain MpJ) mice to investigate the changes in the proteome, immunoglobulins and their glycoproteome as well as protein and immune complexes. Methodologies used included immunohistochemistry, immunoglobulin isotyping, multiplexed proteome profiling, immunoglobulin immunoprecipitation with glycoproteome profiling, and size exclusion chromatography (SEC) profiling to enable a comprehensive proteome profiling of proteins and protein complexes. We also used a novel native multiplexed plasma proteome profiling (NativeMP3) method that relies on native enrichment of plasma proteins enabling ultra-deep single shot profiling where we identified 922 plasma proteins at 1% false discovery rate (FDR) in a single shot mass spectrometry run. We observed many large plasma protein differences between the MRL-lpr and control strain including differences in the immunoglobulins, immunoglobulins against specific antigens, chemokines, and proteases as well as changes in protein complexes such as the immunoproteasome.


Subject(s)
Autoimmune Diseases , Immune Complex Diseases , Mice , Animals , Mice, Inbred MRL lpr , Antigen-Antibody Complex , Proteomics , Proteome , Autoantibodies , Disease Models, Animal , Peptide Hydrolases
5.
Scand J Immunol ; 93(3): e12994, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33151588

ABSTRACT

Some pathogen infections and immune system deficiencies have been linked to a few autoimmune diseases. However, the pathogenesis of most autoimmune diseases is unknown. An explanatory hypothesis for the pathogenesis of infection-initiated autoimmune diseases is provided. Virulent pathogen infections create extensive pathogen antigens that frequently require antibodies. These antibodies create extensive antigen-antibody immune complexes, which some immuno-compromised individuals will not adequately eliminate. This will cause inflammatory type III hypersensitivity symptoms, including protease releases that destroy epithelium, mesothelium and endothelium basement membranes, express new immunogenic antigens from previously sequestered basement membrane constituents, and ultimately induce new autoantibodies. This can continue after the infection ends, if the first wave of protease attacks on basement membranes induces new autoantibodies that cause new uncleared antigen-antibody immune complexes and type III hypersensitivity reactions. The secreted proteases and other enzymes will have preferred substrates and these proteases or other enzymes by themselves, or by their processed protein substrates, can express immunogenic antigens that induce new autoantibodies and initiate various autoimmune diseases. In summary, several autoimmune diseases can be initiated in immuno-compromised individuals during extensive pathogen infections, if these individuals have two immune problems: (a) slow or weak initial immune responses that result in a reliance on antibodies and (b) an inability to eliminate the resulting antigen-antibody immune complexes by phagocytosis. These two immune problems and the resulting immune system type III hypersensitivity reaction can explain the causation of several autoimmune diseases, including the most common and the rarest autoimmune diseases, both their differences and their similarities.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Immunocompromised Host/immunology , Immunologic Deficiency Syndromes/immunology , Infections/immunology , Antigen-Antibody Complex/immunology , Autoantibodies/immunology , Humans , Immune Complex Diseases/immunology , Immune Complex Diseases/pathology
6.
BMC Neurol ; 21(1): 278, 2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34256721

ABSTRACT

BACKGROUND: Idiopathic intracranial hypertension (IIH) is defined by an increased cerebrospinal fluid pressure in the absence of inflammation, structural obstructions, or mass lesions. Although the underlying pathogenesis of IIH is not fully understood, associations with specific risk factors as obesity, obstruction of cerebral venous sinuses, medications, endocrine or systemic conditions and chronic kidney disease have been described. Immune-complex glomerulonephritis as IgA-nephropathy is a frequent cause of chronic kidney failure, which was reported previously in one IIH patient. To date, there is no knowledge about the variable relation of immune-complex nephritis, kidney function and the course of IIH. CASE PRESENTATION: We report three cases (two females) of concurrent diagnosis of IIH and immune-complex glomerulonephritis. All patients presented with typical IIH symptoms of headache and visual disturbances. Two patients had been diagnosed with IgA-nephropathy only few weeks prior to IIH diagnosis. The third patient had been diagnosed earlier with terminal kidney failure due to a cryoglobulin glomerulonephritis. CONCLUSION: We propose a possible link between renal deposition of immune-complexes and increased cerebrospinal fluid pressure. Pathophysiological hypotheses and clinical implications are discussed. We recommend clinical awareness and further systematic research to obtain more information on the association of IIH and immune-complex glomerulonephritis.


Subject(s)
Glomerulonephritis/complications , Glomerulonephritis/diagnostic imaging , Immune Complex Diseases/complications , Immune Complex Diseases/diagnostic imaging , Pseudotumor Cerebri/complications , Pseudotumor Cerebri/diagnostic imaging , Adult , Cranial Sinuses/diagnostic imaging , Female , Headache/diagnostic imaging , Headache/etiology , Humans , Male , Obesity/complications , Obesity/diagnostic imaging , Risk Factors , Vision Disorders/diagnostic imaging , Vision Disorders/etiology , Young Adult
7.
Clin Immunol ; 217: 108487, 2020 08.
Article in English | MEDLINE | ID: mdl-32479986

ABSTRACT

Coronavirus Disease 2019 (COVID-19) is an ongoing public health emergency and new knowledge about its immunopathogenic mechanisms is deemed necessary in the attempt to reduce the death burden, globally. For the first time in worldwide literature, we provide scientific evidence that in COVID-19 vasculitis a life-threatening escalation from type 2 T-helper immune response (humoral immunity) to type 3 hypersensitivity (immune complex disease) takes place. The subsequent deposition of immune complexes inside the vascular walls is supposed to induce a severe inflammatory state and a cytokine release syndrome, whose interleukin-6 is the key myokine, from the smooth muscle cells of blood vessels.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/immunology , Cytokine Release Syndrome/immunology , Immune Complex Diseases/immunology , Pneumonia, Viral/immunology , Severe Acute Respiratory Syndrome/immunology , Th2 Cells/immunology , Vasculitis/immunology , Aged , Antibodies, Viral/biosynthesis , Antigen-Antibody Complex/biosynthesis , Betacoronavirus/immunology , Blood Vessels/immunology , Blood Vessels/pathology , Blood Vessels/virology , COVID-19 , Complement C3/biosynthesis , Coronavirus Infections/complications , Coronavirus Infections/virology , Cytokine Release Syndrome/complications , Cytokine Release Syndrome/virology , Disease Progression , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelial Cells/virology , Humans , Immune Complex Diseases/complications , Immune Complex Diseases/virology , Immunity, Humoral , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Interleukin-6/biosynthesis , Male , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/virology , SARS-CoV-2 , Severe Acute Respiratory Syndrome/complications , Severe Acute Respiratory Syndrome/virology , Th2 Cells/pathology , Th2 Cells/virology , Vasculitis/complications , Vasculitis/virology
8.
Trends Immunol ; 38(6): 383-394, 2017 06.
Article in English | MEDLINE | ID: mdl-28416449

ABSTRACT

Complement dysregulation underlies several inflammatory disorders, and terminal complement inhibition has thus far afforded significant clinical gains. Nonetheless, emerging pathologies, fueled by complement imbalance and therapy-skewing genetic variance, underscore the need for more comprehensive, disease-tailored interventions. Modulation at the level of C3, a multifaceted orchestrator of the complement cascade, opens up prospects for broader therapeutic efficacy by targeting multiple pathogenic pathways modulated by C3-triggered proinflammatory crosstalk. Notably, C3 intervention is emerging as a viable therapeutic strategy for renal disorders with predominantly complement-driven etiology, such as C3 glomerulopathy (C3G). Using C3G as a paradigm, we argue that concerns about the feasibility of long-term C3 intervention need to be placed into perspective and weighed against actual therapeutic outcomes in prospective clinical trials.


Subject(s)
Complement C3/metabolism , Glomerulonephritis, Membranous/drug therapy , Immune Complex Diseases/drug therapy , Inflammation/drug therapy , Animals , Antibodies, Blocking/therapeutic use , Clinical Trials as Topic , Complement Activation , Complement C3/immunology , Evidence-Based Medicine , Glomerulonephritis, Membranous/immunology , Humans , Immune Complex Diseases/immunology , Inflammation/immunology , Models, Immunological , Molecular Targeted Therapy
9.
Clin Nephrol ; 94(2): 97-102, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32567542

ABSTRACT

Staphylococcus infection-associated glomerulonephritis (SAGN) is characterized by the presence of IgA and C3 as the predominant components present in the glomerular immune deposits. Glomerulonephritis frequently resolves after effective treatment of the staphylococcal infection. However, there have been few studies of repeat kidney biopsy after resolution of glomerulonephritis. We present a combined kidney-pancreas transplant patient who developed SAGN due to Staphylococcus aureus bacteremia from an old infected arteriovenous (AV) graft, which occurred after a long period of stable allograft function. Clinical improvement occurred following surgical debridement and appropriate antibiotics with subsequent clearance of bacteremia. However, 3 weeks later he presented with severe acute kidney injury related to rapidly progressive glomerulonephritis. Renal allograft biopsy revealed immune complex glomerulonephritis with predominance of IgA and C3 in subendothelial and mesangial deposits, consistent with SAGN. There was no evidence for recurrent staphylococcal infection. High-dose steroid therapy was followed by resolution of hematuria and improvement in allograft function with gradual return of serum creatinine concentration to near baseline levels. However, 1 year after the diagnosis of SAGN, he developed gradually worsening allograft function with persistent proteinuria. Repeat allograft biopsy showed sclerosing glomerular changes and extensive interstitial fibrosis and tubular atrophy. There was complete resolution of proliferative changes, and IgA and C3 deposits were no longer detectable. Despite transient allograft function stabilization, the patient progressed to end-stage renal disease (ESRD), and maintenance hemodialysis was reinitiated 2.5 years after the diagnosis of SAGN. Pancreatic allograft function remained normal.


Subject(s)
Complement C3/analysis , Glomerulonephritis , Immunoglobulin A/blood , Kidney Transplantation , Staphylococcal Infections , Bacteremia , Humans , Immune Complex Diseases , Male
10.
Semin Immunol ; 28(3): 208-22, 2016 06.
Article in English | MEDLINE | ID: mdl-27321574

ABSTRACT

The complement system is a powerful effector arm of innate immunity that typically confers protection from microbial intruders and accumulating debris. In many clinical situations, however, the defensive functions of complement can turn against host cells and induce or exacerbate immune, inflammatory, and degenerative conditions. Although the value of inhibiting complement in a therapeutic context has long been recognized, bringing complement-targeted drugs into clinical use has proved challenging. This important milestone was finally reached a decade ago, yet the clinical availability of complement inhibitors has remained limited. Still, the positive long-term experience with complement drugs and their proven effectiveness in various diseases has reinvigorated interest and confidence in this approach. Indeed, a broad variety of clinical candidates that act at almost any level of the complement activation cascade are currently in clinical development, with several of them being evaluated in phase 2 and phase 3 trials. With antibody-related drugs dominating the panel of clinical candidates, the emergence of novel small-molecule, peptide, protein, and oligonucleotide-based inhibitors offers new options for drug targeting and administration. Whereas all the currently approved and many of the proposed indications for complement-targeted inhibitors belong to the rare disease spectrum, these drugs are increasingly being evaluated for more prevalent conditions. Fortunately, the growing experience from preclinical and clinical use of therapeutic complement inhibitors has enabled a more evidence-based assessment of suitable targets and rewarding indications as well as related technical and safety considerations. This review highlights recent concepts and developments in complement-targeted drug discovery, provides an overview of current and emerging treatment options, and discusses the new milestones ahead on the way to the next generation of clinically available complement therapeutics.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Complement Activation , Complement System Proteins/immunology , Immune Complex Diseases/therapy , Molecular Targeted Therapy , Animals , Clinical Trials as Topic , Drug Discovery , Drug Evaluation, Preclinical , Evidence-Based Medicine , Humans , Immune Complex Diseases/immunology , Immunity, Innate
11.
Inflammopharmacology ; 28(5): 1219-1222, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32638151

ABSTRACT

SARS-CoV-2, a new virus that appeared in Wuhan, China, in 2019 has approximately an 80% genomic match to the Severe Acute Respiratory Symptom (SARS) virus, which is known to come from a bat virus. Symptoms of Kawasaki disease in general and incomplete Kawasaki disease have been seen in a subset of pediatric patients having a current or previous infection of SARS-CoV-2. A viral infection, such as a SARS-CoV-2 virus infection, could result in extensive antigen-antibody immune complexes that cannot be quickly cleared in a subset of patients and thus create a type III hypersensitivity immune reaction and cause Kawasaki disease or Kawasaki disease symptoms (also known as multisystem inflammatory syndrome) in a subset of patients. Extensive binding of antibodies to viral antigens can create antigen-antibody immune complexes, which, if not eliminated in certain individuals having dysfunctional complement systems, can start inflammatory type III hypersensitivity symptoms, including protease releases that can disrupt epithelium, mesothelium, and endothelium basement membranes, and induce pervasive inflammation throughout the body. This could continue after SARS-CoV-2 infections end if the first wave of protease attacks on basement membranes created new secondary autoantibodies and new uncleared antigen-antibody immune complexes.


Subject(s)
Coronavirus Infections/complications , Coronavirus Infections/virology , Mucocutaneous Lymph Node Syndrome/etiology , Mucocutaneous Lymph Node Syndrome/virology , Pneumonia, Viral/complications , Pneumonia, Viral/virology , Antigen-Antibody Complex , Basement Membrane/immunology , Basement Membrane/pathology , COVID-19 , Child , Humans , Immune Complex Diseases/immunology , Mucocutaneous Lymph Node Syndrome/diagnosis , Pandemics , Peptide Hydrolases/chemistry , Skin/pathology , Systemic Inflammatory Response Syndrome/therapy
12.
Immunol Rev ; 269(1): 175-93, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26683153

ABSTRACT

Mac-1 (CD11b/CD18) is a ß2 integrin classically regarded as a pro-inflammatory molecule because of its ability to promote phagocyte cytotoxic functions and enhance the function of several effector molecules such as FcγR, uPAR, and CD14. Nevertheless, recent reports have revealed that Mac-1 also plays significant immunoregulatory roles, and genetic variants in ITGAM, the gene that encodes CD11b, confer risk for the autoimmune disease systemic lupus erythematosus (SLE). This has renewed interest in the physiological roles of this integrin and raised new questions on how its seemingly opposing biological functions may be regulated. Here, we provide an overview of the CD18 integrins and how their activation may be regulated as this may shed light on how the opposing roles of Mac-1 may be elicited. We then discuss studies that exemplify Mac-1's pro-inflammatory versus regulatory roles particularly in the context of IgG immune complex-mediated inflammation. This includes a detailed examination of molecular mechanisms that could explain the risk-conferring effect of rs1143679, a single nucleotide non-synonymous Mac-1 polymorphism associated with SLE.


Subject(s)
CD11b Antigen/metabolism , Immune Complex Diseases/immunology , Lupus Erythematosus, Systemic/immunology , Animals , CD11b Antigen/genetics , Genetic Predisposition to Disease , Humans , Immunomodulation , Phagocytosis , Polymorphism, Genetic , Risk
13.
BMC Vet Res ; 15(1): 303, 2019 Aug 20.
Article in English | MEDLINE | ID: mdl-31429743

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) has typically a non-immune mediated origin in cats and immune-complex glomerulonephritis (ICGN) is scarcely described. Aims of this study were to characterize ICGN by light and electron microscopy and identify associations with clinico-pathological findings. In addition, comparisons between cats with ICGN and non immune-complex glomerulonephritis (non-ICGN) were performed. Renal samples examined between 2010 and 2019 were considered if both light and electron microscopy were performed. Signalment, feline immunodeficiency virus (FIV) and leukemia virus (FeLV) status, serum creatinine concentration, urine protein-to-creatinine (UPC) ratio, systolic blood pressure (SBP) and International Renal Interest Society (IRIS) stage were retrieved and used for comparisons. RESULTS: Sixty-eight client-owned cats were included. Thirty-seven cats (54.4%) had ICGN and 31 (45.6%) non-ICGN. Eighteen (48.6%) with ICGN had membranous glomerulonephropathy (MGN), 14 (37.8%) membranoproliferative glomerulonephritis (MPGN), and 5 (13.5%) mesangioproliferative glomerulonephritis (MeGN). Clinico-pathological data were not associated with any type of ICGN. Among cats with non-ICGN, 11 (35.5%) had end-stage CKD, 9 (29%) focal segmental glomerulosclerosis, 6 (19.4%) global and multifocal mesangiosclerosis, 2 (6.5%) glomerular atrophy, 2 (6.5%) renal dysplasia and 1 (3.1%) amyloidosis. Eight (25.8%) cats with non-ICGN had chronic interstitial nephritis (CIN) grade 1, 13 (41.9%) grade 2 and 10 (32.3%) grade 3; creatinine and UPC ratio increased with CIN grades (p = 0.001, p < 0.001). Cats with ICGN were more frequently FIV or FeLV-infected (OR:11.4; 95%CI:1.4-94.4; p = 0.024), had higher UPC ratio (OR:6.8; 95%CI:2.5-18.2; p < 0.001) and were younger (OR:0.9; 95%CI:0.7-1.0; p = 0.042) than cats with non-ICGN. CONCLUSIONS: MGN and MPGN were the most common morphological diagnoses of ICGN in cats. Unfortunately, none of the investigated findings differentiated ICGN morphological diagnoses. Serum creatinine concentration and UPC ratio were directly associated with grades of CIN (p = 0.001 and p < 0.001, respectively), confirming previous literature. More ICGN than non-ICGN was observed in cats with retroviral infections, younger cats and higher UPC ratio.


Subject(s)
Cat Diseases/pathology , Glomerulonephritis/veterinary , Immune Complex Diseases/veterinary , Animals , Cats , Female , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Immune Complex Diseases/pathology , Kidney/pathology , Male , Retrospective Studies
14.
J Am Soc Nephrol ; 29(1): 283-294, 2018 01.
Article in English | MEDLINE | ID: mdl-29030465

ABSTRACT

Membranoproliferative GN (MPGN) was recently reclassified as alternative pathway complement-mediated C3 glomerulopathy (C3G) and immune complex-mediated membranoproliferative GN (IC-MPGN). However, genetic and acquired alternative pathway abnormalities are also observed in IC-MPGN. Here, we explored the presence of distinct disease entities characterized by specific pathophysiologic mechanisms. We performed unsupervised hierarchical clustering, a data-driven statistical approach, on histologic, genetic, and clinical data and data regarding serum/plasma complement parameters from 173 patients with C3G/IC-MPGN. This approach divided patients into four clusters, indicating the existence of four different pathogenetic patterns. Specifically, this analysis separated patients with fluid-phase complement activation (clusters 1-3) who had low serum C3 levels and a high prevalence of genetic and acquired alternative pathway abnormalities from patients with solid-phase complement activation (cluster 4) who had normal or mildly altered serum C3, late disease onset, and poor renal survival. In patients with fluid-phase complement activation, those in clusters 1 and 2 had massive activation of the alternative pathway, including activation of the terminal pathway, and the highest prevalence of subendothelial deposits, but those in cluster 2 had additional activation of the classic pathway and the highest prevalence of nephrotic syndrome at disease onset. Patients in cluster 3 had prevalent activation of C3 convertase and highly electron-dense intramembranous deposits. In addition, we provide a simple algorithm to assign patients with C3G/IC-MPGN to specific clusters. These distinct clusters may facilitate clarification of disease etiology, improve risk assessment for ESRD, and pave the way for personalized treatment.


Subject(s)
Complement Activation , Complement C3 Nephritic Factor/metabolism , Complement C3/metabolism , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/immunology , Immune Complex Diseases/complications , Adolescent , Adult , Algorithms , Child , Child, Preschool , Cluster Analysis , Complement C3-C5 Convertases/metabolism , Female , Glomerulonephritis, Membranoproliferative/blood , Humans , Immune Complex Diseases/blood , Male , Nephrotic Syndrome/immunology , Young Adult
15.
Kidney Int ; 93(1): 110-127, 2018 01.
Article in English | MEDLINE | ID: mdl-28754552

ABSTRACT

Ubiquitin C-terminal hydrolase L1 (UCH-L1) is a major deubiquitinating enzyme of the nervous system and associated with the development of neurodegenerative diseases. We have previously shown that UCH-L1 is found in tubular and parietal cells of the kidney and is expressed de novo in injured podocytes. Since the role of UCH-L1 in the kidney is unknown we generated mice with a constitutive UCH-L1-deficiency to determine its role in renal health and disease. UCH-L1-deficient mice developed proteinuria, without gross changes in glomerular morphology. Tubular cells, endothelial cells, and podocytes showed signs of stress with an accumulation of oxidative-modified and polyubiquitinated proteins. Mechanistically, abnormal protein accumulation resulted from an altered proteasome abundance leading to decreased proteasomal activity, a finding exaggerated after induction of anti-podocyte nephritis. UCH-L1-deficient mice exhibited an exacerbated course of disease with increased tubulointerstitial and glomerular damage, acute renal failure, and death, the latter most likely a result of general neurologic impairment. Thus, UCH-L1 is required for regulated protein degradation in the kidney by controlling proteasome abundance. Altered proteasome abundance renders renal cells, particularly podocytes and endothelial cells, susceptible to injury.


Subject(s)
Glomerulonephritis/enzymology , Immune Complex Diseases/enzymology , Podocytes/enzymology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Glomerulonephritis/genetics , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Hypotension/enzymology , Hypotension/genetics , Immune Complex Diseases/genetics , Immune Complex Diseases/immunology , Immune Complex Diseases/pathology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Knockout , Oxidation-Reduction , Podocytes/immunology , Podocytes/pathology , Proteinuria/enzymology , Proteinuria/genetics , Proteolysis , Ubiquitin Thiolesterase/deficiency , Ubiquitin Thiolesterase/genetics , Ubiquitination
16.
Kidney Int ; 93(4): 826-841, 2018 04.
Article in English | MEDLINE | ID: mdl-29395335

ABSTRACT

The atypical chemokine receptor 2 (ACKR2), also named D6, regulates local levels of inflammatory chemokines by internalization and degradation. To explore potential anti-inflammatory functions of ACKR2 in glomerulonephritis, we induced autologous nephrotoxic nephritis in C57/BL6 wild-type and Ackr2-deficient mice. Renal ACKR2 expression increased and localized to interstitial lymphatic endothelium during nephritis. At two weeks Ackr2-/-mice developed increased albuminuria and urea levels compared to wild-type mice. Histological analysis revealed increased structural damage in the glomerular and tubulointerstitial compartments within Ackr2-/- kidneys. This correlated with excessive renal leukocyte infiltration of CD4+ T cells and mononuclear phagocytes with increased numbers in the tubulointerstitium but not glomeruli in knockout mice. Expression of inflammatory mediators and especially markers of fibrotic tissue remodeling were increased along with higher levels of ACKR2 inflammatory chemokine ligands like CCL2 in nephritic Ackr2-/- kidneys. In vitro, Ackr2 deficiency in TNF-stimulated tubulointerstitial tissue but not glomeruli increased chemokine levels. These results are in line with ACKR2 expression in interstitial lymphatic endothelial cells, which also assures efflux of activated leukocytes into regional lymph nodes. Consistently, nephritic Ackr2-/- mice showed reduced adaptive cellular immune responses indicated by decreased regional T-cell activation. However, this did not prevent aggravated injury in the kidneys of Ackr2-/- mice with nephrotoxic nephritis due to simultaneously increased tubulointerstitial chemokine levels, leukocyte infiltration and fibrosis. Thus, ACKR2 is important in limiting renal inflammation and fibrotic remodeling in progressive nephrotoxic nephritis. Hence, ACKR2 may be a potential target for therapeutic interventions in immune complex glomerulonephritis.


Subject(s)
Glomerulonephritis/prevention & control , Immune Complex Diseases/prevention & control , Kidney Glomerulus/metabolism , Kidney Tubules/metabolism , Receptors, Chemokine/metabolism , Adaptive Immunity , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chemotaxis, Leukocyte , Disease Models, Animal , Disease Progression , Fibrosis , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Immune Complex Diseases/immunology , Immune Complex Diseases/metabolism , Immune Complex Diseases/pathology , Inflammation Mediators/metabolism , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Kidney Tubules/immunology , Kidney Tubules/pathology , Lymphocyte Activation , Male , Mice, Inbred C57BL , Mice, Knockout , Mononuclear Phagocyte System/immunology , Mononuclear Phagocyte System/metabolism , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Signal Transduction
17.
Toxicol Pathol ; 46(8): 1013-1019, 2018 12.
Article in English | MEDLINE | ID: mdl-30157708

ABSTRACT

This article summarizes a continuing education presentation on immunogenicity that was part of a continuing education course entitled, "Clinical Pathology of Biotherapeutics." Immunogenicity of a biotherapeutic can have diverse impacts including altered systemic exposure and pharmacologic responses and, in a fraction of the cases, safety concerns including cross-reactive neutralization of endogenous proteins or sequela related to immune complex disease (ICD). In most cases, immune complexes are readily cleared from circulation; however, based on physiochemical properties, insoluble complexes form, activate complement, and deposit in tissues. Using published information and personal experience, a set of repeat-dose monkey toxicity studies with manifestations suggestive of ICD was reviewed to summarize the spectrum of clinical and pathology findings. The most common live-phase observation linked to ICD was an acute postdosing reaction following multiple dose administrations characterized by generalized collapse and attributed to acute complement activation. Less common live-phase observations were related to syndromes such as a consumptive coagulopathy or a protein losing nephropathy. The most common histologic change attributed to ICD was multi-organ vascular/perivascular inflammation followed by glomerulonephritis. The presentation concluded with a description of the challenges in assessing the relevance of immunogenicity-related reaction in monkey to human clinical use.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/immunology , Immune Complex Diseases/chemically induced , Immune Complex Diseases/immunology , Animals , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Humans , Toxicology
18.
Toxicol Pathol ; 46(8): 904-917, 2018 12.
Article in English | MEDLINE | ID: mdl-30089413

ABSTRACT

Prevalence of immune-mediated glomerulonephritis has increased in preclinical toxicity studies, with more frequent use of biotherapeutic agents (especially antigenic humanized molecules) and antisense oligonucleotide (ASO) therapies. Immune complex disease affects a small number of study monkeys, often correlates with antidrug antibody (ADA) titers, and occurs at a dose that favors immune complex formation or impedes clearance. While preclinical glomerulonephritis often fails to correlate with evidence of glomerular or vascular injury in human clinical trials and is not considered predictive, additional animal investigative immunohistochemical work may be performed to substantiate evidence for immune complex pathogenesis. While ADA is most commonly encountered as a predisposing factor with biotherapeutic agents, complement activation may occur without circulating complexes, and other mechanisms of non-ADA immune-mediated glomerulonephritis have been observed including nonendogenous immune aggregates and immunoregulatory pharmacology. Although glomerulonephritis associated with oligonucleotide therapies has been noted occasionally in preclinical studies and more rarely with human patients, pathophysiologic mechanisms involved appear to be different between species and preclinical cases are not considered predictive for humans. ADA is not involved in oligonucleotide-associated cases, and complement fixation plays a more important role in monkeys. Recent screening of ASOs for proinflammatory activity appears to have decreased glomerulonephritis incidence preclinically.


Subject(s)
Biological Therapy/adverse effects , Glomerulonephritis/chemically induced , Oligonucleotides, Antisense/toxicity , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Immune Complex Diseases/chemically induced
19.
Clin Nephrol ; 90(5): 357-362, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30021694

ABSTRACT

A 69-year-old man presented with acute kidney injury, hypocomplementemia, antinuclear antibody, and anti-dsDNA antibody. He had no signs of systemic lupus erythematosus or Sjögren syndrome. He had not begun taking any new drugs in the preceding 6 months. Kidney biopsy revealed 13 glomeruli, 3 with global sclerosis. The remaining glomeruli showed slight mesangial proliferation. The interstitial inflammation was extensive, comprising mainly mature lymphocytes and plasma cells, neutrophils, and a few eosinophils. Remarkable granular and diffuse deposition of IgG and C1q was observed along the tubular basement membranes. Electron microscopy showed electron-dense deposits in the tubular basement membrane. Immunohistochemistry showed only 1 - 4 IgG4-positive plasma cells per high-power field and an IgG4/CD138 ratio of ~ 10%. He was treated with oral prednisolone 35 mg/day, and his kidney function gradually improved. This is a unique case that is not consistent with any known disease entities with immune complex-mediated tubulointerstitial nephritis.
.


Subject(s)
Glomerulonephritis, Membranoproliferative , Immune Complex Diseases , Nephritis, Interstitial , Neutrophils/immunology , Aged , Humans , Male
20.
Blood ; 126(8): 1017-26, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26036804

ABSTRACT

Platelets protect vascular integrity during inflammation. Recent evidence suggests that this action is independent of thrombus formation and requires the engagement of glycoprotein VI (GPVI), but it remains unclear how platelets prevent inflammatory bleeding. We investigated whether platelets and GPVI act primarily by preventing detrimental effects of neutrophils using models of immune complex (IC)-mediated inflammation in mice immunodepleted in platelets and/or neutrophils or deficient in GPVI. Depletion of neutrophils prevented bleeding in thrombocytopenic and GPVI(-/-) mice during IC-mediated dermatitis. GPVI deficiency did not modify neutrophil recruitment, which was reduced by thrombocytopenia. Neutrophil cytotoxic activities were reduced in thrombocytopenic and GPVI(-/-) mice during IC-mediated inflammation. Intravital microscopy revealed that in this setting, intravascular binding sites for platelets were exposed by neutrophils, and GPVI supported the recruitment of individual platelets to these spots. Furthermore, the platelet secretory response accompanying IC-mediated inflammation was partly mediated by GPVI, and blocking of GPVI signaling impaired the vasculoprotective action of platelets. Together, our results show that GPVI plays a dual role in inflammation by enhancing neutrophil-damaging activities while supporting the activation and hemostatic adhesion of single platelets to neutrophil-induced vascular breaches.


Subject(s)
Blood Platelets/metabolism , Immune Complex Diseases/pathology , Inflammation/pathology , Neutrophils/pathology , Platelet Membrane Glycoproteins/metabolism , Animals , Disease Models, Animal , Immune Complex Diseases/complications , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL