Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Cell Rep ; 31(12): 107798, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32579925

ABSTRACT

Multivalent complexes of endothelial adhesion receptors (e.g., selectins) engage leukocytes to orchestrate their migration to inflamed tissues. Proper anchorage and sufficient density (clustering) of endothelial receptors are required for efficient leukocyte capture and rolling. We demonstrate that a polarized spectrin network dictates the stability of the endothelial cytoskeleton, which is attached to the apical membrane, at least in part, by the abundant transmembrane protein CD44. Single-particle tracking revealed that CD44 undergoes prolonged periods of immobilization as it tethers to the cytoskeleton. The CD44-spectrin "picket fence" alters the behavior of bystander molecules-notably, selectins-curtailing their mobility, inducing their apical accumulation, and favoring their clustering within caveolae. Accordingly, depletion of either spectrin or CD44 virtually eliminated leukocyte rolling and adhesion to the endothelium. Our results indicate that a unique spectrin-based apical cytoskeleton tethered to transmembrane pickets-notably, CD44-is essential for proper extravasation of leukocytes in response to inflammation.


Subject(s)
Cytoskeleton/metabolism , Endothelial Cells/metabolism , Hyaluronan Receptors/metabolism , Leukocyte Rolling , Spectrin/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Caveolae/metabolism , Cell Adhesion , Cell Membrane/metabolism , Cell Polarity , Diffusion , Glycocalyx/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immobilized Proteins/metabolism , Neutrophils , Protein Stability , Selectins/metabolism , Single Molecule Imaging
2.
Exp Clin Transplant ; 17(4): 429-434, 2019 08.
Article in English | MEDLINE | ID: mdl-31050613

ABSTRACT

OBJECTIVES: Vaccine-preventable diseases remain a major cause of morbidity and mortality in solid-organ transplant candidates and recipients. Newer recommendations include vaccination of all household members to create a herd immunity around the transplant recipient. This study evaluated the vaccination status of pediatric solid-organ transplant recipients and their household members. MATERIALS AND METHODS: We evaluated 30 pediatric solid-organ transplant recipients (14 kidney, 13 liver, 3 heart) and their household members (26 siblings, 30 parents) at time of transplant. RESULTS: Fourteen recipients (47%) received scheduled vaccinations before solid-organ transplant and were up to date for their age with their diphtheria, tetanus, pertussis; hepatitis B virus; poliomyelitis; Haemophilus influenzae type B; Streptococcus pneumoniae conjugate vaccine; and measles, mumps, and rubella vaccinations. Another 7 recipients (23%) had partially completed their schedules, only missing the second dose of the measles, mumps, and rubella vaccine. Fifteen siblings (58%) had either completed (n = 13, 50%) or partially completed (n = 2, 8%) their vaccinations. All 30 parents were either unaware of their vaccination status (n = 10, 33%) or had only incomplete vaccination records (n = 20, 67%). CONCLUSIONS: We found that most pediatric solid-organ transplant recipients to be appropriately vaccinated. However, vaccination status in household members, especially in parents, was disappointing.


Subject(s)
Infection Control/methods , Organ Transplantation/adverse effects , Parents , Siblings , Vaccination , Vaccine-Preventable Diseases/prevention & control , Adolescent , Adult , Child , Child, Preschool , Humans , Immunity, Herd , Immunization Schedule , Immunocompromised Host , Immunosuppressive Agents/adverse effects , Infant , Male , Middle Aged , Organ Transplantation/mortality , Protective Factors , Retrospective Studies , Risk Factors , Vaccine-Preventable Diseases/immunology , Vaccine-Preventable Diseases/mortality , Vaccine-Preventable Diseases/transmission , Young Adult
3.
Pediatr Transplant ; 23(4): e13407, 2019 06.
Article in English | MEDLINE | ID: mdl-30973671

ABSTRACT

OBJECTIVES: MMA is associated with chronic tubulointerstitial nephritis and a progressive decline in GFR. Optimal management of these children is uncertain. Our objectives were to document the pre-, peri-, and post-transplant course of all children with MMA who underwent liver or combined liver-kidney transplant in our centers. DESIGN AND METHODS: Retrospective chart review of all cases of MMA who underwent organ transplantation over the last 10 years. RESULTS: Five children with MMA underwent liver transplant (4/5) and combined liver-kidney transplant (1/5). Three were Mut0 and two had a cobalamin B disorder. Four of five were transplanted between ages 3 and 5 years. Renal dysfunction prior to transplant was seen in 2/5 patients. Post-transplant (one liver transplant and one combined transplant) renal function improved slightly when using creatinine-based GFR formula. We noticed in 2 patients a big discrepancy between creatinine- and cystatin C-based GFR calculations. One patient with no renal disease developed renal failure post-liver transplantation. Serum MMA levels have decreased in all to <300 µmol/L. Four patients remain on low protein diet, carnitine, coenzyme Q, and vitamin E post-transplant. CONCLUSIONS: MMA is a complex metabolic disorder. Renal disease can continue to progress post-liver transplant and close follow-up is warranted. More research is needed to clarify best screening GFR method in patients with MMA. Whether liver transplant alone, continued protein restriction, or the addition of antioxidants post-transplant can halt the progression of renal disease remains unclear.


Subject(s)
Amino Acid Metabolism, Inborn Errors/complications , Amino Acid Metabolism, Inborn Errors/surgery , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/surgery , Kidney Transplantation , Liver Transplantation , Carnitine/administration & dosage , Child , Child, Preschool , Creatinine/blood , Cystatin C/blood , Disease Progression , Female , Glomerular Filtration Rate , Humans , Infant , Infant, Newborn , Male , Nephritis, Interstitial/complications , Nephritis, Interstitial/surgery , Postoperative Complications , Renal Dialysis , Retrospective Studies , Ubiquinone/administration & dosage , Vitamin B 12/genetics , Vitamin E/administration & dosage
4.
Mol Immunol ; 97: 71-81, 2018 05.
Article in English | MEDLINE | ID: mdl-29571059

ABSTRACT

Neutrophil extracellular traps (NETs) are web-like DNA structures released by activated neutrophils. These structures are decorated with antimicrobial proteins, and considered to trap and kill bacteria extracellularly. However, the exact functions of NETs remain elusive, and contradictory observations have been made with NETs functioning as an antimicrobial or a pathogentrapping mechanism. There is a disconnect in the interpretation of the involvement of other major immune mechanisms, such as the complement system, as effectors of the function of NETs. We have recently shown that NETs activate complement. In this study, we aimed to elucidate the relative antimicrobial roles of NETs in the absence and presence of complement. Using primary human neutrophils, human serum (normal, heat inactivated, and C5-depleted), P. aeruginosa (at multiplicity of infection, MOI, of 1 or 10), S. aureus (MOI of 1), colony-counting assays and confocal microscopy, we demonstrate that most bacteria trapped by NETs remain viable, indicating that NETs have limited bactericidal properties. By contrast, complement effectively killed bacteria, but NETs decreased the bactericidal ability of complement and degrading NETs by DNases restored complement-mediated killing. Experiments with conditions allowing for specific pathway activation showed that the complement classical and lectin, but not the alternative, pathway lead to bacterial killing. NETs under static conditions showed limited killing of bacteria while NETs under dynamic conditions showed enhanced bacteria trapping and reduced killing. Furthermore, NETs incubated with normal human serum depleted complement and reduced the hemolytic capacity of the serum. This report, for the first time, clarifies the relative bactericidal contributions of NETs and complement. We propose that - while NETs can ensnare bacteria such as P. aeruginosa - complement is necessary for efficient bacterial killing.


Subject(s)
Anti-Bacterial Agents , Bacteria/immunology , Complement System Proteins/physiology , Extracellular Traps/physiology , Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/metabolism , Cells, Cultured , Hemolysis , Humans , Neutrophil Activation/physiology , Pseudomonas aeruginosa/immunology , Staphylococcus aureus/immunology
5.
Cell ; 172(1-2): 305-317.e10, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29328918

ABSTRACT

Phagocytic receptors must diffuse laterally to become activated upon clustering by multivalent targets. Receptor diffusion, however, can be obstructed by transmembrane proteins ("pickets") that are immobilized by interacting with the cortical cytoskeleton. The molecular identity of these pickets and their role in phagocytosis have not been defined. We used single-molecule tracking to study the interaction between Fcγ receptors and CD44, an abundant transmembrane protein capable of indirect association with F-actin, hence likely to serve as a picket. CD44 tethers reversibly to formin-induced actin filaments, curtailing receptor diffusion. Such linear filaments predominate in the trailing end of polarized macrophages, where receptor mobility was minimal. Conversely, receptors were most mobile at the leading edge, where Arp2/3-driven actin branching predominates. CD44 binds hyaluronan, anchoring a pericellular coat that also limits receptor displacement and obstructs access to phagocytic targets. Force must be applied to traverse the pericellular barrier, enabling receptors to engage their targets.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Membrane/metabolism , Hyaluronan Receptors/metabolism , Receptors, Immunologic/metabolism , Adult , Animals , Binding Sites , COS Cells , Cells, Cultured , Chlorocebus aethiops , Female , Humans , Hyaluronan Receptors/chemistry , Hyaluronan Receptors/genetics , Hyaluronic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Binding
6.
Pediatr Nephrol ; 33(8): 1297-1307, 2018 08.
Article in English | MEDLINE | ID: mdl-28748411

ABSTRACT

Thrombotic microangiopathy (TMA) is caused by thrombus formation in the microvasculature. The disease spectrum of TMA includes, amongst others, thrombotic thrombocytopenic purpura (TTP) and atypical haemolytic uraemic syndrome (aHUS). TTP is caused by defective cleavage of von Willebrand factor (VWF), whereas aHUS is caused by overshooting complement activation and subsequent endothelial cell (EC) injury. Despite their distinct pathophysiology, the clinical manifestation of TTP and aHUS consisting of microangiopathic haemolytic anaemia and thrombocytopenia is often similar and difficult to distinguish. Recent evidence hints at both a genetic and functional link between TTP and aHUS, especially between VWF and the complement system. There is novel in vitro evidence that complement activation not only results in VWF release from ECs, but that VWF also functions as a negative complement regulator, thus protecting the EC surface from ongoing complement attack. Although contrary to previous experimental work suggesting that complement can be activated on VWF multimers, there may be an explanation in vivo that rationalizes these apparently contradictory findings, whereby a system primarily meant to regulate becomes overwhelmed or pathologic in the disease state. The importance of unravelling these recent findings for our understanding of TMA pathology becomes even more evident considering that glomerular ECs express VWF in a heterogeneous pattern with an overall decreased expression level, thus potentially leaving the glomerular ECs vulnerable to complement-mediated injury. Taken together, these findings support the concept that TTP and aHUS represent two extreme ends of a TMA disease spectrum rather than isolated disease entities.


Subject(s)
Atypical Hemolytic Uremic Syndrome/pathology , Kidney Glomerulus/pathology , Microvessels/pathology , Purpura, Thrombotic Thrombocytopenic/pathology , von Willebrand Factor/metabolism , ADAMTS13 Protein/genetics , ADAMTS13 Protein/immunology , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/immunology , Blood Coagulation/immunology , Complement Activation/immunology , Endothelial Cells/immunology , Endothelial Cells/pathology , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/immunology , Microvessels/cytology , Microvessels/immunology , Mutation , Purpura, Thrombotic Thrombocytopenic/genetics , Purpura, Thrombotic Thrombocytopenic/immunology , von Willebrand Factor/immunology
7.
Kidney Int Rep ; 2(1): 66-75, 2017 Jan.
Article in English | MEDLINE | ID: mdl-29142942

ABSTRACT

INTRODUCTION: Atypical hemolytic uremic syndrome is a thrombotic microangiopathy, which is linked to hereditary or autoimmune defects in complement activators or regulators present in blood and on vascular endothelial cells. Acute thrombotic microangiopathy episodes are typically preceded by infections, which by themselves would not be expected to manifest HUS. Thus, it is possible that the host immune response contributes to the precipitation of aHUS. However, the mechanisms involved are not fully understood. We hypothesized that neutrophils trigger aHUS via initiating platelet aggregate formation on complement-activated endothelial cells. METHODS: We investigated neutrophil adhesion to complement-activated endothelial cells under static and flow conditions in vitro and ex vivo. RESULTS: Our results show that complement activation on endothelial cells promotes neutrophil adhesion, which is significantly reduced when the complement terminal pathway is blocked. When neutrophils and platelets are perfused simultaneously, neutrophils adhering to endothelial cells also induce the formation of platelet-neutrophil aggregates on these cells. Sera from patients with aHUS recapitulated these results. DISCUSSION: Therefore, our findings of (i) neutrophils adhering to complement-activated endothelial cells, (ii) the formation of neutrophil-platelet aggregates on endothelial cells, and (iii) the ability of aHUS serum to induce similar effects identify a possible role for neutrophils in aHUS manifestation.

8.
Clin J Am Soc Nephrol ; 12(12): 1974-1983, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29146700

ABSTRACT

BACKGROUND AND OBJECTIVES: Genetic heterogeneity and phenotypic variability are major challenges in familial nephronophthisis and related ciliopathies. To date, mutations in 20 different genes (NPHP1 to -20) have been identified causing either isolated kidney disease or complex multiorgan disorders. In this study, we provide a comprehensive and detailed characterization of 152 children with a special focus on extrarenal organ involvement and the long-term development of ESRD. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: We established an online-based registry (www.nephreg.de) to assess the clinical course of patients with nephronophthisis and related ciliopathies on a yearly base. Cross-sectional and longitudinal data were collected. Mean observation time was 7.5±6.1 years. RESULTS: In total, 51% of the children presented with isolated nephronophthisis, whereas the other 49% exhibited related ciliopathies. Monogenetic defects were identified in 97 of 152 patients, 89 affecting NPHP genes. Eight patients carried mutations in other genes related to cystic kidney diseases. A homozygous NPHP1 deletion was, by far, the most frequent genetic defect (n=60). We observed a high prevalence of extrarenal manifestations (23% [14 of 60] for the NPHP1 group and 66% [61 of 92] for children without NPHP1). A homozygous NPHP1 deletion not only led to juvenile nephronophthisis but also was able to present as a predominantly neurologic phenotype. However, irrespective of the initial clinical presentation, the kidney function of all patients carrying NPHP1 mutations declined rapidly between the ages of 8 and 16 years, with ESRD at a mean age of 11.4±2.4 years. In contrast within the non-NPHP1 group, there was no uniform pattern regarding the development of ESRD comprising patients with early onset and others preserving normal kidney function until adulthood. CONCLUSIONS: Mutations in NPHP genes cause a wide range of ciliopathies with multiorgan involvement and different clinical outcomes.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Ciliopathies/genetics , Kidney Diseases, Cystic/congenital , Kidney Failure, Chronic/genetics , Membrane Proteins/genetics , Phenotype , Adolescent , Anemia/genetics , Antigens, Neoplasm/genetics , Calmodulin-Binding Proteins/genetics , Carrier Proteins/genetics , Cell Cycle Proteins , Child , Ciliopathies/complications , Cross-Sectional Studies , Cytoskeletal Proteins , Female , Glomerular Filtration Rate/genetics , Homozygote , Humans , Kidney/diagnostic imaging , Kidney Diseases, Cystic/complications , Kidney Diseases, Cystic/diagnostic imaging , Kidney Diseases, Cystic/genetics , Kidney Failure, Chronic/physiopathology , Kinesins/genetics , Longitudinal Studies , Male , Neoplasm Proteins/genetics , Nervous System Diseases/genetics , Polyuria/genetics , Proteins/genetics , Ultrasonography , Young Adult
9.
J Clin Apher ; 32(6): 494-500, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28485072

ABSTRACT

BACKGROUND: Therapeutic plasma exchange (TPE) has evolved to an accepted therapy for selected indications. However, it is technically challenging in children. Moreover, data on safety and efficacy are mainly derived from adult series. The aim of this study was to review the procedure in the context of clinical indications, effectiveness, and safety. STUDY DESIGN AND METHODS: All TPE procedures performed at a tertiary care hospital during a 12-year period (2005-2016) were retrospectively evaluated. RESULTS: Eighteen patients with a median age of 8.5 (0.2-17) years underwent a total of 280 TPE sessions. Eleven (61%) patients were treated for renal diseases. Three (17%) patients were diagnosed with neurological diseases, two had liver failure, and one patient each had sepsis and stem cell transplant-associated thrombotic microangiopathy. Seven patients (39%) were classified as American Society for Apheresis Category I, four (22%) as Category II, two (13%) each as Category III and IV, and two (13%) were not classified. Two patients with atypical hemolytic-uremic syndrome received TPE as long-term therapy over 2 and 5 years. All procedures were performed using the filtration technique and heparin anticoagulation. Twelve (67%) patients showed full or partial recovery after TPE, six had no response or an uncertain response. Minor adverse events occurred in 30/280 (10.6%) procedures, and one major complication (0.4%) was reported. CONCLUSION: TPE is a safe apheresis method in children, even when performed as a long-term therapy. Efficacy is high under selected conditions. A highly skilled and experienced staff is mandatory to ensure patient safety and efficacy.


Subject(s)
Plasma Exchange/standards , Adolescent , Blood Component Removal , Child , Child, Preschool , Filtration , Heparin/therapeutic use , Humans , Infant , Plasma Exchange/adverse effects , Remission Induction , Retrospective Studies , Tertiary Care Centers , Treatment Outcome
10.
Pediatr Nephrol ; 32(1): 43-57, 2017 01.
Article in English | MEDLINE | ID: mdl-27056062

ABSTRACT

Recent advances in our understanding of the disease pathology of membranoproliferative glomerulonephritis has resulted in its re-classification as complement C3 glomerulopathy (C3G) and immune complex-mediated glomerulonephritis (IC-GN). The new consensus is based on its underlying pathomechanism, with a key pathogenetic role for the complement alternative pathway (AP), rather than on histomorphological characteristics. In C3G, loss of AP regulation leads to predominant glomerular C3 deposition, which distinguishes C3G from IC-GN with predominant immunoglobulin G staining. Electron microscopy further subdivides C3G into C3 glomerulonephritis and dense deposit disease depending on the presence and distribution pattern of electron-dense deposits within the glomerular filter. Mutations or autoantibodies affecting the function of AP activators or regulators, in particular the decay of the C3 convertase (C3 nephritic factor), have been detected in up to 80 % of C3G patients. The natural outcome of C3G is heterogeneous, but 50 % of patients progress slowly and reach end-stage renal disease within 10-15 years. The new classification not only marks significant advancement in the pathogenic understanding of this rare disease, but also opens doors towards more specific treatment with the potential for improved outcomes.


Subject(s)
Complement C3 , Glomerulonephritis, Membranoproliferative/therapy , Animals , Complement C3 Nephritic Factor , Disease Models, Animal , Glomerulonephritis, Membranoproliferative/diagnosis , Glomerulonephritis, Membranoproliferative/pathology , Humans , Treatment Outcome
11.
Front Immunol ; 7: 137, 2016.
Article in English | MEDLINE | ID: mdl-27148258

ABSTRACT

Neutrophils deposit antimicrobial proteins, such as myeloperoxidase and proteases on chromatin, which they release as neutrophil extracellular traps (NETs). Neutrophils also carry key components of the complement alternative pathway (AP) such as properdin or complement factor P (CFP), complement factor B (CFB), and C3. However, the contribution of these complement components and complement activation during NET formation in the presence and absence of bacteria is poorly understood. We studied complement activation on NETs and a Gram-negative opportunistic bacterial pathogen Pseudomonas aeruginosa (PA01, PAKwt, and PAKgfp). Here, we show that anaphylatoxin C5a, formyl-methionyl-leucyl-phenylalanine (fMLP) and phorbol myristate acetate (PMA), which activates NADPH oxidase, induce the release of CFP, CFB, and C3 from neutrophils. In response to PMA or P. aeruginosa, neutrophils secrete CFP, deposit it on NETs and bacteria, and induce the formation of terminal complement complexes (C5b-9). A blocking anti-CFP antibody inhibited AP-mediated but not non-AP-mediated complement activation on NETs and P. aeruginosa. Therefore, NET-mediated complement activation occurs via both AP- and non AP-based mechanisms, and AP-mediated complement activation during NETosis is dependent on CFP. These findings suggest that neutrophils could use their "AP tool kit" to readily activate complement on NETs and Gram-negative bacteria, such as P. aeruginosa, whereas additional components present in the serum help to fix non-AP-mediated complement both on NETs and bacteria. This unique mechanism may play important roles in host defense and help to explain specific roles of complement activation in NET-related diseases.

12.
Transfus Apher Sci ; 54(2): 220-31, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27160864

ABSTRACT

Thrombotic microangiopathies (TMA) are disorders defined by microangiopathic hemolytic anemia, non-immune thrombocytopenia and have multi-organ involvement including the kidneys, brain, gastrointestinal, respiratory tract and skin. Emerging evidence points to the central role of complement dysregulation in leading to microvascular endothelial injury which is crucial for the development of TMAs. This key insight has led to the development of complement-targeted therapy. Eculizumab is an anti-C5 monoclonal antibody, which has revolutionized the treatment of atypical hemolytic uremic syndrome. Several other anti-complement therapeutic agents are currently in development, offering a potential armamentarium of therapies available to treat complement-mediated TMAs. The development of sensitive, reliable and easy to perform assays to monitor complement activity and therapeutic efficacy will be key to devising an individualized treatment regime with the potential of safely weaning or discontinuing treatment in the appropriate clinical setting.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement Pathway, Alternative/drug effects , Thrombotic Microangiopathies/blood , Thrombotic Microangiopathies/drug therapy , Anemia, Hemolytic/blood , Anemia, Hemolytic/complications , Anemia, Hemolytic/drug therapy , Humans , Thrombocytopenia/blood , Thrombocytopenia/complications , Thrombocytopenia/drug therapy , Thrombotic Microangiopathies/etiology
14.
Kidney Int ; 90(1): 123-34, 2016 07.
Article in English | MEDLINE | ID: mdl-27236750

ABSTRACT

Atypical hemolytic uremic syndrome and thrombotic thrombocytopenic purpura have traditionally been considered separate entities. Defects in the regulation of the complement alternative pathway occur in atypical hemolytic uremic syndrome, and defects in the cleavage of von Willebrand factor (VWF)-multimers arise in thrombotic thrombocytopenic purpura. However, recent studies suggest that both entities are related as defects in the disease-causing pathways overlap or show functional interactions. Here we investigate the possible functional link of VWF-multimers and the complement system on endothelial cells. Blood outgrowth endothelial cells (BOECs) were obtained from 3 healthy individuals and 2 patients with Type 3 von Willebrand disease lacking VWF. Cells were exposed to a standardized complement challenge via the combination of classical and alternative pathway activation and 50% normal human serum resulting in complement fixation to the endothelial surface. Under these conditions we found the expected release of VWF-multimers causing platelet adhesion onto BOECs from healthy individuals. Importantly, in BOECs derived from patients with von Willebrand disease complement C3c deposition and cytotoxicity were more pronounced than on BOECs derived from normal individuals. This is of particular importance as primary glomerular endothelial cells display a heterogeneous expression pattern of VWF with overall reduced VWF abundance. Thus, our results support a mechanistic link between VWF-multimers and the complement system. However, our findings also identify VWF as a new complement regulator on vascular endothelial cells and suggest that VWF has a protective effect on endothelial cells and complement-mediated injury.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Complement Pathway, Alternative/immunology , Endothelial Cells/immunology , Purpura, Thrombotic Thrombocytopenic/immunology , von Willebrand Factor/metabolism , Blood Platelets/immunology , Cell Adhesion/immunology , Complement C3c/metabolism , Humans , Kidney Glomerulus/cytology , Primary Cell Culture , von Willebrand Disease, Type 3/blood
15.
Pediatr Nephrol ; 31(11): 2079-86, 2016 11.
Article in English | MEDLINE | ID: mdl-27008643

ABSTRACT

BACKGROUND: Post-infectious glomerulonephritis (PIGN) usually follows a benign course, but few children have an atypical, severe presentation, and these exceptional cases have been linked to the dysregulation of the complement alternative pathway (CAP). There is a considerable overlap in the histopathological features of PIGN and C3 glomerulopathy (C3G), which is also associated with CAP dysregulation but has a poorer outcome. We hypothesized that PIGN and C3G define a disease spectrum, and that in the past there may be some children with C3G who were misclassified with PIGN before C3G was described as a separate disease entity. METHODS: Children with PIGN (n = 33) diagnosed between 1985 and 2010 who underwent a renal biopsy due to their unusual course were reviewed and of them, 8 were reclassified into C3G based on the current classification criteria. Outcome was based on the degree of proteinuria, C3 level, and renal function at follow-up. RESULTS: Sixteen (72.7%) children with typical PIGN recovered completely as compared to only 2 (25%) with C3G. Of note, children with "typical" PIGN had a more severe disease course at onset; however, the outcome at last follow up was favorable. CONCLUSIONS: Our results support the hypothesis that PIGN and C3G form a disease spectrum and have different long-term clinical implications and management strategies.


Subject(s)
Communicable Diseases/complications , Complement C3/analysis , Complement Pathway, Alternative , Glomerulonephritis, Membranoproliferative/metabolism , Glomerulonephritis/metabolism , Adolescent , Biopsy , Child , Child, Preschool , Complement C3/metabolism , Female , Glomerulonephritis/blood , Glomerulonephritis/etiology , Glomerulonephritis/pathology , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/pathology , Humans , Kidney Glomerulus/pathology , Longitudinal Studies , Male , Proteinuria/urine
16.
Pediatr Nephrol ; 31(1): 15-39, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25859752

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) emerged during the last decade as a disease largely of complement dysregulation. This advance facilitated the development of novel, rational treatment options targeting terminal complement activation, e.g., using an anti-C5 antibody (eculizumab). We review treatment and patient management issues related to this therapeutic approach. We present consensus clinical practice recommendations generated by HUS International, an international expert group of clinicians and basic scientists with a focused interest in HUS. We aim to address the following questions of high relevance to daily clinical practice: Which complement investigations should be done and when? What is the importance of anti-factor H antibody detection? Who should be treated with eculizumab? Is plasma exchange therapy still needed? When should eculizumab therapy be initiated? How and when should complement blockade be monitored? Can the approved treatment schedule be modified? What approach should be taken to kidney and/or combined liver-kidney transplantation? How should we limit the risk of meningococcal infection under complement blockade therapy? A pressing question today regards the treatment duration. We discuss the need for prospective studies to establish evidence-based criteria for the continuation or cessation of anticomplement therapy in patients with and without identified complement mutations.


Subject(s)
Atypical Hemolytic Uremic Syndrome/therapy , Nephrology/standards , Adolescent , Age Factors , Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/epidemiology , Atypical Hemolytic Uremic Syndrome/immunology , Child , Child, Preschool , Combined Modality Therapy , Complement Activation/drug effects , Consensus , Cooperative Behavior , Drug Monitoring , Humans , Immunologic Factors/therapeutic use , Infant , Infant, Newborn , International Cooperation , Kidney Transplantation , Liver Transplantation , Monitoring, Immunologic , Patient Selection , Plasma Exchange , Predictive Value of Tests , Risk Factors , Treatment Outcome
17.
J Immunol ; 193(10): 4895-903, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25339666

ABSTRACT

Th17 cells infiltrate the kidneys of patients with lupus nephritis (LN) and are critical for the pathogenesis of this disease. In this study, we show that enhanced activity of Stat3 in CD4(+)CD45RA(-)Foxp3(-) and Foxp3(low) effector T cells from children with LN correlates with increased frequencies of IL-17-producing cells within these T cell populations. The levels of retinoic acid-related orphan receptor c and IL-17 mRNA are significantly higher in PBMCs from children with LN than in those from controls. Mammalian target of rapamycin inhibition by rapamycin reduces both Stat3 activation in effector T cells and the frequency of IL-17-producing T cells in lupus patients. Complement factor C5a slightly increases the expression of IL-17 and induces activation of Akt in anti-CD3-activated lupus effector T cells. Th17 cells from children with LN exhibit high Akt activity and enhanced migratory capacity. Inhibition of the Akt signaling pathway significantly decreases Th17 cell migration. These findings indicate that the Akt signaling pathway plays a significant role in the migratory activity of Th17 cells from children with LN and suggest that therapeutic modulation of the Akt activity may inhibit Th17 cell trafficking to sites of inflammation and thus suppress chronic inflammatory processes in children with LN.


Subject(s)
Lupus Nephritis/immunology , Proto-Oncogene Proteins c-akt/genetics , STAT3 Transcription Factor/genetics , Th17 Cells/immunology , Adolescent , Cell Movement/drug effects , Child , Complement C5a/genetics , Complement C5a/immunology , Female , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Humans , Immunosuppressive Agents/pharmacology , Interleukin-17/genetics , Interleukin-17/immunology , Kidney/immunology , Kidney/pathology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Lupus Nephritis/genetics , Lupus Nephritis/pathology , Male , Primary Cell Culture , Proto-Oncogene Proteins c-akt/immunology , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/immunology , STAT3 Transcription Factor/immunology , Signal Transduction , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , Th17 Cells/drug effects , Th17 Cells/pathology
19.
Semin Thromb Hemost ; 40(4): 444-64, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24911558

ABSTRACT

Thrombotic microangiopathy (TMA) is a rare but severe disorder characterized by endothelial cell activation and thrombus formation. It manifests with the triad of hemolytic anemia, thrombocytopenia, and organ failure. Prompt diagnosis and treatment initiation are crucial for long-term outcome. TMA often manifests subsequent to infectious events, of which (enterohemorrhagic) Escherichia coli is the most frequently reported. TMA also occurs on the background of genetic/autoimmune defects in the complement system (atypical hemolytic uremic syndrome [aHUS]) and underlying conditions, such as pregnancy, transplantation, drugs, other glomerulopathies, vasculitides, or metabolic defects. Complement activation or defects in its regulation have now been described in an increasing number of acquired diseases with TMA. Coinciding with this expanding spectrum of complement-mediated diseases, the question arises which patients might benefit from a complement-targeted therapy. Success of therapy depends on the individual contribution of complement activation in disease pathogenesis. The advent of eculizumab, a monoclonal antibody that blocks terminal complement activation, has markedly improved outcome and quality of life in patients with aHUS. This review discusses the contribution of complement and highlights its complex interaction with inflammation, coagulation, and the endothelium. Treatment experiences focusing on eculizumab therapy are discussed in detail across the emerging spectrum of complement-mediated thrombotic microangiopathies.


Subject(s)
Complement System Proteins/immunology , Thrombotic Microangiopathies/immunology , Thrombotic Microangiopathies/therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Antiphospholipid Syndrome , Atypical Hemolytic Uremic Syndrome/immunology , Autoimmune Diseases , Bone Marrow Transplantation , Complement Activation , Diacylglycerol Kinase/immunology , Enterohemorrhagic Escherichia coli , Escherichia coli Infections/complications , Female , Homeostasis , Humans , Inflammation , Kidney Transplantation , Male , Pregnancy , Pregnancy Complications , Quality of Life , Recurrence , Stem Cell Transplantation , Thrombotic Microangiopathies/microbiology , Treatment Outcome
20.
Pediatr Nephrol ; 29(10): 1967-78, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24817340

ABSTRACT

BACKGROUND: In 2009, the European Paediatric Study Group for Haemolytic Uraemic Syndrome (HUS) published a clinical practice guideline for the investigation and initial therapy of diarrhea-negative HUS (now more widely referred to as atypical HUS, aHUS). The therapeutic component of the guideline (comprising early, high-volume plasmapheresis) was derived from anecdotal evidence and expert consensus, and the authors committed to auditing outcome. METHODS: Questionnaires were distributed to pediatric nephrologists across Europe, North America, and the Middle East, who were asked to complete one questionnaire per patient episode of aHUS between July 1, 2009 and December 31, 2010. Comprehensive, anonymous demographic and clinical data were collected. RESULTS: Seventy-one children were reported with an episode of aHUS during the audit period. Six cases occurred on a background of influenza A H1N1 infection. Of 71 patients, 59 (83 %) received plasma therapy within the first 33 days, of whom ten received plasma infusion only. Complications of central venous catheters occurred in 16 out of 51 patients with a catheter in-situ (31 %). Median time to enter hematological remission was 11.5 days, and eight of 71 (11 %) patients did not enter hematological remission by day 33. Twelve patients (17 %) remained dialysis dependent at day 33. CONCLUSIONS: This audit provides a snapshot of the early outcome of a group of children with aHUS in the months prior to more widespread use of eculizumab.


Subject(s)
Atypical Hemolytic Uremic Syndrome/therapy , Guideline Adherence/statistics & numerical data , Adolescent , Child , Child, Preschool , Clinical Audit , Female , Humans , Infant , Infant, Newborn , Male , Practice Guidelines as Topic , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL
...