Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Pediatr Nephrol ; 39(8): 2459-2465, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38589699

ABSTRACT

BACKGROUND: Hemolytic uremic syndrome (HUS) is an important cause of acute kidney injury in children. HUS is known as an acute disease followed by complete recovery, but patients may present with kidney abnormalities after long periods of time. This study evaluates the long-term outcome of Shiga toxin-producing Escherichia coli-associated HUS (STEC-HUS) in pediatric patients, 10 years after the acute phase of disease to identify risk factors for long-term sequelae. METHODS: Over a 6-year period, 619 patients under 18 years of age with HUS (490 STEC-positive, 79%) were registered in Austria and Germany. Long-term follow-up data of 138 STEC-HUS-patients were available after 10 years for analysis. RESULTS: A total of 66% (n = 91, 95% CI 0.57-0.73) of patients fully recovered showing no sequelae after 10 years. An additional 34% (n = 47, 95% CI 0.27-0.43) presented either with decreased glomerular filtration rate (24%), proteinuria (23%), hypertension (17%), or neurological symptoms (3%). Thirty had sequelae 1 year after STEC-HUS, and the rest presented abnormalities unprecedented at the 2-year (n = 2), 3-year (n = 3), 5-year (n = 3), or 10-year (n = 9) follow-up. A total of 17 patients (36.2%) without kidney abnormalities at the 1-year follow-up presented with either proteinuria, hypertension, or decreased eGFR in subsequent follow-up visits. Patients needing extracorporeal treatments during the acute phase were at higher risk of presenting symptoms after 10 years (p < 0.05). CONCLUSIONS: Patients with STEC-HUS should undergo regular follow-up, for a minimum of 10 years following their index presentation, due to the risk of long-term sequelae of their disease. An initial critical illness, marked by need of kidney replacement therapy or plasma treatment may help predict poor long-term outcome.


Subject(s)
Escherichia coli Infections , Hemolytic-Uremic Syndrome , Shiga-Toxigenic Escherichia coli , Humans , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/therapy , Hemolytic-Uremic Syndrome/complications , Hemolytic-Uremic Syndrome/epidemiology , Shiga-Toxigenic Escherichia coli/isolation & purification , Male , Female , Child , Escherichia coli Infections/microbiology , Escherichia coli Infections/complications , Escherichia coli Infections/epidemiology , Escherichia coli Infections/diagnosis , Child, Preschool , Follow-Up Studies , Adolescent , Infant , Germany/epidemiology , Risk Factors , Glomerular Filtration Rate , Austria/epidemiology , Time Factors , Proteinuria/etiology , Proteinuria/microbiology , Proteinuria/diagnosis
2.
Pediatr Nephrol ; 36(4): 917-925, 2021 04.
Article in English | MEDLINE | ID: mdl-33025207

ABSTRACT

BACKGROUND: The complement factor H antibody (CFH-Ab)-associated hemolytic uremic syndrome (HUS) forms a distinct subgroup within the complement-mediated HUS disease spectrum. The autoimmune nature of this HUS subgroup implies the potential benefit of a targeted immunosuppressive therapy. Data on long-term outcome are scarce. METHODS: This observational study evaluates the clinical outcome of 19 pediatric CFH-Ab HUS patients from disease onset until their 5-year follow-up. RESULTS: All but one relapse occurred during the first 2 years, and patients who had no relapse within the first 6 months were relapse-free until the end of the observation period. Kidney function at disease onset determines long-term kidney function: all individuals with normal kidney function at disease onset had normal kidney function after 5 years, and all patients with reduced kidney function at onset had impaired kidney function at the last follow-up. Level of CFH-Ab titer at disease onset was not correlated with a higher risk of recurrences or worse long-term outcome after 5 years. Resolution of CFH-Ab titers after 5 years was common. CONCLUSIONS: CFH-Ab HUS patients have a varied overall long-term course. Early relapses are common, making close surveillance during the first years essential, regardless of the initial CFH-Ab titer.


Subject(s)
Complement Factor H/immunology , Hemolytic-Uremic Syndrome , Renal Insufficiency , Autoantibodies , Child , Chronic Disease , Follow-Up Studies , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/therapy , Humans , Recurrence , Treatment Outcome
3.
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
4.
Am J Transplant ; 18(11): 2818-2822, 2018 11.
Article in English | MEDLINE | ID: mdl-29962080

ABSTRACT

Primary focal segmental glomerulosclerosis (FSGS) recurs in up to 55% of patients after kidney transplantation. Herein we report the successful management of recurrent FSGS. A 5-year-old boy with primary FSGS received a deceased donor renal transplant. Immediate and fulminant recurrence of FSGS caused anuric graft failure that was resistant to plasmapheresis and rituximab. After exclusion of structural or immunologic damage to the kidney by repeated biopsies, the allograft was retrieved from the first recipient on day 27 and transplanted into a 52-year-old second recipient who had vascular nephropathy. Immediately after retransplantation, the allograft regained function with excellent graft function persistent now at 3 years after transplant. After 2 years on hemodialysis, the boy was listed for kidney retransplantation. To prevent FSGS recurrence, pretreatment with ofatumumab was performed. Nephrotic range proteinuria still occurred after the second transplantation, which responded, however, to daily plasma exchange in combination with ofatumumab. At 8 months after kidney retransplantation graft function is good. The clinical course supports the hypothesis of a circulating permeability factor in the pathogenesis of FSGS. Successful ofatumumab pretreatment implicates a key role of B cells. Herein we provide a description of successful management of kidney failure by FSGS, carefully avoiding waste of organs.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Glomerulosclerosis, Focal Segmental/surgery , Graft Rejection/prevention & control , Kidney Transplantation/adverse effects , Child, Preschool , Graft Rejection/etiology , Humans , Male , Middle Aged , Prognosis , Recurrence
6.
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
7.
Hum Mutat ; 38(4): 365-372, 2017 04.
Article in English | MEDLINE | ID: mdl-28181337

ABSTRACT

We identified two unrelated consanguineous families with three children affected by the rare association of congenital nephrotic syndrome (CNS) diagnosed in the first days of life, of hypogonadism, and of prenatally detected adrenal calcifications, associated with congenital adrenal insufficiency in one case. Using exome sequencing and targeted Sanger sequencing, two homozygous truncating mutations, c.1513C>T (p.Arg505*) and c.934delC (p.Leu312Phefs*30), were identified in SGPL1-encoding sphingosine-1-phosphate (S1P) lyase 1. SGPL1 catalyzes the irreversible degradation of endogenous and dietary S1P, the final step of sphingolipid catabolism, and of other phosphorylated long-chain bases. S1P is an intracellular and extracellular signaling molecule involved in angiogenesis, vascular maturation, and immunity. The levels of SGPL1 substrates, S1P, and sphingosine were markedly increased in the patients' blood and fibroblasts, as determined by liquid chromatography-tandem mass spectrometry. Vascular alterations were present in a patient's renal biopsy, in line with changes seen in Sgpl1 knockout mice that are compatible with a developmental defect in vascular maturation. In conclusion, loss of SGPL1 function is associated with CNS, adrenal calcifications, and hypogonadism.


Subject(s)
Adrenal Gland Diseases/genetics , Aldehyde-Lyases/genetics , Calcinosis/genetics , Mutation , Nephrotic Syndrome/genetics , Adrenal Gland Diseases/congenital , Adrenal Gland Diseases/enzymology , Adult , Aldehyde-Lyases/deficiency , Animals , Base Sequence , Calcinosis/enzymology , Consanguinity , Female , Humans , Infant , Lysophospholipids/blood , Lysophospholipids/metabolism , Male , Mice, Knockout , Nephrotic Syndrome/congenital , Nephrotic Syndrome/enzymology , Pedigree , Sequence Analysis, DNA/methods , Sphingosine/analogs & derivatives , Sphingosine/blood , Sphingosine/metabolism
9.
Pediatr Transplant ; 19(5): E121-5, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26037622

ABSTRACT

CFH-Ab-associated aHUS requires different diagnostic and therapeutic approaches and then the genetically defined aHUS forms. The risk of post-transplant recurrence with graft dysfunction in CFH-Ab aHUS is not well documented. It is suggested that recurrence can be expected if a significant CFH-Ab load persists at the time of transplantation. A pretransplant procedure to reduce CFH-Ab titer seems reasonable, but accurate recommendations are lacking. Whether further prophylactic interventions after transplantation are necessary has to be decided on an individual basis. We report the case of a late diagnosed CFH-Ab HUS with initial ESRD and a successful living-related renal transplantation over a post-transplant period of four and a half years on the basis of a prophylactic pretransplant IVIG admission.


Subject(s)
Antibodies/immunology , Atypical Hemolytic Uremic Syndrome/complications , Atypical Hemolytic Uremic Syndrome/surgery , Complement Factor H/immunology , Kidney Transplantation/methods , Renal Insufficiency/surgery , Child , Graft Survival , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunosuppressive Agents/chemistry , Living Donors , Male , Recurrence , Renal Insufficiency/complications , Treatment Outcome
10.
Pediatr Int ; 57(2): e42-4, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25868960

ABSTRACT

A 15-year-old girl presented with acute bilateral loss of central visual acuity due to hypertensive retinopathy level IV. She was found to have unrecognized malignant arterial hypertension associated with end-stage renal failure. At the time of diagnosis she also had severe left ventricular hypertrophy (LVH). Hypertension was successfully treated with combined anti-hypertensive therapy, but renal function did not recover. The patient underwent successful kidney transplant 4 months later and over a period of 20 months hypertensive retinopathy and LVH gradually resolved. This report emphasizes the importance of routine measurement of blood pressure and describes the possible consequences of unrecognized arterial hypertension in children. Early diagnosis and appropriate treatment are necessary to avoid development and progression of target organ damage and promote better long-term cardiovascular prognosis.


Subject(s)
Blindness/etiology , Hypertension, Malignant/complications , Hypertensive Retinopathy/complications , Kidney Failure, Chronic/complications , Adolescent , Antihypertensive Agents/therapeutic use , Blood Pressure , Female , Humans , Hypertension, Malignant/drug therapy , Hypertensive Retinopathy/diagnosis , Hypertensive Retinopathy/drug therapy , Hypertrophy, Left Ventricular/etiology , Kidney Failure, Chronic/surgery , Kidney Transplantation
11.
Front Pediatr ; 2: 97, 2014.
Article in English | MEDLINE | ID: mdl-25250305

ABSTRACT

Thrombotic microangiopathies (TMA) are rare but severe disorders, characterized by endothelial cell activation and thrombus formation leading to hemolytic anemia, thrombocytopenia, and organ failure. Complement over activation in combination with defects in its regulation is described in an increasing number of TMA and if primary for the disease denominated as atypical hemolytic-uremic syndrome. Although TMA predominantly affects the renal microvasculature, extra-renal manifestations are observed in 20% of patients including involvement of the central nerve system, cardiovascular system, lungs, skin, skeletal muscle, and gastrointestinal tract. Prompt diagnosis and treatment initiation are therefore crucial for the prognosis of disease acute phase and the long-term outcome. This review summarizes the available evidence on extra-renal TMA manifestations and discusses the role of acute and chronic complement activation by highlighting its complex interaction with inflammation, coagulation, and endothelial homeostasis.

12.
Semin Thromb Hemost ; 40(4): 431-43, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24799303

ABSTRACT

The presence of circulating autoantibodies, primarily to complement factor H antibodies (CFH-Abs) in plasma characterizes the autoimmune form of atypical hemolytic uremic syndrome (aHUS). This acquired form of aHUS defines a distinct subgroup of aHUS patients, which requires diagnostic and treatment approaches in part different from those of the genetically defined forms. The mechanisms leading to CFH-Ab production and disease onset are not completely understood, but CFH-Ab HUS seems to be secondary to a combination of genetic predisposition and environmental factors. Early diagnosis of this specific aHUS entity is important, as prompt induction of plasma exchange and concomitant immunosuppression leads to a favorable outcome. Nevertheless, information on clinical features and outcome in children is limited. Here, we review the literature on the biological and clinical features of CFH-Ab HUS and discuss therapeutic options.


Subject(s)
Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/therapy , Complement Factor H/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/immunology , Autoantibodies/blood , Binding Sites , Clinical Trials as Topic , Complement C3b Inactivator Proteins/immunology , Genetic Predisposition to Disease , Humans , Kidney Transplantation , Protein Structure, Tertiary , Treatment Outcome
13.
Semin Thromb Hemost ; 40(4): 478-86, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24830813

ABSTRACT

There has been rapid progress in the understanding of the pathophysiology of the hemolytic uremic syndrome (HUS) disease spectrum; thus, complex diagnostic and therapeutic requirements have emerged in parallel. Current recommendations for diagnosis and therapy were rapidly adapted from the prior skilled scientific groundwork. However, such recommendations can be realized only when highly specialized laboratories and sufficient financial resources are available. Thus, many recommendations are not feasible for patients living and working in developing countries. More than one-third of the world's population has no access to essential drugs and more than half of this group lives in the poorest regions of Africa and Asia. From this perspective, distinct initial diagnostic and therapeutic recommendations, as well as international cooperations are needed to complete proper diagnostic work-ups in a stringent and cost-efficient manner and to enable patients to be adequately treated with available resources. However, while costs for complement-targeted drugs remain tremendously high, state-of-the-art treatment options remain unavailable for the vast majority of patients in developing areas.


Subject(s)
Complement System Proteins/immunology , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/therapy , Africa , Asia , Developing Countries , Enterohemorrhagic Escherichia coli , Hemolytic-Uremic Syndrome/epidemiology , Humans , International Cooperation , Shiga Toxin/chemistry , South America , Treatment Outcome
15.
Pediatr Rheumatol Online J ; 12(1): 37, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-27643389

ABSTRACT

BACKGROUND: Kawasaki disease (KD) is a rare vasculitis seen predominantly in children. In developing countries, it is the leading cause of childhood-acquired heart disease. Besides a case report from 1981 there have been no data published dealing with the epidemiology and clinical aspects of KD in Austria. METHODS: The purpose of the present study was to investigate the clinical spectrum of KD in a geographically determined cohort of infants, children, and adolescents that were diagnosed and treated at the University Hospital of Innsbruck from 2003-2012. RESULTS: Thirty-two patients were included in the study with a median age of 32.96 months (2-192). 59.4% of the patients were aged between six months and four years. The male-to-female ratio was 1:1.13. Clinical examination revealed non-purulent conjunctivitis and exanthema as the most common symptoms (84.4%). 75% showed oropharyngeal changes, 21.9% had gastrointestinal complaints such as diarrhoe, stomachache or vomiting prior to diagnosis. One third of the patients were admitted with a preliminary diagnosis, whereas 78.1% were pre-treated with antibiotics. The median fever duration at the time of presentation was estimated with 4.96 days (1-14), at time of diagnosis 6.76 days (3-15).75% were diagnosed with complete KD, and 25% with an incomplete form of the disease. There was no significant difference in the duration of fever neither between complete and incomplete KD, nor between the different age groups. Typical laboratory findings included increased C-reactive protein (CRP) (80.6%) and erythrocyte sedimentation rate (ESR) (96%),leukocytosis (48.4%) and thrombocytosis (40.6%) without any significant quantitative difference between complete and incomplete KD. Coronary complications could be observed in six patients: one with a coronary aneurysm and five with tubular dilatation of the coronary arteries. Our patient cohort represents the age distribution as described in literature and emphasizes that KD could affect persons of any age. The frequency of occurrence of the clinical symptoms differs from previous reports - in our study, we predominantly observed non-purulent conjunctivitis and exanthema. CONCLUSION: KD should always be considered as a differential diagnosis in a child with fever of unknown origin, as treatment can significantly decrease the frequency of coronary complications.


Subject(s)
Coronary Disease , Mucocutaneous Lymph Node Syndrome , Adolescent , Age Distribution , Austria/epidemiology , Child , Coronary Disease/diagnosis , Coronary Disease/etiology , Coronary Disease/prevention & control , Diagnosis, Differential , Early Diagnosis , Early Medical Intervention/methods , Female , Humans , Infant , Male , Mucocutaneous Lymph Node Syndrome/complications , Mucocutaneous Lymph Node Syndrome/diagnosis , Mucocutaneous Lymph Node Syndrome/epidemiology , Mucocutaneous Lymph Node Syndrome/physiopathology , Symptom Assessment/methods
16.
Clin J Am Soc Nephrol ; 8(3): 407-15, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23243267

ABSTRACT

BACKGROUND AND OBJECTIVES: This study evaluated the relevance of complement factor H (CFH)-related protein (CFHR) 1 deficiency in pediatric patients with atypical hemolytic uremic syndrome (aHUS) by evaluating both the frequency of deletions in CFHR1 and the presence of complement factor H (CFH) antibodies. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: A total of 116 patients (mainly from central Europe) and 118 healthy blood donors were included from 2001 to 2012. The presence of CFHR1 gene deletions was determined in 90 pediatric patients with aHUS and 118 controls by an easy, fast, and cheap PCR assay; 100 patients with aHUS and 42 controls were tested for CFH antibodies by ELISA. Questionnaires were administered to evaluate the clinical and laboratory data. RESULTS: Homozygous deletion in CFHR1 was detected in 32% of the patients with aHUS tested, compared with 2.5% of controls (P<0.001). CFH antibodies were present in 25% of the patients and none of the controls. CFH antibodies were detected in 82% of patients with homozygous CFHR1 gene deletion and in 6% of patients without. CFH antibody-positive patients with aHUS showed a significantly lower platelet nadir at disease onset and significantly less frequent involvement of the central nervous system than did antibody-negative patients. Antibody-positive patients also received plasma therapy more often. CONCLUSION: Homozygous deletion in CFHR1 is strongly associated with occurrence of CFH antibodies in pediatric patients with aHUS. However, despite this apparent genetic disease predisposition, it cannot be considered an exclusive cause for aHUS. Initial presentation of Shiga toxin-negative HUS with severe thrombocytopenia and no central nervous system complications in pediatric patients is especially suspicious for CFH antibody aHUS.


Subject(s)
Autoantibodies/blood , Autoimmunity , Complement C3b Inactivator Proteins/deficiency , Complement C3b Inactivator Proteins/genetics , Gene Deletion , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/immunology , Age Factors , Atypical Hemolytic Uremic Syndrome , Chi-Square Distribution , Child , Child, Preschool , Comparative Genomic Hybridization , Complement Factor H/immunology , Enzyme-Linked Immunosorbent Assay , Europe , Female , Gene Frequency , Genetic Predisposition to Disease , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/diagnosis , Heterozygote , Homozygote , Humans , Infant , Male , Odds Ratio , Phenotype , Polymerase Chain Reaction , Prospective Studies , Retrospective Studies , Risk Factors , Surveys and Questionnaires
17.
Clin Infect Dis ; 54(10): 1413-21, 2012 May.
Article in English | MEDLINE | ID: mdl-22412065

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the long-term prognosis of children with hemolytic uremic syndrome (HUS). METHODS: Over a 6-year period, 619 pediatric patients with the clinical diagnosis of HUS were registered in Austria and Germany, and a subset (n = 274) was prospectively followed up for 5 years. RESULTS: Infection with enterohemorrhagic Escherichia coli (EHEC) was confirmed in 79% of cases. Five years after diagnosis, 70% of EHEC-infected patients (95% confidence interval [CI], .63-.76) were fully recovered. The remaining 30% had persistent hypertension (9%), neurological symptoms (4%), decreased glomerular filtration rate (7%), and/or proteinuria (18%). Hypertension and proteinuria developed in a total of 18% of patients who had no sequelae 1 year after the acute phase (95% CI, 12-26). Multivariate logistic regression analysis demonstrated an association between the use of plasma therapy during acute phase and poor long-term outcome (odds ratio, 2.9-13; 95% CI, 2.4-33; P < .05), but this treatment was also used more frequently in severe cases. In contrast, the use of antibiotic therapy in the diarrheal phase and other established risk factors for developing HUS, such as Shiga toxin 2 and EHEC serotypes traditionally considered to be "high risk," were not associated with adverse long-term outcome. In particular, there was no difference between O157 and non-O157 EHEC. CONCLUSIONS: This study identified an association between the use of plasma treatment and poor long-term outcome and confirms already known risk factors for poor prognosis. Follow-up investigations for at least 5 years are recommended to detect late-emerging sequelae.


Subject(s)
Delayed Diagnosis/statistics & numerical data , Enterohemorrhagic Escherichia coli/isolation & purification , Escherichia coli Infections/complications , Escherichia coli Infections/microbiology , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/epidemiology , Anti-Bacterial Agents/therapeutic use , Austria/epidemiology , Child, Preschool , Escherichia coli Infections/therapy , Female , Follow-Up Studies , Germany/epidemiology , Humans , Infant , Male , Prospective Studies , Transfusion Reaction , Treatment Outcome
18.
Rheumatol Int ; 32(6): 1815-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21647665

ABSTRACT

The complement system plays a role in the pathogenesis of some autoimmunopathies. This longitudinal study evaluates the contribution of the complement system in the pathogenesis of oligoarticular juvenile idiopathic arthritis (JIA). Serum of the peripheral blood and the synovial fluid were investigated for the activity of the classical (CP), the mannose binding lectin (MBL), and the alternative pathway (AP). A total of 12 samples from peripheral blood (PB) and two samples from synovial fluid (SF) of girls with oligoarticular JIA were investigated in a longitudinal observation from the time point of the diagnosis of JIA. The differences between the complement activity in the PB and in the SF were extremely statistically significant (CP and MBL: P < 0.0001; AP: < 0.0087). The activity of the CP and the MBL pathway was reduced. The AP is the main contributor in the pathogenesis of oligoarticular JIA. Anti-C5 therapy may be an option to avoid the creation of the membrane attack complex.


Subject(s)
Arthritis, Juvenile/immunology , Complement Activation , Complement Pathway, Alternative , Complement Pathway, Classical , Complement Pathway, Mannose-Binding Lectin , Complement System Proteins/metabolism , Arthritis, Juvenile/blood , Arthritis, Juvenile/diagnosis , Austria , Biomarkers/blood , Female , Humans , Longitudinal Studies , Severity of Illness Index , Synovial Fluid/immunology , Time Factors
19.
Clin J Am Soc Nephrol ; 4(8): 1356-62, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19590060

ABSTRACT

BACKGROUND AND OBJECTIVES: Atypical hemolytic uremic syndrome (aHUS) is associated with mutations in genes encoding complement-regulatory proteins factor H, I and B and membrane cofactor protein. Recently, heterozygous gain-of-function mutations in the complement C3 gene have been found in patients with aHUS. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: A large family with a C3 R570Q mutation is described. Clinical and laboratory findings of carriers of the mutation and unaffected family members are reported. RESULTS: The index patient suffered from recurrent aHUS at age 22 and developed end-stage renal failure. Of 24 family members, nine harbored the C3 R570Q mutation. Carriers showed reduced or borderline C3 levels. Arterial hypertension was found in six family members, microhematuria in five and chronic kidney disease stage 3 in two elderly carrier patients. Despite marked consumption of C3, serum terminal complement complex levels were not elevated in carriers compared with other family members. CONCLUSIONS: The penetrance of the C3 R570Q mutation to induce aHUS is incomplete and lower compared with mutations in other genes predisposing to the disease. The mutation is possibly also associated with hypertension, hematuria and chronic kidney disease, all of which may represent consequences of long-term complement activation in the renal vasculature.


Subject(s)
Complement C3/genetics , Hematuria/genetics , Hemolytic-Uremic Syndrome/genetics , Hypertension/genetics , Kidney Failure, Chronic/genetics , Mutation , Adult , Aged , Child , Complement C3/immunology , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Hematuria/immunology , Hemolytic-Uremic Syndrome/immunology , Heterozygote , Humans , Hypertension/immunology , Kidney Failure, Chronic/immunology , Male , Middle Aged , Pedigree , Polymorphism, Single Nucleotide , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...