Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters

Publication year range
1.
Euro Surveill ; 29(30)2024 Jul.
Article in English | MEDLINE | ID: mdl-39056198

ABSTRACT

BackgroundHaemolytic uremic syndrome (HUS) is a severe complication of infection with Shiga toxin-producing Escherichia coli (STEC). Although the reservoirs of STEC are known, the source of the infection of sporadic cases is often unknown. In 2023, we observed several cases of bloody diarrhoea with STEC infection in children and adolescents returning from vacations.AimWe aimed to explore the association between travel and bloody diarrhoea with STEC infection in children and adolescents.MethodsWe included all children and adolescents with bloody diarrhoea with STEC infection identified in 2023 by the ItalKid-HUS Network surveillance system in northern Italy. We interviewed children's families and sent a questionnaire on recent travels abroad. The exposure time was between 3 days after arrival abroad and 5 days after return home. A self-controlled case series (SCCS) design was used in the analysis.ResultsOf the 43 cases, 11 developed HUS. Twenty-three cases did not travel abroad, while 20 had travelled to several destinations. The incidence rate ratio (IRR) associated with travel to Egypt was 88.6 (95% confidence interval (CI): 17.0-462). Serotype analysis excluded the possibility of a single strain causing the infections. We did not find the source of the infections.ConclusionThere is an elevated risk of acquiring STEC infection with bloody diarrhoea and HUS associated with travel to Egypt. Specific investigations to identify the source are needed to implement effective preventive measures.


Subject(s)
Diarrhea , Escherichia coli Infections , Hemolytic-Uremic Syndrome , Shiga-Toxigenic Escherichia coli , Travel , Humans , Egypt/epidemiology , Shiga-Toxigenic Escherichia coli/isolation & purification , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Escherichia coli Infections/diagnosis , Adolescent , Child , Female , Male , Hemolytic-Uremic Syndrome/epidemiology , Hemolytic-Uremic Syndrome/microbiology , Italy/epidemiology , Diarrhea/microbiology , Diarrhea/epidemiology , Child, Preschool , Infant , Incidence , Population Surveillance
2.
Br J Clin Pharmacol ; 89(5): 1682-1685, 2023 05.
Article in English | MEDLINE | ID: mdl-36480744

ABSTRACT

AIMS: Interferon-beta (IFNß), the most widely prescribed medication for multiple sclerosis, is generally considered safe. Nevertheless, rare serious and/or life-threatening side effects have been reported such as thrombotic microangiopathy. A few mechanisms have been proposed to explain how interferon causes thrombotic microangiopathy, but immunological studies have been unable to pin this phenomenon down to a single pathophysiologic pathway. The aim of this article was to report a new mechanism explaining Interferon beta related thrombotic microangiopathy. METHODS: We report thrombotic microangiopathy in a 28-year-old male receiving interferon-beta treatment for multiple sclerosis. RESULTS: After three years of starting interferon beta therapy, the patient presented with malignant hypertension causing seizures, rapidly progressive renal failure requiring haemodialysis and haemolytic anaemia. Corticosteroid and plasma exchange sessions permitted haemolysis control. Nonetheless, the patient remained hemodialysis-dependent. Exploration of the complement system found a complement factor I deficiency whose activity normalized at the control carried out after 2 years. CONCLUSION: IFNß treatment may cause complement factor I deficit, which can lead to thrombotic microangiopathy and severe renal failure.


Subject(s)
Multiple Sclerosis , Renal Insufficiency , Thrombotic Microangiopathies , Male , Humans , Adult , Interferon-beta/adverse effects , Multiple Sclerosis/drug therapy , Thrombotic Microangiopathies/chemically induced , Renal Insufficiency/complications
3.
BMC Nephrol ; 24(1): 127, 2023 05 05.
Article in English | MEDLINE | ID: mdl-37147581

ABSTRACT

BACKGROUND: We present this challenging case report of Atypical Haemolytic Uremic Syndrome (aHUS) presenting with multi-organ involvement in a patient and heterozygous CFHR1/CFHR3 gene variant, which was refractory to initial eculizumab therapy. CASE PRESENTATION: A forty-three year old female presented with aHUS and had heterozygous disease-associated deletions in the complement genes CFHR1/CFHR3. She had progressive kidney failure and severe extra-renal manifestations including cardiomyopathy and haemorrhagic cystitis; as well as pulmonary, gastrointestinal and neurological involvement. The initial kidney biopsy revealed thrombotic microangiopathy (TMA) changes involving all glomeruli. Clinical improvement was initially seen during eculizumab initiation with suppressed CH50 level, but a new rhinovirus/enterovirus upper respiratory tract infection triggered further severe multi-organ disease activity. The extra-renal manifestations stabilised, then ultimately improved after a period of eculizumab dose intensification. However, the impact on dose intensification on this improvement is unclear. Despite the extra-renal clinical improvement, she ultimately progressed to end-stage kidney disease (ESKD), commencing peritoneal dialysis for three years before undergoing a successful uncomplicated cadaveric kidney transplant without prophylactic eculizumab. Two years after transplant, she has excellent transplant graft function without any further disease recurrence. CONCLUSIONS: This case highlights the concept of extra-renal manifestations in aHUS initially resistant to eculizumab, which potentially responded to dose intensification. Whilst organ injuries are potentially reversible with timely targeted treatment, it appears that the kidneys are most vulnerable to injury.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Kidney Failure, Chronic , Kidney Transplantation , Female , Humans , Adult , Gene Deletion , Atypical Hemolytic Uremic Syndrome/complications , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/drug therapy , Kidney , Kidney Failure, Chronic/genetics , Blood Proteins , Complement C3b Inactivator Proteins/genetics
4.
BMC Anesthesiol ; 23(1): 94, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36977996

ABSTRACT

BACKGROUND: Atypical Haemolytic Uremic Syndrome is an acute life-threatening condition, characterized by the clinical triad of microangiopathic hemolytic anaemia, thrombocytopenia, kidney injury. Management of pregnants affected by Atypical Haemolytic Uremic Syndrome can be a serious concern for obstetric anesthesiologist in the delivery room and in the intensive care unit. CASE PRESENTATION: A 35-year-old primigravida with a monochorionic diamniotic twin pregnancy, presented with an acute haemorrhage due to retained placenta after elective caesarean section and underwent surgical exploration. In the postoperative period, the patient progressively developed hypoxemic respiratory failure and, later on, anaemia, severe thrombocytopenia, and acute kidney injury. A timely diagnosis of Atypical Haemolytic Uremic Syndrome was made. Non-invasive ventilation and high-flow nasal cannula oxygen therapy sessions were initially required. Hypertensive crisis and fluid overload were aggressively treated with a combination of beta and alpha adrenergic blockers (labetalol 0,3 mg/kg/h by continuous intravenous infusion for the first 24 hours, bisoprolol 2,5 mg twice daily for the first 48 hours, doxazosin 2 mg twice daily), central sympatholytics (methyldopa 250 mg twice daily for the first 72 hours, transdermal clonidine 5 mg by the third day), diuretics (furosemide 20 mg three times daily), calcium antagonists (amlodipine 5 mg twice daily). Eculizumab 900 mg was administered via intravenous infusion once per week, attaining hematological and renal remissions. The patient also received several blood transfusion units and anti- meningococcal B, anti-pneumococcal, anti-haemophilus influenzae type B vaccination. Her clinical condition progressively improved, and she was finally discharged from intensive care unit 5 days after admission. CONCLUSIONS: The clinical course of this report underlines how crucial it is for the obstetric anaesthesiologist to promptly identify Atypical Haemolytic Uremic Syndrome, since early initiation of eculizumab, together with supportive therapy, has a direct effect on patient outcome.


Subject(s)
Acute Kidney Injury , Atypical Hemolytic Uremic Syndrome , Complement Inactivating Agents , Pregnancy Complications , Adult , Female , Humans , Pregnancy , Acute Kidney Injury/etiology , Atypical Hemolytic Uremic Syndrome/therapy , Atypical Hemolytic Uremic Syndrome/drug therapy , Cesarean Section/adverse effects , Hemorrhage , Kidney , Complement Inactivating Agents/therapeutic use
5.
Vnitr Lek ; 68(8): 498-507, 2022.
Article in English | MEDLINE | ID: mdl-36575067

ABSTRACT

Haemophagocytic syndrome, diffuse alveolar haemorrhage, catastrophic antiphospholipid syndrome and various types of thrombotic microangiopathies are rare conditions with significant morbidity and mortality. A common feature is late diagnosis, which can affect the success of treatment. The aim of this review article is to summarize the basic diagnostic and therapeutic steps of the present subpopulation of critically ill patients.


Subject(s)
Antiphospholipid Syndrome , Lymphohistiocytosis, Hemophagocytic , Thrombotic Microangiopathies , Humans , Adult , Thrombotic Microangiopathies/diagnosis , Antiphospholipid Syndrome/complications , Antiphospholipid Syndrome/diagnosis , Antiphospholipid Syndrome/therapy
6.
BMC Nephrol ; 22(1): 86, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33691638

ABSTRACT

BACKGROUND: Thrombotic microangiopathy (TMA), a rare but serious complication of systemic lupus erythematosus (SLE), is associated with poor outcomes to conventional immunosuppressive therapy. Recently, eculizumab, a humanised monoclonal antibody that blocks the complement factor 5, has been known to effectively treat atypical haemolytic uremic syndrome (aHUS). Here, we report a case of aHUS co-existing with lupus nephritis that was successfully treated with eculizumab. CASE PRESENTATION: A 23-year-old man presented with abdominal pain and diarrhoea. Initial laboratory tests have shown thrombocytopaenia, microangiopathic haemolytic anaemia, and acute kidney injury. Immunologic tests were consistent with SLE. Kidney biopsy have revealed lupus nephritis class IV-G with TMA. Genetic analysis have shown complement C3 gene mutations, which hints the co-existence of lupus nephritis with aHUS, a form of complement-mediated TMA. Although initial treatment with haemodialysis, plasma exchange, and conventional immunosuppressive therapy (steroid and cyclophosphamide) did not appreciably improve kidney function and thrombocytopaenia, the patient was able to respond to eculizumab therapy. CONCLUSIONS: Due to the similar features of TMA and SLE, clinical suspicion of aHUS in patients with lupus nephritis is important for early diagnosis and prompt management. Timely administration of eculizumab should be considered as a treatment option for aHUS in lupus nephritis patients to yield optimal therapeutic outcomes.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/drug therapy , Complement Inactivating Agents/therapeutic use , Mutation , Atypical Hemolytic Uremic Syndrome/complications , Atypical Hemolytic Uremic Syndrome/genetics , Complement C3/genetics , Humans , Lupus Nephritis/complications , Male , Young Adult
7.
Br J Haematol ; 189(1): 182-186, 2020 04.
Article in English | MEDLINE | ID: mdl-31879952

ABSTRACT

Atypical haemolytic uremic syndrome (aHUS) is associated with complement system abnormality, such as production of complement factor H (CFH) autoantibodies. The growing evidence indicates complement overactivation on platelets is intimately involved in aHUS pathogenesis, besides endothelial injury. We here showed plasma from patients with anti-CFH antibodies induced aggregation of washed platelets, while purified anti-CFH antibodies suppressed aggregation. This suggested anti-CFH antibody itself suppressed thrombosis, while other plasma factor including complement factors could overactivate the platelets, leading to aggregation, which augmented the notion the state of complement activation influenced by anti-CFH antibodies is important in the aggregation of platelets in aHUS.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Autoantibodies , Blood Platelets , Complement Factor H/immunology , Platelet Aggregation/immunology , Atypical Hemolytic Uremic Syndrome/blood , Atypical Hemolytic Uremic Syndrome/immunology , Atypical Hemolytic Uremic Syndrome/pathology , Autoantibodies/blood , Autoantibodies/immunology , Blood Platelets/immunology , Blood Platelets/metabolism , Blood Platelets/pathology , Child , Child, Preschool , Female , Humans , Male
8.
Nephrology (Carlton) ; 25(7): 518-521, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31900968

ABSTRACT

Atypical haemolytic uraemic syndrome (aHUS) is a severe, life-threatening condition that requires early recognition and urgent treatment. In aHUS rare genetic variants in CFH, CFI, CD46, C3 and CFB predispose to complement over activation. This case describes a case of aHUS in which there was a strong temporal association between disease onset and the use of smoked cocaine. The patient was found to have a rare genetic variant in the CFI gene which may have been unmasked by first-time exposure to cocaine. The patient stabilized and improved with early administration of eculizumab, supporting the notion of an underlying immunological pathogenesis and the importance of early intervention.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Atypical Hemolytic Uremic Syndrome , Cocaine Smoking , Complement Factor I/genetics , Renal Insufficiency , Thrombocytopenia , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/physiopathology , Atypical Hemolytic Uremic Syndrome/therapy , Biopsy/methods , Cocaine Smoking/adverse effects , Cocaine Smoking/prevention & control , Humans , Kidney/pathology , Kidney/physiopathology , Kidney Function Tests/methods , Male , Middle Aged , Mutation , Prognosis , Renal Dialysis/methods , Renal Insufficiency/diagnosis , Renal Insufficiency/therapy , Thrombocytopenia/diagnosis , Thrombocytopenia/etiology , Thrombocytopenia/therapy , Thrombotic Microangiopathies/etiology , Thrombotic Microangiopathies/pathology , Treatment Outcome
9.
BMC Nephrol ; 21(1): 488, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33203373

ABSTRACT

BACKGROUND: Pregnancy-related Atypical Haemolytic Uremic Syndrome (P-aHUS) is a rare condition affecting genetically predisposed women during pregnancy. It is often difficult to diagnose and has a significant impact on maternal and foetal outcomes. It is characterised by microangiopathic haemolytic anaemia and kidney injury from thrombotic microangiopathy. CASE PRESENTATION: A 27-year-old female of Lebanese descent presented at 36 weeks' gestation with foetal death in-utero (FDIU) with placental abruption on a background of previously normal antenatal visits. She was coagulopathic and anaemic with anuric acute kidney injury, requiring emergency Caesarean section, intubation and dialysis. Her coagulopathy rapidly resolved, however, her anaemia and renal dysfunction persisted. A diagnosis of P-aHUS was made, and she was empirically treated with Eculizumab. Her ADAMTS13 level was normal, effectively excluding thrombotic thrombocytopenic purpura. Within 2 weeks of treatment her haematological parameters improved, and her renal function began to recover and within 2 months she became dialysis independent. CONCLUSION: This case highlights the challenges of a timely diagnosis of P-aHUS from other pregnancy-related diseases. Although our patient is dialysis-independent, her risk of relapse remains high with subsequent pregnancies. Currently we are awaiting her genetic sequencing to complete her assessment for underlying mutations and are determining the safest approach to a future planned pregnancy.


Subject(s)
Abruptio Placentae , Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/diagnosis , Complement Inactivating Agents/therapeutic use , Pregnancy Complications/diagnosis , Acute Kidney Injury/etiology , Adult , Anuria/etiology , Atypical Hemolytic Uremic Syndrome/complications , Atypical Hemolytic Uremic Syndrome/drug therapy , Diagnosis, Differential , Female , Fetal Death , Humans , Kidney/diagnostic imaging , Pregnancy , Pregnancy Complications/drug therapy , Thrombotic Microangiopathies/diagnosis
10.
J Paediatr Child Health ; 56(4): 577-580, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31705764

ABSTRACT

The late outcome in 55 children with infection-mediated haemolytic uremic syndrome (Shiga Toxin E Coli (STEC)-HUS and pneumococcal HUS) observed in 1979-1995 was followed up 23 years after disease onset. Of these, two were later confirmed to have atypical HUS (aHUS). Furthermore, of this population, five children had impaired kidney function at 3-months follow-up, which continued to deteriorate. These children had significant oliguria/anuria and hypertension during their illness requiring early dialysis and antihypertensive therapy. At 23 years post-disease onset, all five (100%) of these children have developed end-stage kidney disease or chronic kidney disease. Eculizumab is a monoclonal antibody that binds with high affinity to the C5 protein of the complement pathway, a major component of the pathophysiology of infection-mediated HUS. There are no long-term randomised controlled trials in the literature to support its use in such cases. Our 23-year follow-up of a population of severely affected children with infection-mediated HUS demonstrates a high percentage of chronic kidney disease and end-stage kidney disease (19%). Randomised controlled trials with eculizumab are now being conducted in this affected cohort.


Subject(s)
Escherichia coli , Hemolytic-Uremic Syndrome , Antibodies, Monoclonal, Humanized , Child , Follow-Up Studies , Hemolytic-Uremic Syndrome/complications , Hemolytic-Uremic Syndrome/drug therapy , Humans , Prognosis
11.
Epidemiol Infect ; 147: e164, 2019 01.
Article in English | MEDLINE | ID: mdl-31063106

ABSTRACT

In 2014-2016, we conducted a cross-sectional survey in 115 sheep, 104 beef and 82 dairy cattle herds to estimate Shiga toxin-producing Escherichia coli (STEC) prevalence, and collected data on human clinical cases of infection. Isolates were characterised (stx1, stx2, eae, ehxA) and serogroups O157 and O111 identified by PCR, and their antimicrobial resistance (AMR) profiles were determined by broth microdilution. STEC were more frequently isolated from beef cattle herds (63.5%) and sheep flocks (56.5%) than from dairy cattle herds (30.5%) (P < 0.001). A similar but non-significant trend was observed for O157:H7 STEC. In humans, mean annual incidence rate was 1.7 cases/100 000 inhabitants for O157 STEC and 4.7 for non-O157 STEC, but cases concentrated among younger patients. Distribution of virulence genes in STEC strains from ruminants differed from those from human clinical cases. Thus, stx2 was significantly associated with animal STEC isolates (O157 and non-O157), ehxA to ruminant O157 STEC (P = 0.004) and eae to human non-O157 STEC isolates (P < 0.001). Resistance was detected in 21.9% of human and 5.2% of animal O157 STEC isolates, whereas all non-O157 isolates were fully susceptible. In conclusion, STEC were widespread in ruminants, but only some carried virulence genes associated with severe disease in humans; AMR in ruminants was low but profiles were similar to those found in human isolates.


Subject(s)
Cattle Diseases/epidemiology , Escherichia coli Infections/epidemiology , Escherichia coli Infections/veterinary , Sheep Diseases/epidemiology , Shiga-Toxigenic Escherichia coli/classification , Virulence Factors/genetics , Zoonoses/epidemiology , Animals , Cattle , Cattle Diseases/microbiology , Cross-Sectional Studies , Escherichia coli Infections/microbiology , Genotype , Humans , Incidence , Prevalence , Sheep , Sheep Diseases/microbiology , Shiga-Toxigenic Escherichia coli/genetics , Shiga-Toxigenic Escherichia coli/isolation & purification , Shiga-Toxigenic Escherichia coli/pathogenicity , Virulence , Zoonoses/microbiology
12.
Rheumatol Int ; 39(2): 359-365, 2019 02.
Article in English | MEDLINE | ID: mdl-30554307

ABSTRACT

The correct diagnosis, classification and therapeutic management of thrombotic microangiopathies (TMA) continue to be a challenge for the clinician. We report a rare case of eosinophilic granulomatosis with polyangiitis (EGPA) as a trigger for complement-mediated TMA in a 57-year-old man who was successfully treated with corticoids, cyclophosphamide and therapeutic plasma exchange. Additionally, we review few other cases reported in the literature and the pathophysiological pathway of association between TMA and EGPA. We found that the mutual relationships between the inflammation triggered by vasculitis, the exacerbated complement activation, together with hypereosinophilia and endothelial damage seem to be the key in explaining the connection between both entities. We suggest that an understanding of the multi-causal nature of TMAs is crucial for the correct diagnosis and treatment of these patients.


Subject(s)
Eosinophilia/complications , Granulomatosis with Polyangiitis/complications , Thrombotic Microangiopathies/etiology , Complement Activation , Humans , Male , Middle Aged , Thrombotic Microangiopathies/drug therapy
13.
Scand J Immunol ; 88(1): e12671, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29706017

ABSTRACT

Factor H is an important regulator of complement activation in plasma and on cell surfaces in both humans and mice. If FH function is compromised, inappropriate complement activation on self-surfaces can have disastrous effects as seen in the kidney diseases atypical haemolytic uremic syndrome (aHUS) and C3 glomerulopathy. As FH constructs have been proposed to be used in treatment for these diseases, we studied the distribution of exogenous FH fragments in mice. Full-length mFH, mFH1-5 and mFH18-20 fragments were radiolabelled, and their distribution was examined in WT, FH-/- and FH-/- C3-/- mice in vivo. Whole body scintigraphy revealed accumulation of radioactivity in the abdominal part of the mice, but also to the thyroid gland and urinary bladder. At organ level in WT mice, some full-length FH accumulated in internal organs, but most of it remained in the circulation. Both of the mFH fragments accumulated in the kidneys and were excreted in urine. For mFH1-5, urinary secretion is the likely cause for the accumulation. Concentration of mFH18-20 to kidneys was slower, and at tissue level, mFH18-20 was localized at the proximal tubuli in WT and FH-/- C3-/- mice. No C3-independent binding to glomeruli was detected. In conclusion, these results show that glomerular glycosaminoglycans and sialic acids alone do not collect FH in kidneys. Deposition of C3 fragments is also needed, which implies that in aHUS, the problem is in simultaneous recognition of C3 fragments and glycosaminoglycans or sialic acids by FH, not just the inability of FH to recognize glomerular endothelium as such.


Subject(s)
Complement Factor H/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/metabolism , Tissue Distribution
14.
Epidemiol Infect ; 147: e6, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30182864

ABSTRACT

In Belgium, it is mandatory to report Shiga toxin-producing Escherichia coli (STEC) infections to the health inspection authorities. To facilitate the decision making regarding infection control measures, information about the risk factors for the development of the haemolytic uremic syndrome (HUS) can be helpful. We performed statistical analyses on a dataset of 411 Belgian STEC strains. Demographic and clinical patient characteristics as well as phenotypical and genotypical STEC strain characteristics were taken into account. Multivariate logistic regression models indicated that age categories ⩽5, 6-12 and ⩾75; the stx2 gene; and the eae gene were significant HUS development risk determinants. The stx2a subtype had the highest risk (OR 29.6, 95% CI 7.0-125.1), while all stx1 subtypes encompassed a significant lower risk (OR 0.3, 95% CI 0.1-0.5). Presence of the stx1 gene without stx2 encompassed a lower risk than the combined presence of stx1 and stx2, or stx2 solely. Based on these results, we propose a new virulence typing algorithm that will enable the National Reference Centre to provide the physicians and health inspection authorities with a risk classification for the development of HUS. We believe this will contribute to a more efficient STEC infection control management in Belgium.

15.
Epidemiol Infect ; 145(14): 2998-3006, 2017 10.
Article in English | MEDLINE | ID: mdl-28925349

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) is a significant cause of gastrointestinal infection and the haemolytic-uremic syndrome (HUS). STEC outbreaks are commonly associated with food but animal contact is increasingly being implicated in its transmission. We report an outbreak of STEC affecting young infants at a nursery in a rural community (three HUS cases, one definite case, one probable case, three possible cases and five carriers, based on the combination of clinical, epidemiological and laboratory data) identified using culture-based and molecular techniques. The investigation identified repeated animal contact (animal farming and petting) as a likely source of STEC introduction followed by horizontal transmission. Whole genome sequencing (WGS) was used for real-time investigation of the incident and revealed a unique strain of STEC O26:H11 carrying stx2a and intimin. Following a public health intervention, no additional cases have occurred. This is the first STEC outbreak reported from Israel. WGS proved as a useful tool for rapid laboratory characterization and typing of the outbreak strain and informed the public health response at an early stage of this unusual outbreak.


Subject(s)
Disease Outbreaks , Escherichia coli Infections/epidemiology , Hemolytic-Uremic Syndrome/epidemiology , Shiga-Toxigenic Escherichia coli/physiology , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Genomics , Hemolytic-Uremic Syndrome/microbiology , Humans , Infant , Israel/epidemiology , Nurseries, Infant , Phylogeny , Public Health , Sequence Analysis, DNA , Shiga-Toxigenic Escherichia coli/genetics
16.
Nephrology (Carlton) ; 22 Suppl 1: 4-6, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28176471

ABSTRACT

A 25-year-old man presented with microangiopathic haemolytic anaemia and acute kidney injury. With a normal ADAMTS-13 level, negative faecal shiga-toxin test and strong family history of atypical haemolytic uremic syndrome, he was commenced on eculizumab to good clinical response. Subsequent genetic testing revealed a heterozygous complement factor H mutation. Eculizumab was discontinued after 44 months of treatment, and he relapsed within 6 months, with the first sign being downtrending haptoglobin levels, with no other markers of haemolysis or thrombocytopaenia, 5 weeks prior to development of acute kidney injury. He was recommenced on eculizumab and to date still remains on it. This case highlights the unusual pattern of relapse and discusses the considerations for eculizumab discontinuation in patients with stable atypical haemolytic uremic syndrome receiving maintenance therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/drug therapy , Adult , Atypical Hemolytic Uremic Syndrome/etiology , Humans , Male , Recurrence , Withholding Treatment
17.
Euro Surveill ; 22(47)2017 Nov.
Article in English | MEDLINE | ID: mdl-29183554

ABSTRACT

IntroductionAt the beginning of 2016, an increase in paediatric haemolytic uremic syndrome (HUS) cases was observed in Romania. The microbiological investigations allowed isolation of Shiga toxin-producing Escherichia coli (STEC) O26 as the causative agent from most cases. Methods: An enhanced national surveillance of HUS and severe diarrhoea was established across the country following the identification of the first cases and was carried out until August 2016. A total of 15 strains were isolated from 10 HUS and five diarrhoea cases. Strains were characterised by virulence markers (i.e. stx type/subtype, eae, ehxA genes), phylogroup, genetic relatedness and clonality using PCR-based assays, PFGE and multilocus sequence typing (MLST). The first six strains were further characterised by whole genome sequencing (WGS). Results: Five PCR-defined genotypes were distinguished. All strains from HUS cases harboured stx2a and eae, with or without stx1a, while strains from diarrhoea cases carried exclusively stx1a and eae genes. PFGE resolved strains into multiple pulsotypes, compatible with a certain geographic segregation of the cases, and strains were assigned to phylogroup B1 and sequence type (ST) 21. WGS confirmed the results of conventional molecular methods, brought evidence of O26:H11 serotype, and complemented the virulence profiles. Discussion/conclusion: This first description of STEC O26 strains from cases in Romania showed that the isolates belonged to a diverse population. The virulence content of most strains highlighted a high risk for severe outcome in infected patients. Improving the national surveillance strategy for STEC infections in Romania needs to be further considered.


Subject(s)
Diarrhea/microbiology , Disease Outbreaks , Hemolytic-Uremic Syndrome/diagnosis , Shiga-Toxigenic Escherichia coli/isolation & purification , Virulence/genetics , Child, Preschool , Diarrhea/epidemiology , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Escherichia coli O157/genetics , Escherichia coli Proteins/genetics , Female , Hemolytic-Uremic Syndrome/epidemiology , Hemolytic-Uremic Syndrome/microbiology , Humans , Infant , Male , Multilocus Sequence Typing , Polymerase Chain Reaction , Population Surveillance , Romania/epidemiology , Serogroup , Shiga-Toxigenic Escherichia coli/genetics , Whole Genome Sequencing
18.
J Ayub Med Coll Abbottabad ; 29(2): 360-362, 2017.
Article in English | MEDLINE | ID: mdl-28718268

ABSTRACT

BACKGROUND: Atypical Haemolytic Uremic Syndrome (aHUS) is a rare life threatening entity characterized by thrombocytopenia, haemolytic anaemia and renal dysfunction. It is a thrombotic microangiopathy related to genetic mutations in the alternate complement pathway and has a distinct pathophysiology which makes it harder to distinguish from other microangiopathies. We present a case of a 25-year-old male patient with history of polysubstance abuse who presented with chest pain and dyspnoea. He admitted to using injectable oxymorphone (Opana) two weeks before presentation. Patient's vital signs were stable except for tachycardia and high blood pressure. On physical examination, epigastric tenderness and mild splenomegaly was appreciated. Urine Drug Screen was positive for oxycodone and opiates. Laboratory work up revealed haemolytic anaemia, thrombocytopenia and acute kidney injury. Extensive evaluation resulted in our impression of the disease being atypical haemolytic-uremic syndrome. He was managed with dialysis, intravenous steroids and plasmapheresis with improvement in his hematologic parameters.


Subject(s)
Atypical Hemolytic Uremic Syndrome/diagnosis , Oxymorphone/poisoning , Thrombotic Microangiopathies/chemically induced , Adult , Analgesics, Opioid/poisoning , Humans , Male , Renal Dialysis , Thrombotic Microangiopathies/diagnosis
19.
J Ayub Med Coll Abbottabad ; 29(3): 502-505, 2017.
Article in English | MEDLINE | ID: mdl-29076694

ABSTRACT

We report a case of a 38-year-old U.A.E national who presented with malignant hypertension and features of thrombotic microangiopathy. He presented with oliguria, renal failure, thrombocytopenia and haemolytic anaemia. He required several sessions of renal replacement therapy. ADAM 13 mutational analysis was sent to differentiate Thrombotic micro angiopathy due to thrombotic thrombocytopenic purpura (TTP) or malignant hypertension. Renal biopsy revealed histopathological features of malignant arteriolar nephrosclerosis (MANS). Haemolytic parameters improved after control of blood pressure and he was subsequently discharged with early nephrology follow up.


Subject(s)
ADAM Proteins/genetics , Hypertension, Malignant/diagnosis , Membrane Proteins/genetics , Mutation , Thrombotic Microangiopathies/diagnosis , Adult , Anemia, Hemolytic/diagnosis , Humans , Male
20.
Eur J Pediatr ; 175(4): 465-73, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26498648

ABSTRACT

UNLABELLED: Despite the severity of HUS and the fact that it represents a leading cause of acute kidney injury in children, the general epidemiology of HUS is all but well documented. The present study provides updated, population-based, purely epidemiological information on HUS in childhood from a large and densely populated area of northern Italy (9.6 million inhabitants, 1.6 million children). We systematically reviewed the files concerning patients with STEC-HUS and atypical HUS (aHUS) over a 10-year observation period (January 2003-December 2012). We included all incident cases with a documented first episode of HUS before the age of 18 years. We identified 101 cases of HUS during the 10 years. The overall mean annual incidence was 6.3 cases/million children aged <18 years (range 1.9-11.9), and 15.7/million of age-related population (MARP) among subjects aged <5 years; aHUS accounted for 11.9 % of the cases (mean incidence 0.75/MARP). The overall case fatality rate was 4.0 % (3.4 % STEC-HUS, 8.3 % aHUS). CONCLUSION: Given the public health impact of HUS, this study provides recent, population-based epidemiological data useful for healthcare planning and particularly for estimating the financial burden that healthcare providers might have to face in treating HUS, whose incidence rate seems to increase in Northern Italy. WHAT IS KNOWN: • HUS is a rare disease, but it represents the leading cause of acute kidney injury in children worldwide. • STEC-HUS (also called typical, D + HUS) is more common compared to atypical HUS, but recent, population-based epidemiological data (incidence) are scanty. What is New: • Comprehensive, population-based epidemiological data concerning both typical and atypical HUS based on a long observational period.


Subject(s)
Escherichia coli Infections/epidemiology , Escherichia coli/isolation & purification , Hemolytic-Uremic Syndrome/epidemiology , Adolescent , Child , Child, Preschool , Escherichia coli Infections/complications , Hemolytic-Uremic Syndrome/microbiology , Humans , Incidence , Infant , Italy/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL