Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 469
Filter
1.
Toxins (Basel) ; 15(12)2023 11 23.
Article in English | MEDLINE | ID: mdl-38133173

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli (STEC) infections cause outbreaks of severe disease in children ranging from bloody diarrhea to hemolytic uremic syndrome (HUS). The adherent factor intimin, encoded by eae, can facilitate the colonization process of strains and is frequently associated with severe disease. The purpose of this study was to examine and analyze the prevalence and polymorphisms of eae in clinical STEC strains from pediatric patients under 17 years old with and without HUS, and to assess the pathogenic risk of different eae subtypes. We studied 240 STEC strains isolated from pediatric patients in Finland with whole genome sequencing. The gene eae was present in 209 (87.1%) strains, among which 49 (23.4%) were from patients with HUS, and 160 (76.6%) were from patients without HUS. O157:H7 (126, 60.3%) was the most predominant serotype among eae-positive STEC strains. Twenty-three different eae genotypes were identified, which were categorized into five eae subtypes, i.e., γ1, ß3, ε1, θ and ζ3. The subtype eae-γ1 was significantly overrepresented in strains from patients aged 5-17 years, while ß3 and ε1 were more commonly found in strains from patients under 5 years. All O157:H7 strains carried eae-γ1; among non-O157 strains, strains of each serotype harbored one eae subtype. No association was observed between the presence of eae/its subtypes and HUS. However, the combination of eae-γ1+stx2a was significantly associated with HUS. In conclusion, this study demonstrated a high occurrence and genetic variety of eae in clinical STEC from pediatric patients under 17 years old in Finland, and that eae is not essential for STEC-associated HUS. However, the combination of certain eae subtypes with stx subtypes, i.e., eae-γ1+stx2a, may be used as risk predictors for the development of severe disease in children.


Subject(s)
Adhesins, Bacterial , Escherichia coli Infections , Escherichia coli Proteins , Hemolytic-Uremic Syndrome , Shiga-Toxigenic Escherichia coli , Adolescent , Child , Humans , Adhesins, Bacterial/genetics , Escherichia coli Infections/epidemiology , Escherichia coli O157/genetics , Escherichia coli Proteins/genetics , Finland/epidemiology , Hemolytic-Uremic Syndrome/epidemiology , Hemolytic-Uremic Syndrome/genetics , Serotyping , Shiga-Toxigenic Escherichia coli/genetics , Scandinavians and Nordic People
2.
Pediatr Nephrol ; 38(4): 1195-1203, 2023 04.
Article in English | MEDLINE | ID: mdl-35969277

ABSTRACT

BACKGROUND: Hemolytic uremic syndrome secondary to Shiga-toxin-producing Escherichia coli infection (STEC-HUS) generally shows a favorable outcome. Few cases develop extra-renal complications, since neurological involvement is an important cause of morbidity and mortality. The role of complement in STEC-HUS has been recently highlighted, and the use of eculizumab in severe cases has been communicated. HUS results from environmental and genetic factors, but the simultaneous occurrence of STEC and complement mutations remains undetermined. METHODS: A pediatric case with severe STEC-HUS carrying CFH mutations, with favorable response to eculizumab is analyzed. RESULTS: STEC-HUS was diagnosed in a 4-year-old girl with classic HUS, including low C3. Peritoneal dialysis was started due to hypertension, oligoanuria, and pleural effusion. She evolved with generalized tonic-clonic seizures and required mechanical ventilation. MRI reported multiple supra- and infratentorial ischemic lesions with laminar/striatal cortical necrosis and leukoencephalopathy. After two eculizumab doses, a significative stabilization in diuresis, blood pressure, creatinine, and C3 was achieved. At the third week, episodes of massive digestive bleeding and a life-threatening condition required a colectomy thus preserving the ileocecal valve. Due to atypical evolution, a genetic study was considered, identifying two heterozygous variants (CFH S1191L/V1197A). CONCLUSION: STEC-HUS in patients with a genetic predisposition has been previously reported, but the low frequency of occurrence makes it a rare disease. As in the present case, patients with atypical course might benefit from genetic analysis to evaluate early eculizumab initiation and to better understand its phenotype. A higher resolution version of the Graphical abstract is available as Supplementary information.


Subject(s)
Escherichia coli Infections , Hemolytic-Uremic Syndrome , Shiga-Toxigenic Escherichia coli , Female , Humans , Escherichia coli Infections/complications , Shiga-Toxigenic Escherichia coli/genetics , Hemolytic-Uremic Syndrome/complications , Hemolytic-Uremic Syndrome/drug therapy , Hemolytic-Uremic Syndrome/genetics , Complement System Proteins , Mutation
3.
Int J Hematol ; 116(5): 812-818, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35790695

ABSTRACT

Complement-mediated hemolytic uremic syndrome (CM-HUS) following chemotherapy for pediatric acute lymphoid neoplasms has rarely been reported. We report the case of an 8-year-old boy with T-lymphoblastic lymphoma (T-LBL) who developed CM-HUS with complement factor H (CFH) mutations (S1191L, V1197A) during induction therapy. Safe administration of chemotherapy after CM-HUS recovery was challenging. By closely monitoring hemolytic and renal parameters during the 2-year treatment period, we observed four episodes of microangiopathic hemolytic anemia (MAHA) with hypocomplementemia and low haptoglobin but no renal dysfunction or thrombocytopenia. Here, we describe the MAHA and CM-HUS episodes in the hopes of elucidating the complex pathophysiology of disorders associated with CFH mutation.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Hemolytic-Uremic Syndrome , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Purpura, Thrombotic Thrombocytopenic , Male , Humans , Child , Complement Factor H/genetics , Complement Factor H/therapeutic use , Hemolysis , Haptoglobins/therapeutic use , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/pathology , Hemolytic-Uremic Syndrome/therapy , Purpura, Thrombotic Thrombocytopenic/therapy , Complement System Proteins , Atypical Hemolytic Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome/genetics
4.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 30(2): 636-640, 2022 Apr.
Article in Chinese | MEDLINE | ID: mdl-35396010

ABSTRACT

Hemolytic uremic syndrome (HUS) is clinically rare, with high mortality and case fatality rates. In recent years, the research on HUS has been intensified and the pathophysiological mechanism has been continuously improved. At present, the main mechanism of pathogenesis is the excessive activation of complement alternative pathways mediated by complement-related gene mutations or the existence of antibodies. The treatment methods and strategies are also constantly updated, mainly including complement-blocking drugs such as Eculizumab, Lavalizumab, and Ravulizumab. In this review, the new developments in the pathogenesis and treatment of HUS is summarized, and provide references for the clinical treatment of HUS.


Subject(s)
Hemolytic-Uremic Syndrome , Complement System Proteins/genetics , Complement System Proteins/therapeutic use , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/therapy , Humans , Mutation
5.
Physiol Genomics ; 54(5): 153-165, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35384732

ABSTRACT

Shiga toxin 2 (Stx2) and lipopolysaccharide (LPS) contribute to the development of hemolytic uremic syndrome (HUS). Mouse models of HUS induced by LPS/Stx2 have been used for elucidating HUS pathophysiology and for therapeutic development. However, the underlying molecular mechanisms and detailed injury sites in this model remain unknown. We analyzed mouse kidneys after LPS/Stx2 administration using microarrays. Decreased urinary osmolality and urinary potassium were observed after LPS/Stx2 administration, suggestive of distal nephron disorders. A total of 1,212 and 1,016 differentially expressed genes were identified in microarrays at 6 h and 72 h after LPS/Stx2 administration, respectively, compared with those in controls. Ingenuity pathway analysis revealed activation of TNFR1/2, iNOS, and IL-6 signaling at both time points, and inhibition of pathways associated with lipid metabolism at 72 h only. The strongly downregulated genes in the 72-h group were expressed in the distal nephrons. In particular, genes associated with distal convoluted tubule (DCT) 2/connecting tubule (CNT) and principal cells of the cortical collecting duct (CCD) were downregulated to a greater extent than those associated with DCT1 and intercalated cells. Stx receptor globotriaosylceramide 3 (Gb3) revealed no colocalization with DCT1-specific PVALB and intercalated cell-specific SLC26A4 but did present colocalization with SLC12A3 (present in both DCT1 and DCT2), and AQP2 in principal cells. Gb3 localization tended to coincide with the segment in which the downregulated genes were present. Thus, the LPS/Stx2-induced kidney injury model represents damage to DCT2/CNT and principal cells in the CCD, based on molecular, biological, and physiological findings.


Subject(s)
Hemolytic-Uremic Syndrome , Shiga Toxin 2 , Animals , Aquaporin 2/metabolism , Hemolytic-Uremic Syndrome/chemically induced , Hemolytic-Uremic Syndrome/genetics , Lipopolysaccharides/pharmacology , Male , Mice , Shiga Toxin/metabolism , Shiga Toxin 2/genetics , Shiga Toxin 2/metabolism , Solute Carrier Family 12, Member 3/metabolism , Transcriptome/genetics
6.
Pediatr Nephrol ; 37(9): 1967-1980, 2022 09.
Article in English | MEDLINE | ID: mdl-35041041

ABSTRACT

The syndrome of thrombotic microangiopathy (TMA) is a clinical-pathological entity characterized by microangiopathic hemolytic anemia, thrombocytopenia, and end organ involvement. It comprises a spectrum of underlying etiologies that may differ in children and adults. In children, apart from ruling out shigatoxin-associated hemolytic uremic syndrome (HUS) and other infection-associated TMA like Streptococcus pneumoniae-HUS, rare inherited causes including complement-associated HUS, cobalamin defects, and mutations in diacylglycerol kinase epsilon gene must be investigated. TMA should also be considered in the setting of solid organ or hematopoietic stem cell transplantation. In this review, acquired and inherited causes of TMA are described with a focus on particularities of the main causes of TMA in children. A pragmatic approach that may help the clinician tailor evaluation and management is provided. The described approach will allow for early initiation of treatment while waiting for the definitive diagnosis of the underlying TMA.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hemolytic-Uremic Syndrome , Purpura, Thrombotic Thrombocytopenic , Thrombotic Microangiopathies , Child , Complement System Proteins , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/genetics , Humans , Purpura, Thrombotic Thrombocytopenic/diagnosis , Thrombotic Microangiopathies/diagnosis , Thrombotic Microangiopathies/etiology , Thrombotic Microangiopathies/therapy
7.
Nephrology (Carlton) ; 27(3): 231-237, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34796567

ABSTRACT

AIM: Pregnancy-associated hemolytic uremic syndrome (P-aHUS) is an important cause of peripartum acute kidney injury. Studies from Europe have described mutations in complement regulator genes, and data in Indian patients is scarce. Hence this study used multiplex ligation-dependent probe amplification (MLPA) to identify variants in complement genes in P-aHUS patients. METHODS: We present 17 patients of P-aHUS who were investigated for complement protein levels and genetic analysis with MLPA for complement genes. Plasma exchange therapy was offered to all patients presenting in acute phase. RESULTS: Mean age 26.74 (3.36) years with 15/17 delivered by caesarean section. Eleven patients received early (within 7 days) plasma exchange, three were dialysis-dependent at 3 months and seven were dialysis-free. Only one of the three patients receiving late (after 7 days) plasma exchange was dialysis-free. MLPA showed that 11 patients had heterozygous deletions of exons 3, 5, 6 of CFHR1 and upstream region of exons 1, 2, 3, 6 and intron 4 of CFHR3 gene while four patients had homozygous deletions at the same loci. Two patients had no MLPA-detectable variations. CONCLUSION: This study reports a high proportion of deletions of exons of CFHR1 & CFHR3 genes in Indian P-aHUS patients detectable by MLPA by copy number variations. This needs confirmation in large multicentre studies. Plasma exchange can be an effective therapy in the non-availability of Eculizumab.


Subject(s)
Blood Proteins/genetics , Complement C3b Inactivator Proteins/genetics , Gene Deletion , Hemolytic-Uremic Syndrome/genetics , Pregnancy Complications/genetics , Adult , Female , Humans , India , Pregnancy , Prospective Studies , Young Adult
8.
Curr Opin Microbiol ; 65: 183-190, 2022 02.
Article in English | MEDLINE | ID: mdl-34929548

ABSTRACT

The formation of attaching and effacing (A/E) lesions on intestinal epithelium, combined with Shiga toxin production, are hallmarks of enterohemorrhagic Escherichia coli (EHEC) infection that can lead to lethal hemolytic uremic syndrome. Although an animal infection model that fully recapitulates human disease remains elusive, mice orally infected with Citrobacter rodentium(ϕStx2dact), a natural murine pathogen lysogenized with an EHEC-derived Shiga toxin 2-producing bacteriophage, develop intestinal A/E lesions and toxin-dependent systemic disease. This model has facilitated investigation of how: (A) phage gene expression and prophage induction contribute to disease and are potentially triggered by antibiotic treatment; (B) virulence gene expression is altered by microbiota and the colonic metabolomic milieu; and (C) innate immune signaling is affected by Stx. Thus, the model provides a unique tool for accessing diverse aspects of EHEC pathogenesis.


Subject(s)
Bacteriophages , Enterohemorrhagic Escherichia coli , Escherichia coli Infections , Hemolytic-Uremic Syndrome , Animals , Bacteriophages/metabolism , Citrobacter rodentium/genetics , Citrobacter rodentium/metabolism , Disease Models, Animal , Enterohemorrhagic Escherichia coli/metabolism , Female , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/pathology , Humans , Intestinal Mucosa/metabolism , Male , Mice
9.
Toxins (Basel) ; 13(3)2021 03 18.
Article in English | MEDLINE | ID: mdl-33803852

ABSTRACT

Shiga toxins (Stxs) are classic bacterial toxins and major virulence factors of toxigenic Shigella dysenteriae and enterohemorrhagic Escherichia coli (EHEC). These toxins recognize a glycosphingolipid globotriaosylceramide (Gb3/CD77) as their receptor and inhibit protein synthesis in cells by cleaving 28S ribosomal RNA. They are the major cause of life-threatening complications such as hemolytic uremic syndrome (HUS), associated with severe cases of EHEC infection, which is the leading cause of acute kidney injury in children. The threat of Stxs is exacerbated by the lack of toxin inhibitors and effective treatment for HUS. Here, we briefly summarize the Stx structure, subtypes, in vitro and in vivo models, Gb3 expression and HUS, and then introduce recent studies using CRISPR-Cas9-mediated genome-wide screens to identify the host cell factors required for Stx action. We also summarize the latest progress in utilizing and engineering Stx components for biomedical applications.


Subject(s)
Escherichia coli Infections/metabolism , Hemolytic-Uremic Syndrome/metabolism , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Trihexosylceramides/metabolism , Animals , CRISPR-Cas Systems , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/microbiology , Host-Pathogen Interactions , Humans , Immunotoxins/therapeutic use , Models, Molecular , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Protein Conformation , Shiga Toxins/chemistry , Shiga Toxins/genetics , Shiga Toxins/therapeutic use , Shiga-Toxigenic Escherichia coli/genetics , Structure-Activity Relationship
10.
Methods Mol Biol ; 2291: 381-397, 2021.
Article in English | MEDLINE | ID: mdl-33704765

ABSTRACT

Shiga toxin-producing E. coli (STEC) is a common foodborne pathogen in developed countries. STEC generates "attaching and effacing" (AE) lesions on colonic epithelium, characterized by effacement of microvilli and the formation of actin "pedestals" beneath intimately attached bacteria. In addition, STEC are lysogenized with a phage that, upon induction, can produce potent Shiga toxins (Stx), potentially leading to both hemorrhagic colitis and hemolytic uremic syndrome. Investigation of the pathogenesis of this disease has been challenging because STEC does not readily colonize conventional mice.Citrobacter rodentium (CR) is a related mouse pathogen that also generates AE lesions. Whereas CR does not produce Stx, a murine model for STEC utilizes CR lysogenized with an E. coli-derived Stx phage, generating CR(Φstx), which both colonizes conventional mice and readily gives rise to systemic disease. We present here key methods for the use of CR(Φstx) infection as a highly predictable murine model for infection and disease by STEC. Importantly, we detail CR(Φstx) inoculation by feeding, determination of pathogen colonization, production of phage and toxin, and assessment of intestinal and renal pathology. These methods provide a framework for studying STEC-mediated systemic disease that may aid in the development of efficacious therapeutics.


Subject(s)
Bacteriophages , Citrobacter rodentium , Colitis , Gastrointestinal Hemorrhage , Hemolytic-Uremic Syndrome , Intestinal Mucosa , Lysogeny , Shiga Toxins , Shiga-Toxigenic Escherichia coli , Animals , Bacteriophages/genetics , Bacteriophages/metabolism , Citrobacter rodentium/genetics , Citrobacter rodentium/metabolism , Citrobacter rodentium/pathogenicity , Citrobacter rodentium/virology , Colitis/genetics , Colitis/metabolism , Colitis/microbiology , Disease Models, Animal , Gastrointestinal Hemorrhage/genetics , Gastrointestinal Hemorrhage/metabolism , Gastrointestinal Hemorrhage/microbiology , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Shiga Toxins/biosynthesis , Shiga Toxins/genetics
11.
Proc Natl Acad Sci U S A ; 117(40): 25055-25065, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32968018

ABSTRACT

Enterohemorrhagic Escherichia coli is a significant human pathogen that causes disease ranging from hemorrhagic colitis to hemolytic uremic syndrome. The latter can lead to potentially fatal renal failure and is caused by the release of Shiga toxins that are encoded within lambdoid bacteriophages. The toxins are encoded within the late transcript of the phage and are regulated by antitermination of the PR' late promoter during lytic induction of the phage. During lysogeny, the late transcript is prematurely terminated at tR' immediately downstream of PR', generating a short RNA that is a byproduct of antitermination regulation. We demonstrate that this short transcript binds the small RNA chaperone Hfq, and is processed into a stable 74-nt regulatory small RNA that we have termed StxS. StxS represses expression of Shiga toxin 1 under lysogenic conditions through direct interactions with the stx1AB transcript. StxS acts in trans to activate expression of the general stress response sigma factor, RpoS, through direct interactions with an activating seed sequence within the 5' UTR. Activation of RpoS promotes high cell density growth under nutrient-limiting conditions. Many phages utilize antitermination to regulate the lytic/lysogenic switch and our results demonstrate that short RNAs generated as a byproduct of this regulation can acquire regulatory small RNA functions that modulate host fitness.


Subject(s)
Enterohemorrhagic Escherichia coli/genetics , Hemolytic-Uremic Syndrome/genetics , RNA, Small Untranslated/genetics , Shiga Toxin/genetics , Bacterial Proteins/genetics , Bacteriophage lambda/genetics , Bacteriophage lambda/pathogenicity , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/genetics , Hemolytic-Uremic Syndrome/microbiology , Host Factor 1 Protein/genetics , Humans , Lysogeny/genetics , Promoter Regions, Genetic/genetics , Regulatory Sequences, Ribonucleic Acid/genetics , Sigma Factor/genetics
12.
Toxins (Basel) ; 12(7)2020 07 03.
Article in English | MEDLINE | ID: mdl-32635212

ABSTRACT

Hemolytic uremic syndrome (HUS) is a severe renal disease that is often preceded by infection with Shiga toxin (Stx)-producing Escherichia coli (STEC). The exact mechanism of Stx-mediated inflammation on human glomerular microvascular endothelial cells (HGMVECs) during HUS is still not well understood. In this study, we investigated the effect of Stx1 on the gene expression of proteins involved in leucocyte-mediated and complement-mediated inflammation. Our results showed that Stx1 enhances the mRNA and protein expression of heparan sulfate proteoglycan (HSPG) syndecan-4 in HGMVECs pre-stimulated with tumor necrosis factor α (TNFα). CD44 was upregulated on mRNA but not on protein level; no effect on the mRNA expression of other tested HSPGs glypican-1 and betaglycan was observed. Furthermore, Stx1 upregulated the mRNA, cell surface expression, and supernatant levels of the intercellular adhesion molecule-1 (ICAM-1) in HGMVECs. Interestingly, no effect on the protein levels of alternative pathway (AP) components was observed, although C3 mRNA was upregulated. All observed effects were much stronger in HGMVECs than in human umbilical endothelial cells (HUVECs), a common model cell type used in endothelial studies. Our results provide new insights into the role of Stx1 in the pathogenesis of HUS. Possibilities to target the overexpression of syndecan-4 and ICAM-1 for STEC-HUS therapy should be investigated in future studies.


Subject(s)
Endothelial Cells/drug effects , Hemolytic-Uremic Syndrome/etiology , Intercellular Adhesion Molecule-1/metabolism , Kidney Glomerulus/blood supply , Microvessels/drug effects , Shiga Toxin 1/toxicity , Syndecan-4/metabolism , Cells, Cultured , Complement System Proteins/genetics , Complement System Proteins/metabolism , Endothelial Cells/metabolism , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/metabolism , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Humans , Intercellular Adhesion Molecule-1/genetics , Microvessels/metabolism , Syndecan-4/genetics , Up-Regulation
13.
Eur J Med Genet ; 63(8): 103953, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32413569

ABSTRACT

Atypical haemolytic uraemic syndrome and steroid-resistant nephrotic syndrome are highly rare kidney diseases that can occur in childhood. In some cases, genetic variants may trigger these conditions, although in atypical haemolytic uraemic syndrome they mostly confer only a predisposition to the disease. Most variants causing atypical haemolytic uraemic syndrome were identified in genes encoding proteins regulating the complement pathway; on the other hand, there are approximately 58 genes encoding distinct proteins primarily causing steroid-resistant nephrotic syndrome. We present a child with steroid-resistant nephrotic syndrome and a confirmed homozygous c.966G > A, p.Trp322Ter pathogenic variant in DGKE. This variant was also found in compound with a novel DGKE heterozygous deletion c.171delG, p.Ser58Alafs*111 in a patient from our paediatric cohort with atypical haemolytic uraemic syndrome. Both cases presented with hypertension, nephrotic proteinuria and severe acute kidney injury followed by renal recovery; however, their renal histology was different. In this paper, we deal with the clinical course of children with disrupted DGKE, including the steroid-resistant nephrotic syndrome and atypical haemolytic uraemic syndrome overlap.


Subject(s)
Diacylglycerol Kinase/genetics , Hemolytic-Uremic Syndrome/genetics , Nephrotic Syndrome/congenital , Phenotype , Adolescent , Child , Female , Hemolytic-Uremic Syndrome/pathology , Homozygote , Humans , Kidney/pathology , Mutation , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology
14.
Crit Care Clin ; 36(2): 333-356, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32172817

ABSTRACT

Hemolytic uremic syndrome is characterized by microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. Disseminated intravascular coagulation, thrombotic thrombocytopenic purpura, and hemolytic uremic syndrome have a similar clinical presentation. Diagnostic needs to be prompt to decrease mortality, because identifying the different disorders can help to tailor specific, effective therapies. However, diagnosis is challenging and morbidity and mortality remain high, especially in the critically ill population. Development of clinical prediction scores and rapid diagnostic tests for hemolytic uremic syndrome based on mechanistic knowledge are needed to facilitate early diagnosis and assign timely specific treatments to patients with hemolytic uremic syndrome variants.


Subject(s)
Critical Illness , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/etiology , Algorithms , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/etiology , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/therapy , Diagnosis, Differential , Early Diagnosis , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/therapy , Humans , Prognosis , Risk Factors , Shiga Toxin/toxicity
15.
Crit Care Clin ; 36(2): 379-390, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32172819

ABSTRACT

Thrombocytopenia-associated multiple organ failure is a clinical phenotype encompassing a spectrum of syndromes associated with disseminated microvascular thromboses. Autopsies performed in patients that died with thrombotic thrombocytopenic purpura, hemolytic uremic syndrome, or disseminated intravascular coagulation reveal specific findings that can differentiate these 3 entities. Significant advancements have been made in our understanding of the pathologic mechanisms of these syndromes. Von Willebrand factor and ADAMTS-13 play a central role in thrombotic thrombocytopenic purpura. Shiga toxins and the complement pathway drive the hemolytic uremic syndrome pathology. Tissue factor activity is vital in the development of disseminated intravascular coagulation.


Subject(s)
Multiple Organ Failure/etiology , Thrombocytopenia/complications , Disseminated Intravascular Coagulation/complications , Disseminated Intravascular Coagulation/genetics , Disseminated Intravascular Coagulation/therapy , Genetic Predisposition to Disease , Hemolytic-Uremic Syndrome/complications , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/therapy , Humans , Multiple Organ Failure/genetics , Multiple Organ Failure/therapy , Phenotype , Plasma Exchange , Thrombocytopenia/genetics , Thrombocytopenia/therapy
16.
Sci Rep ; 9(1): 14362, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31591425

ABSTRACT

Shiga toxin (Stx) is the main virulence factor of enterohemorrhagic Escherichia coli (EHEC), that cause gastrointestinal infection leading to hemolytic uremic syndrome. The aim of this study was to investigate if Stx signals via ATP and if blockade of purinergic receptors could be protective. Stx induced ATP release from HeLa cells and in a mouse model. Toxin induced rapid calcium influx into HeLa cells, as well as platelets, and a P2X1 receptor antagonist, NF449, abolished this effect. Likewise, the P2X antagonist suramin blocked calcium influx in Hela cells. NF449 did not affect toxin intracellular retrograde transport, however, cells pre-treated with NF449 exhibited significantly higher viability after exposure to Stx for 24 hours, compared to untreated cells. NF449 protected HeLa cells from protein synthesis inhibition and from Stx-induced apoptosis, assayed by caspase 3/7 activity. The latter effect was confirmed by P2X1 receptor silencing. Stx induced the release of toxin-positive HeLa cell- and platelet-derived microvesicles, detected by flow cytometry, an effect significantly reduced by NF449 or suramin. Suramin decreased microvesicle levels in mice injected with Stx or inoculated with Stx-producing EHEC. Taken together, we describe a novel mechanism of Stx-mediated cellular injury associated with ATP signaling and inhibited by P2X receptor blockade.


Subject(s)
Escherichia coli Infections/drug therapy , Hemolytic-Uremic Syndrome/drug therapy , Receptors, Purinergic P2X1/genetics , Shiga Toxin/genetics , Adenosine Triphosphate/metabolism , Animals , Benzenesulfonates/pharmacology , Blood Platelets/microbiology , Enterohemorrhagic Escherichia coli/drug effects , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , HeLa Cells , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Humans , Mice , Purinergic P2X Receptor Antagonists/pharmacology , Shiga Toxin/antagonists & inhibitors
17.
Am J Physiol Renal Physiol ; 317(6): F1420-F1429, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31482731

ABSTRACT

Typical hemolytic uremic syndrome is caused by Shiga toxin (Stx2) and lipopolysaccharide (LPS) of Escherichia coli and leads to acute kidney injury. The role of innate immunity in this pathogenesis is unclear. We analyzed the role of high mobility group box 1 (HMGB1) at the onset of disease in a murine model. C57BL/6 mice were intraperitoneally administered saline (group A), anti-HMGB1 monoclonal antibody (group B), Stx2 and LPS to elicit severe disease (group C), or Stx2, LPS, and anti-HMGB1 antibody (group D). While all mice in group C died by day 5 of the experiment, all mice in group D survived. Anemia and thrombocytopenia were pronounced and plasma creatinine levels were significantly elevated in group C only at 72 h. While at 72 h after toxin administration the glomerulus tissue in group C showed pathology similar to that of humans, mesangial cell proliferation was seen in group D. Plasma HMGB1 levels in group C peaked 3 h after administration and were higher than those in other groups. Expression of the receptor of advanced glycation end products and NF-κB, involved in HMGB1 signaling, was significantly elevated in group C but not in group D. Administration of anti-HMGB1 antibody in a murine model of severe disease inhibited plasma HMGB1 and promoted amelioration of tissue damage. HMGB1 was found to be involved in the disease pathology; therefore, controlling HMGB1 activity might inhibit disease progression.


Subject(s)
HMGB1 Protein/genetics , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/pathology , Anemia/etiology , Animals , Antibodies, Blocking , Creatinine/blood , Cytokines/analysis , Cytokines/metabolism , Glycation End Products, Advanced/metabolism , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/immunology , Hemolytic-Uremic Syndrome/chemically induced , Kidney Glomerulus/pathology , Lipopolysaccharides , Male , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Survival Analysis , Syntaxin 1/metabolism , Thrombocytopenia/etiology
18.
Indian J Med Res ; 149(3): 412-417, 2019 03.
Article in English | MEDLINE | ID: mdl-31249208

ABSTRACT

Background & objectives: : Shiga toxin (Stx) is produced by Shigella dysenteriae, a Gram-negative, facultative anaerobic bacillus that causes shigellosis, haemolytic uraemic syndrome (HUS) and Reiter's syndrome. The detection methods for shiga toxin needs to be rapid, accurate, reliable and must be extensively evaluated under field conditions. The aim of this study was to develop rapid, sensitive and specific detection method for Stx. Methods: : Mice and rabbits were immunized with purified recombinant Shiga toxin B (rStxB). Using these antibodies dot ELISA, sandwich ELISA and flow through assay were developed. Results: : The high-titre antibodies specifically reacted with purified rStxB. Dot-ELISA, sandwich ELISA and flow-through assay were developed and standardized that could detect StxB with limit of detection (LOD) of 9.75, 9.7 ng/ml and 0.46 µg/cassette, respectively. Interpretation & conclusions: : The rStxB was used to produce antibodies to avoid handling of pathogen. The Flow through assay 'developed was specific, rapid and field amenable.


Subject(s)
Dysentery, Bacillary/diagnosis , Hemolytic-Uremic Syndrome/diagnosis , Shiga Toxin/isolation & purification , Shigella dysenteriae/genetics , Animals , Antibodies, Bacterial/genetics , Antibodies, Bacterial/immunology , Arthritis, Reactive/diagnosis , Arthritis, Reactive/genetics , Arthritis, Reactive/microbiology , Dysentery, Bacillary/genetics , Dysentery, Bacillary/microbiology , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/microbiology , Humans , Mice , Shiga Toxin/genetics , Shigella dysenteriae/pathogenicity
19.
CEN Case Rep ; 8(4): 266-270, 2019 11.
Article in English | MEDLINE | ID: mdl-31177384

ABSTRACT

Thrombotic microangiopathies include hemolytic uremic syndrome (HUS) and thrombotic thrombocytopenic purpura (TTP). Measurement of plasma levels of "a disintegrin-like and metalloproteinase with thrombospondin type 1 motifs 13" (ADAMTS13) activity can distinguish HUS from TTP. Reduced plasma ADAMTS13 activity (< 10% normal range) is atypical for HUS, but not for TTP. However, we detected reduced ADAMTS13 activity in a patient with Shiga toxin-producing Escherichia coli-associated HUS caused by non-IgG anti-ADMTS13 autoantibodies. Furthermore, the patient exhibited possible genetic abnormalities associated with atypical HUS. The patient fully recovered after administration of supportive therapy. To the best of our knowledge, very few cases of STEC-HUS with reduced ADAMTS13 activity have been reported; thus far, none have described the presence of non-IgG anti-ADMTS13 autoantibodies. Therefore, we suggest that anti-ADAMTS13 analyses should be performed in patients diagnosed with STEC-HUS, especially in those who present with prolonged healing or unexpected clinical symptoms.


Subject(s)
ADAMTS13 Protein/blood , Disintegrins/blood , Hemolytic-Uremic Syndrome/microbiology , Shiga-Toxigenic Escherichia coli/isolation & purification , Autoantibodies/blood , Erythrocyte Transfusion/methods , Female , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/therapy , Humans , Infusions, Intravenous/methods , Membrane Cofactor Protein/genetics , Purpura, Thrombotic Thrombocytopenic/diagnosis , Renal Insufficiency/diagnosis , Renal Insufficiency/etiology , Severity of Illness Index , Thrombotic Microangiopathies/complications , Thrombotic Microangiopathies/diagnosis , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL