Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 698
Filter
1.
Microb Pathog ; 186: 106482, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38086442

ABSTRACT

BACKGROUND: Shiga toxin-producing Escherichia coli is the main cause of post-diarrheal hemolytic uremic syndrome (HUS) which produces acute kidney injury mainly in children, although it can also affect adults. The kidneys are the organs most affected by Shiga toxin type 2 (Stx2) in patients with HUS. However, previous studies in pregnant rats showed that a sublethal dose of Stx2 causes severe damage in the uteroplacental unit and induces abortion, whereas produces mild to moderate renal damage. The aim of the present work was to study the progression of renal injury caused by a sublethal dose of Stx2, as well as renal recovery, in pregnant and non-pregnant rats, and to investigate whether pregnancy physiology may affect renal damage progression mediated by Stx2. METHODS: Renal function and histopathology was evaluated in pregnant rats intraperitoneally injected with a sublethal dose of Stx2 (0.5 ng/g bwt) at the early stage of gestation (day 8 of gestation), and results in these rats were compared over time with those observed in non-pregnant female rats injected with the same Stx2 dose. Hence, progression of cell proliferation and dedifferentiation in renal tubular epithelia was also investigated. RESULTS: The sublethal dose of Stx2 induced abortion in pregnant rats as well as a significant more extended functional and histological renal injury in non-pregnant rats than in pregnant rats. Stx2 also caused decreased ability to concentrate urine in non-pregnant rats compared to their controls. However, renal water handling in pregnant rats was not altered by Stx2, and was significantly different than in non-pregnant rats. The greatest renal injury in both pregnant and non-pregnant rats was observed at 4 days post-Stx2 injection, and coincided with a significant increase in tubular epithelial proliferation. Expression of mesenchymal marker vimentin in tubular epithelia was consistent with the level of tubular damage, being higher in non-pregnant rats than in pregnant rats. Recovery from Stx2-induced kidney injury was faster in pregnant rats than in non-pregnant rats. CONCLUSIONS: Adaptive mechanisms developed during pregnancy such as changes in water handle and renal hemodynamic may contribute to lessen the Stx2-induced renal injury, perhaps at the expense of fetal loss.


Subject(s)
Hemolytic-Uremic Syndrome , Shiga-Toxigenic Escherichia coli , Humans , Pregnancy , Child , Adult , Rats , Female , Animals , Shiga Toxin 2/toxicity , Kidney/pathology , Hemolytic-Uremic Syndrome/pathology , Water , Regeneration
2.
Toxicon ; 236: 107349, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37979924

ABSTRACT

Shiga toxin producing Escherichia coli (STEC) are foodborne pathogens that release Shiga toxin (Stx), virulence factor responsible for the development of Hemolytic Uremic Syndrome (HUS). Stx causes endothelial cell damage, which leads to platelets deposition and thrombi formation within the microvasculature. It has been described that Stx activates blood cells and induces the shedding of proinflammatory and prothrombotic microvesicles (MVs) containing the toxin. In this sense, it has been postulated that MVs containing Stx2 (MVs-Stx2+) can contribute to the physiopathology of HUS, allowing Stx2 to reach the target organs while evading the immune system. In this work, we propose that circulating MVs-Stx2+ can be a potential biomarker for the diagnosis and prognosis of STEC infections and HUS progression. We developed a rat HUS model by the intraperitoneal injection of a sublethal dose of Stx2 and observed: decrease in body weight, increase of creatinine and urea levels, decrease of creatinine clearance and histological renal damages. After characterization of renal damages, we investigated circulating total MVs and MVs-Stx2+ by flow cytometry at different times after Stx2 injection. Additionally, we evaluated the correlation of biochemical parameters such as creatinine and urea in plasma with MVs-Stx2+. As a result, we found a significant circulation of MVs-Stx2+ at 72 and 96 h after Stx2 injection, nevertheless no correlation with creatinine and urea plasma levels were detected. Our results suggest that MVs-Stx2+ may be an additional biomarker for the characterization and diagnosis of HUS progression. A further analysis is required in order to validate MVs-Stx2+ as biomarker of the disease.


Subject(s)
Escherichia coli Infections , Hemolytic-Uremic Syndrome , Shiga-Toxigenic Escherichia coli , Rats , Animals , Shiga Toxin 2/toxicity , Creatinine , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/pathology , Urea , Biomarkers
3.
Med ; 4(11): 761-777.e8, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37863058

ABSTRACT

BACKGROUND: Shiga toxin (Stx)-producing Escherichia coli hemolytic uremic syndrome (STEC-HUS) is the leading cause of acute kidney injury in children, with an associated mortality of up to 5%. The mechanisms underlying STEC-HUS and why the glomerular microvasculature is so susceptible to injury following systemic Stx infection are unclear. METHODS: Transgenic mice were engineered to express the Stx receptor (Gb3) exclusively in their kidney podocytes (Pod-Gb3) and challenged with systemic Stx. Human glomerular cell models and kidney biopsies from patients with STEC-HUS were also studied. FINDINGS: Stx-challenged Pod-Gb3 mice developed STEC-HUS. This was mediated by a reduction in podocyte vascular endothelial growth factor A (VEGF-A), which led to loss of glomerular endothelial cell (GEnC) glycocalyx, a reduction in GEnC inhibitory complement factor H binding, and local activation of the complement pathway. Early therapeutic inhibition of the terminal complement pathway with a C5 inhibitor rescued this podocyte-driven, Stx-induced HUS phenotype. CONCLUSIONS: This study potentially explains why systemic Stx exposure targets the glomerulus and supports the early use of terminal complement pathway inhibition in this devastating disease. FUNDING: This work was supported by the UK Medical Research Council (MRC) (grant nos. G0901987 and MR/K010492/1) and Kidney Research UK (grant nos. TF_007_20151127, RP42/2012, and SP/FSGS1/2013). The Mary Lyon Center is part of the MRC Harwell Institute and is funded by the MRC (A410).


Subject(s)
Escherichia coli Infections , Hemolytic-Uremic Syndrome , Kidney Diseases , Podocytes , Shiga-Toxigenic Escherichia coli , Child , Humans , Mice , Animals , Podocytes/metabolism , Podocytes/pathology , Shiga Toxin/genetics , Shiga Toxin/metabolism , Shiga Toxin/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/therapeutic use , Escherichia coli Infections/complications , Escherichia coli Infections/drug therapy , Escherichia coli Infections/metabolism , Hemolytic-Uremic Syndrome/drug therapy , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/pathology , Shiga-Toxigenic Escherichia coli/metabolism , Complement Activation , Kidney Diseases/pathology
4.
mSphere ; 8(4): e0014223, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37358300

ABSTRACT

Streptococcus pneumoniae-induced hemolytic uremic syndrome (Sp-HUS) is a kidney disease characterized by microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. This disease is frequently underdiagnosed and its pathophysiology is poorly understood. In this work, we compared clinical strains, isolated from infant Sp-HUS patients, with a reference pathogenic strain D39, for host cytotoxicity and further explored the role of Sp-derived extracellular vesicles (EVs) in the pathogenesis of an HUS infection. In comparison with the wild-type strain, pneumococcal HUS strains caused significant lysis of human erythrocytes and increased the release of hydrogen peroxide. Isolated Sp-HUS EVs were characterized by performing dynamic light-scattering microscopy and proteomic analysis. Sp-HUS strain released EVs at a constant concentration during growth, yet the size of the EVs varied and several subpopulations emerged at later time points. The cargo of the Sp-HUS EVs included several virulence factors at high abundance, i.e., the ribosomal subunit assembly factor BipA, the pneumococcal surface protein A, the lytic enzyme LytC, several sugar utilization, and fatty acid synthesis proteins. Sp-HUS EVs strongly downregulated the expression of the endothelial surface marker platelet endothelial cell adhesion molecule-1 and were internalized by human endothelial cells. Sp-HUS EVs elicited the release of pro-inflammatory cytokines (interleukin [IL]-1ß, IL-6) and chemokines (CCL2, CCL3, CXCL1) by human monocytes. These findings shed new light on the overall function of Sp-EVs, in the scope of infection-mediated HUS, and suggest new avenues of research for exploring the usefulness of Sp-EVs as therapeutic and diagnostic targets. IMPORTANCE Streptococcus pneumoniae-associated hemolytic uremic syndrome (Sp-HUS) is a serious and underdiagnosed deadly complication of invasive pneumococcal disease. Despite the introduction of the pneumococcal vaccine, cases of Sp-HUS continue to emerge, especially in children under the age of 2. While a lot has been studied regarding pneumococcal proteins and their role on Sp-HUS pathophysiology, little is known about the role of extracellular vesicles (EVs). In our work, we isolate and initially characterize EVs from a reference pathogenic strain (D39) and a strain isolated from a 2-year-old patient suffering from Sp-HUS. We demonstrate that despite lacking cytotoxicity toward human cells, Sp-HUS EVs are highly internalized by endothelial cells and can trigger cytokine and chemokine production in monocytes. In addition, this work specifically highlights the distinct morphological characteristics of Sp-HUS EVs and their unique cargo. Overall, this work sheds new light into potentially relevant players contained in EVs that might elucidate about pneumococcal EVs biogenesis or pose as interesting candidates for vaccine design.


Subject(s)
Extracellular Vesicles , Hemolytic-Uremic Syndrome , Infant , Child , Humans , Child, Preschool , Streptococcus pneumoniae , Endothelial Cells/pathology , Proteomics , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/pathology , Cytokines , Pneumococcal Vaccines
5.
Am J Ther ; 30(3): e209-e219, 2023 May 01.
Article in English | MEDLINE | ID: mdl-37104648

ABSTRACT

BACKGROUND: Complement-mediated HUS (CM-HUS) and paroxysmal nocturnal hemoglobinuria (PNH) are rare hematologic disorders that cause dysregulation and hyperactivation of the complement system. Historically, treatment of CM-HUS involved plasma exchange (PLEX), often with limited benefit and variable tolerance. Conversely, PNH was treated with supportive care or hemopoietic stem cell transplant. Within the last decade, monoclonal antibody therapies that block terminal complement pathway activation, have emerged as less invasive and more efficacious options for management of both disorders. This manuscript seeks to discuss a relevant clinical case of CM-HUS and the evolving landscape of complement inhibitor therapies for CM-HUS and PNH. AREAS OF UNCERTAINTY: Eculizumab, the first humanized anti-C5 monoclonal antibody, has been the standard of care in treating CM-HUS and PNH for over a decade. Although eculizumab has remained an effective agent, the variability in ease and frequency of administration has remained an obstacle for patients. The development of novel complement inhibitor therapies with longer half-lives, has allowed for changes in frequency and route of administration, thus improving patient QOL. However, there are limited prospective clinical trial data given disease rarity, and limited information on variable infusion frequency and length of treatment. THERAPEUTIC ADVANCES: Recently, there has been a push to formulate complement inhibitors that improve QOL while maintaining efficacy. Ravulizumab, a derivative of eculizumab, was developed to allow for less frequent administration, while remaining efficacious. In addition, the novel oral and subcutaneous therapies, danicopan and crovalimab, respectively, along with pegcetacoplan are currently undergoing active clinical trials, and poised to further reduce treatment burden. CONCLUSION: Complement inhibitor therapies have changed the treatment landscape for CM-HUS and PNH. With a significant emphasis on patient QOL, novel therapies continue to emerge and require an in-depth review of their appropriate use and efficacy in these rare disorders. CLINICAL CASE: A 47-year-old woman with hypertension and hyperlipidemia presented with shortness of breath and was found to have hypertensive emergency in the setting of acute renal failure. Her serum creatinine was 13.9 mg/dL; elevated from 1.43 mg/dL 2 years before. The differential diagnosis for her acute kidney injury (AKI) included infectious, autoimmune, and hematologic processes. Infectious work-up was negative. ADAMTS13 activity level was not low at 72.9%, ruling out thrombotic thrombocytopenic purpura (TTP). Patient underwent a renal biopsy, which revealed acute on chronic thrombotic microangiopathy (TMA). A trial of eculizumab was initiated with concurrent hemodialysis. The diagnosis of CM-HUS was later confirmed by a heterozygous mutation in complement factor I (CFI), resulting in increased membrane attack complex (MAC) cascade activation. The patient was maintained on biweekly eculizumab and was eventually transitioned to ravulizumab infusions as an outpatient. Her renal failure did not recover, and the patient remains on hemodialysis while awaiting kidney transplantation.


Subject(s)
Antibodies, Monoclonal, Humanized , Complement Inactivating Agents , Hemoglobinuria, Paroxysmal , Hemolytic-Uremic Syndrome , Humans , Female , Middle Aged , Thrombotic Microangiopathies/diagnosis , Thrombotic Microangiopathies/therapy , Hemoglobinuria, Paroxysmal/diagnosis , Hemoglobinuria, Paroxysmal/pathology , Hemoglobinuria, Paroxysmal/therapy , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/pathology , Hemolytic-Uremic Syndrome/therapy , Complement Inactivating Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Clinical Trials as Topic
6.
Rev. Hosp. Ital. B. Aires (2004) ; 43(1): 17-20, mar. 2023. ilus
Article in Spanish | LILACS, UNISALUD, BINACIS | ID: biblio-1434216

ABSTRACT

El síndrome urémico hemolítico (SUH), descripto en 1955, se caracteriza por la tríada de anemia hemolítica no inmunomediada, trombocitopenia y lesión renal aguda. En su patogenia interviene la toxina Shiga, producida con mayor frecuencia por E. coli O157:H. Puede manifestarse a cualquier edad, aunque es infrecuente en adultos, y se desarrolla en forma esporádica o en brote. Se presenta con un cuadro de dolor abdominal, diarrea, fiebre y vómitos. Puede afectar el sistema nervioso central, pulmones, páncreas y corazón. En adultos, el síndrome evoluciona tras un período de incubación de 1 semana posterior a la diarrea y tiene alta morbimortalidad, a diferencia de los casos pediátricos. Presentamos el caso de una paciente adulta, que cursó internación por síndrome urémico hemolítico. (AU)


Hemolytic uremic syndrome (HUS), described in 1955, is characterized by the triad of non-immune mediated hemolytic anemia, thrombocytopenia, and acute kidney injury. Shiga toxin, produced most frequently by E coli O157:H, is involved in its pathogenesis. Hus can manifest at any age, although it is rare in adults and develops sporadically or in outbreaks. HUS presents with a picture of abdominal pain, diarrhea, fever and vomiting. It can affect the central nervous system, lungs, pancreas, and heart.In adults, the syndrome evolves after an incubation period of 1 week after diarrhea, with high morbidity and mortality, unlike pediatric cases.We present the case of an adult patient who was hospitalized for hemolytic uremic syndrome. (AU)


Subject(s)
Humans , Female , Middle Aged , Escherichia coli O157/isolation & purification , Escherichia coli Infections/complications , Hemolytic-Uremic Syndrome/pathology , Hemolytic-Uremic Syndrome/diagnostic imaging , Polymerase Chain Reaction , Diarrhea/etiology , Hemolytic-Uremic Syndrome/diet therapy , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/therapy , Infusions, Parenteral , Kidney Function Tests
7.
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
8.
EMBO Mol Med ; 14(1): e14678, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34842355

ABSTRACT

Shiga toxins (Stxs) produced by enterohemorrhagic Escherichia coli (EHEC) are the major virulence factors responsible for hemorrhagic colitis, which can lead to life-threatening systemic complications including acute renal failure (hemolytic uremic syndrome) and neuropathy. Here, we report that O-GlcNAcylation, a type of post-translational modification, was acutely increased upon induction of endoplasmic reticulum (ER) stress in host cells by Stxs. Suppression of the abnormal Stx-mediated increase in O-GlcNAcylation effectively inhibited apoptotic and inflammatory responses in Stx-susceptible cells. The protective effect of O-GlcNAc inhibition for Stx-mediated pathogenic responses was also verified using three-dimensional (3D)-cultured spheroids or organoids mimicking the human kidney. Treatment with an O-GlcNAcylation inhibitor remarkably improved the major disease symptoms and survival rate for mice intraperitoneally injected with a lethal dose of Stx. In conclusion, this study elucidates O-GlcNAcylation-dependent pathogenic mechanisms of Stxs and demonstrates that inhibition of aberrant O-GlcNAcylation is a potential approach to treat Stx-mediated diseases.


Subject(s)
Escherichia coli Infections , Hemolytic-Uremic Syndrome , Animals , Endoplasmic Reticulum Stress , Hemolytic-Uremic Syndrome/pathology , Kidney/pathology , Mice , Shiga Toxin/metabolism , Shiga Toxins
9.
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
10.
Toxins (Basel) ; 13(8)2021 07 29.
Article in English | MEDLINE | ID: mdl-34437406

ABSTRACT

Shiga toxin-producing E. coli (STEC) produces Stx1 and/or Stx2, and Subtilase cytotoxin (SubAB). Since these toxins may be present simultaneously during STEC infections, the purpose of this work was to study the co-action of Stx2 and SubAB. Stx2 + SubAB was assayed in vitro on monocultures and cocultures of human glomerular endothelial cells (HGEC) with a human proximal tubular epithelial cell line (HK-2) and in vivo in mice after weaning. The effects in vitro of both toxins, co-incubated and individually, were similar, showing that Stx2 and SubAB contribute similarly to renal cell damage. However, in vivo, co-injection of toxins lethal doses reduced the survival time of mice by 24 h and mice also suffered a strong decrease in the body weight associated with a lowered food intake. Co-injected mice also exhibited more severe histological renal alterations and a worsening in renal function that was not as evident in mice treated with each toxin separately. Furthermore, co-treatment induced numerous erythrocyte morphological alterations and an increase of free hemoglobin. This work shows, for the first time, the in vivo effects of Stx2 and SubAB acting together and provides valuable information about their contribution to the damage caused in STEC infections.


Subject(s)
Escherichia coli Proteins/toxicity , Hemolytic-Uremic Syndrome/etiology , Shiga Toxin 2/toxicity , Subtilisins/toxicity , Animals , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Endothelial Cells/drug effects , Epithelial Cells/drug effects , Hemolytic-Uremic Syndrome/pathology , Humans , Kidney/drug effects , Kidney/pathology , Kidney Glomerulus/cytology , Kidney Tubules, Proximal/cytology , Male , Mice, Inbred BALB C
11.
Am J Pathol ; 191(5): 795-804, 2021 05.
Article in English | MEDLINE | ID: mdl-33652019

ABSTRACT

This review focuses on typical hemolytic uremic syndrome (HUS), a life-threatening sequela of human infections caused, particularly in children, by Shiga toxin-producing Escherichia coli strains. Thrombotic microangiopathy of the brain and the kidney is the end point of toxin action, resulting in the hallmarks of HUS (ie, thrombocytopenia, anemia, and acute renal failure). A growing body of evidence points to the role of extracellular vesicles released in the blood of patients by toxin-challenged circulating cells (monocytes, neutrophils, and erythrocytes) and platelets, as a key factor in the pathogenesis of HUS. This review provides i) an updated description of the pathogenesis of Shiga toxin-producing E. coli infections; ii) an analysis of blood cell-derived extracellular vesicles, and of their parent cells, as triggering factors in HUS; and iii) a model explaining why Shiga toxin-containing vesicles dock preferentially to the endothelia of target organs.


Subject(s)
Escherichia coli Infections/pathology , Hemolytic-Uremic Syndrome/pathology , Shiga-Toxigenic Escherichia coli/physiology , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Anemia/etiology , Anemia/pathology , Endothelial Cells/pathology , Erythrocytes/pathology , Extracellular Vesicles/pathology , Hemolytic-Uremic Syndrome/complications , Humans , Monocytes/pathology , Neutrophils/pathology , Thrombocytopenia/etiology , Thrombocytopenia/pathology
12.
Ren Fail ; 43(1): 382-387, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33641616

ABSTRACT

BACKGROUND: Insulin-like growth factor-binding protein (IGFBP) 2 plays an important role in the regulation of cell adhesion, migration, growth, and apoptosis. This study aimed to investigate the clinical significance of serum IGFBP2 as a biomarker for disease activity and severity in hemolytic uremic syndrome (HUS) induced by enterohemorrhagic Escherichia coli (EHEC). METHODS: IGFBP2 production by human renal glomerular endothelial cells (RGECs) after exposure to Shiga toxin 2 (Stx-2) was investigated in vitro. Serum IGFBP2 levels in blood samples obtained from 22 patients with HUS and 10 healthy controls (HCs) were quantified using an enzyme-linked immunosorbent assay. The results were compared to the clinical features of HUS and serum tau and cytokine levels. RESULTS: Stx-2 induced the production of IGFBP2 in RGECs in a dose-dependent manner. Serum IGFBP2 levels were significantly higher in patients with HUS than in HCs and correlated with disease severity. Additionally, serum IGFBP2 levels were significantly higher in patients with encephalopathy than in those without encephalopathy. A serum IGFBP2 level above 3585 pg/mL was associated with a high risk of encephalopathy. Furthermore, serum IGFBP2 levels significantly correlated with serum levels of tau and inflammatory cytokines associated with the development of HUS. CONCLUSIONS: Correlation of serum IGFBP2 level with disease activity in patients with HUS suggests that IGFBP2 may be considered as a possible indicator for disease activity and severity in HUS. Larger studies and additional experiments using various cells in central nervous system should elucidate the true value of IGFBP2 as a clinical diagnostic marker. ABBREVIATIONS: IGFBP: insulin-like growth factor-binding protein; HUS: hemolytic uremic syndrome; EHEC: enterohemorrhagic Escherichia coli; RGECs: renal glomerular endothelial cells; STx-2: Shiga toxin 2; HCs: healthy controls; LPS: lipopolysaccharide; ROC: receiver operating characteristic; sTNFR: soluble tumor necrosis factor receptor.


Subject(s)
Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/microbiology , Insulin-Like Growth Factor Binding Protein 2/blood , Adolescent , Adult , Biomarkers/blood , Case-Control Studies , Child , Child, Preschool , Escherichia coli Infections/complications , Female , Hemolytic-Uremic Syndrome/pathology , Humans , Infant , Male , ROC Curve , Severity of Illness Index , Young Adult
13.
Clin Sci (Lond) ; 135(3): 575-588, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33496327

ABSTRACT

Hemolytic Uremic Syndrome (HUS), a disease triggered by Shiga toxin (Stx), is characterized by hemolytic anemia, thrombocytopenia and renal failure. The inflammatory response mediated by polymorphonuclear neutrophils (PMNs) and monocytes is essential to HUS onset. Still, the role of anti-inflammatory cytokines is less clear. The deficiency of IL-10, an anti-inflammatory cytokine, leads to severe pathology in bacterial infections but also to beneficial effects in models of sterile injury. The aim of this work was to analyze the role of IL-10 during HUS. Control and IL-10 lacking mice (IL-10-/-) were intravenously injected with Stx type 2 (Stx2) and survival rate was evaluated. PMN and circulating and renal pro- and anti-inflammatory factors were analyzed by FACS and enzyme-linked immunosorbent assay (ELISA) respectively. IL-10-/- mice showed a higher survival associated with lower renal damage reflected by reduced plasma urea and creatinine levels than control mice. Circulating PMN increased at 72 h in both mouse strains accompanied by an up-regulation of CD11b in control mice. In parallel, renal PMN were significantly increased only in control mice after toxin. Plasma TNF-α, IL-6 and corticosterone levels were higher increased in IL-10-/- than control mice. Simultaneously renal TNF-α raised constantly but was accompanied by increased TGF-ß levels in IL-10-/- mice. These results demonstrate that the profile of circulating and renal cytokines after Stx2 differed between strains suggesting that balance of these factors could participate in renal protection. We conclude that IL-10 absence has a protective role in an experimental model of HUS by reducing PMN recruitment into kidney and renal damage, and increasing mice survival.


Subject(s)
Hemolytic-Uremic Syndrome/chemically induced , Interleukin-10/metabolism , Shiga Toxin 2/toxicity , Animals , Corticosterone/blood , Hemolytic-Uremic Syndrome/pathology , Interleukin-10/genetics , Interleukin-6/blood , Kidney/chemistry , Kidney/pathology , Mice, Inbred BALB C , Mice, Knockout , Neutrophils , Survival Rate , Transforming Growth Factor beta , Tumor Necrosis Factor-alpha/blood
15.
Int J Mol Sci ; 21(17)2020 Aug 19.
Article in English | MEDLINE | ID: mdl-32824988

ABSTRACT

Glomerulonephritis (GN) continues to be one of the main causes of end-stage kidney disease (ESKD) with an incidence rating from 10.5% to 38.2%. Therefore, recurrent GN, previously considered to be a minor contributor to graft loss, is the third most common cause of graft failure 10 years after renal transplantation. However, the incidence, pathogenesis, and natural course of recurrences are still not completely understood. This review focuses on the most frequent diseases that recur after renal transplantation, analyzing rate of recurrence, epidemiology and risk factors, pathogenesis and bimolecular mechanisms, clinical presentation, diagnosis, and therapy, taking into consideration the limited data available in the literature. First of all, the risk for recurrence depends on the type of glomerulonephritis. For example, recipient patients with anti-glomerular basement membrane (GBM) disease present recurrence rarely, but often exhibit rapid graft loss. On the other hand, recipient patients with C3 glomerulonephritis present recurrence in more than 50% of cases, although the disease is generally slowly progressive. It should not be forgotten that every condition that can lead to chronic graft dysfunction should be considered in the differential diagnosis of recurrence. Therefore, a complete workup of renal biopsy, including light, immunofluorescence and electron microscopy study, is essential to provide the diagnosis, excluding alternative diagnosis that may require different treatment. We will examine in detail the biomolecular mechanisms of both native and transplanted kidney diseases, monitoring the risk of recurrence and optimizing the available treatment options.


Subject(s)
Glomerulonephritis, IGA/pathology , Glomerulonephritis, Membranous/pathology , Glomerulonephritis/pathology , Hemolytic-Uremic Syndrome/pathology , Kidney Transplantation/adverse effects , Postoperative Complications/pathology , Glomerulonephritis/epidemiology , Glomerulonephritis/etiology , Glomerulonephritis, IGA/epidemiology , Glomerulonephritis, IGA/etiology , Glomerulonephritis, Membranous/epidemiology , Glomerulonephritis, Membranous/etiology , Hemolytic-Uremic Syndrome/epidemiology , Hemolytic-Uremic Syndrome/etiology , Humans , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Recurrence
16.
BMC Nephrol ; 21(1): 260, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32646497

ABSTRACT

BACKGROUND: The seroprevalence of human Parvovirus B19 (PVB19) is 70-85% in adults worldwide. PVB19 is the etiologic agent of the fifth disease, is a cause of aplastic anemia, and can be associated with kidney injury. We aimed to describe the cases of 4 patients with kidney injury related to PVB19 primary infection, and to evaluate the seroprevalence of PVB19 and the incidence of PVB19 primary infection in patients undergoing a native kidney biopsy. METHODS: Cases of PVB19 infection with kidney injury were reviewed from the archives of the department of Nephrology. A systematic screening of anti-PVB19 IgG and IgM antibodies and viral DNA was performed in sera from 100 consecutive patients with a kidney biopsy in 2017-2018. RESULTS: The 4 patients with PVB19 infection-associated kidney disease displayed: one lupus-like glomerulonephritis (GN) without lupus auto-antibodies, one minimal change disease with tubular necrosis, one secondary hemolytic and uremic syndrome and one membrano-proliferative GN. In the 100 patients biopsied, 67 had elevated anti-PVB19 IgG, among whom 8 had elevated IgM, without circulating viral DNA, without any particular renal pathological pattern. One additional patient showed a seroconversion at the time of kidney biopsy, which revealed a class V lupus nephritis. CONCLUSION: PVB19 primary infection can be associated with different kidney diseases. The seroprevalence of PVB19 among patients with a kidney biopsy is similar to the overall population, and primary infection is rarely documented (1%) after systematic screening. Whether PV19 is nephrotoxic, or triggers renal endothelial injury and immune activation, remains to be elucidated.


Subject(s)
Acute Kidney Injury/virology , Antibodies, Viral/immunology , DNA, Viral/blood , Erythema Infectiosum/immunology , Parvovirus B19, Human/immunology , Acute Kidney Injury/blood , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy , Erythema Infectiosum/blood , Erythema Infectiosum/complications , Female , Glomerulonephritis/blood , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Glomerulonephritis/virology , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/immunology , Glomerulonephritis, Membranoproliferative/pathology , Glomerulonephritis, Membranoproliferative/virology , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/pathology , Hemolytic-Uremic Syndrome/virology , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Incidence , Kidney , Kidney Tubular Necrosis, Acute/blood , Kidney Tubular Necrosis, Acute/immunology , Kidney Tubular Necrosis, Acute/pathology , Kidney Tubular Necrosis, Acute/virology , Male , Middle Aged , Nephrosis, Lipoid/blood , Nephrosis, Lipoid/immunology , Nephrosis, Lipoid/pathology , Nephrosis, Lipoid/virology , Parvovirus B19, Human/genetics , Seroepidemiologic Studies , Viremia/blood , Young Adult
17.
Toxins (Basel) ; 12(6)2020 06 04.
Article in English | MEDLINE | ID: mdl-32512916

ABSTRACT

The global emergence of clinical diseases caused by enterohemorrhagic Escherichia coli (EHEC) is an issue of great concern. EHEC release Shiga toxins (Stxs) as their key virulence factors, and investigations on the cell-damaging mechanisms toward target cells are inevitable for the development of novel mitigation strategies. Stx-mediated hemolytic uremic syndrome (HUS), characterized by the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute renal injury, is the most severe outcome of an EHEC infection. Hemolytic anemia during HUS is defined as the loss of erythrocytes by mechanical disruption when passing through narrowed microvessels. The formation of thrombi in the microvasculature is considered an indirect effect of Stx-mediated injury mainly of the renal microvascular endothelial cells, resulting in obstructions of vessels. In this review, we summarize and discuss recent data providing evidence that HUS-associated hemolytic anemia may arise not only from intravascular rupture of erythrocytes, but also from the extravascular impairment of erythropoiesis, the development of red blood cells in the bone marrow, via direct Stx-mediated damage of maturing erythrocytes, leading to "non-hemolytic" anemia.


Subject(s)
Erythrocytes/microbiology , Erythropoiesis , Escherichia coli Infections/microbiology , Hemolytic-Uremic Syndrome/microbiology , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Animals , Down-Regulation , Erythrocytes/metabolism , Erythrocytes/pathology , Escherichia coli Infections/blood , Escherichia coli Infections/pathology , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/pathology , Host-Pathogen Interactions , Humans , Shiga Toxins/blood , Shiga-Toxigenic Escherichia coli/pathogenicity , Stress, Mechanical
18.
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
19.
Toxins (Basel) ; 12(5)2020 05 23.
Article in English | MEDLINE | ID: mdl-32456125

ABSTRACT

Shiga toxins (Stxs), syn. Vero(cyto)toxins, are potent bacterial exotoxins and the principal virulence factor of enterohemorrhagic Escherichia coli (EHEC), a subset of Shiga toxin-producing E. coli (STEC). EHEC strains, e.g., strains of serovars O157:H7 and O104:H4, may cause individual cases as well as large outbreaks of life-threatening diseases in humans. Stxs primarily exert a ribotoxic activity in the eukaryotic target cells of the mammalian host resulting in rapid protein synthesis inhibition and cell death. Damage of endothelial cells in the kidneys and the central nervous system by Stxs is central in the pathogenesis of hemolytic uremic syndrome (HUS) in humans and edema disease in pigs. Probably even more important, the toxins also are capable of modulating a plethora of essential cellular functions, which eventually disturb intercellular communication. The review aims at providing a comprehensive overview of the current knowledge of the time course and the consecutive steps of Stx/cell interactions at the molecular level. Intervention measures deduced from an in-depth understanding of this molecular interplay may foster our basic understanding of cellular biology and microbial pathogenesis and pave the way to the creation of host-directed active compounds to mitigate the pathological conditions of STEC infections in the mammalian body.


Subject(s)
Escherichia coli Infections/microbiology , Hemolytic-Uremic Syndrome/microbiology , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Virulence Factors/metabolism , Animals , Apoptosis , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/pathology , Host-Pathogen Interactions , Humans , Shiga-Toxigenic Escherichia coli/pathogenicity , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...