Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 686
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.
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
3.
Semin Immunol ; 45: 101331, 2019 10.
Article in English | MEDLINE | ID: mdl-31711769

ABSTRACT

Glomerulonephritis (GN) refers to a group of renal diseases affecting the glomeruli due to the damage mediated by immunological mechanisms. A large proportion of the disease manifestations are caused by disturbances in the complement system. They can be due to genetic errors, autoimmunity, microbes or abnormal immunoglobulins, like modified IgA or paraproteins. The common denominator in most of the problems is an overactive or misdirected alternative pathway complement activation. An assessment of kidney function, amount of proteinuria and hematuria are crucial elements to evaluate, when glomerulonephritis is suspected. However, the cornerstones of the diagnoses are renal biopsy and careful examination of the complement abnormality. Differential diagnostics between the various forms of GN is not possible based on clinical features, as they may vary greatly. This review describes the known mechanisms of complement dysfunction leading to different forms of primary GN (like IgA glomerulonephritis, dense deposit disease, C3 glomerulonephritis, post-infectious GN, membranous GN) and differences to atypical hemolytic uremic syndrome. It also covers the basic elements of etiology-directed therapy and prognosis of the most common forms of GN. Common principles in the management of GN include treatment of hypertension and reduction of proteinuria, some require immunomodulating treatment. Complement inhibition is an emerging treatment option. A thorough understanding of the basic disease mechanism and a careful follow-up are needed for optimal therapy.


Subject(s)
Complement Activation/immunology , Complement System Proteins/immunology , Disease Susceptibility/immunology , Glomerulonephritis/etiology , Glomerulonephritis/metabolism , Animals , Bacterial Infections/complications , Biomarkers , Complement Activation/genetics , Complement System Proteins/metabolism , Glomerulonephritis/diagnosis , Glomerulonephritis, IGA/etiology , Glomerulonephritis, IGA/metabolism , Glomerulonephritis, IGA/pathology , Glomerulonephritis, Membranoproliferative/etiology , Glomerulonephritis, Membranoproliferative/metabolism , Glomerulonephritis, Membranoproliferative/pathology , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/pathology , Humans
4.
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
5.
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
6.
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
7.
Nephrol Dial Transplant ; 35(12): 2147-2153, 2020 12 04.
Article in English | MEDLINE | ID: mdl-31411695

ABSTRACT

BACKGROUND: Treatment with eculizumab in Shiga toxin-associated haemolytic and uraemic syndrome (STEC-HUS) remains controversial despite its increasing utilization. The aim of our study was to evaluate the outcomes of children treated with eculizumab for STEC-HUS in a single-centre matched cohort study. METHODS: Data were retrospectively collected from medical records of children diagnosed with STEC-HUS. The outcomes of patients treated with eculizumab for STEC-HUS were compared with those of a control group of untreated patients matched for age, sex and severity of acute kidney injury with a 1:2 matching scheme. RESULTS: Eighteen children (median age 40.6 months) with STEC-HUS treated with eculizumab were compared with 36 matched control patients (median age 36.4 months) who did not receive eculizumab. All patients survived in the two groups. Within 1 month of HUS onset, the evolution of haematological and renal parameters did not differ between the two groups. At 12 months of follow-up, renal outcome was not significantly different between the two groups. At the last follow-up, the prevalence of decreased glomerular filtration rate in the eculizumab group (27%) was not statistically different from that in controls (38%), as was the prevalence of proteinuria and high blood pressure. Children who received eculizumab more often had extrarenal sequelae during follow-up. Eculizumab treatment appeared to be safe in children with STEC-HUS. CONCLUSION: The benefit of eculizumab on renal and extrarenal outcomes in STEC-HUS could not be established based on our findings. However, efficacy and safety are not best assessed by the observational design and small sample size of our study. Randomized controlled trials are thus required to determine the efficacy of eculizumab in this indication.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement Inactivating Agents/therapeutic use , Escherichia coli Infections/complications , Hemolytic-Uremic Syndrome/drug therapy , Shiga Toxin/toxicity , Shiga-Toxigenic Escherichia coli/pathogenicity , Case-Control Studies , Child , Child, Preschool , Escherichia coli Infections/chemically induced , Escherichia coli Infections/microbiology , Female , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Humans , Infant , Male , Retrospective Studies , Treatment Outcome
8.
BMC Nephrol ; 21(1): 96, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32164588

ABSTRACT

BACKGROUND: Thrombotic microangiopathy (TMA) syndromes are characterized by the association of hemolytic anemia, thrombocytopenia and organ injury due to arteriolar and capillary thrombosis. CASE PRESENTATION: We report the first case of adult onset cobalamin C (Cbl C) disease associated with anti-factor H antibody-associated hemolytic uremic syndrome (HUS). A 19-year-old woman was admitted to the nephrology department owing to acute kidney failure, proteinuria, and hemolytic anemia with schizocytes. TMA was diagnosed and plasma exchanges were started in emergency. Exhaustive analyses showed 1) circulating anti factor H antibody and 2) hyperhomocysteinemia, hypomethioninemia and high levels of methylmalonic aciduria pointing towards Clb C disease. Cbl C disease has been confirmed by methylmalonic aciduria and homocystinuria type C protein gene sequencing revealing two heterozygous pathogenic variants. The kidney biopsy showed 1) intraglomerular and intravascular thrombi 2) noticeable thickening of the capillary wall with a duplication aspect of the glomerular basement membrane and a glomerular capillary wall IgM associated with Cbl C disease related TMA. We initiated treatment including hydroxycobalamin, folinic acid, betaine and levocarnitine and Eculizumab. Rituximab infusions were performed allowing a high decrease in anti-factor H antibody rate. Six month after the disease onset, Eculizumab was weaning and vitaminotherapy continued. Outcome was favorable with a dramatic improvement in kidney function. CONCLUSION: TMA with renal involvement can have a complex combination of risk factors including anti-FH autoantibody in the presence of cblC deficiency.


Subject(s)
Autoantibodies/blood , Complement Factor H/immunology , Hemolytic-Uremic Syndrome/complications , Hemolytic-Uremic Syndrome/immunology , Vitamin B 12 Deficiency/complications , Adult , Biopsy , Female , Hemolytic-Uremic Syndrome/pathology , Hemolytic-Uremic Syndrome/therapy , Humans
9.
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
10.
Pediatr Int ; 62(3): 308-315, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31742829

ABSTRACT

Hemolytic - uremic syndrome (HUS) is a severe complication of infection by Shiga toxin (STx)-producing enterohemorrhagic Escherichia coli. Hemolytic - uremic syndrome is defined clinically as a triad of non-immune microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injuries. Neurologic complications such as acute encephalopathy are also observed. In humans, endothelial cells, proximal tubular epithelial cells, mesangial cells, podocytes, intestinal epithelial cells, and monocytes / macrophages are susceptible to STx-mediated injury. Shiga toxin induces the secretion of inflammatory cytokines and chemokines from susceptible cells, including tumor necrosis factor-α interleukin (IL)-1, IL-6, and IL-8. These cytokines and chemokines contribute to the pathogenesis of HUS and encephalopathy by enhancing STx-induced cytotoxicity and inducing inflammatory cell infiltration. Serum cytokine/chemokine levels are therefore useful as indicators of disease activity and predictors of progression from acute kidney injury to chronic kidney disease. Anti-inflammation therapy combined with apheresis to remove excessive cytokines / chemokines and methylprednisolone pulse therapy to suppress cytokine/chemokine production may be an effective treatment regimen for severe E. coli-associated HUS. However, this regimen requires careful monitoring of potential side effects, such as infections, thrombus formation, and hypertension.


Subject(s)
Chemokines/blood , Cytokines/blood , Enterohemorrhagic Escherichia coli/isolation & purification , Escherichia coli Infections/complications , Hemolytic-Uremic Syndrome/etiology , Acute Kidney Injury/blood , Acute Kidney Injury/etiology , Biomarkers/blood , Brain Diseases/blood , Brain Diseases/etiology , Escherichia coli Infections/blood , Escherichia coli Infections/pathology , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/pathology , Humans , Prognosis , Severity of Illness Index , Shiga Toxins/adverse effects
11.
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
12.
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
13.
Kidney Int ; 95(6): 1443-1452, 2019 06.
Article in English | MEDLINE | ID: mdl-30982675

ABSTRACT

Secondary hemolytic uremic syndrome (HUS) is a heterogeneous group of thrombotic microangiopathies associated with various underlying conditions. Whether it belongs to the spectrum of complement-mediated HUS remains controversial. We analysed the presentation, outcome, and frequency of complement gene rare variants in a cohort of 110 patients with secondary HUS attributed to drugs (29%), autoimmune diseases (24%), infections (17%), malignancies (10%), glomerulopathies (9%), extra-renal organ transplantation (8%), and pancreatitis (3%). The frequency of complement gene rare variants was similar in patients with secondary HUS (5%) and in healthy individuals (6% and 8% in French and European controls, respectively). At diagnosis, 40% of patients required dialysis and 18% had neurological manifestations. Fifty percent of patients received plasmatherapy and 35% were treated with eculizumab. Haematological and complete renal remission was achieved in 80% and 24% of patients, respectively. Thirty-nine percent of patients progressed to chronic kidney disease (stages 3-4) and an additional 37% reached end-stage renal disease. Eleven percent of patients died, most often from complications of the underlying cause of HUS. Only one patient experienced an HUS relapse. Patients treated with eculizumab presented with more severe HUS and were more likely to require dialysis at the time of diagnosis as compared to patients not treated with eculizumab. Rates of hematological remission, chronic kidney disease (stages 3-4), and end-stage renal disease were similar in the two groups. Secondary HUS is an acute nonrelapsing form of HUS, not related to complement dysregulation. The efficacy of eculizumab in this setting is not yet established.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Complement System Proteins/genetics , Hemolytic-Uremic Syndrome/etiology , Kidney Failure, Chronic/epidemiology , Renal Insufficiency, Chronic/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/mortality , Atypical Hemolytic Uremic Syndrome/pathology , Atypical Hemolytic Uremic Syndrome/therapy , Child , Child, Preschool , Complement Activation/genetics , Complement Inactivating Agents/therapeutic use , Complement System Proteins/immunology , Disease Progression , Female , France/epidemiology , Hemolytic-Uremic Syndrome/mortality , Hemolytic-Uremic Syndrome/pathology , Hemolytic-Uremic Syndrome/therapy , Humans , Kidney/immunology , Kidney/pathology , Kidney Failure, Chronic/pathology , Male , Middle Aged , Plasmapheresis/statistics & numerical data , Registries/statistics & numerical data , Renal Dialysis/statistics & numerical data , Renal Insufficiency, Chronic/pathology , Retrospective Studies , Risk Factors , Treatment Outcome , Young Adult
14.
Epidemiol Rev ; 41(1): 121-129, 2019 01 31.
Article in English | MEDLINE | ID: mdl-31616910

ABSTRACT

Shiga toxin-producing Escherichia coli are pathogenic bacteria found in the gastrointestinal tract of humans. Severe infections could lead to life-threatening complications, especially in young children and the elderly. Understanding the distribution of the incubation period, which is currently inconsistent and ambiguous, can help in controlling the burden of disease. We conducted a systematic review of outbreak investigation reports, extracted individual incubation data and summary estimates, tested for heterogeneity, classified studies into subgroups with limited heterogeneity, and undertook a meta-analysis to identify factors that may contribute to the distribution of the pathogen's incubation period. Twenty-eight studies were identified for inclusion in the review (1 of which included information on 2 outbreaks), and the resulting I2 value was 77%, indicating high heterogeneity. Studies were classified into 5 subgroups, with the mean incubation period ranging from 3.5 to 8.1 days. The length of the incubation period increased with patient age and decreased by 7.2 hours with every 10% increase in attack rate.


Subject(s)
Escherichia coli Infections/pathology , Infectious Disease Incubation Period , Shiga-Toxigenic Escherichia coli , Adult , Age Factors , Child , Disease Outbreaks , Escherichia coli Infections/epidemiology , Escherichia coli Infections/transmission , Hemolytic-Uremic Syndrome/epidemiology , Hemolytic-Uremic Syndrome/pathology , Humans
15.
Cytokine ; 121: 154732, 2019 09.
Article in English | MEDLINE | ID: mdl-31153054

ABSTRACT

BACKGROUND: The inflammatory response of the host to Shiga toxin and/or lipopolysaccharide (LPS) of Escherichia coli (E. coli) is included in (HUS). The TLR4-LPS complex is internalized and TLR4 induced inflammatory signaling is stopped by targeting the complex for degradation. Rab7b, a small guanosine triphosphatase (GTPase) expressed in monocytes, regulates the later stages of the endocytic pathway. OBJECTIVE: we studied the Rab7b participation on the TLR4 endocytic pathway and its effect on monocyte cytokine production along the acute course of pediatric Shiga toxin-associated HUS. METHODS AND RESULTS: Monocytes were identified according to their positivity in CD14 expression. Surface TLR4 expression in monocytes from 18 HUS patients significantly increased by day 1 to 6, showing the highest increase on day 4 compared to monocytes of 10 healthy children. Significant higher surface TLR4 expression was accompanied by increased proinflammatory intracellular cytokines, tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6). In contrast, after these time points, surface TLR4 expression and intracellular TNF-α levels, returned to near control levels after 10 days. Furthermore, confocal immunofluorescence microscopy proved colocalization of increased intracellular TLR4/Rab7b determined by Pearson's coefficient in monocytes from HUS patients from day 1 on the highest colocalization of both proteins by day 4. Decreased TLR4/Rab7b colocalization was shown 10 days after HUS onset. CONCLUSION: The colocalization of TLR4 and Rab7b allows us to suggest Rab7b participation in the control of the TLR4 endocytic pathway in HUS patient monocytes. A consequential fall in cytokine production throughout the early follow up of HUS is demonstrated.


Subject(s)
Endocytosis , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/pathology , Shiga Toxin/metabolism , Toll-Like Receptor 4/metabolism , rab GTP-Binding Proteins/metabolism , Acute Disease , Child , Child, Preschool , Cytokines/blood , Follow-Up Studies , Hemolytic-Uremic Syndrome/blood , Humans , Infant , Lipopolysaccharide Receptors/metabolism , Monocytes/metabolism , rab7 GTP-Binding Proteins
16.
BMC Gastroenterol ; 19(1): 156, 2019 Aug 28.
Article in English | MEDLINE | ID: mdl-31462210

ABSTRACT

BACKGROUND: Haemolytic uraemic syndrome is a rarely seen in adults often leading to critical illness. This case highlights how difficult it can be to establish a diagnosis and treat when a patient presents with bloody diarrhoea. CASE PRESENTATION: A 17-year-old Iraqi man presented to the emergency department with abdominal pain and bloody diarrhoea. He was initially treated as acute appendicitis, undergoing an appendectomy but following a recurrence in his symptoms a colonoscopy was performed. A diagnosis of shiga toxin-producing Escherichia coli leading to HUS was suspected following histology obtained at colonoscopy and this was confirmed on antibody testing. Despite intravenous fluids and supportive therapy the patient's symptoms and condition deteriorated. He developed seizures and acute renal failure requiring intubation and plasma exchange in the intensive care setting. He eventually required treatment with ecluzimab therapy; a monoclonal antibody and subsequently made a full recovery. CONCLUSIONS: Haemolytic uraemic syndrome is a triad of progressive renal failure, thrombocytopenia and haemolytic anaemia which is a condition rarely seen in adults. It is usually associated with an E. coli infection and supportive therapy remains the mainstay of treatment.


Subject(s)
Escherichia coli Infections/diagnosis , Hemolytic-Uremic Syndrome/diagnosis , Acute Kidney Injury/etiology , Acute Kidney Injury/therapy , Adolescent , Colonoscopy , Diarrhea/etiology , Escherichia coli Infections/complications , Escherichia coli Infections/pathology , Escherichia coli Infections/therapy , Escherichia coli O157 , Fluid Therapy , Gastrointestinal Hemorrhage/etiology , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/pathology , Hemolytic-Uremic Syndrome/therapy , Humans , Intubation, Intratracheal , Male , Plasma Exchange , Seizures/etiology
17.
Pediatr Nephrol ; 34(3): 379-388, 2019 03.
Article in English | MEDLINE | ID: mdl-29214442

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli (STEC) is the offending agent in post-diarrhea-associated hemolytic uremic syndrome (HUS), a disorder characterized by thrombocytopenia, microangiopathic hemolytic anemia, and acute kidney failure, with thrombi occluding the renal microvasculature. Endothelial dysfunction has been recognized as the trigger event in the development of microangiopathic processes. Glomerular endothelial cells are susceptible to the toxic effects of Stxs that, via nuclear factor kappa B (NF-κB) activation, induce the expression of genes encoding for adhesion molecules and chemokines, culminating in leukocyte adhesion and platelet thrombus formation on the activated endothelium. Complement activation via the alternative pathway has been seen in patients during the acute phase of STEC-associated HUS. Experimental evidence has highlighted the role of complement proteins in driving glomerular endothelium toward a thrombogenic phenotype. At the glomerular level, podocytes are also an important target of Stx-induced complement activation. Glomerular injury as a consequence of podocyte dysfunction and loss is thus a mechanism that might affect long-term renal outcomes in the disease. New approaches to targeting the complement system may be useful therapeutic options for patients with STEC-HUS.


Subject(s)
Endothelial Cells/pathology , Hemolytic-Uremic Syndrome/immunology , Podocytes/pathology , Shiga Toxin/toxicity , Shiga-Toxigenic Escherichia coli/pathogenicity , Animals , Colon/microbiology , Complement Pathway, Alternative/drug effects , Complement Pathway, Alternative/immunology , Diarrhea/complications , Diarrhea/microbiology , Disease Models, Animal , Endothelial Cells/immunology , Hemolytic-Uremic Syndrome/drug therapy , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Intestinal Mucosa/microbiology , Microvessels/cytology , Microvessels/immunology , Microvessels/pathology , Podocytes/immunology , Shiga Toxin/immunology , Shiga Toxin/metabolism , Shiga-Toxigenic Escherichia coli/immunology , Shiga-Toxigenic Escherichia coli/metabolism
18.
Wilderness Environ Med ; 30(1): 66-78, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30711421

ABSTRACT

Thrombotic microangiopathy (TMA), which includes the spectrum of hemolytic uremic syndrome and thrombotic thrombocytopenic purpura, is an uncommon complication of hump-nosed pit viper envenomation. We describe 4 cases of TMA following hump-nosed pit viper (Hypnale spp) bites in Sri Lanka. The first case is a typical TMA that spontaneously resolved with supportive treatments. The second and third cases are related to hemolytic uremic syndrome complicated with acute kidney injury that required hemodialysis. The fourth case is thrombotic thrombocytopenic purpura associated with acute kidney injury that required hemodialysis and therapeutic plasma exchange. For each patient we describe the circumstances of the bite, clinical features, laboratory findings, and management.


Subject(s)
Hemolytic-Uremic Syndrome/etiology , Purpura, Thrombotic Thrombocytopenic/etiology , Snake Bites/therapy , Thrombotic Microangiopathies/etiology , Viper Venoms/toxicity , Viperidae/classification , Aged , Animals , Female , Hemolytic-Uremic Syndrome/pathology , Hemolytic-Uremic Syndrome/therapy , Humans , Male , Middle Aged , Purpura, Thrombotic Thrombocytopenic/pathology , Purpura, Thrombotic Thrombocytopenic/therapy , Sri Lanka , Thrombotic Microangiopathies/pathology , Viperidae/physiology
19.
J Infect Dis ; 217(6): 1000-1010, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29216383

ABSTRACT

Background: Postdiarrheal hemolytic-uremic syndrome (D+HUS) following Shiga toxin-producing Escherichia coli (STEC) infection is a serious condition lacking specific treatment. Host immune dysregulation and genetic susceptibility to complement hyperactivation are implicated in non-STEC-related HUS. However, genetic susceptibility to D+HUS remains largely uncharacterized. Methods: Patients with culture-confirmed STEC diarrhea, identified through the Centers for Disease Control and Prevention FoodNet surveillance system (2007-2012), were serotyped and classified by laboratory and/or clinical criteria as having suspected, probable, or confirmed D+HUS or as controls and underwent genotyping at 200 loci linked to nondiarrheal HUS or similar pathologies. Genetic associations with D+HUS were explored by multivariable regression, with adjustment for known risk factors. Results: Of 641 enrollees with STEC O157:H7, 80 had suspected D+HUS (41 with probable and 32 with confirmed D+HUS). Twelve genes related to cytokine signaling, complement pathways, platelet function, pathogen recognition, iron transport, and endothelial function were associated with D+HUS in multivariable-adjusted analyses (P ≤ .05). Of 12 significant single-nucleotide polymorphisms (SNPs), 5 were associated with all levels of D+HUS (intergenic SNP rs10874639, TFRC rs3804141, EDN1 rs5370, GP1BA rs121908064, and B2M rs16966334), and 7 SNPs (6 non-complement related) were associated with confirmed D+HUS (all P < .05). Conclusions: Polymorphisms in many non-complement-related genes may contribute to D+HUS susceptibility. These results require replication, but they suggest novel therapeutic targets in patients with D+HUS.


Subject(s)
Centers for Disease Control and Prevention, U.S. , Escherichia coli Infections/complications , Escherichia coli Infections/microbiology , Genetic Predisposition to Disease , Hemolytic-Uremic Syndrome/genetics , Shiga-Toxigenic Escherichia coli/pathogenicity , Adolescent , Child , Child, Preschool , Diarrhea/complications , Diarrhea/microbiology , Female , Hemolytic-Uremic Syndrome/pathology , Humans , Male , Risk Factors , United States
20.
Int J Med Microbiol ; 308(7): 940-946, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29983334

ABSTRACT

The life-threatening sequela of hemorrhagic colitis induced by Shiga toxins (Stx)-producing Escherichia coli (STEC) infections in humans is hemolytic uremic syndrome (HUS), the main cause of acute renal failure in early childhood. The key step in the pathogenesis of HUS is the appearance of Stx in the blood of infected patients because these powerful virulence factors are capable of inducing severe microangiopathic lesions in the kidney. During precocious toxemia, which occurs in patients before the onset of HUS during the intestinal phase, Stx bind to several different circulating cells. An early response of these cells might include the release of proinflammatory mediators associated with the development of HUS. Here, we show that primary human monocytes stimulated with Shiga toxin 1a (Stx1a) through the glycolipid receptor globotriaosylceramide released larger amounts of proinflammatory molecules (IL-1ß, TNFα, IL-6, G-CSF, CXCL8, CCL2, CCL4) than Stx1a-treated neutrophils. The mediators (except IL-1ß) are among the top six proinflammatory mediators found in the sera from patients with HUS in different studies. The molecules appear to be involved in different pathogenetic steps of HUS, i.e. sensitization of renal endothelial cells to the toxin actions (IL-1ß, TNFα), activation of circulating monocytes and neutrophils (CXCL8, CCL2, CCL4) and increase in neutrophil counts in patients with poor prognosis (G-CSF). Hence, a role of circulating monocytes in the very early phases of the pathogenetic process culminating with HUS can be envisaged. Impairment of the events of precocious toxemia would prevent or reduce the risk of HUS in STEC-infected children.


Subject(s)
Cytokines/blood , Hemolytic-Uremic Syndrome/pathology , Monocytes/metabolism , Shiga Toxin 1/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , Trihexosylceramides/metabolism , Cells, Cultured , Cytokines/metabolism , Hemolytic-Uremic Syndrome/microbiology , Humans , Interleukin-8/blood , Neutrophils/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL