1
|
Mazzotta C, Ingelfinger JR, Grabowski EF. Shiga toxin down-regulates ERG protein in endothelial cells and impairs angiogenesis. Thromb Res 2024; 240:109038. [PMID: 38850807 DOI: 10.1016/j.thromres.2024.109038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Shiga toxin (Stx) can activate inflammatory signaling, leading to vascular dysfunction and promotion of a pro-thrombotic tissue microenvironment. Stx can trigger the development of the enterohemorrhagic (childhood) hemolytic uremic syndrome (eHUS), a triad of thrombocytopenia, hemolytic anemia, and acute kidney injury, often requiring dialysis. Additional features may include damage to other organs, including the gastrointestinal tract, pancreas, brain and cardiovascular system; death occurs in 2-5 %. eHUS is a thrombotic microangiopathy; thus, endothelial cell (EC) injury and platelet fibrin thrombus formation in glomerular arterioles and in the arterioles of other affected organs are likely. To elucidate mechanisms of this microangiopathy, we examined in human ECs the regulation of the platelet adhesion proteins P-selectin and von Willebrand factor (VWF), along with the downregulation of erythroblast-transformation-specific transcription factor (ERG) a key regulator of angiogenesis and megakaryocyte development. METHODS VWF, P-selectin, and ERG levels were determined using immunofluorescence and Western blot in human umbilical endothelial cells (HUVECs). HUVECs were treated with tumor necrosis factor-alpha (TNF-α), Stx-1 or both, versus normal controls. Capillary morphogenesis on Matrigel was performed using HUVECs treated, for 22 h with TNF-α, Stx-1, or both, or treated 4 h with Stx-1 alone or in combination with TNF-α for 22 h. RESULTS Stx-1 significantly reduced ERG and VWF expression on HUVECs, but upregulated P-selectin expression. ERG levels decreased with Stx-1 alone or in combination with TNF-α, in the nuclear, perinuclear and cytoplasmatic regions. Stx-1 reduced capillary morphogenesis, while Stx-1-TNF-α combined treatment reduced capillary morphogenesis still further. CONCLUSIONS In the presence of Stx-1 or TNF-α or both treatments, ECs were activated, expressing higher levels of P-selectin and lower levels of VWF. Our findings, further, provide evidence that Stx-1 downregulates ERG, repressing angiogenesis in vitro.
Collapse
Affiliation(s)
- Celestina Mazzotta
- Cardiovascular Thrombosis Laboratory, Hematology/Oncology Division, Department of Pediatrics, *Massachusetts General Hospital for Children, Massachusetts General Hospital, and Harvard Medical School, United States
| | - Julie R Ingelfinger
- Nephology Division, Department of Pediatrics, Massachusetts General Hospital for Children, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, United States
| | - Eric F Grabowski
- Cardiovascular Thrombosis Laboratory, Hematology/Oncology Division, Department of Pediatrics, *Massachusetts General Hospital for Children, Massachusetts General Hospital, and Harvard Medical School, United States.
| |
Collapse
|
2
|
Mansour MA, Khalil DF, Hasham MA, Youssef A, Rashad M, Awadallah M, Ali H. Hemolytic uremic syndrome with central nervous system manifestations, a case report and literature review. Radiol Case Rep 2023; 18:2268-2273. [PMID: 37128253 PMCID: PMC10147953 DOI: 10.1016/j.radcr.2023.02.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/19/2023] [Accepted: 02/16/2023] [Indexed: 05/03/2023] Open
Abstract
Hemolytic uremic syndrome (HUS) is a multisystem disorder generally seen in children and young adults, manifesting with the symptomatic triad of thrombocytopenia, hemolytic anemia, and acute kidney injury. These symptoms are often preceded by a prodrome of bloody diarrhea, vomiting, fever, and weakness. HUS is an exceedingly rare entity, with less than 1.5 per 100,000 people affected annually. HUS with central nervous system (CNS) manifestations constitutes approximately 20%-50% of cases and often presents with seizures, altered level of consciousness, and brainstem symptoms. CNS involvement in HUS is a major cause of acute morbidity and mortality; therefore, timely diagnosis and treatment are crucial in the management of these cases. Neuroimaging plays a critical role in the diagnosis; however, it might be very challenging in a large number of cases because studies that report the typical neuroradiologic features of brain injury in cases with HUS are not commonly available. Herein, we demonstrate in a case-based approach, the importance of combining clinical suspicion with different radiologic modalities to better characterize HUS cases with CNS involvement, as well as demonstrate how the early start of meticulous supportive therapy can lead to a favorable outcome even when severe brain involvement is evident on acute imaging studies. Furthermore, we provide an illustrated overview of the current theories that explain the neurologic involvement in HUS, as well as the commonly affected brain areas and how this entity can be radiologically differentiated from other potential diagnoses.
Collapse
Affiliation(s)
- Moustafa A. Mansour
- Department of Neurology and Neurologic Surgery, Faculty of Medicine, Al-Azhar University, 1 Al-Mokhyam Al-Daem St., Nasr City, Cairo 11884, Egypt
- Department of Neurology and Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Division of Neuro-Intensive Care, Dar Al-Fouad Medical Corporation, Cairo, Egypt
- Department of Emergency Medicine and Critical Care, Faculty of Medicine, Al-Azhar University, 1 Al-Mokhyam Al-Daem St., Nasr City, Cairo 11884, Egypt
- Corresponding author.
| | - Dyana F. Khalil
- Department of Emergency Medicine and Critical Care, Dubai Healthcare City, Dubai, UAE
| | - Mohab A. Hasham
- Department of Emergency Medicine and Critical Care, Faculty of Medicine, Al-Azhar University, 1 Al-Mokhyam Al-Daem St., Nasr City, Cairo 11884, Egypt
| | - Ahmed Youssef
- Department of Emergency Medicine and Critical Care, Faculty of Medicine, Al-Azhar University, 1 Al-Mokhyam Al-Daem St., Nasr City, Cairo 11884, Egypt
| | - Mohamed Rashad
- Department of Pediatrics, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Muhammad Awadallah
- Department of Pediatrics, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Hassan Ali
- Department of Pediatrics, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
- Division of Neurology and Neurodevelopmental Disorders, Department of Pediatrics, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
3
|
Tissue-resident macrophages mediate neutrophil recruitment and kidney injury in shiga toxin-induced hemolytic uremic syndrome. Kidney Int 2021; 100:349-363. [PMID: 33930412 DOI: 10.1016/j.kint.2021.03.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 01/01/2023]
Abstract
Enterohaemorrhagic E. coli cause major epidemics worldwide with significant organ damage and very high percentages of death. Due to the ability of enterohaemorrhagic E. coli to produce shiga toxin these bacteria damage the kidney leading to the hemolytic uremic syndrome. A therapy against this serious kidney disease has not been developed yet and the impact and mechanism of leukocyte activation and recruitment are unclear. Tissue-resident macrophages represent the main leukocyte population in the healthy kidney, but the role of this important cell population in shiga toxin-producing E. coli-hemolytic uremic syndrome is incompletely understood. Using state of the art microscopy and mass spectrometry imaging, our preclinical study demonstrated a phenotypic and functional switch of tissue-resident macrophages after disease induction in mice. Kidney macrophages produced the inflammatory molecule TNFα and depletion of tissue-resident macrophages via the CSF1 receptor abolished TNFα levels in the kidney and significantly diminished disease severity. Furthermore, macrophage depletion did not only attenuate endothelial damage and thrombocytopenia, but also activation of thrombocytes and neutrophils. Moreover, we observed that neutrophils infiltrated the kidney cortex and depletion of macrophages significantly reduced the recruitment of neutrophils and expression of the neutrophil-attracting chemokines CXCL1 and CXCL2. Intravital microscopy revealed that inhibition of CXCR2, the receptor for CXCL1 and CXCL2, significantly reduced the infiltration of neutrophils and reduced kidney injury. Thus, our study shows activation of tissue-resident macrophages during shiga toxin-producing E. coli-hemolytic uremic syndrome leading to the production of disease-promoting TNFα and CXCR2-dependent recruitment of neutrophils.
Collapse
|
4
|
Plavec TV, Zahirović A, Zadravec P, Sabotič J, Berlec A. Lectin-Mediated Binding of Engineered Lactococcus lactis to Cancer Cells. Microorganisms 2021; 9:microorganisms9020223. [PMID: 33499141 PMCID: PMC7911926 DOI: 10.3390/microorganisms9020223] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 12/28/2022] Open
Abstract
Lectins have been increasingly utilized as carriers for targeted drug delivery based on their specific binding to glycans located on mammalian cells. This study employed two lectins, B subunit of bacterial Shiga holotoxin (Stx1B) and fungal Clitocybe nebularis lectin (CNL), for surface display on the lactic acid bacterium Lactococcus lactis. The specific adhesion of these engineered, lectin-displaying L. lactis to cancer cells was evaluated. The expression and surface display of both lectins on L. lactis were demonstrated by western blotting and flow cytometry, respectively. MTS assays revealed that recombinant Stx1B had no effect on Caco-2 cell viability at concentrations of ≤25 µg/mL, whereas CNL was non-toxic even at relatively high concentrations of ≤250 µg/mL. Stx1B bound to Caco-2, HT-29 and HeLa cells after 1 h of incubation. CNL bound to Caco-2 cells and recognized several glycoproteins in HT-29 and Caco-2 cell homogenates of which a 70 kDa protein predominated. Confocal microscopy revealed adhesion of Stx1B-displaying L. lactis to HeLa, Caco-2, and, to a lesser extent, HT-29 cells; CNL-displaying L. lactis showed a relatively similar level of adherence to HT-29 and Caco-2 cells. Thus, lectin-displaying L. lactis might serve as a carrier in targeted drug delivery when coupled to a therapeutic moiety.
Collapse
Affiliation(s)
- Tina Vida Plavec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (T.V.P.); (A.Z.); (P.Z.); (J.S.)
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Abida Zahirović
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (T.V.P.); (A.Z.); (P.Z.); (J.S.)
| | - Petra Zadravec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (T.V.P.); (A.Z.); (P.Z.); (J.S.)
- Lek d.d., Kolodvorska 27, 1234 Mengeš, Slovenia
| | - Jerica Sabotič
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (T.V.P.); (A.Z.); (P.Z.); (J.S.)
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (T.V.P.); (A.Z.); (P.Z.); (J.S.)
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
5
|
Goldstein J, Nuñez-Goluboay K, Pinto A. Therapeutic Strategies to Protect the Central Nervous System against Shiga Toxin from Enterohemorrhagic Escherichia coli. Curr Neuropharmacol 2021; 19:24-44. [PMID: 32077828 PMCID: PMC7903495 DOI: 10.2174/1570159x18666200220143001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 01/20/2020] [Accepted: 02/19/2020] [Indexed: 11/23/2022] Open
Abstract
Infection with Shiga toxin-producing Escherichia coli (STEC) may cause hemorrhagic colitis, hemolytic uremic syndrome (HUS) and encephalopathy. The mortality rate derived from HUS adds up to 5% of the cases, and up to 40% when the central nervous system (CNS) is involved. In addition to the well-known deleterious effect of Stx, the gram-negative STEC releases lipopolysaccharides (LPS) and may induce a variety of inflammatory responses when released in the gut. Common clinical signs of severe CNS injury include sensorimotor, cognitive, emotional and/or autonomic alterations. In the last few years, a number of drugs have been experimentally employed to establish the pathogenesis of, prevent or treat CNS injury by STEC. The strategies in these approaches focus on: 1) inhibition of Stx production and release by STEC, 2) inhibition of Stx bloodstream transport, 3) inhibition of Stx entry into the CNS parenchyma, 4) blockade of deleterious Stx action in neural cells, and 5) inhibition of immune system activation and CNS inflammation. Fast diagnosis of STEC infection, as well as the establishment of early CNS biomarkers of damage, may be determinants of adequate neuropharmacological treatment in time.
Collapse
Affiliation(s)
- Jorge Goldstein
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica “Houssay” (IFIBIO), Laboratorio de Neurofisiopatología, Facultad de Medicina, Argentina
| | - Krista Nuñez-Goluboay
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica “Houssay” (IFIBIO), Laboratorio de Neurofisiopatología, Facultad de Medicina, Argentina
| | - Alipio Pinto
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica “Houssay” (IFIBIO), Laboratorio de Neurofisiopatología, Facultad de Medicina, Argentina
| |
Collapse
|
6
|
Molecular Biology of Escherichia Coli Shiga Toxins' Effects on Mammalian Cells. Toxins (Basel) 2020; 12:toxins12050345. [PMID: 32456125 PMCID: PMC7290813 DOI: 10.3390/toxins12050345] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
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.
Collapse
|
7
|
Joseph A, Cointe A, Mariani Kurkdjian P, Rafat C, Hertig A. Shiga Toxin-Associated Hemolytic Uremic Syndrome: A Narrative Review. Toxins (Basel) 2020; 12:E67. [PMID: 31973203 PMCID: PMC7076748 DOI: 10.3390/toxins12020067] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/13/2020] [Accepted: 01/17/2020] [Indexed: 01/28/2023] Open
Abstract
The severity of human infection by one of the many Shiga toxin-producing Escherichia coli (STEC) is determined by a number of factors: the bacterial genome, the capacity of human societies to prevent foodborne epidemics, the medical condition of infected patients (in particular their hydration status, often compromised by severe diarrhea), and by our capacity to devise new therapeutic approaches, most specifically to combat the bacterial virulence factors, as opposed to our current strategies that essentially aim to palliate organ deficiencies. The last major outbreak in 2011 in Germany, which killed more than 50 people in Europe, was evidence that an effective treatment was still lacking. Herein, we review the current knowledge of STEC virulence, how societies organize the prevention of human disease, and how physicians treat (and, hopefully, will treat) its potentially fatal complications. In particular, we focus on STEC-induced hemolytic and uremic syndrome (HUS), where the intrusion of toxins inside endothelial cells results in massive cell death, activation of the coagulation within capillaries, and eventually organ failure.
Collapse
Affiliation(s)
- Adrien Joseph
- Department of Nephrology, AP-HP, Hôpital Tenon, F-75020 Paris, France; (A.J.); (C.R.)
| | - Aurélie Cointe
- Department of Microbiology, AP-HP, Hôpital Robert Debré, F-75019 Paris, France; (A.C.); (P.M.K.)
| | | | - Cédric Rafat
- Department of Nephrology, AP-HP, Hôpital Tenon, F-75020 Paris, France; (A.J.); (C.R.)
| | - Alexandre Hertig
- Department of Renal Transplantation, Sorbonne Université, AP-HP, Hôpital Pitié Salpêtrière, F-75013 Paris, France
| |
Collapse
|
8
|
Kushak RI, Boyle DC, Rosales IA, Ingelfinger JR, Stahl GL, Ozaki M, Colvin RB, Grabowski EF. Platelet thrombus formation in eHUS is prevented by anti-MBL2. PLoS One 2019; 14:e0220483. [PMID: 31881024 PMCID: PMC6934323 DOI: 10.1371/journal.pone.0220483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/05/2019] [Indexed: 11/29/2022] Open
Abstract
E. coli associated Hemolytic Uremic Syndrome (epidemic hemolytic uremic syndrome, eHUS) caused by Shiga toxin-producing bacteria is characterized by thrombocytopenia, microangiopathic hemolytic anemia, and acute kidney injury that cause acute renal failure in up to 65% of affected patients. We hypothesized that the mannose-binding lectin (MBL) pathway of complement activation plays an important role in human eHUS, as we previously demonstrated that injection of Shiga Toxin-2 (Stx-2) led to fibrin deposition in mouse glomeruli that was blocked by co-injection of the anti-MBL-2 antibody 3F8. However, the markers of platelet thrombosis in affected mouse glomeruli were not delineated. To investigate the effect of 3F8 on markers of platelet thrombosis, we used kidney sections from our mouse model (MBL-2+/+ Mbl-A/C-/-; MBL2 KI mouse). Mice in the control group received PBS, while mice in a second group received Stx-2, and those in a third group received 3F8 and Stx-2. Using double immunofluorescence (IF) followed by digital image analysis, kidney sections were stained for fibrin(ogen) and CD41 (marker for platelets), von-Willebrand factor (marker for endothelial cells and platelets), and podocin (marker for podocytes). Electron microscopy (EM) was performed on ultrathin sections from mice and human with HUS. Injection of Stx-2 resulted in an increase of both fibrin and platelets in glomeruli, while administration of 3F8 with Stx-2 reduced both platelet and fibrin to control levels. EM studies confirmed that CD41-positive objects observed by IF were platelets. The increases in platelet number and fibrin levels by injection of Stx-2 are consistent with the generation of platelet-fibrin thrombi that were prevented by 3F8.
Collapse
Affiliation(s)
- R. I. Kushak
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| | - D. C. Boyle
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - I. A. Rosales
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - J. R. Ingelfinger
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - G. L. Stahl
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States of America
| | - M. Ozaki
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States of America
- Department of Emergency and Critical Care Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - R. B. Colvin
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - E. F. Grabowski
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
9
|
Üçeyler N, Urlaub D, Mayer C, Uehlein S, Held M, Sommer C. Tumor necrosis factor-α links heat and inflammation with Fabry pain. Mol Genet Metab 2019; 127:200-206. [PMID: 31221509 DOI: 10.1016/j.ymgme.2019.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/08/2019] [Accepted: 05/17/2019] [Indexed: 02/06/2023]
Abstract
Fabry disease (FD) is an X-linked lysosomal storage disorder associated with pain triggered by heat or febrile infections. We modelled this condition by measuring the cytokine expression of peripheral blood mononuclear cells (PBMC) from FD patients in vitro upon stimulation with heat and lipopolysaccharide (LPS). We enrolled 67 FD patients and 37 healthy controls. We isolated PBMC, assessed their gene expression of selected pro- and anti-inflammatory cytokines, incubated them with heat, LPS, globotriaosylceramide (Gb3), and tumor necrosis factor-α (TNF), and measured TNF secretion in the supernatant and intracellular Gb3 accumulation, respectively. We found increased TNF, interleukin (IL-)1β, and toll-like receptor 4 (TLR4) gene expression in FD men (p < .05 to p < .01). TNF and IL-10 were higher, and IL-4 was lower in the subgroup of FD men with pain compared to controls (p < .05 to p < .01). Hereby, TNF was only increased in FD men with pain and classical mutations (p < .05) compared to those without pain. PBMC from FD patients secreted more TNF upon stimulation with LPS (p < .01) than control PBMC. Incubation with Gb3 and an additional α-galactosidase A inhibitor did not further increase TNF secretion, but incubation with TNF greatly increased the Gb3 load in FD PBMC compared to controls (p < .01). Also, LPS incubation and heat challenge (40 °C) increased Gb3 accumulation in PBMC of patients compared to baseline (p < .05 each), while no alterations were observed in control PBMC. Our data show that TNF holds a crucial role in the pathophysiology of FD associated pain, which may open a novel perspective for analgesic treatment in FD pain.
Collapse
Affiliation(s)
- Nurcan Üçeyler
- Department of Neurology, University of Würzburg, Germany; Würzburg Fabry Center for Interdisciplinary Therapy (FAZIT), University of Würzburg, Germany.
| | - Daniela Urlaub
- Department of Neurology, University of Würzburg, Germany
| | | | | | - Melissa Held
- Department of Neurology, University of Würzburg, Germany
| | - Claudia Sommer
- Department of Neurology, University of Würzburg, Germany; Würzburg Fabry Center for Interdisciplinary Therapy (FAZIT), University of Würzburg, Germany
| |
Collapse
|
10
|
Giordano P, Netti GS, Santangelo L, Castellano G, Carbone V, Torres DD, Martino M, Sesta M, Di Cuonzo F, Resta MC, Gaeta A, Milella L, Chironna M, Germinario C, Scavia G, Gesualdo L, Giordano M. A pediatric neurologic assessment score may drive the eculizumab-based treatment of Escherichia coli-related hemolytic uremic syndrome with neurological involvement. Pediatr Nephrol 2019; 34:517-527. [PMID: 30362078 DOI: 10.1007/s00467-018-4112-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND Thrombotic microangiopathy (TMA) is a clinical syndrome encompassing a large group of rare but severe disorders including thrombotic thrombocytopenic purpura (TTP) and both typical and atypical forms of hemolytic uremic syndrome (HUS). The key role of the complement system is well known in TTP and atypical HUS, but recent reports describe its involvement in the pathogenesis of HUS secondary to gastrointestinal infections due to Shiga toxin-producing Escherichia coli (STEC). METHODS TMA mainly affects the kidney, but extra-renal complications are frequently described. The involvement of the central nervous system (CNS) represents often a life-threatening condition and it can result in serious long-term disability in HUS patients who overcome the acute phase of illness. In the present study, we retrospectively analyzed a pediatric cohort of a single tertiary pediatric hospital in Southern Italy, in which this complication occurred in 12/54 children (22% of cases), of whom five with severe neurological involvement had been successfully treated with eculizumab. RESULTS The great clinical variability of brain injury in our cohort has led us to retrospectively build a "neurological score" useful to assess the clinical severity of neurologic involvement. Subjects with higher neurologic score due to the most severe CNS involvement resulted in the group of patients early treated with eculizumab, obtaining a good clinical response (four out five patients). In conclusion, the early treatment with eculizumab in children with severe neurological involvement during STEC-HUS was associated with complete regression of both acute kidney injury (AKI) and neurological lesions observed at magnetic resonance imaging (MRI). CONCLUSIONS A "neurological score" may be a useful tool to drive the early treatment of CNS complications in STEC-HUS with eculizumab, although future perspective controlled studies are urgently needed to validate this therapeutic approach.
Collapse
Affiliation(s)
- Paolo Giordano
- Pediatric Nephrology and Dialysis Unit, Pediatric Hospital "Giovanni XXIII", Bari, Italy
| | - Giuseppe Stefano Netti
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Luisa Santangelo
- Pediatric Nephrology and Dialysis Unit, Pediatric Hospital "Giovanni XXIII", Bari, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Bari, Italy
| | - Vincenza Carbone
- Pediatric Nephrology and Dialysis Unit, Pediatric Hospital "Giovanni XXIII", Bari, Italy
| | | | - Marida Martino
- Pediatric Nephrology and Dialysis Unit, Pediatric Hospital "Giovanni XXIII", Bari, Italy
| | - Michela Sesta
- Pediatric Neurology Unit, Pediatric Hospital "Giovanni XXIII", Bari, Italy
| | - Franca Di Cuonzo
- Department of Neuroradiology, University of Bari "Aldo Moro", Bari, Italy
| | - Maria Chiara Resta
- Department of Neuroradiology, University of Bari "Aldo Moro", Bari, Italy
| | - Alberto Gaeta
- Pediatric Radiology Unit, Pediatric Hospital "Giovanni XXIII", Bari, Italy
| | - Leonardo Milella
- Intensive Care Unit, Pediatric Hospital "Giovanni XXIII", Bari, Italy
| | - Maria Chironna
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Cinzia Germinario
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Gaia Scavia
- Food Safety, Nutrition and Veterinary Public Health Department, National Institute of Health, Rome, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Bari, Italy
| | - Mario Giordano
- Pediatric Nephrology and Dialysis Unit, Pediatric Hospital "Giovanni XXIII", Bari, Italy.
| |
Collapse
|
11
|
Zhang T, de Waard AA, Wuhrer M, Spaapen RM. The Role of Glycosphingolipids in Immune Cell Functions. Front Immunol 2019; 10:90. [PMID: 30761148 PMCID: PMC6361815 DOI: 10.3389/fimmu.2019.00090] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/14/2019] [Indexed: 01/06/2023] Open
Abstract
Glycosphingolipids (GSLs) exhibit a variety of functions in cellular differentiation and interaction. Also, they are known to play a role as receptors in pathogen invasion. A less well-explored feature is the role of GSLs in immune cell function which is the subject of this review article. Here we summarize knowledge on GSL expression patterns in different immune cells. We review the changes in GSL expression during immune cell development and differentiation, maturation, and activation. Furthermore, we review how immune cell GSLs impact membrane organization, molecular signaling, and trans-interactions in cellular cross-talk. Another aspect covered is the role of GSLs as targets of antibody-based immunity in cancer. We expect that recent advances in analytical and genome editing technologies will help in the coming years to further our knowledge on the role of GSLs as modulators of immune cell function.
Collapse
Affiliation(s)
- Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Antonius A de Waard
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
12
|
Anti-inflammatory agents reduce microglial response, demyelinating process and neuronal toxin uptake in a model of encephalopathy produced by Shiga Toxin 2. Int J Med Microbiol 2018; 308:1036-1042. [PMID: 30314914 DOI: 10.1016/j.ijmm.2018.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 09/07/2018] [Accepted: 09/30/2018] [Indexed: 11/20/2022] Open
Abstract
Infections by Enterohemorrhagic Escherichia coli may cause in addition to hemolytic uremic syndrome neurological disorders which may lead to fatal outcomes in patients. The brain striatum is usually affected during this outcome. The aim of this study was to determine in this area the role of the microglia in pro-inflammatory events that may occur during Shiga toxin 2 intoxication and consequently to this, whether oligodendrocytes were being affected. In the present paper we demonstrated that anti-inflammatory treatments reduced deleterious effects in brain striatal cells exposed to Shiga toxin 2 and LPS. While dexamethasone treatment decreased microglial activation and recovered myelin integrity in the mice striatum, etanercept treatment decreased neuronal uptake of Stx2 in rat striatal neurons, improving the affected area from toxin-derived injury. In conclusion, microglial activation is related to pro-inflammatory events that may deteriorate the brain function during intoxication with Stx2 and LPS. Consequently, the role of anti-inflammatory agents in the treatment of EHEC-derived encephalopathy should be studied in clinical trials.
Collapse
|
13
|
Legros N, Pohlentz G, Steil D, Müthing J. Shiga toxin-glycosphingolipid interaction: Status quo of research with focus on primary human brain and kidney endothelial cells. Int J Med Microbiol 2018; 308:1073-1084. [PMID: 30224239 DOI: 10.1016/j.ijmm.2018.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/28/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022] Open
Abstract
Shiga toxin (Stx)-mediated injury of the kidneys and the brain represent the major extraintestinal complications in humans upon infection by enterohemorrhagic Escherichia coli (EHEC). Damage of renal and cerebral endothelial cells is the key event in the pathogenesis of the life-threatening hemolytic uremic syndrome (HUS). Stxs are AB5 toxins and the B-pentamers of the two clinically important Stx subtypes Stx1a and Stx2a preferentially bind to the glycosphingolipid globotriaosylceramide (Gb3Cer, Galα4Galβ4Glcβ1Cer) and to less extent to globotetraosylceramide (Gb4Cer, GalNAcβ3Galα4Galβ4Glcβ1), which are expected to reside in lipid rafts in the plasma membrane of the human endothelium. This review summarizes the current knowledge on the Stx glycosphingolipid receptors and their lipid membrane ensemble in primary human brain microvascular endothelial cells (pHBMECs) and primary human renal glomerular endothelial cells (pHRGECs). Increasing knowledge on the precise initial molecular mechanisms by which Stxs interact with cellular targets will help to develop specific therapeutics and/or preventive measures to combat EHEC-caused diseases.
Collapse
Affiliation(s)
- Nadine Legros
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | | | - Daniel Steil
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany; Interdisciplinary Center for Clinical Research (IZKF), University of Münster, D-48149 Münster, Germany.
| |
Collapse
|
14
|
Pohl J, Volke JK, Thiebes S, Brenzel A, Fuchs K, Beziere N, Ehrlichmann W, Pichler BJ, Squire A, Gueler F, Engel DR. CCR2‐dependent Gr1
high
monocytes promote kidney injury in shiga toxin‐induced hemolytic uremic syndrome in mice. Eur J Immunol 2018; 48:990-1000. [DOI: 10.1002/eji.201747138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 12/18/2017] [Accepted: 02/05/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Judith‐Mira Pohl
- Institute of Experimental Immunology and ImagingUniversity Duisburg‐Essen and University Hospital Essen Essen Germany
| | - Julia K. Volke
- Institute of Experimental Immunology and ImagingUniversity Duisburg‐Essen and University Hospital Essen Essen Germany
| | - Stephanie Thiebes
- Institute of Experimental Immunology and ImagingUniversity Duisburg‐Essen and University Hospital Essen Essen Germany
| | - Alexandra Brenzel
- Institute of Experimental Immunology and ImagingUniversity Duisburg‐Essen and University Hospital Essen Essen Germany
| | - Kerstin Fuchs
- Werner Siemens Imaging CenterDepartment of Preclinical Imaging and RadiopharmacyEberhard Karls University of Tuebingen Tuebingen Germany
| | - Nicolas Beziere
- Werner Siemens Imaging CenterDepartment of Preclinical Imaging and RadiopharmacyEberhard Karls University of Tuebingen Tuebingen Germany
| | - Walter Ehrlichmann
- Werner Siemens Imaging CenterDepartment of Preclinical Imaging and RadiopharmacyEberhard Karls University of Tuebingen Tuebingen Germany
| | - Bernd J. Pichler
- Werner Siemens Imaging CenterDepartment of Preclinical Imaging and RadiopharmacyEberhard Karls University of Tuebingen Tuebingen Germany
| | - Anthony Squire
- Institute of Experimental Immunology and ImagingUniversity Duisburg‐Essen and University Hospital Essen Essen Germany
| | - Faikah Gueler
- Department of Nephrology and HypertensionHannover Medical School Hannover Germany
| | - Daniel R. Engel
- Institute of Experimental Immunology and ImagingUniversity Duisburg‐Essen and University Hospital Essen Essen Germany
| |
Collapse
|
15
|
Shiga Toxin Glycosphingolipid Receptors in Human Caco-2 and HCT-8 Colon Epithelial Cell Lines. Toxins (Basel) 2017; 9:toxins9110338. [PMID: 29068380 PMCID: PMC5705953 DOI: 10.3390/toxins9110338] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/11/2017] [Accepted: 10/19/2017] [Indexed: 12/21/2022] Open
Abstract
Shiga toxins (Stxs) released by enterohemorrhagic Escherichia coli (EHEC) into the human colon are the causative agents for fatal outcome of EHEC infections. Colon epithelial Caco-2 and HCT-8 cells are widely used for investigating Stx-mediated intestinal cytotoxicity. Only limited data are available regarding precise structures of their Stx receptor glycosphingolipids (GSLs) globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer), and lipid raft association. In this study we identified Gb3Cer and Gb4Cer lipoforms of serum-free cultivated Caco-2 and HCT-8 cells, chiefly harboring ceramide moieties composed of sphingosine (d18:1) and C16:0, C22:0 or C24:0/C24:1 fatty acid. The most significant difference between the two cell lines was the prevalence of Gb3Cer with C16 fatty acid in HCT-8 and Gb4Cer with C22–C24 fatty acids in Caco-2 cells. Lipid compositional analysis of detergent-resistant membranes (DRMs), which were used as lipid raft-equivalents, indicated slightly higher relative content of Stx receptor Gb3Cer in DRMs of HCT-8 cells when compared to Caco-2 cells. Cytotoxicity assays revealed substantial sensitivity towards Stx2a for both cell lines, evidencing little higher susceptibility of Caco-2 cells versus HCT-8 cells. Collectively, Caco-2 and HCT-8 cells express a plethora of different receptor lipoforms and are susceptible towards Stx2a exhibiting somewhat lower sensitivity when compared to Vero cells.
Collapse
|
16
|
Müller SK, Wilhelm I, Schubert T, Zittlau K, Imberty A, Madl J, Eierhoff T, Thuenauer R, Römer W. Gb3-binding lectins as potential carriers for transcellular drug delivery. Expert Opin Drug Deliv 2016; 14:141-153. [PMID: 27935765 DOI: 10.1080/17425247.2017.1266327] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Epithelial cell layers as well as endothelia forming the blood-brain barrier can drastically reduce the efficiency of drug targeting. Our goal was to investigate lectins recognizing the glycosphingolipid globotriaosylceramide (Gb3) for their potential as carriers for transcytotic drug delivery. METHODS We utilized an in vitro model based on Madin-Darby canine kidney cells transfected with Gb3 synthase to characterize transcytosis of the Gb3-binding lectins LecA from Pseudomonas aeruginosa and the B-subunit of Shiga toxin (StxB). RESULTS Both lectins were rapidly transcytosed from the apical to the basolateral plasma membrane and vice versa. Whereas StxB proceeded on retrograde and transcytotic routes, LecA avoided retrograde transport. This differential trafficking could be explained by our observation that LecA and StxB segregated into different domains during endocytosis. Furthermore, inhibiting the small GTPase Rab11a, which organizes trafficking through apical recycling endosomes, blocked basolateral to apical transcytosis of both lectins. CONCLUSIONS Gb3-binding lectins are promising candidates for transcytotic drug delivery. Our findings highlight that LecA and StxB, which both bind Gb3 but exhibit dissimilar valence and molecular structures of their carbohydrate binding sites and can take divergent intracellular trafficking routes. This opens up the possibility of developing tailor-made glycosphingolipid-binding carrier lectins, which take optimized trafficking pathways.
Collapse
Affiliation(s)
- Stefan K Müller
- a Faculty of Biology , Albert-Ludwigs-University Freiburg , Freiburg , Germany.,b BIOSS - Centre for Biological Signalling Studies , Albert-Ludwigs-University Freiburg , Freiburg , Germany
| | - Isabel Wilhelm
- a Faculty of Biology , Albert-Ludwigs-University Freiburg , Freiburg , Germany.,b BIOSS - Centre for Biological Signalling Studies , Albert-Ludwigs-University Freiburg , Freiburg , Germany.,c Spemann Graduate School of Biology and Medicine , Albert-Ludwigs University of Freiburg , Freiburg , Germany
| | - Thomas Schubert
- a Faculty of Biology , Albert-Ludwigs-University Freiburg , Freiburg , Germany.,b BIOSS - Centre for Biological Signalling Studies , Albert-Ludwigs-University Freiburg , Freiburg , Germany
| | - Katharina Zittlau
- a Faculty of Biology , Albert-Ludwigs-University Freiburg , Freiburg , Germany.,b BIOSS - Centre for Biological Signalling Studies , Albert-Ludwigs-University Freiburg , Freiburg , Germany
| | - Anne Imberty
- d Centre de Recherches sur les Macromolécules Végétales, UPR5301 , CNRS and Université Grenoble Alpes , Grenoble , France
| | - Josef Madl
- a Faculty of Biology , Albert-Ludwigs-University Freiburg , Freiburg , Germany.,b BIOSS - Centre for Biological Signalling Studies , Albert-Ludwigs-University Freiburg , Freiburg , Germany.,c Spemann Graduate School of Biology and Medicine , Albert-Ludwigs University of Freiburg , Freiburg , Germany
| | - Thorsten Eierhoff
- a Faculty of Biology , Albert-Ludwigs-University Freiburg , Freiburg , Germany.,b BIOSS - Centre for Biological Signalling Studies , Albert-Ludwigs-University Freiburg , Freiburg , Germany
| | - Roland Thuenauer
- a Faculty of Biology , Albert-Ludwigs-University Freiburg , Freiburg , Germany.,b BIOSS - Centre for Biological Signalling Studies , Albert-Ludwigs-University Freiburg , Freiburg , Germany
| | - Winfried Römer
- a Faculty of Biology , Albert-Ludwigs-University Freiburg , Freiburg , Germany.,b BIOSS - Centre for Biological Signalling Studies , Albert-Ludwigs-University Freiburg , Freiburg , Germany.,c Spemann Graduate School of Biology and Medicine , Albert-Ludwigs University of Freiburg , Freiburg , Germany
| |
Collapse
|
17
|
Pradhan S, Pellino C, MacMaster K, Coyle D, Weiss AA. Shiga Toxin Mediated Neurologic Changes in Murine Model of Disease. Front Cell Infect Microbiol 2016; 6:114. [PMID: 27747196 PMCID: PMC5040725 DOI: 10.3389/fcimb.2016.00114] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 09/12/2016] [Indexed: 12/22/2022] Open
Abstract
Seizures and neurologic involvement have been reported in patients infected with Shiga toxin (Stx) producing E. coli, and hemolytic uremic syndrome (HUS) with neurologic involvement is associated with more severe outcome. We investigated the extent of renal and neurologic damage in mice following injection of the highly potent form of Stx, Stx2a, and less potent Stx1. As observed in previous studies, Stx2a brought about moderate to acute tubular necrosis of proximal and distal tubules in the kidneys. Brain sections stained with hematoxylin and eosin (H&E) appeared normal, although some red blood cell congestion was observed. Microglial cell responses to neural injury include up-regulation of surface-marker expression (e.g., Iba1) and stereotypical morphological changes. Mice injected with Stx2a showed increased Iba1 staining, mild morphological changes associated with microglial activation (thickening of processes), and increased microglial staining per unit area. Microglial changes were observed in the cortex, hippocampus, and amygdala regions, but not the nucleus. Magnetic resonance imaging (MRI) of Stx2a-treated mice revealed no hyper-intensities in the brain, although magnetic resonance spectroscopy (MRS) revealed significantly decreased levels of phosphocreatine in the thalamus. Less dramatic changes were observed following Stx1 challenge. Neither immortalized microvascular endothelial cells from the cerebral cortex of mice (bEnd.3) nor primary human brain microvascular endothelial cells were found to be susceptible to Stx1 or Stx2a. The lack of susceptibility to Stx for both cell types correlated with an absence of receptor expression. These studies indicate Stx causes subtle, but identifiable changes in the mouse brain.
Collapse
Affiliation(s)
- Suman Pradhan
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati Cincinnati, OH, USA
| | | | - Kayleigh MacMaster
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati Cincinnati, OH, USA
| | - Dennis Coyle
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati Cincinnati, OH, USA
| | - Alison A Weiss
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati Cincinnati, OH, USA
| |
Collapse
|
18
|
Legros N, Dusny S, Humpf HU, Pohlentz G, Karch H, Müthing J. Shiga toxin glycosphingolipid receptors and their lipid membrane ensemble in primary human blood-brain barrier endothelial cells. Glycobiology 2016; 27:99-109. [PMID: 27558838 DOI: 10.1093/glycob/cww090] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/17/2016] [Accepted: 08/19/2016] [Indexed: 12/22/2022] Open
Abstract
Shiga toxin (Stx)-mediated injury to microvascular endothelial cells in the brain significantly contributes to the pathogenesis of the hemolytic-uremic syndrome caused by enterohemorrhagic Escherichia coli (EHEC). Stxs are AB5 toxins and the B-pentamers of the two major Stx subtypes Stx1a and Stx2a preferentially bind to the glycosphingolipid (GSL) globotriaosylceramide (Gb3Cer) expressed by human endothelial cells. Here we report on comprehensive structural analysis of the different lipoforms of Gb3Cer (Galα4Galβ4Glcβ1Cer) and globotetraosylceramide (Gb4Cer, GalNAcβ3Galα4Galβ4Glcβ1Cer, the less effective Stx receptor) of primary human brain microvascular endothelial cells and their association with lipid rafts. Detergent-resistant membranes (DRMs), obtained by sucrose density gradient ultracentrifugation, were used as lipid raft-analogous microdomains of the liquid-ordered phase and nonDRM fractions were employed as equivalents for the liquid-disordered phase of cell membranes. Structures of the prevalent lipoforms of Gb3Cer and Gb4Cer were those with Cer (d18:1, C16:0), Cer (d18:1, C22:0) and Cer (d18:1, C24:1/C24:0) determined by electrospray ionization mass spectrometry that was combined with thin-layer chromatography immunodetection using anti-Gb3Cer and anti-Gb4Cer antibodies as well as Stx1a and Stx2a subtypes. Association of Stx receptor GSLs was determined by co-localization with lipid raft-specific membrane protein flotillin-2 and canonical lipid raft marker sphingomyelin with Cer (d18:1, C16:0) and Cer (d18:1, C24:1/C24:0) in the liquid-ordered phase, whereas lyso-phosphatidylcholine was detectable exclusively in the liquid-disordered phase. Defining the precise microdomain structures of primary endothelial cells may help to unravel the initial mechanisms by which Stxs interact with their target cells and will help to develop novel preventive and therapeutic measures for EHEC-mediated diseases.
Collapse
Affiliation(s)
- Nadine Legros
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany
| | - Stefanie Dusny
- Institute for Food Chemistry, University of Münster, Corrensstr. 45, D-48149 Münster, Germany
| | - Hans-Ulrich Humpf
- Institute for Food Chemistry, University of Münster, Corrensstr. 45, D-48149 Münster, Germany
| | - Gottfried Pohlentz
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany
| | - Helge Karch
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany .,Interdisciplinary Center for Clinical Research (IZKF), University of Münster, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| |
Collapse
|
19
|
Thuenauer R, Müller SK, Römer W. Pathways of protein and lipid receptor-mediated transcytosis in drug delivery. Expert Opin Drug Deliv 2016; 14:341-351. [DOI: 10.1080/17425247.2016.1220364] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
20
|
Abstract
Post-infectious hemolytic uremic syndrome (HUS) is caused by specific pathogens in patients with no identifiable HUS-associated genetic mutation or autoantibody. The majority of episodes is due to infections by Shiga toxin (Stx) producing Escherichia coli (STEC). This chapter reviews the epidemiology and pathogenesis of STEC-HUS, including bacterial-derived factors and host responses. STEC disease is characterized by hematological (microangiopathic hemolytic anemia), renal (acute kidney injury) and extrarenal organ involvement. Clinicians should always strive for an etiological diagnosis through the microbiological or molecular identification of Stx-producing bacteria and Stx or, if negative, serological assays. Treatment of STEC-HUS is supportive; more investigations are needed to evaluate the efficacy of putative preventive and therapeutic measures, such as non-phage-inducing antibiotics, volume expansion and anti-complement agents. The outcome of STEC-HUS is generally favorable, but chronic kidney disease, permanent extrarenal, mainly cerebral complication and death (in less than 5 %) occur and long-term follow-up is recommended. The remainder of this chapter highlights rarer forms of (post-infectious) HUS due to S. dysenteriae, S. pneumoniae, influenza A and HIV and discusses potential interactions between these pathogens and the complement system.
Collapse
Affiliation(s)
- Denis F. Geary
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Franz Schaefer
- Division of Pediatric Nephrology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
21
|
Abstract
Enterohemorrhagic Escherichia coli (EHEC) is a highly pathogenic bacterial strain capable of causing watery or bloody diarrhea, the latter termed hemorrhagic colitis, and hemolytic-uremic syndrome (HUS). HUS is defined as the simultaneous development of non-immune hemolytic anemia, thrombocytopenia, and acute renal failure. The mechanism by which EHEC bacteria colonize and cause severe colitis, followed by renal failure with activated blood cells, as well as neurological symptoms, involves the interaction of bacterial virulence factors and specific pathogen-associated molecular patterns with host cells as well as the host response. The innate immune host response comprises the release of antimicrobial peptides as well as cytokines and chemokines in addition to activation and/or injury to leukocytes, platelets, and erythrocytes and activation of the complement system. Some of the bacterial interactions with the host may be protective in nature, but, when excessive, contribute to extensive tissue injury, inflammation, and thrombosis, effects that may worsen the clinical outcome of EHEC infection. This article describes aspects of the host response occurring during EHEC infection and their effects on specific organs.
Collapse
|
22
|
Grabowski EF, Liu B, Gerace MR, Kushak RI, Ingelfinger JR. Shiga toxin-1 Decreases Endothelial Cell Tissue Factor Pathway Inhibitor Not Co-localized with Tissue Factor on the Cell Membrane. Thromb Res 2015; 135:1214-7. [PMID: 25864889 DOI: 10.1016/j.thromres.2015.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 03/08/2015] [Accepted: 03/09/2015] [Indexed: 02/04/2023]
|
23
|
A symptomatic Fabry disease mouse model generated by inducing globotriaosylceramide synthesis. Biochem J 2015; 456:373-83. [PMID: 24094090 DOI: 10.1042/bj20130825] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Fabry disease is a lysosomal storage disorder in which neutral glycosphingolipids, predominantly Gb3 (globotriaosylceramide), accumulate due to deficient α-Gal A (α-galactosidase A) activity. The GLAko (α-Gal A-knockout) mouse has been used as a model for Fabry disease, but it does not have any symptomatic abnormalities. In the present study, we generated a symptomatic mouse model (G3Stg/GLAko) by cross-breeding GLAko mice with transgenic mice expressing human Gb3 synthase. G3Stg/GLAko mice had high Gb3 levels in major organs, and their serum Gb3 level at 5-25 weeks of age was 6-10-fold higher than that in GLAko mice of the same age. G3Stg/GLAko mice showed progressive renal impairment, with albuminuria at 3 weeks of age, decreased urine osmolality at 5 weeks, polyuria at 10 weeks and increased blood urea nitrogen at 15 weeks. The urine volume and urinary albumin concentration were significantly reduced in the G3Stg/GLAko mice when human recombinant α-Gal A was administered intravenously. These data suggest that Gb3 accumulation is a primary pathogenic factor in the symptomatic phenotype of G3Stg/GLAko mice, and that this mouse line is suitable for studying the pathogenesis of Fabry disease and for preclinical studies of candidate therapies.
Collapse
|
24
|
Moazzezy N, Oloomi M, Bouzari S. Effect of shiga toxin and its subunits on cytokine induction in different cell lines. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2014; 3:108-17. [PMID: 25035861 PMCID: PMC4082813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 04/22/2014] [Accepted: 05/20/2014] [Indexed: 11/01/2022]
Abstract
Shiga toxins (Stxs) are bacterial virulence factors produced by Shigella dysenteriae serotype 1 and Escherichia coli strains. Stxs are critical factors for the development of diseases such as severe bloody diarrhea and hemolytic uremic syndrome. Additionally, Stxs trigger the secretion of pro- inflammatory cytokines and chemokines, particularly in monocytes or macrophages. The inflammatory cytokines result in the modulation of the immune system, local inflammations and enhancement of cytotoxicity. In this study, stimulation of the pro- inflammatory cytokines IL-1α, IL-1β, IL-6, IL-8, and TNF-α was assessed by recombinant Stx (rStx) and its subunits (rStxA and rStxB). Cytokines expression at mRNA level was investigated by Reverse Transcription-Polymerase Chain Reaction (RT-PCR) method in HeLa cells and THP1 monocyte/ macrophage cell lines. After incubation with rStx and its recombinant subunits, the expression of IL-1α, IL- 6 and IL- 8 mRNAs was strongly induced in HeLa cells. In HeLa cells, low expression of IL-1α mRNA was shown by rStxB induction. Furthermore, the expression of IL-1α and IL-1β mRNAs in undifferentiated THP1 cells was only induced by rStx. In differentiated THP1 cells, rStx and its recombinant subunits elicited the expression of IL-1α, IL-1β, IL-8 and IL- 6 mRNAs. On the other hand, expression of TNF-α mRNA was only induced by rStx. Based on the data, the profile of cytokine induction in response to the rStx, and its subunits differs depending on the cell types.
Collapse
Affiliation(s)
- Neda Moazzezy
- Molecular Biology Unit, Pasteur Institute of Iran, Pasteur Ave. 13164 Tehran, Iran.
| | - Mana Oloomi
- Molecular Biology Unit, Pasteur Institute of Iran, Pasteur Ave. 13164 Tehran, Iran.,Corresponding author: Molecular Biology Unit, Pasteur Institute of Iran, Pasteur Ave. 13164, Tehran, Iran. E. mail:
| | - Saeid Bouzari
- Molecular Biology Unit, Pasteur Institute of Iran, Pasteur Ave. 13164 Tehran, Iran.
| |
Collapse
|
25
|
Sasaki N, Toyoda M. Glycoconjugates and related molecules in human vascular endothelial cells. Int J Vasc Med 2013; 2013:963596. [PMID: 24171112 PMCID: PMC3793293 DOI: 10.1155/2013/963596] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 08/12/2013] [Indexed: 11/18/2022] Open
Abstract
Vascular endothelial cells (ECs) form the inner lining of blood vessels. They are critically involved in many physiological functions, including control of vasomotor tone, blood cell trafficking, hemostatic balance, permeability, proliferation, survival, and immunity. It is considered that impairment of EC functions leads to the development of vascular diseases. The carbohydrate antigens carried by glycoconjugates (e.g., glycoproteins, glycosphingolipids, and proteoglycans) mainly present on the cell surface serve not only as marker molecules but also as functional molecules. Recent studies have revealed that the carbohydrate composition of the EC surface is critical for these cells to perform their physiological functions. In this paper, we consider the expression and functional roles of endogenous glycoconjugates and related molecules (galectins and glycan-degrading enzymes) in human ECs.
Collapse
Affiliation(s)
- Norihiko Sasaki
- Research Team for Geriatric Medicine (Vascular Medicine), Tokyo Metropolitan Institute of Gerontology, Sakaecho 35-2, Itabashi-ku, Tokyo 173-0015, Japan
| | - Masashi Toyoda
- Research Team for Geriatric Medicine (Vascular Medicine), Tokyo Metropolitan Institute of Gerontology, Sakaecho 35-2, Itabashi-ku, Tokyo 173-0015, Japan
| |
Collapse
|
26
|
Abstract
Ribosome-inactivating proteins (RIPs) were first isolated over a century ago and have been shown to be catalytic toxins that irreversibly inactivate protein synthesis. Elucidation of atomic structures and molecular mechanism has revealed these proteins to be a diverse group subdivided into two classes. RIPs have been shown to exhibit RNA N-glycosidase activity and depurinate the 28S rRNA of the eukaryotic 60S ribosomal subunit. In this review, we compare archetypal RIP family members with other potent toxins that abolish protein synthesis: the fungal ribotoxins which directly cleave the 28S rRNA and the newly discovered Burkholderia lethal factor 1 (BLF1). BLF1 presents additional challenges to the current classification system since, like the ribotoxins, it does not possess RNA N-glycosidase activity but does irreversibly inactivate ribosomes. We further discuss whether the RIP classification should be broadened to include toxins achieving irreversible ribosome inactivation with similar turnovers to RIPs, but through different enzymatic mechanisms.
Collapse
Affiliation(s)
- Matthew J Walsh
- RNA Biology Laboratory; Sheffield Institute for Translational Neuroscience (SITraN); Department of Neuroscience; University of Sheffield; Sheffield, UK
| | - Jennifer E Dodd
- RNA Biology Laboratory; Sheffield Institute for Translational Neuroscience (SITraN); Department of Neuroscience; University of Sheffield; Sheffield, UK
| | - Guillaume M Hautbergue
- RNA Biology Laboratory; Sheffield Institute for Translational Neuroscience (SITraN); Department of Neuroscience; University of Sheffield; Sheffield, UK
| |
Collapse
|
27
|
Grabowski EF, Kushak RI, Liu B, Ingelfinger JR. Shiga toxin downregulates tissue factor pathway inhibitor, modulating an increase in the expression of functional tissue factor on endothelium. Thromb Res 2013; 131:521-8. [PMID: 23642803 DOI: 10.1016/j.thromres.2013.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 03/04/2013] [Accepted: 03/08/2013] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Endothelial expression of tissue factor (TF) may play a major role in (Stx)-related hemolytic uremic syndrome. We examined human umbilical vein endothelial cell (HUVEC) monolayers to determine the interaction between TF and TF pathway inhibitor (TFPI), hypothesizing that changes in TFPI modulate TF expression. MATERIALS AND METHODS We studied 1) cell surface expression of globotriasylceramide (Gb3, the receptor for Stx) with Stx-1 (10 pM), TNFα (20 Ng/ml), or Stx-1 plus TNFα compared to control, 2) gene expression of TF and TFPI, 3) total cellular and cell surface antigenic TF and TFPI, 4) TFPI secretion into supernatant, and 5) factor Xa production. RESULTS AND CONCLUSIONS Gb3 expression, negligible with control and Stx-1 alone, increased significantly with TNFα and with Stx-1 plus TNFα. TF mRNA increased 1.25 ± 0.32- fold (N = 9; p = 0.041) with Stx-1 alone vs. 2.82 ± 0.92-fold (N = 13; p < 0.0005) with TNFα alone. However, Stx-1 plus TNFα yielded a 6.51 ± 3.48-fold increase (N = 17; p < 0.0005). TFPI mRNA decreased with TNFα (p < 0.001) and Stx-1 plus TNFα (p < 0.0005). Total cellular and cell surface TF antigen increased significantly with TNFα, but no further with Stx-1 plus TNFα. Total TFPI cellular and cell surface antigen levels, and TFPI secretion decreased significantly with Stx-1 plus TNFα. Median factor Xa production for Stx-1 plus TNFα vs TNFα alone increased (p < 0.001) 3.24-fold. Our results indicate that a subinhibitory concentration of Stx-1 plus TNFα impairs TFPI gene expression, synthesis, cell-surface association, and secretion, leading to augmented functional TF.
Collapse
Affiliation(s)
- Eric F Grabowski
- Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | | | | | | |
Collapse
|
28
|
Bauwens A, Betz J, Meisen I, Kemper B, Karch H, Müthing J. Facing glycosphingolipid-Shiga toxin interaction: dire straits for endothelial cells of the human vasculature. Cell Mol Life Sci 2013; 70:425-57. [PMID: 22766973 PMCID: PMC11113656 DOI: 10.1007/s00018-012-1060-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 05/25/2012] [Accepted: 06/14/2012] [Indexed: 12/23/2022]
Abstract
The two major Shiga toxin (Stx) types, Stx1 and Stx2, produced by enterohemorrhagic Escherichia coli (EHEC) in particular injure renal and cerebral microvascular endothelial cells after transfer from the human intestine into the circulation. Stxs are AB(5) toxins composed of an enzymatically active A subunit and the pentameric B subunit, which preferentially binds to the glycosphingolipid globotriaosylceramide (Gb3Cer/CD77). This review summarizes the current knowledge on Stx-caused cellular injury and the structural diversity of Stx receptors as well as the initial molecular interaction of Stxs with the human endothelium of different vascular beds. The varying lipoforms of Stx receptors and their spatial organization in lipid rafts suggest a central role in different modes of receptor-mediated endocytosis and intracellular destiny of the toxins. The design and development of tailored Stx neutralizers targeting the oligosaccharide-toxin recognition event has become a very real prospect to ameliorate or prevent life-threatening renal and neurological complications.
Collapse
Affiliation(s)
- Andreas Bauwens
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, 48149 Münster, Germany
| | - Josefine Betz
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, 48149 Münster, Germany
| | - Iris Meisen
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, 48149 Münster, Germany
- Interdisciplinary Center for Clinical Research, University of Münster, Domagkstr. 3, 48149 Münster, Germany
| | - Björn Kemper
- Center for Biomedical Optics and Photonics, University of Münster, Robert-Koch-Str. 45, 48149 Münster, Germany
| | - Helge Karch
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, 48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, 48149 Münster, Germany
- Interdisciplinary Center for Clinical Research, University of Münster, Domagkstr. 3, 48149 Münster, Germany
| |
Collapse
|
29
|
Chemokine expression in human astrocytes in response to shiga toxin 2. Int J Inflam 2012; 2012:135803. [PMID: 23304632 PMCID: PMC3529876 DOI: 10.1155/2012/135803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 11/07/2012] [Accepted: 11/07/2012] [Indexed: 12/11/2022] Open
Abstract
Infection with Shiga toxin- (Stx-) producing Escherichia coli can lead to hemolytic uremic syndrome (HUS). Approximately, 30% of patients with HUS suffer from complications in the central nervous system (CNS), which is an important determinant of mortality in such patients. Autopsy shows mostly edema and hypoxic-ischemic changes in the CNS, often with microhemorrhages. It has been suggested that Stx-induced damage to human brain endothelial cells, which are essential constituents of the blood-brain barrier, plays a crucial role in the development of the CNS complications. However, it is unclear whether Stx affects brain neuroglial cells. In the present study, we investigated the direct involvement of Stx in the inflammatory responses of human astrocytes (HASTs) treated with Stx. Immunohistochemistry and real-time PCR revealed that the expression of globotriaosylceramide (Gb3), the receptor for Stx2, and Gb3 synthase (GalT6) in HASTs was increased by interleukin-1β (IL-1β). Expression of both interleukin-8 (IL-8) and monocyte chemoattractant protein-1 (MCP-1) mRNA in HASTs was significantly upregulated by Stx2. These results suggest that Stx2 induces inflammatory responses, particularly through expression of chemokines, in HASTs expressing Gb3 and may, thus, affect brain glial cells, playing a key role in the pathogenesis of CNS manifestations associated with HUS.
Collapse
|
30
|
Trachtman H, Austin C, Lewinski M, Stahl RAK. Renal and neurological involvement in typical Shiga toxin-associated HUS. Nat Rev Nephrol 2012; 8:658-69. [PMID: 22986362 DOI: 10.1038/nrneph.2012.196] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Shiga toxin-producing Escherichia coli-associated haemolytic uraemic syndrome (STEC-HUS) is one of the most important causes of acute kidney injury in patients of all ages, especially in children. It can occur sporadically or in outbreaks. STEC-HUS is a systemic illness caused by toxin-mediated injury to the vascular endothelium and a generalized inflammatory response. The kidney and the brain are the two primary target organs. Nearly 40% of patients with STEC-HUS require at least temporary renal replacement therapy and up to 20% will have permanent residual kidney dysfunction. Neurological injury can be sudden and severe and is the most frequent cause of acute mortality in patients with STEC-HUS. Over the past 30 years, a wide range of inflammatory mediators have been linked to the pathogenesis of STEC-HUS and associated renal and neurological complications. Recently, evidence has accumulated that abnormal activation of the alternative pathway of complement occurs in patients with STEC-HUS. In the large outbreak of STEC-HUS caused by E. coli O104:H4 that occurred in Germany in May 2011, a large number of patients received eculizumab, a monoclonal antibody directed against C5, in an open-label manner. We describe the experience with eculizumab under these emergent circumstances at one large centre.
Collapse
Affiliation(s)
- Howard Trachtman
- Department of Pediatrics, Division of Nephrology, NYU Langone Medical Center, Clinical Translational Science Institute, Room 712, 227 East 30th Street, New York, NY 10016-9196, USA.
| | | | | | | |
Collapse
|
31
|
Bergan J, Dyve Lingelem AB, Simm R, Skotland T, Sandvig K. Shiga toxins. Toxicon 2012; 60:1085-107. [PMID: 22960449 DOI: 10.1016/j.toxicon.2012.07.016] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 06/19/2012] [Accepted: 07/25/2012] [Indexed: 02/03/2023]
Abstract
Shiga toxins are virulence factors produced by the bacteria Shigella dysenteriae and certain strains of Escherichia coli. There is currently no available treatment for disease caused by these toxin-producing bacteria, and understanding the biology of the Shiga toxins might be instrumental in addressing this issue. In target cells, the toxins efficiently inhibit protein synthesis by inactivating ribosomes, and they may induce signaling leading to apoptosis. To reach their cytoplasmic target, Shiga toxins are endocytosed and transported by a retrograde pathway to the endoplasmic reticulum, before the enzymatically active moiety is translocated to the cytosol. The toxins thereby serve as powerful tools to investigate mechanisms of intracellular transport. Although Shiga toxins are a serious threat to human health, the toxins may be exploited for medical purposes such as cancer therapy or imaging.
Collapse
Affiliation(s)
- Jonas Bergan
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Norway
| | | | | | | | | |
Collapse
|
32
|
Lucero MS, Mirarchi F, Goldstein J, Silberstein C. Intraperitoneal administration of Shiga toxin 2 induced neuronal alterations and reduced the expression levels of aquaporin 1 and aquaporin 4 in rat brain. Microb Pathog 2012; 53:87-94. [PMID: 22610042 DOI: 10.1016/j.micpath.2012.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 05/03/2012] [Accepted: 05/10/2012] [Indexed: 11/28/2022]
Abstract
Shiga toxin-producing Escherichia coli produces watery and hemorrhagic diarrhea, and hemolytic uremic syndrome (HUS) characterized by thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure. Central nervous system (CNS) complications are observed in around 30% of infant population with HUS. Common signs of severe CNS involvement leading to death include seizures, alteration of consciousness, hemiparesis, visual disturbances, and brain stem symptoms. The purpose of the present work was to study the effects of Shiga toxin 2 (Stx2) in the brain of rats intraperitoneally (i.p.) injected with a supernatant from recombinant E. coli expressing Stx2 (sStx2). Neurological alterations such as postural and motor abnormalities including lethargy, abnormal walking, and paralysis of hind legs, were observed in this experimental model of HUS in rats. Neuronal damage, as well as significant decrease in aquaporin 1 (AQP1) and aquaporin 4 (AQP4) expression levels were observed in the brain of rats, 2 days after sStx2 injection, compared to controls. Downregulation of aquaporin protein levels, and neuronal alterations, observed in brain of rats injected with sStx2, may be involved in edema formation and in neurological manifestations characteristic of HUS.
Collapse
Affiliation(s)
- María Soledad Lucero
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
33
|
Mohawk KL, O'Brien AD. Mouse models of Escherichia coli O157:H7 infection and shiga toxin injection. J Biomed Biotechnol 2011; 2011:258185. [PMID: 21274267 PMCID: PMC3022220 DOI: 10.1155/2011/258185] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 11/03/2010] [Indexed: 01/19/2023] Open
Abstract
Escherichia coli O157:H7 has been responsible for multiple food- and waterborne outbreaks of diarrhea and/or hemorrhagic colitis (HC) worldwide. More importantly, a portion of E. coli O157:H7-infected individuals, particularly young children, develop a life-threatening sequela of infection called hemolytic uremic syndrome (HUS). Shiga toxin (Stx), a potent cytotoxin, is the major virulence factor linked to the presentation of both HC and HUS. Currently, treatment of E. coli O157:H7 and other Stx-producing E. coli (STEC) infections is limited to supportive care. To facilitate development of therapeutic strategies and vaccines for humans against these agents, animal models that mimic one or more aspect of STEC infection and disease are needed. In this paper, we focus on the characteristics of various mouse models that have been developed and that can be used to monitor STEC colonization, disease, pathology, or combinations of these features as well as the impact of Stx alone.
Collapse
Affiliation(s)
- Krystle L. Mohawk
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Alison D. O'Brien
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
34
|
Biochemical, pathological and oncological relevance of Gb3Cer receptor. Med Oncol 2010; 28 Suppl 1:S675-84. [PMID: 21069478 DOI: 10.1007/s12032-010-9732-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Accepted: 10/21/2010] [Indexed: 10/18/2022]
Abstract
Glycosphingolipids are amphipathic molecules composed of hydrophilic oligosaccharide chain and a hydrophobic ceramide part, located primarily in the membrane microdomains of animal cells. Their oligosaccharide chains make them excellent candidates for the cell surface recognition molecules. Natural glycosphingolipid, globotriaosylceramide (Gal α1-4, Gal β1-4, Glc β1-1, ceramide), is also called CD77 and its expression was previously associated with proliferating centroblasts undergoing somatic hypermutation, but it has been demonstrate that globotriaosylceramide is not a reliable marker to discriminate human centroblasts from centrocytes. Globotriaosylceramide constitutes rare P k blood group antigen on erythrocytes, and it is also known as Burkitt's lymphoma antigen. On endothelial cells, globotriaosylceramide plays as the receptor for bacterial toxins of the Shiga family, also called verotoxins. Precise biological function and significance of globotriaosylceramide expression on endothelial cells remains to be the subject of many studies and it is believed globotriaosylceramide represents an example of a glycolipid antigen able to transduce a signal leading to apoptosis. In past decade, cancer researches put a great afford in determining new therapeutic agents such as bacterial toxins against tumor malignancies. Reports have demonstrated that verotoxin-1 induces apoptosis in solid tumor cell lines expressing globotriaosylceramide such as astrocytoma, renal cell carcinoma, colon cancer and breast cancer due to verotoxin-1 high specificity and apoptosis-inducing properties, and therefore, it is suggested to be an anticancer agent. Verotoxins have been investigated weather they could reduce treatment side-effects and toxicity to normal tissues and become a new oncological tool in cancer labeling.
Collapse
|
35
|
Higashi N, Matsumura Y, Mizuno F, Kasahara K, Sugiura S, Mikasa K, Kita E. Enhanced expression of ATP-binding cassette transporter A1 in non-rafts decreases the sensitivity of vascular endothelial cells to Shiga toxin. Microb Pathog 2010; 49:141-52. [DOI: 10.1016/j.micpath.2010.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 05/06/2010] [Accepted: 05/17/2010] [Indexed: 11/16/2022]
|
36
|
Johannes L, Römer W. Shiga toxins--from cell biology to biomedical applications. Nat Rev Microbiol 2009; 8:105-16. [PMID: 20023663 DOI: 10.1038/nrmicro2279] [Citation(s) in RCA: 359] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Shiga toxin-producing Escherichia coli is an emergent pathogen that can induce haemolytic uraemic syndrome. The toxin has received considerable attention not only from microbiologists but also in the field of cell biology, where it has become a powerful tool to study intracellular trafficking. In this Review, we summarize the Shiga toxin family members and their structures, receptors, trafficking pathways and cellular targets. We discuss how Shiga toxin affects cells not only by inhibiting protein biosynthesis but also through the induction of signalling cascades that lead to apoptosis. Finally, we discuss how Shiga toxins might be exploited in cancer therapy and immunotherapy.
Collapse
Affiliation(s)
- Ludger Johannes
- Institut Curie - Centre de Recherche and CNRS UMR144, Traffic, Signalling and Delivery Laboratory, 26 rue d'Ulm, 75248 Paris Cedex 05, France.
| | | |
Collapse
|
37
|
Shiga toxin 1-induced inflammatory response in lipopolysaccharide-sensitized astrocytes is mediated by endogenous tumor necrosis factor alpha. Infect Immun 2009; 78:1193-201. [PMID: 20008539 DOI: 10.1128/iai.00932-09] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hemolytic-uremic syndrome (HUS) is generally caused by Shiga toxin (Stx)-producing Escherichia coli. Endothelial dysfunction mediated by Stx is a central aspect in HUS development. However, inflammatory mediators such as bacterial lipopolysaccharide (LPS) and polymorphonuclear neutrophils (PMN) contribute to HUS pathophysiology by potentiating Stx effects. Acute renal failure is the main feature of HUS, but in severe cases, patients can develop neurological complications, which are usually associated with death. Although the mechanisms of neurological damage remain uncertain, alterations of the blood-brain barrier associated with brain endothelial injury is clear. Astrocytes (ASTs) are the most abundant inflammatory cells of the brain that modulate the normal function of brain endothelium and neurons. The aim of this study was to evaluate the effects of Stx type 1 (Stx1) alone or in combination with LPS in ASTs. Although Stx1 induced a weak inflammatory response, pretreatment with LPS sensitized ASTs to Stx1-mediated effects. Moreover, LPS increased the level of expression of the Stx receptor and its internalization. An early inflammatory response, characterized by the release of tumor necrosis factor alpha (TNF-alpha) and nitric oxide and PMN-chemoattractant activity, was induced by Stx1 in LPS-sensitized ASTs, whereas activation, evidenced by higher levels of glial fibrillary acid protein and cell death, was induced later. Furthermore, increased adhesion and PMN-mediated cytotoxicity were observed after Stx1 treatment in LPS-sensitized ASTs. These effects were dependent on NF-kappaB activation or AST-derived TNF-alpha. Our results suggest that TNF-alpha is a pivotal effector molecule that amplifies Stx1 effects on LPS-sensitized ASTs, contributing to brain inflammation and leading to endothelial and neuronal injury.
Collapse
|
38
|
Kamioka I, Yoshiya K, Satomura K, Kaito H, Fujita T, Iijima K, Nakanishi K, Yoshikawa N, Nozu K, Matsuo M. Risk factors for developing severe clinical course in HUS patients: a national survey in Japan. Pediatr Int 2008; 50:441-6. [PMID: 19143964 DOI: 10.1111/j.1442-200x.2008.02605.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Hemolytic uremic syndrome (HUS) is characterized by acute renal failure, thrombocytopenia and hemolytic anemia. Cases accompanied by prodromal gastrointestinal tract symptoms are referred to as typical HUS. Some severe HUS patients require dialysis or develop central nervous system (CNS) disorders after the onset of HUS. METHODS Patients who developed typical HUS in 2001 and 2002 in Japan, 127 in all, were the study subjects. To identify the risk factors for the development of a severe clinical course, clinical and laboratory data were analyzed on logistic regression. RESULTS Two of the 127 patients died (1.6%): one from acute cardiac failure and the other from a CNS disorder. Thirty-five patients required dialysis (28%) and 30 had CNS symptoms (24%). Multivariate analysis indicated that the risk factors for need for dialysis were serum sodium and alanine aminotransferase (ALT) levels of </=130 mEq/L and >/=70 IU/L, respectively, at the onset of HUS and those for developing CNS disorders were dialysis and C-reactive protein (CRP) >/=5.0 mg/dL at the onset of HUS. CONCLUSIONS Because patients with these risk factors, such as low serum sodium, high ALT or high CRP levels, may require dialysis or develop CNS disorders, they should be treated carefully in the early stage of HUS.
Collapse
Affiliation(s)
- Ichiro Kamioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Takahashi K, Funata N, Ikuta F, Sato S. Neuronal apoptosis and inflammatory responses in the central nervous system of a rabbit treated with Shiga toxin-2. J Neuroinflammation 2008; 5:11. [PMID: 18355415 PMCID: PMC2330034 DOI: 10.1186/1742-2094-5-11] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Accepted: 03/21/2008] [Indexed: 12/19/2022] Open
Abstract
Background Shiga toxins (Stxs) are the major agents responsible for hemorrhagic colitis and hemolytic-uremic syndrome (HUS) during infections caused by Stx-producing Escherichia coli (STEC) such as serotype O157:H7. Central nervous system (CNS) involvement is an important determinant of mortality in diarrhea associated-HUS. It has been suggested that vascular endothelial injuries caused by Stxs play a crucial role in the development of the disease. The current study investigates the relationship between the cytotoxic effects of Stxs and inflammatory responses in a rabbit brain treated with Stx2. Methods In a rabbit model treated with purified Stx2 or PBS(-), we examined the expression of the Stx receptor globotriaosylceramide (Gb3)/CD77 in the CNS and microglial activation using immunohistochemistry. The relationship between inflammatory responses and neuronal cell death was analyzed by the following methods: real time quantitative reverse transcriptase (RT)-polymerase chain reaction (PCR) to determine the expression levels of pro-inflammatory cytokines, and the terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) method to detect apoptotic changes. Results Gb3/CD77 expression was detected in endothelial cells but not in neurons or glial cells. In the spinal cord gray matter, significant levels of Gb3/CD77 expression were observed. Severe endothelial injury and microvascular thrombosis resulted in extensive necrotic infarction, which led to acute neuronal damage. Conversely, in the brain, Stx receptor expression was much lower. The observed neuropathology was less severe. However, neuronal apoptosis was observed at the onset of neurological symptoms, and the number of apoptotic cells significantly increased in the brain at a later stage, several days after onset. Microglial activation was observed, and tumor necrosis factor (TNF)-α and interleukin (IL)-1β mRNA in the CNS parenchyma was significantly up-regulated. There was significant overexpression of TNF-α transcripts in the brain. Conclusion This study indicates that Stx2 may not directly damage neural cells, but rather inflammatory responses occur in the brain parenchyma in response to primary injury by Stx2 in vascular endothelial cells expressing Gb3/CD77. These findings suggest that neuroinflammation may play a critical role in neurodegenerative processes during STEC infection and that anti-inflammatory intervention may have therapeutic potential.
Collapse
Affiliation(s)
- Kiyomi Takahashi
- Department of Microbiology, Iwate Medical University School of Medicine, 19-1 Uchimaru, Morioka, Iwate 020-8505, Japan.
| | | | | | | |
Collapse
|
40
|
Glycosphingolipids in vascular endothelial cells: relationship of heterogeneity in Gb3Cer/CD77 receptor expression with differential Shiga toxin 1 cytotoxicity. Glycoconj J 2008; 25:291-304. [DOI: 10.1007/s10719-007-9091-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Revised: 10/18/2007] [Accepted: 11/15/2007] [Indexed: 11/27/2022]
|
41
|
p38 mitogen-activated protein kinase mediates lipopolysaccharide and tumor necrosis factor alpha induction of shiga toxin 2 sensitivity in human umbilical vein endothelial cells. Infect Immun 2007; 76:1115-21. [PMID: 18086809 DOI: 10.1128/iai.01300-07] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Escherichia coli O157:H7 Shiga toxin 2 (Stx2), one of the causative agents of hemolytic-uremic syndrome, is toxic to endothelial cells, including primary cultured human umbilical vein endothelial cells (HUVEC). This sensitivity of cells to Stx2 can be increased with either lipopolysaccharide (LPS) or tumor necrosis factor alpha (TNF-alpha). The goal of the present study was to identify the intracellular signaling pathway(s) by which LPS and TNF-alpha sensitize HUVEC to the cytotoxic effects of Stx2. To identify these pathways, specific pharmacological inhibitors and small interfering RNAs were tested with cell viability endpoints. A time course and dose response experiment for HUVEC exposure to LPS and TNF-alpha showed that a relatively short exposure to either agonist was sufficient to sensitize the cells to Stx2 and that both agonists stimulated intracellular signaling pathways within a short time. Cell viability assays indicated that the p38 mitogen-activated protein kinase (MAPK) inhibitors SB202190 and SB203580 and the general protein synthesis inhibitor cycloheximide inhibited both the LPS and TNF-alpha sensitization of HUVEC to Stx2, while all other inhibitors tested did not inhibit this sensitization. Additionally, SB202190 reduced the cellular globotriaosylceramide content under LPS- and TNF-alpha-induced conditions. In conclusion, our results show that LPS and TNF-alpha induction of Stx2 sensitivity in HUVEC is mediated through a pathway that includes p38 MAPK. These results indicate that inhibition of p38 MAPK in endothelial cells may protect a host from the deleterious effects of Stx2.
Collapse
|
42
|
Moake JL. Thrombotic Thrombocytopenic Purpura and the Hemolytic-Uremic Syndrome. Platelets 2007. [DOI: 10.1016/b978-012369367-9/50812-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
43
|
Chen TG, Chen TL, Chang HC, Tai YT, Cherng YG, Chang YT, Chen RM. Oxidized low-density lipoprotein induces apoptotic insults to mouse cerebral endothelial cells via a Bax-mitochondria-caspase protease pathway. Toxicol Appl Pharmacol 2006; 219:42-53. [PMID: 17239413 DOI: 10.1016/j.taap.2006.11.031] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2006] [Revised: 11/16/2006] [Accepted: 11/19/2006] [Indexed: 11/16/2022]
Abstract
Cerebral endothelial cells (CECs) are crucial components of the blood-brain barrier. Oxidized low-density lipoprotein (oxLDL) can induce cell injuries. In this study, we attempted to evaluate the effects of oxLDL on mouse CECs and its possible mechanisms. Mouse CECs were isolated from brain tissues and identified by immunocytochemical staining of vimentin and Factor VIII. oxLDL was prepared from LDL oxidation by copper sulfate. Exposure of mouse CECs to oxLDL decreased cell viability in concentration- and time-dependent manners. oxLDL time-dependently caused shrinkage of cell morphologies. Administration of oxLDL to CECs induced DNA fragmentation in concentration- and time-dependent manners. Analysis of the cell cycle revealed that oxLDL concentration- and time-dependently increased the proportion of CECs which underwent apoptosis. Analysis of confocal microscopy and immunoblot revealed that oxLDL significantly increased cellular and mitochondrial Bax levels as well as the translocation of this proapoptotic protein from the cytoplasm to mitochondria. In parallel with the increase in the levels and translocation of Bax, oxLDL time-dependently decreased the mitochondrial membrane potential. Exposure of mouse CECs to oxLDL decreased the amounts of mitochondrial cytochrome c, but enhanced cytosolic cytochrome c levels. The amounts of intracellular reactive oxygen species were significantly augmented after oxLDL administration. Sequentially, oxLDL increased activities of caspase-9, -3, and -6 in time-dependent manners. Pretreatment with Z-VEID-FMK, an inhibitor of caspase-6, significantly decreased caspase-6 activity and the oxLDL-induced DNA fragmentation and cell apoptosis. This study showed that oxLDL induces apoptotic insults to CECs via signal-transducing events, including enhancing Bax translocation, mitochondrial dysfunction, cytochrome c release, increases in intracellular reactive oxygen species, and cascade activation of caspase-9, -3, and -6. Therefore, oxLDL can damage the blood-brain barrier through induction of CEC apoptosis via a Bax-mitochondria-caspase protease pathway.
Collapse
Affiliation(s)
- Tyng-Guey Chen
- Department of Anesthesiology, Wan-Fang Medical Center, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
44
|
Guessous F, Marcinkiewicz M, Polanowska-Grabowska R, Kongkhum S, Heatherly D, Obrig T, Gear ARL. Shiga toxin 2 and lipopolysaccharide induce human microvascular endothelial cells to release chemokines and factors that stimulate platelet function. Infect Immun 2006; 73:8306-16. [PMID: 16299328 PMCID: PMC1307066 DOI: 10.1128/iai.73.12.8306-8316.2005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Shiga toxins (Stxs) produced by Shigella dysenteriae type 1 and enterohemorrhagic Escherichia coli are the most common cause of hemolytic-uremic syndrome (HUS). It is well established that vascular endothelial cells, mainly those located in the renal microvasculature, are targets for Stxs. The aim of the present research was to evaluate whether E. coli-derived Shiga toxin 2 (Stx2) incubated with human microvascular endothelial cells (HMEC-1) induces release of chemokines and other factors that might stimulate platelet function. HMEC-1 were exposed for 24 h in vitro to Stx2, lipopolysaccharide (LPS), or the Stx2-LPS combination, and chemokine production was assessed by immunoassay. More interleukin-8 was released than stromal cell-derived factor 1alpha (SDF-1alpha) or SDF-1beta and RANTES. The Stx2-LPS combination potentiated chemokine release, but Stx2 alone caused more release of SDF-1alpha at 24 h than LPS or Stx2-LPS did. In the presence of low ADP levels, HMEC-1 supernatants activated platelet function assessed by classical aggregometry, single-particle counting, granule secretion, P-selectin exposure, and the formation of platelet-monocyte aggregates. Supernatants from HMEC-1 exposed only to Stx2 exhibited enhanced exposure of platelet P-selectin and platelet-THP-1 cell interactions. Blockade of platelet cyclooxygenase by indomethacin prevented functional activation. The chemokine RANTES enhanced platelet aggregation induced by SDF-1alpha, macrophage-derived chemokine, or thymus and activation-regulated chemokine in the presence of very low ADP levels. These data support the hypothesis that microvascular endothelial cells exposed to E. coli O157:H7-derived Stx2 and LPS release chemokines and other factors, which when combined with low levels of primary agonists, such as ADP, cause platelet activation and promote the renal thrombosis associated with HUS.
Collapse
Affiliation(s)
- Fadila Guessous
- Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, P.O. Box 700833, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The emergence of enterohaemorrhagic Escherichia coli (EHEC) as major human pathogens began with the identification of serotype O157:H7 in the early 1980s as the cause of various food-borne outbreaks of severe intestinal disease. The key virulence factors include verocytotoxins (Vt) and effectors and adhesins associated with type III secretion systems. Tracing the origins of human outbreaks reveals that the primary source of this organism is the ruminant gastro-intestinal tract and a variety of transmission routes to humans have been identified. The epidemiology of E. coli O157:H7 within cattle and other ruminants has been studied extensively and the prevalence of non-O157:H7 serotypes contrasts with the observed dominance of E. coli O157:H7 amongst human EHEC isolates. Although there is some evidence that EHEC cause disease in young animals, the high prevalence of Vt within healthy ruminants suggests that this is not a virulence factor within these species. An understanding of the mechanisms underpinning EHEC persistence within their natural reservoir hosts and the development of a molecular understanding of EHEC biology and evolution could eventually allow a reduction in the incidence of human disease and may reduce future threats. The use of animal models to replicate and study human EHEC pathogenesis is described.
Collapse
Affiliation(s)
- Stuart W Naylor
- Animal Health Group, Scottish Agricultural College, West Mains Road, Edinburgh, EH9 3JG, UK.
| | | | | |
Collapse
|
46
|
Nolasco LH, Turner NA, Bernardo A, Tao Z, Cleary TG, Dong JF, Moake JL. Hemolytic uremic syndrome-associated Shiga toxins promote endothelial-cell secretion and impair ADAMTS13 cleavage of unusually large von Willebrand factor multimers. Blood 2005; 106:4199-209. [PMID: 16131569 PMCID: PMC1895236 DOI: 10.1182/blood-2005-05-2111] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Shiga toxin 1 (Stx-1) and Stx-2 produced by enterohemorrhagic Escherichia coli cause the diarrhea-associated hemolytic uremic syndrome (HUS). This type of HUS is characterized by obstruction of the glomeruli and renal microvasculature by platelet-fibrin thrombi, acute renal failure, thrombocytopenia, microvascular hemolytic anemia, and plasma levels of von Willebrand factor (VWF)-cleaving protease (ADAMTS13) activity that are within a broad normal range. We investigated the mechanism of initial platelet accumulation on Stx-stimulated endothelial cells. Stx-1 or Stx-2 (1-10 nM) stimulated the rapid secretion of unusually large (UL) VWF multimeric strings from human umbilical vein endothelial cells (HUVECs) or human glomerular microvascular endothelial cells (GMVECs). Perfused normal human platelets immediately adhered to the secreted ULVWF multimeric strings. Nanomolar concentrations (1-10 nM) of the Shiga toxins were as effective in inducing the formation of ULVWF-platelet strings as millimolar concentrations (0.1-20 mM) of histamine. The rate of ULVWF-platelet string cleavage by plasma or recombinant ADAMTS13 was delayed by 3 to 10 minutes (or longer) in the presence of 10 nM Stx-1 or Stx-2 compared with 20 mM histamine. Stx-induced formation of ULVWF strings, and impairment of ULVWF-platelet string cleavage by ADAMTS13, may promote initial platelet adhesion above glomerular endothelial cells. These processes may contribute to the evolution of glomerular occlusion by platelet and fibrin thrombi in diarrhea-associated HUS.
Collapse
Affiliation(s)
- Leticia H Nolasco
- Hematology Research Section, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Viebig NK, Wulbrand U, Förster R, Andrews KT, Lanzer M, Knolle PA. Direct activation of human endothelial cells by Plasmodium falciparum-infected erythrocytes. Infect Immun 2005; 73:3271-7. [PMID: 15908351 PMCID: PMC1111820 DOI: 10.1128/iai.73.6.3271-3277.2005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cytoadherence of Plasmodium falciparum-infected erythrocytes (PRBC) to endothelial cells causes severe clinical disease, presumably as a of result perfusion failure and tissue hypoxia. Cytoadherence to endothelial cells is increased by endothelial cell activation, which is believed to occur in a paracrine fashion by mediators such as tumor necrosis factor alpha (TNF-alpha) released from macrophages that initially recognize PRBC. Here we provide evidence that PRBC directly stimulate human endothelial cells in the absence of macrophages, leading to increased expression of adhesion-promoting molecules, such as intercellular adhesion molecule 1. Endothelial cell stimulation by PRBC required direct physical contact for a short time (30 to 60 min) and was correlated with parasitemia. Gene expression profiling of endothelial cells stimulated by PRBC revealed increased expression levels of chemokine and adhesion molecule genes. PRBC-stimulated endothelial cells especially showed increased expression of molecules involved in parasite adhesion but failed to express molecules promoting leukocyte adhesion, such as E-selectin and vascular cell adhesion molecule 1, even after challenge with TNF-alpha. Collectively, our data suggest that stimulation of endothelial cells by PRBC may have two effects: prevention of parasite clearance through increased cytoadherence and attenuation of leukocyte binding to endothelial cells, thereby preventing deleterious immune reactivity.
Collapse
Affiliation(s)
- Nicola K Viebig
- Hygiene Institut, Abteilung Parasitologie, Universität Heidelberg, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
48
|
Pohlenz JF, Winter KR, Dean-Nystrom EA. Shiga-toxigenic Escherichia coli-inoculated neonatal piglets develop kidney lesions that are comparable to those in humans with hemolytic-uremic syndrome. Infect Immun 2005; 73:612-6. [PMID: 15618202 PMCID: PMC538958 DOI: 10.1128/iai.73.1.612-616.2005] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Kidney lesions similar to those in humans with hemolytic-uremic syndrome were observed histologically in 82 of 122 piglets inoculated intragastrically with Shiga-toxigenic Escherichia coli but not in 29 controls. The locations of lesions matched locations where Stx-2 binding and Gb3 (globotriasylceramide receptors for Stx) were identified immunohistochemically.
Collapse
Affiliation(s)
- J F Pohlenz
- National Animal Disease Center, USDA, ARS, P.O. Box 70, Ames, IA 50010-0070, USA
| | | | | |
Collapse
|
49
|
Cleary TG. The role of Shiga-toxin-producing Escherichia coli in hemorrhagic colitis and hemolytic uremic syndrome. ACTA ACUST UNITED AC 2004; 15:260-5. [PMID: 15494950 DOI: 10.1053/j.spid.2004.07.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The Shiga-toxin-producing E. coli represent a major class of pathogens that have been defined over the last twenty years. They cause distinctive clinical manifestations such as afebrile bloody diarrhea with severe abdominal pain (hemorrhagic colitis) and microangiopathic hemolytic anemia with renal failure (hemolytic uremic syndrome). The most common Shiga-toxin-producing E. coli is serotype O157:H7, although at least one hundred different serotypes share the virulence traits and clinical manifestations with this organism. Understanding the pathophysicology, improving diagnostic tools, and developing a treatment strategy are important areas of ongoing investigations.
Collapse
Affiliation(s)
- Thomas G Cleary
- Center for Infectious Diseases, School of Public Health, University of Texas Medical School, Houston, TX 77030, USA.
| |
Collapse
|
50
|
Harrison LM, van Haaften WCE, Tesh VL. Regulation of proinflammatory cytokine expression by Shiga toxin 1 and/or lipopolysaccharides in the human monocytic cell line THP-1. Infect Immun 2004; 72:2618-27. [PMID: 15102770 PMCID: PMC387879 DOI: 10.1128/iai.72.5.2618-2627.2004] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2003] [Revised: 01/06/2004] [Accepted: 02/02/2004] [Indexed: 11/20/2022] Open
Abstract
Infection with Shiga toxin (Stx)-producing bacteria and the subsequent release of Stxs and endotoxins into the bloodstream may damage blood vessels in the colon, kidneys, and central nervous system, leading to bloody diarrhea, acute renal failure, and neurological complications. The proinflammatory cytokines tumor necrosis factor alpha (TNF-alpha) and interleukin-1beta (IL-1beta) may contribute to the pathogenesis of Stx-induced vascular lesions by up-regulating toxin receptor expression on endothelial cells. We previously showed that macrophages treated with purified Shiga toxin 1 (Stx1) or lipopolysaccharides (LPS) secrete TNF-alpha and IL-1beta. Northern blot analysis revealed that treatment of the human monocytic cell line THP-1 with LPS induced a rapid and transient increase in steady-state TNF-alpha and IL-1beta transcripts. In contrast, Stx1 induced slower but prolonged elevations in cytokine transcripts. The presence of both stimulants resulted in optimal cytokine mRNA induction in terms of kinetics and prolonged expression. Compared to LPS, Stx1 was a poor inducer of IL-1beta protein expression, although levels of soluble IL-1beta induced by all treatments continually increased over 72 h. IL-1beta transcripts were not induced by Stx1 B-subunits. Using the transcriptional inhibitor actinomycin D, we determined that treatment with Stx1 or Stx1 plus LPS induced cytokine transcripts with increased stability compared to transcripts induced by LPS alone. For all treatments, IL-1beta mRNA decay was slower than TNF-alpha. Collectively, our data suggest that Stxs affect cytokine expression, in part, at the posttranscriptional level by stabilizing mRNAs. Optimal TNF-alpha expression occurs when both Stxs and LPS are present.
Collapse
Affiliation(s)
- Lisa M Harrison
- Department of Medical Microbiology and Immunology, Texas A&M University System Health Science Center, College Station, Texas 77843-1114, USA
| | | | | |
Collapse
|