1
|
Roxas PB, Cruz J, Horelka NR, Burgos C, Radwanski J, Baires F, Sierra-Hoffman M, Hesse H, Madril AC. Typhus group Rickettsia community-acquired bacterial central nervous system infections: We must think outside the box! J Neurol Sci 2024; 466:123281. [PMID: 39447222 DOI: 10.1016/j.jns.2024.123281] [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: 05/27/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
Typhus group rickettsiosis (TGR), caused by Rickettsia typhi and Rickettsia prowazekii, are globally distributed vector-borne diseases with increasing cases. Diagnosis is usually clinical, confirmed by seroconversion of IgG antibodies. Human infection occurs in diverse geographic areas with some developing CNS infection characterized by fever, headache, meningismus, and/or focal signs - usually beyond the first week of initial symptomatology. Seizures and other CNS manifestations have been observed. When untreated, infection may result in neurologic sequelae and even death. This study presents a systematic review of all documented cases of Rickettsia typhi meningoencephalitis published since 2015 with the addition of five cases of TGR in South Coastal Texas, USA. This review followed the guidelines outlined in PRISMA. A schematic explanation of the pathophysiology is offered. CSF may present with high opening pressure, mild to moderate pleocytosis, mildly elevated protein levels, and low csf/serum glucose ratio, or normal findings. Meningeal enhancement, intracranial hypertension, and focal abnormalities have been described in imaging studies, but can be normal. Treatment with doxycycline leads to prompt resolution of symptoms. Failure to initiate early empiric treatment can lead to serious consequences. The study recommends routine testing for TGR in patients from endemic areas with classical symptoms when other diagnoses are inconclusive or in cases with atypical presentations. The authors advocate for incorporating empiric treatment for murine typhus into community-acquired bacterial meningitis guidelines in endemic areas; and stress the importance of enhancing laboratory diagnostic capabilities in public health entities world-wide. Further studies of community acquired mengingoencephalitis caused by TGR are highly encouraged.
Collapse
Affiliation(s)
- Pauline B Roxas
- Family Medicine Residency, Detar Healthcare System - Texas A&M University School of Medicine, Victoria, TX, USA
| | - Justice Cruz
- Victoria College, Department of Science, Victoria, TX, USA.
| | | | - Cesar Burgos
- Universidad Nacional Autónoma de Honduras, Instituto Nacional Cardiopulmonar El Tórax, Tegucigalpa, Honduras
| | | | - Fernando Baires
- Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Miguel Sierra-Hoffman
- El Campo Memorial Hospital, El Campo, Texas, USA; Clinical Professor of Texas A&M, Rural Health Medicine Residency Program, Victoria, Texas, USA
| | - Heike Hesse
- Instituto de Investigaciones One Health, Universidad Tecnológica Centroamericana, Tegucigalpa, Honduras.
| | - Amy C Madril
- Department of Hospital Medicine, El Campo Memorial Hospital, El Campo, TX, USA
| |
Collapse
|
2
|
Li Z, Zhang Z, Yu P, Ni Y. Microbial communities, antibiotic resistance genes, and virulence factors in urinary infectious stone-associated urinary tract infections. Urolithiasis 2024; 52:88. [PMID: 38874649 DOI: 10.1007/s00240-024-01588-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024]
Abstract
Urinary infectious stones are challenging due to bacterial involvement, necessitating a comprehensive understanding of these conditions. Antibiotic-resistant urease-producing bacteria further complicate clinical management. In this study, analysis of urine and stone samples from urinary tract infection (UTI) patients revealed microbial shifts, gene enrichment in stones, and metabolic pathway disparities; antibiotic resistance gene trends were phylum-specific, urease-producing bacteria are at risk of acquiring AMR carried by Enterobacteriaceae under antibiotic, emphasizing potential AMR dissemination between them; Correlations of key pathogenic species in kidney stone and urine microbial communities highlight the need for targeted therapeutic strategies to manage complexities in UTIs; Stones and urine contain a variety of deleterious genes even before antibiotic use, and piperacillin/tazobactam better reduced the abundance of antibiotic resistance genes in stones and urine. The presence of diverse antibiotic resistance and virulence genes underscores challenges in clinical management and emphasizes the need for effective treatment strategies to mitigate risks associated with UTIs and urinary infectious stone formation. Ongoing research is vital for advancing knowledge and developing innovative approaches to address these urological conditions.
Collapse
Affiliation(s)
- Ziyun Li
- Center for Translational Medicine Research, Shandong Provincial Third Hospital, Shandong University, Jinan, China
| | - Zhaocun Zhang
- Urology Department, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Peng Yu
- Urology Department, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China
- Urology Department, Weihai Municipal Hospital, Shandong University, Weihai, China
| | - Yongliang Ni
- Urology Department, Shandong Provincial Third Hospital, Shandong University, Jinan, China.
| |
Collapse
|
3
|
Fitzsimmons L, Bublitz D, Clark T, Hackstadt T. Rickettsia rickettsii virulence determinants RARP2 and RapL mitigate IFN- β signaling in primary human dermal microvascular endothelial cells. mBio 2024; 15:e0345023. [PMID: 38445878 PMCID: PMC11005427 DOI: 10.1128/mbio.03450-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/13/2024] [Indexed: 03/07/2024] Open
Abstract
We compared the growth characteristics of a virulent Rickettsia rickettsii strain (Sheila Smith) to an attenuated R. rickettsii stain (Iowa) and a non-pathogenic species (R. montanensis) in primary human dermal microvascular endothelial cells (HDMEC). All replicated in Vero cells, however, only the Sheila Smith strain productively replicated in HDMECs. The Iowa strain showed minimal replication over a 24-h period, while R. montanensis lost viability and induced lysis of the HDMECs via a rapid programmed cell death response. Both the virulent and attenuated R. rickettsii strains, but not R. montanensis, induced an interferon-1 response, although the response was of lesser magnitude and delayed in the Sheila Smith strain. IFN-β secretion correlated with increased host cell lysis, and treatment with anti-IFNAR2 antibody decreased lysis from Iowa-infected but not Sheila Smith-infected cells. Both Sheila Smith- and Iowa-infected cells eventually lysed, although the response from Sheila Smith was delayed and showed characteristics of apoptosis. We, therefore, examined whether reconstitution of the Iowa strain with two recently described putative virulence determinants might enhance survival of Iowa within HDMECs. Reconstitution with RARP2, which is inhibitory to anterograde trafficking through the Golgi apparatus, reduced IFN-β secretion but had no effect on cell lysis. RapL, which proteolytically processes surface exposed autotransporters and enhances replication of Iowa in Guinea pigs, suppressed both IFN-β production and host cell lysis. These findings suggest distinct mechanisms by which virulent spotted fever group rickettsiae may enhance intracellular survival and replication.IMPORTANCEWe examined a naturally occurring non-pathogenic rickettsial species, R. montanensis, a laboratory-attenuated R. rickettsii strain (Iowa), and a fully virulent R. rickettsii strain (Sheila Smith) for growth in human dermal microvascular endothelial cells. The two avirulent strains replicated poorly or not at all. Only the virulent Sheila Smith strain replicated. IFN-β production correlated with the inhibition of R. rickettsii Iowa. Reconstitution of Iowa with either of two recently described putative virulence determinants altered the IFN-β response. A rickettsial ankyrin repeat protein, RARP2, disrupts the trans-Golgi network and inhibits IFN-β secretion. An autotransporter peptidase, RapL, restores proteolytic maturation of outer membrane autotransporters and diminishes the IFN-β response to enhance cell survival and permit replication of the recombinant strain. These studies point the way toward discovery of mechanisms for innate immune response avoidance by virulent rickettsia.
Collapse
Affiliation(s)
- Liam Fitzsimmons
- Host-Parasite Interactions Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - DeAnna Bublitz
- Host-Parasite Interactions Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Tina Clark
- Host-Parasite Interactions Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Ted Hackstadt
- Host-Parasite Interactions Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
4
|
Sankar P, Mishra BB. Early innate cell interactions with Mycobacterium tuberculosis in protection and pathology of tuberculosis. Front Immunol 2023; 14:1260859. [PMID: 37965344 PMCID: PMC10641450 DOI: 10.3389/fimmu.2023.1260859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/26/2023] [Indexed: 11/16/2023] Open
Abstract
Tuberculosis (TB) remains a significant global health challenge, claiming the lives of up to 1.5 million individuals annually. TB is caused by the human pathogen Mycobacterium tuberculosis (Mtb), which primarily infects innate immune cells in the lungs. These immune cells play a critical role in the host defense against Mtb infection, influencing the inflammatory environment in the lungs, and facilitating the development of adaptive immunity. However, Mtb exploits and manipulates innate immune cells, using them as favorable niche for replication. Unfortunately, our understanding of the early interactions between Mtb and innate effector cells remains limited. This review underscores the interactions between Mtb and various innate immune cells, such as macrophages, dendritic cells, granulocytes, NK cells, innate lymphocytes-iNKT and ILCs. In addition, the contribution of alveolar epithelial cell and endothelial cells that constitutes the mucosal barrier in TB immunity will be discussed. Gaining insights into the early cellular basis of immune reactions to Mtb infection is crucial for our understanding of Mtb resistance and disease tolerance mechanisms. We argue that a better understanding of the early host-pathogen interactions could inform on future vaccination approaches and devise intervention strategies.
Collapse
Affiliation(s)
| | - Bibhuti Bhusan Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| |
Collapse
|
5
|
Vaca DJ, Frenzel F, Ballhorn W, Torres SG, Leisegang MS, Günther S, Bender D, Kraiczy P, Göttig S, Kempf VAJ. Adhesion of human pathogenic bacteria to endothelial cells is facilitated by fibronectin interaction. Microbes Infect 2023; 25:105172. [PMID: 37343664 DOI: 10.1016/j.micinf.2023.105172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 06/07/2023] [Accepted: 06/10/2023] [Indexed: 06/23/2023]
Abstract
Human pathogenic bacteria circulating in the bloodstream need to find a way to interact with endothelial cells (ECs) lining the blood vessels to infect and colonise the host. The extracellular matrix (ECM) of ECs might represent an attractive initial target for bacterial interaction, as many bacterial adhesins have reported affinities to ECM proteins, in particular to fibronectin (Fn). Here, we analysed the general role of EC-expressed Fn for bacterial adhesion. For this, we evaluated the expression levels of ECM coding genes in different ECs, revealing that Fn is the highest expressed gene and thereby, it is highly abundant in the ECM environment of ECs. The role of Fn as a mediator in bacterial cell-host adhesion was evaluated in adhesion assays of Acinetobacter baumannii, Bartonella henselae, Borrelia burgdorferi, and Staphylococcus aureus to ECs. The assays demonstrated that bacteria colocalised with Fn fibres, as observed by confocal laser scanning microscopy. Fn removal from the ECM environment (FN1 knockout ECs) diminished bacterial adherence to ECs in both static and dynamic adhesion assays to varying extents, as evaluated via absolute quantification using qPCR. Interactions between adhesins and Fn might represent the crucial step for the adhesion of human-pathogenic Gram-negative and Gram-positive bacteria targeting the ECs as a niche of infection.
Collapse
Affiliation(s)
- Diana J Vaca
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Fabienne Frenzel
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Wibke Ballhorn
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Sara Garcia Torres
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Matthias S Leisegang
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, Parkstraße 1, 61231, Bad Nauheim, Germany.
| | - Daniela Bender
- Federal Institute for Vaccines and Biomedicines, Department of Virology, Paul Ehrlich Institute, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany.
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Stephan Göttig
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Volkhard A J Kempf
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| |
Collapse
|
6
|
Buggert M, Price DA, Mackay LK, Betts MR. Human circulating and tissue-resident memory CD8 + T cells. Nat Immunol 2023:10.1038/s41590-023-01538-6. [PMID: 37349380 DOI: 10.1038/s41590-023-01538-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/04/2023] [Indexed: 06/24/2023]
Abstract
Our current knowledge of human memory CD8+ T cells is derived largely from studies of the intravascular space. However, emerging data are starting to challenge some of the dogmas based on this work, suggesting that a conceptual revision may be necessary. In this review, we provide a brief history of the field and summarize the biology of circulating and tissue-resident memory CD8+ T cells, which are ultimately responsible for effective immune surveillance. We also incorporate recent findings into a biologically integrated model of human memory CD8+ T cell differentiation. Finally, we address how future innovative human studies could improve our understanding of anatomically localized CD8+ T cells to inform the development of more effective immunotherapies and vaccines, the need for which has been emphasized by the global struggle to contain severe acute respiratory syndrome coronavirus 2.
Collapse
Affiliation(s)
- Marcus Buggert
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, UK
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Michael R Betts
- Institute for Immunology and Center for AIDS Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Kosak L, Satz N, Jutzi M, Dobec M, Schlagenhauf P. Spotted fever group rickettsiae and Anaplasma phagocytophilum in Borrelia burgdorferi sensu lato seropositive individuals with or without Lyme disease: A retrospective analysis. New Microbes New Infect 2023; 53:101139. [PMID: 37168237 PMCID: PMC10165448 DOI: 10.1016/j.nmni.2023.101139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/13/2023] [Accepted: 03/26/2023] [Indexed: 05/13/2023] Open
Abstract
Background The Ixodes ricinus tick is the main vector of Borrelia burgdorferi and tick-borne encephalitis virus in Switzerland. Spotted fever group Rickettsiae (SFG) and Anaplasma phagocytophilum have been detected in Swiss ticks, however, information about the extent and clinical presentation of these infections in humans is scant. Methods Indirect fluorescent antibody tests for SFG rickettsiae and Anaplasma phagocytophilum were performed on serum samples of 121 Borrelia burgdorferi seropositive patients with and without Lyme disease and 43 negative controls. Results Out of 121 Borrelia burgdorferi seropositive individuals, 65 (53.7%) were seropositive for IgG and 15 (12.4%) for IgM antibodies to SFG rickettsiae. IgM antibodies were detected more frequently in early-than in late-stage of Lyme disease (12 out of 51 and 2 out of 49; respectively; p = 0.0078). Significantly higher IgG antibody titers against SFG rickettsiae were found in patients with late-stage compared to patients with early-stage Lyme disease (mean titer 1:261 and 1:129, respectively; p = 0.038). This difference was even more pronounced in patients with acrodermatitis chronica atrophicans compared to patients with early stage of Lyme disease (mean titer 1:337 and 1:129, respectively; p = 0.009).In patients presenting with fatigue, headache and myalgia, the prevalence of IgG antibodies against SFG rickettsiae was significantly higher (7 out of 11; 63.6%) than in Borrelia burgdorferi seropositive individuals without clinical illness (1 out of 10; 10%; p = 0.024). IgG antibodies to Anaplasma phagocytophilum were detected in 12 out of 121 individuals (9.9%), no IgM antibodies were found. Conclusion Infections with SFG rickettsiae and Anaplasma phagocytophilum are underdiagnosed and should be ruled out after a tick bite. Further studies are needed to elucidate the possible causative role of SFG rickettsiae for myalgia, headache and long-lasting fatigue after a tick bite and to determine the necessity for an antibiotic treatment.
Collapse
Affiliation(s)
- Leonie Kosak
- University of Zürich, Institute for Epidemiology, Biostatistics and Prevention, Zürich, Switzerland
- Corresponding author.
| | | | - Markus Jutzi
- Analytica Medizinische Laboratorien AG, Zürich, Switzerland
| | - Marinko Dobec
- Analytica Medizinische Laboratorien AG, Zürich, Switzerland
| | - Patricia Schlagenhauf
- University of Zürich, Institute for Epidemiology, Biostatistics and Prevention, WHO Collaborating Centre for Travellers' Health, Department of Global and Public Health, MilMedBiol Competence Centre, Zürich, Switzerland
| |
Collapse
|
8
|
Moran TE, Hammers DE, Lee SW. The Role of Host-Cellular Responses in COVID-19 Endothelial Dysfunction. Curr Drug Targets 2022; 23:1555-1566. [PMID: 35748550 DOI: 10.2174/1389450123666220624094940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/11/2022] [Accepted: 05/11/2022] [Indexed: 01/25/2023]
Abstract
SARS-CoV2, Severe acute respiratory syndrome coronavirus 2, is a novel member of the human coronavirus family that has recently emerged worldwide to cause COVID-19 disease. COVID-19 disease has been declared a worldwide pandemic with over 270 million total cases, and >5 million deaths as of this writing. Although co-morbidities and preexisting conditions have played a significant role in the severity of COVID-19, the hallmark feature of severe disease associated with SARS-CoV2 is respiratory failure. Recent findings have demonstrated a key role for endothelial dysfunction caused by SARS-CoV2 in these clinical outcomes, characterized by endothelial inflammation, the persistence of a pro-coagulative state, and major recruitment of leukocytes and other immune cells to localized areas of endothelial dysfunction. Though it is generally recognized that endothelial impairment is a major contributor to COVID-19 disease, studies to examine the initial cellular events involved in triggering endothelial dysfunction are needed. In this article, we review the general strategy of pathogens to exploit endothelial cells and the endothelium to cause disease. We discuss the role of the endothelium in COVID-19 disease and highlight very recent findings that identify key signaling and cellular events that are associated with the initiation of SARS-CoV2 infection. These studies may reveal specific molecular pathways that can serve as potential means of therapeutic development against COVID-19 disease.
Collapse
Affiliation(s)
- Thomas E Moran
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Daniel E Hammers
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.,Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA
| | - Shaun W Lee
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.,Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA.,W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN, USA.,Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
9
|
Zhu J, Chen H, Le Y, Guo J, Liu Z, Dou X, Lu D. Salvianolic acid A regulates pyroptosis of endothelial cells via directly targeting PKM2 and ameliorates diabetic atherosclerosis. Front Pharmacol 2022; 13:1009229. [PMID: 36425580 PMCID: PMC9679534 DOI: 10.3389/fphar.2022.1009229] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2023] Open
Abstract
Rescuing endothelial cells from pyroptotic cell death emerges as a potential therapeutic strategy to combat diabetic atherosclerosis. Salvianolic acid A (SAA) is a major water-soluble phenolic acid in the Salvia miltiorrhiza Bunge, which has been used in traditional Chinese medicine (TCM) and health food products for a long time. This study investigated whether SAA-regulated pyruvate kinase M2 (PKM2) functions to protect endothelial cells. In streptozotocin (STZ)-induced diabetic ApoE-/- mice subjected to a Western diet, SAA attenuated atherosclerotic plaque formation and inhibited pathological changes in the aorta. In addition, SAA significantly prevented NLRP3 inflammasome activation and pyroptosis of endothelial cells in the diabetic atherosclerotic aortic sinus or those exposed to high glucose. Mechanistically, PKM2 was verified to be the main target of SAA. We further revealed that SAA directly interacts with PKM2 at its activator pocket, inhibits phosphorylation of Y105, and hinders the nuclear translocation of PKM2. Also, SAA consistently decreased high glucose-induced overproduction of lactate and partially lactate-dependent phosphorylation of PKR (a regulator of the NLRP3 inflammasome). Further assay on Phenylalanine (PKM2 activity inhibitor) proved that SAA exhibits the function in high glucose-induced pyroptosis of endothelial cells dependently on PKM2 regulation. Furthermore, an assay on c16 (inhibitor of PKR activity) with co-phenylalanine demonstrated that the regulation of the phosphorylated PKR partially drives PKM2-dependent SAA modulation of cell pyroptosis. Therefore, this article reports on the novel function of SAA in the pyroptosis of endothelial cells and diabetic atherosclerosis, which provides important insights into immunometabolism reprogramming that is important for diabetic cardiovascular disease complications therapy.
Collapse
Affiliation(s)
- Ji Zhu
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Zhejiang Chinese Medical University, Hangzhou, China
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hang Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yifei Le
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianan Guo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijun Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaobing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dezhao Lu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
10
|
Falcinelli E, Petito E, Gresele P. The role of platelets, neutrophils and endothelium in COVID-19 infection. Expert Rev Hematol 2022; 15:727-745. [PMID: 35930267 DOI: 10.1080/17474086.2022.2110061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION COVID-19 is associated to an increased risk of thrombosis, as a result of a complex process that involves the activation of vascular and circulating cells, the release of soluble inflammatory and thrombotic mediators and blood clotting activation. AREAS COVERED This article reviews the pathophysiological role of platelets, neutrophils and the endothelium, and of their interactions, in the thrombotic complications of COVID-19 patients, and the current and future therapeutic approaches targeting these cell types. EXPERT OPINION Virus-induced platelet, neutrophil and endothelial cell changes are crucial triggers of the thrombotic complications and of the adverse evolution of COVID-19. Both the direct interaction with the virus and the associated cytokine storm concur to trigger cell activation in a classical thromboinflammatory vicious circle. Although heparin has proven to be an effective prophylactic and therapeutic weapon for the prevention and treatment of COVID-19-associated thrombosis, it acts downstream of the cascade of events triggered by SARS-CoV-2. The identification of specific molecular targets interrupting the thromboinflammatory cascade upstream, and more specifically acting either on the interaction of SARS-CoV-2 with blood and vascular cells or on the specific signalling mechanisms associated with their COVID-19-associated activation, might theoretically offer greater protection with potentially lesser side effects.
Collapse
Affiliation(s)
- E Falcinelli
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - E Petito
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - P Gresele
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
11
|
Stolarz AJ, Mu S, Zhang H, Fouda AY, Rusch NJ, Ding Z. Opinion: Endothelial Cells - Macrophage-Like Gatekeepers? Front Immunol 2022; 13:902945. [PMID: 35619719 PMCID: PMC9127206 DOI: 10.3389/fimmu.2022.902945] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/19/2022] [Indexed: 01/08/2023] Open
Affiliation(s)
- Amanda J Stolarz
- Department of Pharmaceutical Sciences, College of Pharmacy, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Shengyu Mu
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Huiliang Zhang
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Abdelrahman Y Fouda
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Nancy J Rusch
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Zufeng Ding
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
12
|
Oxidative Stress Evaluation in Dogs Affected with Canine Monocytic Ehrlichiosis. Antioxidants (Basel) 2022; 11:antiox11020328. [PMID: 35204210 PMCID: PMC8868293 DOI: 10.3390/antiox11020328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 01/06/2023] Open
Abstract
The study aimed to evaluate the concentration of reactive oxidative metabolites (R-OOHs), the antioxidant barrier (OXY), and the ratio between R-OOHs and OXY (OSi) and thiol groups of plasma compounds (SHp) in in canine monocytic ehrlichiosis. Thirty dogs affected with monocytic ehrlichiosis (canine monocytic ehrlichiosis group—CME group) and ten healthy dogs (control group—CTR group) were evaluated. CME was diagnosed by the presence of clinical signs and the detection of anti-Ehlichia canis antibodies. Oxidative stress parameters of two groups were compared using the Mann–Whitney test. Significance was set at p < 0.05. Spearman rank correlation was performed to analyze oxidative stress, and hematological and biochemical variables in the CME group. All dogs affected with CME showed a wide spectrum of clinical signs such as lethargy, anorexia, fever, weight loss, lymph adenomegaly, splenomegaly, subcutaneous and mucosal petechial and ecchymosis, and vomiting. Anaemia, leukocytosis, thrombocytopenia, hyperglobulinemia, hypoalbuminemia and an increase of blood urea nitrogen and creatinine are also detected. Results showed significantly lower values of SHp in the CME group than in CTR. A statistically significant difference in the number of white blood cells, platelets, and blood urea nitrogen concentration was assayed comparing to the two groups. A negative correlation between SHp and hemoglobin concentration was recorded. These preliminary results may suggest a possible function of oxidative stress in the onset of clinical signs during the course of CME.
Collapse
|
13
|
Loterio RK, Zamboni DS, Newton HJ. Keeping the host alive - lessons from obligate intracellular bacterial pathogens. Pathog Dis 2021; 79:6424899. [PMID: 34755855 DOI: 10.1093/femspd/ftab052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/04/2021] [Indexed: 01/20/2023] Open
Abstract
Mammals have evolved sophisticated host cell death signaling pathways as an important immune mechanism to recognize and eliminate cell intruders before they establish their replicative niche. However, intracellular bacterial pathogens that have co-evolved with their host have developed a multitude of tactics to counteract this defense strategy to facilitate their survival and replication. This requires manipulation of pro-death and pro-survival host signaling pathways during infection. Obligate intracellular bacterial pathogens are organisms that absolutely require an eukaryotic host to survive and replicate, and therefore they have developed virulence factors to prevent diverse forms of host cell death and conserve their replicative niche. This review encapsulates our current understanding of these host-pathogen interactions by exploring the most relevant findings of Anaplasma spp., Chlamydia spp., Rickettsia spp. and Coxiella burnetii modulating host cell death pathways. A detailed comprehension of the molecular mechanisms through which these obligate intracellular pathogens manipulate regulated host cell death will not only increase the current understanding of these difficult-to-study pathogens but also provide insights into new tools to study regulated cell death and the development of new therapeutic approaches to control infection.
Collapse
Affiliation(s)
- Robson Kriiger Loterio
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto Medical School, FMRP/USP. Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil.,Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, 3000, Victoria, Australia
| | - Dario S Zamboni
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto Medical School, FMRP/USP. Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil
| | - Hayley J Newton
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, 3000, Victoria, Australia
| |
Collapse
|
14
|
Šikić J, Planinić Z, Matišić V, Friščić T, Molnar V, Jagačić D, Vujičić L, Tudorić N, Postružin Gršić L, Ljubičić Đ, Primorac D. COVID-19: The Impact on Cardiovascular System. Biomedicines 2021; 9:1691. [PMID: 34829920 PMCID: PMC8615470 DOI: 10.3390/biomedicines9111691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 has been circulating in population worldwide for the past year and a half, and thus a vast amount of scientific literature has been produced in order to study the biology of the virus and the pathophysiology of COVID-19, as well as to determine the best way to prevent infection, treat the patients and eliminate the virus. SARS-CoV-2 binding to the ACE2 receptor is the key initiator of COVID-19. The ability of SARS-CoV-2 to infect various types of cells requires special attention to be given to the cardiovascular system, as it is commonly affected. Thorough diagnostics and patient monitoring are beneficial in reducing the risk of cardiovascular morbidity and to ensure the most favorable outcomes for the infected patients, even after they are cured of the acute disease. The multidisciplinary nature of the fight against the COVID-19 pandemic requires careful consideration from the attending clinicians, in order to provide fast and reliable treatment to their patients in accordance with evidence-based medicine principles. In this narrative review, we reviewed the available literature on cardiovascular implications of COVID-19; both the acute and the chronic.
Collapse
Affiliation(s)
- Jozica Šikić
- Department of Cardiology, Clinical Hospital Sveti Duh, 10000 Zagreb, Croatia; (Z.P.); (T.F.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | - Zrinka Planinić
- Department of Cardiology, Clinical Hospital Sveti Duh, 10000 Zagreb, Croatia; (Z.P.); (T.F.)
| | - Vid Matišić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | - Tea Friščić
- Department of Cardiology, Clinical Hospital Sveti Duh, 10000 Zagreb, Croatia; (Z.P.); (T.F.)
| | - Vilim Molnar
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | - Dorijan Jagačić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | - Lovro Vujičić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | - Neven Tudorić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
| | | | - Đivo Ljubičić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
- Clinical Hospital Dubrava, 10000 Zagreb, Croatia
| | - Dragan Primorac
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (V.M.); (D.J.); (L.V.); (N.T.); (Đ.L.); (D.P.)
- Eberly College of Science, The Pennsylvania State University, University Park, State College, PA 16802, USA
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
- Medical School, University of Split, 21000 Split, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School REGIOMED, 96 450 Coburg, Germany
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| |
Collapse
|
15
|
The Identity of Human Tissue-Emigrant CD8 + T Cells. Cell 2020; 183:1946-1961.e15. [PMID: 33306960 DOI: 10.1016/j.cell.2020.11.019] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 10/02/2020] [Accepted: 11/12/2020] [Indexed: 11/23/2022]
Abstract
Lymphocyte migration is essential for adaptive immune surveillance. However, our current understanding of this process is rudimentary, because most human studies have been restricted to immunological analyses of blood and various tissues. To address this knowledge gap, we used an integrated approach to characterize tissue-emigrant lineages in thoracic duct lymph (TDL). The most prevalent immune cells in human and non-human primate efferent lymph were T cells. Cytolytic CD8+ T cell subsets with effector-like epigenetic and transcriptional signatures were clonotypically skewed and selectively confined to the intravascular circulation, whereas non-cytolytic CD8+ T cell subsets with stem-like epigenetic and transcriptional signatures predominated in tissues and TDL. Moreover, these anatomically distinct gene expression profiles were recapitulated within individual clonotypes, suggesting parallel differentiation programs independent of the expressed antigen receptor. Our collective dataset provides an atlas of the migratory immune system and defines the nature of tissue-emigrant CD8+ T cells that recirculate via TDL.
Collapse
|
16
|
Plasencia-Muñoz B, Avelar-González FJ, De la Garza M, Jacques M, Moreno-Flores A, Guerrero-Barrera AL. Actinobacillus pleuropneumoniae Interaction With Swine Endothelial Cells. Front Vet Sci 2020; 7:569370. [PMID: 33195549 PMCID: PMC7658479 DOI: 10.3389/fvets.2020.569370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/28/2020] [Indexed: 12/26/2022] Open
Abstract
Actinobacillus pleuropneumonia is a swine (host) specific respiratory pathogen and the etiological agent of swine pleuropneumonia which affects pigs of all ages, many being asymptomatic carriers. This pathogen has high morbidity and mortality rates which generates large economic losses for the pig industry. Actinobacillus pleuropneumoniae is a widely studied bacterium, however its pathogenesis is not yet fully understood. The prevalence of the 18 serotypes of A. pleuropneumoniae varies by geographic region, in North American area, more specifically in Mexico, serotypes 1, 3, 5b, and 7 show higher prevalence. Actinobacillus pleuropneumoniae is described as a strict extracellular pathogen with tropism for lower respiratory tract. However, this study depicts the ability of these serotypes to adhere to non-phagocytic cells, using an endothelial cell model, as well as their ability to internalize them, proposing it could be considered as an intracellular pathogen.
Collapse
Affiliation(s)
- Berenice Plasencia-Muñoz
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Francisco J Avelar-González
- Laboratorio de Estudios Ambientales, Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Mireya De la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Mario Jacques
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Adriana Moreno-Flores
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Alma L Guerrero-Barrera
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| |
Collapse
|
17
|
Kamel M, Pavulraj S, Fauler B, Mielke T, Azab W. Equid Herpesvirus-1 Exploits the Extracellular Matrix of Mononuclear Cells to Ensure Transport to Target Cells. iScience 2020; 23:101615. [PMID: 33015592 PMCID: PMC7521387 DOI: 10.1016/j.isci.2020.101615] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/27/2020] [Accepted: 09/23/2020] [Indexed: 12/21/2022] Open
Abstract
Mononuclear cells are the first line of defense against microbial infection. Yet, several viruses have evolved different mechanisms to overcome host defenses to ensure their spread. Here, we show unique mechanisms of how equid herpesvirus-1 manipulates peripheral blood mononuclear cells (PBMC) to travel further in the body. (1) "PBMC-hitching": at the initial contact, herpesviruses lurk in the extracellular matrix (ECM) of PBMC without entering the cells. The virus exploits the components of the ECM to bind, transport, and then egress to infect other cells. (2) "Intracellular delivery": transendothelial migration is a physiological mechanism where mononuclear cells can transmigrate through the endothelial cells. The virus was intangible and probably did not interfere with such a mechanism where the infected PBMC can probably deliver the virus inside the endothelium. (3) "Classical-fusion": this process is well mastered by herpesviruses due to a set of envelope glycoproteins that facilitate cell-cell fusion and virus spread.
Collapse
Affiliation(s)
- Mohamed Kamel
- Institut für Virologie, Robert von Ostertag-Haus, Zentrum für Infektionsmedizin, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163 Berlin, Germany.,Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, 12211 Cairo, Egypt
| | - Selvaraj Pavulraj
- Institut für Virologie, Robert von Ostertag-Haus, Zentrum für Infektionsmedizin, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163 Berlin, Germany
| | - Beatrix Fauler
- Max-Planck-Institut für Molekulare Genetik, Mikroskopie und Kryo-Elektronenmikroskopie Servicegruppe, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Thorsten Mielke
- Max-Planck-Institut für Molekulare Genetik, Mikroskopie und Kryo-Elektronenmikroskopie Servicegruppe, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Walid Azab
- Institut für Virologie, Robert von Ostertag-Haus, Zentrum für Infektionsmedizin, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163 Berlin, Germany
| |
Collapse
|
18
|
Varikuti S, Jha BK, Holcomb EA, McDaniel JC, Karpurapu M, Srivastava N, McGwire BS, Satoskar AR, Parinandi NL. The role of vascular endothelium and exosomes in human protozoan parasitic diseases. ACTA ACUST UNITED AC 2020; 4. [PMID: 33089078 PMCID: PMC7575144 DOI: 10.20517/2574-1209.2020.27] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The vascular endothelium is a vital component in maintaining the structure and function of blood vessels. The endothelial cells (ECs) mediate vital regulatory functions such as the proliferation of cells, permeability of various tissue membranes, and exchange of gases, thrombolysis, blood flow, and homeostasis. The vascular endothelium also regulates inflammation and immune cell trafficking, and ECs serve as a replicative niche for many bacterial, viral, and protozoan infectious diseases. Endothelial dysfunction can lead to vasodilation and pro-inflammation, which are the hallmarks of many severe diseases. Exosomes are nanoscale membrane-bound vesicles that emerge from cells and serve as important extracellular components, which facilitate communication between cells and maintain homeostasis during normal and pathophysiological states. Exosomes are also involved in gene transfer, inflammation and antigen presentation, and mediation of the immune response during pathogenic states. Protozoa are a diverse group of unicellular organisms that cause many infectious diseases in humans. In this regard, it is becoming increasingly evident that many protozoan parasites (such as Plasmodium, Trypanosoma, Leishmania, and Toxoplasma) utilize exosomes for the transfer of their virulence factors and effector molecules into the host cells, which manipulate the host gene expression, immune responses, and other biological activities to establish and modulate infection. In this review, we discuss the role of the vascular endothelium and exosomes in and their contribution to pathogenesis in malaria, African sleeping sickness, Chagas disease, and leishmaniasis and toxoplasmosis with an emphasis on their actions on the innate and adaptive immune mechanisms of resistance.
Collapse
Affiliation(s)
- Sanjay Varikuti
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH 43201, USA.,Department of Bioscience & Biotechnology, Banasthali University, Banasthali 304022, India
| | - Bijay Kumar Jha
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Erin A Holcomb
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Jodi C McDaniel
- College of Nursing, The Ohio State University, Columbus, OH 43201, USA
| | - Manjula Karpurapu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Nidhi Srivastava
- Department of Bioscience & Biotechnology, Banasthali University, Banasthali 304022, India
| | - Bradford S McGwire
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Abhay R Satoskar
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Narasimham L Parinandi
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| |
Collapse
|
19
|
Huynh T, Gary JM, Welch SR, Coleman-McCray J, Harmon JR, Kainulainen MH, Bollweg BC, Ritter JM, Shieh WJ, Nichol ST, Zaki SR, Spiropoulou CF, Spengler JR. Lassa virus antigen distribution and inflammation in the ear of infected strain 13/N Guinea pigs. Antiviral Res 2020; 183:104928. [PMID: 32898586 DOI: 10.1016/j.antiviral.2020.104928] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/25/2020] [Accepted: 09/01/2020] [Indexed: 10/23/2022]
Abstract
Sudden-onset sensorineuronal hearing loss (SNHL) is reported in approximately one-third of survivors of Lassa fever (LF) and remains the most prominent cause of Lassa virus (LASV)-associated morbidity in convalescence. Using a guinea pig model of LF, and incorporating animals from LASV vaccine trials, we investigated viral antigen distribution and histopathology in the ear of infected animals to elucidate the pathogenesis of hearing loss associated with LASV infection. Antigen was detected only in animals that succumbed to disease and was found within structures of the inner ear that are intimately associated with neural detection and/or translation of auditory stimuli and in adjacent vasculature. No inflammation or viral cytopathic changes were observed in the inner ear or surrounding structures in these animals. In contrast, no viral antigen was detected in the ear of surviving animals. However, all survivors that exhibited clinical signs of disease during the course of infection developed perivascular mononuclear inflammation within and adjacent to the ear, indicating an ongoing inflammatory response in these animals that may contribute to hearing loss. These data contribute to the knowledge of LASV pathogenesis in the auditory system, support an immune-mediated process resulting in LASV-associated hearing loss, and demonstrate that vaccination protecting animals from clinical disease can also prevent infection-associated auditory pathology.
Collapse
Affiliation(s)
- Thanhthao Huynh
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Joy M Gary
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Stephen R Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - JoAnn Coleman-McCray
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Jessica R Harmon
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Markus H Kainulainen
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Brigid C Bollweg
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Jana M Ritter
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Wun-Ju Shieh
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Sherif R Zaki
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA.
| |
Collapse
|
20
|
Wang S, Liang T, Luo Q, Li P, Zhang R, Xu M, Su J, Xu T, Wu Q. H9N2 swine influenza virus infection-induced damage is mediated by TRPM2 channels in mouse pulmonary microvascular endothelial cells. Microb Pathog 2020; 148:104408. [PMID: 32707310 DOI: 10.1016/j.micpath.2020.104408] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 01/10/2023]
Abstract
Oxidative stress is implicated in the pathogenesis of influenza virus infection. Increasing evidences show that transient receptor potential melastatin 2 (TRPM2), a Ca2+-permeable non-selective cation channel, plays an important role in the pathomechanism of reactive oxygen species (ROS)-coupled diseases. The present study investigated the role of TRPM2 in pulmonary microvascular endothelial cells (PMVECs) during H9N2 influenza virus infection. We knocked down TRPM2 in PMVECs using TRPM2 shRNA lentiviral particles. Subsequently, we utilized enzyme-linked immunosorbent assay and flow cytometry to compare ROS levels, DNA damage, mitochondrial integrity, apoptosis, and inflammatory factors between control and TRPM2-knockdown PMVECs following H9N2 influenza virus infection. Inhibition of TRPM2 channels reduced H9N2 virus-induced intracellular ROS production, decreased DNA damage, and inhibited H9N2-induced cellular apoptosis. This study shows that the inhibition of TRPM2 channels may protect PMVECs from the damage caused by H9N2 virus infection. Our results highlight the importance of TRPM2 in modulating ROS production, apoptosis, mitochondrial dysfunction, cytokine expression, and DNA damage in H9N2 virus-infected PMVECs, and suggest that TRPM2 may be a potential antiviral target.
Collapse
Affiliation(s)
- Shaohua Wang
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, PR China
| | - Ting Liang
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou, 075131, Hebei, PR China
| | - Qiang Luo
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou, 075131, Hebei, PR China
| | - Peiyao Li
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou, 075131, Hebei, PR China
| | - Ruihua Zhang
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou, 075131, Hebei, PR China
| | - Mingju Xu
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou, 075131, Hebei, PR China
| | - Jingliang Su
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, PR China
| | - Tong Xu
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou, 075131, Hebei, PR China.
| | - Qingmin Wu
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, PR China.
| |
Collapse
|
21
|
Pulmonary endothelium-derived PD-L1 induced by the H9N2 avian influenza virus inhibits the immune response of T cells. Virol J 2020; 17:92. [PMID: 32631356 PMCID: PMC7336647 DOI: 10.1186/s12985-020-01341-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 05/10/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The PD-1/PD-L1 pathway is an inhibitory signaling pathway that maintains the balance between the immune response and immunotolerance, and its overactivation in cancer and viral infections inhibits T cell function. The target cells of various viruses, microvascular endothelial cells (MECs) have been shown to be key regulatory points in immune regulation and virion diffusion in vivo during infection with multiple influenza virus subtypes. Furthermore, avian influenza virus (AIV) infection can induce immunosuppression by causing imbalances in immune responses and immune organ damage. Thus, the aim of this study was to investigate whether the H9N2 virus inhibited the immune function of T cells that migrated across MECs by upregulating PD-L1 expression on MECs. METHODS The susceptibility of rat pulmonary microvascular endothelial cells (RPMECs) to the H9N2 virus was evaluated by a plaque-forming assay and immunofluorescence staining. Then, we quantified the mRNA and protein levels of PD-L1 in RPMECs induced by H9N2 virus infection using quantitative real-time PCR and flow cytometry. The interaction between the activated T cells and RPMECs infected with the H9N2 virus was revealed using a coculture system. The effect of endothelial-derived PD-L1 on T cell function was investigated by using ELISA and flow cytometry with or without a PD-L1-specific antibody. RESULTS Surface staining and the plaque-forming assay showed that the H9N2 virus infected and replicated in RPMECs. Both the PD-L1 mRNA level and PD-L1 protein level were upregulated in RPMECs infected with the H9N2 virus. H9N2 virus-induced PD-L1 expression significantly reduced the secretions of IL-2, IFN-γ and granzyme B and perforin expression in T cells. The above data were significantly increased after treatment with an anti-PD-L1 antibody, confirming the above mentioned findings. In addition, the induction of PD-L1 expression decreased the proliferative capacity of the cocultured T cells but did not affect the apoptosis rate of T cells. CONCLUSIONS Taken together, the results suggest that the H9N2 virus is able to inhibit the T cell immune response by upregulating PD-L1 expression in pulmonary microvascular endothelial cells.
Collapse
|
22
|
Møller-Olsen C, Ross T, Leppard KN, Foisor V, Smith C, Grammatopoulos DK, Sagona AP. Bacteriophage K1F targets Escherichia coli K1 in cerebral endothelial cells and influences the barrier function. Sci Rep 2020; 10:8903. [PMID: 32483257 PMCID: PMC7264188 DOI: 10.1038/s41598-020-65867-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/07/2020] [Indexed: 12/22/2022] Open
Abstract
Bacterial neonatal meningitis results in high mortality and morbidity rates for those affected. Although improvements in diagnosis and treatment have led to a decline in mortality rates, morbidity rates have remained relatively unchanged. Bacterial resistance to antibiotics in this clinical setting further underlines the need for developing other technologies, such as phage therapy. We exploited an in vitro phage therapy model for studying bacterial neonatal meningitis based on Escherichia coli (E. coli) EV36, bacteriophage (phage) K1F and human cerebral microvascular endothelial cells (hCMECs). We show that phage K1F is phagocytosed and degraded by constitutive- and PAMP-dependent LC3-assisted phagocytosis and does not induce expression of inflammatory cytokines TNFα, IL-6, IL-8 or IFNβ. Additionally, we observed that phage K1F temporarily decreases the barrier resistance of hCMEC cultures, a property that influences the barrier permeability, which could facilitate the transition of immune cells across the endothelial vessel in vivo. Collectively, we demonstrate that phage K1F can infect intracellular E. coli EV36 within hCMECs without themselves eliciting an inflammatory or defensive response. This study illustrates the potential of phage therapy targeting infections such as bacterial neonatal meningitis and is an important step for the continued development of phage therapy targeting antibiotic-resistant bacterial infections generally.
Collapse
Affiliation(s)
| | - Toby Ross
- School of Life Sciences, University of Warwick, Gibbet Hill Road, CV4 7AL, Coventry, UK
| | - Keith N Leppard
- School of Life Sciences, University of Warwick, Gibbet Hill Road, CV4 7AL, Coventry, UK
| | - Veronica Foisor
- Department of Chemistry, University of Warwick, Gibbet Hill Road, CV4 7AL, Coventry, UK
| | - Corinne Smith
- School of Life Sciences, University of Warwick, Gibbet Hill Road, CV4 7AL, Coventry, UK
| | - Dimitris K Grammatopoulos
- Warwick Medical School, University of Warwick, Gibbet Hill Road, CV4 7AL, Coventry, UK
- Institute of Precision Diagnostics and Translational Medicine, Dept of Pathology, UHCW NHS Trust, Clifford Bridge Road, CV2 2DX, Coventry, UK
| | - Antonia P Sagona
- School of Life Sciences, University of Warwick, Gibbet Hill Road, CV4 7AL, Coventry, UK.
- Warwick Integrative Synthetic Biology Centre, University of Warwick, Coventry, CV47AL, UK.
| |
Collapse
|
23
|
A new role for host annexin A2 in establishing bacterial adhesion to vascular endothelial cells: lines of evidence from atomic force microscopy and an in vivo study. J Transl Med 2019; 99:1650-1660. [PMID: 31253864 PMCID: PMC6913097 DOI: 10.1038/s41374-019-0284-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/08/2019] [Accepted: 05/20/2019] [Indexed: 01/27/2023] Open
Abstract
Understanding bacterial adhesion is challenging and critical to our understanding of the initial stages of the pathogenesis of endovascular bacterial infections. The vascular endothelial cell (EC) is the main target of Rickettsia, an obligately intracellular bacterium that causes serious systemic disease in humans and animals. But the mechanism(s) underlying bacterial adherence to ECs under shear stress from flowing blood prior to activation are unknown for any bacteria. Although host surface annexin a2 (ANXA2) has been identified to participate in efficient bacterial invasion of epithelial cells, direct evidence is lacking in the field of bacterial infections of ECs. In the present study, we employ a novel, anatomically based, in vivo quantitative bacterial-adhesion-to-vascular-EC system, combined with atomic force microscopy (AFM), to examine the role of endothelial luminal surface ANXA2 during rickettsial adherence to ECs. We also examined whether ANXA2 antibody affected binding of Staphylococcus aureus to ECs. We found that deletion of ANXA2 impeded rickettsial attachment to the ECs in vitro and blocked rickettsial adherence to the blood vessel luminal surface in vivo. The AFM studies established that EC surface ANXA2 acts as an adherence receptor for rickettsiae, and that rickettsial adhesin OmpB is the associated bacterial ligand. Furthermore, pretreatment of ECs with anti-ANXA2 antibody reduced EC surface-associated S. aureus. We conclude that the endothelial surface ANXA2 plays an important role in initiating pathogen-host interactions, ultimately leading to bacterial anchoring on the vascular luminal surface.
Collapse
|
24
|
Khaiboullina SF, Ribeiro FM, Uppal T, Martynova EV, Rizvanov AA, Verma SC. Zika Virus Transmission Through Blood Tissue Barriers. Front Microbiol 2019; 10:1465. [PMID: 31333605 PMCID: PMC6621930 DOI: 10.3389/fmicb.2019.01465] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 06/11/2019] [Indexed: 01/12/2023] Open
Abstract
The recent Zika virus (ZIKV) epidemic in the Americas and the Caribbean revealed a new deadly strain of the mosquito-borne virus, which has never been associated with previous outbreaks in Asia. For the first time, widespread ZIKV infection was shown to cause microcephaly and death of newborns, which was most likely due to the mutation acquired during the large outbreak recorded in French Polynesia in 2013–2014. Productive ZIKV replication and persistence has been demonstrated in placenta and fetal brains. Possible association between ZIKV and microcephaly and fetal death has been confirmed using immunocompetent mouse models in vitro and in vivo. Having crossed the placenta, ZIKV directly targets neural progenitor cells (NPCs) in developing human fetus and triggers apoptosis. The embryonic endothelial cells are exceptionally susceptible to ZIKV infection, which causes cell death and tissue necrosis. On the contrary, ZIKV infection does not affect the adult brain microvascular cell morphology and blood–brain barrier function. ZIKV is transmitted primarily by Aedes mosquito bite and is introduced into the placenta/blood through replication at the site of the entry. Also, virus can be transmitted through unprotected sex. Although, multiple possible routes of virus infection have been identified, the exact mechanism(s) utilized by ZIKV to cross the placenta still remain largely unknown. In this review, the current understanding of ZIKV infection and transmission through the placental and brain barriers is summarized.
Collapse
Affiliation(s)
- Svetlana F Khaiboullina
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, Reno, NV, United States.,Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Fabiola M Ribeiro
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Timsy Uppal
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, Reno, NV, United States
| | - Ekaterina V Martynova
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Albert A Rizvanov
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Subhash C Verma
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, Reno, NV, United States
| |
Collapse
|
25
|
Sahni A, Narra HP, Patel J, Sahni SK. MicroRNA-Regulated Rickettsial Invasion into Host Endothelium via Fibroblast Growth Factor 2 and Its Receptor FGFR1. Cells 2018; 7:cells7120240. [PMID: 30513762 PMCID: PMC6315532 DOI: 10.3390/cells7120240] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/20/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023] Open
Abstract
Microvascular endothelial cells (ECs) represent the primary target cells during human rickettsioses and respond to infection via the activation of immediate–early signaling cascades and the resultant induction of gene expression. As small noncoding RNAs dispersed throughout the genome, microRNAs (miRNAs) regulate gene expression post-transcriptionally to govern a wide range of biological processes. Based on our recent findings demonstrating the involvement of fibroblast growth factor receptor 1 (FGFR1) in facilitating rickettsial invasion into host cells and published reports suggesting miR-424 and miR-503 as regulators of FGF2/FGFR1, we measured the expression of miR-424 and miR-503 during R. conorii infection of human dermal microvascular endothelial cells (HMECs). Our results revealed a significant decrease in miR-424 and miR-503 expression in apparent correlation with increased expression of FGF2 and FGFR1. Considering the established phenomenon of endothelial heterogeneity and pulmonary and cerebral edema as the prominent pathogenic features of rickettsial infections, and significant pathogen burden in the lungs and brain in established mouse models of disease, we next quantified miR-424 and miR-503 expression in pulmonary and cerebral microvascular ECs. Again, R. conorii infection dramatically downregulated both miRNAs in these tissue-specific ECs as early as 30 min post-infection in correlation with higher FGF2/FGFR1 expression. Changes in the expression of both miRNAs and FGF2/FGFR1 were next confirmed in a mouse model of R. conorii infection. Furthermore, miR-424 overexpression via transfection of a mimic into host ECs reduced the expression of FGF2/FGFR1 and gave a corresponding decrease in R. conorii invasion, while an inhibitor of miR-424 had the expected opposite effect. Together, these findings implicate the rickettsial manipulation of host gene expression via regulatory miRNAs to ensure efficient cellular entry as the critical requirement to establish intracellular infection.
Collapse
Affiliation(s)
- Abha Sahni
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609, USA.
| | - Hema P Narra
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609, USA.
| | - Jignesh Patel
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609, USA.
| | - Sanjeev K Sahni
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609, USA.
| |
Collapse
|
26
|
Sahni A, Fang R, Sahni SK, Walker DH. Pathogenesis of Rickettsial Diseases: Pathogenic and Immune Mechanisms of an Endotheliotropic Infection. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:127-152. [PMID: 30148688 DOI: 10.1146/annurev-pathmechdis-012418-012800] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obligately intracytosolic rickettsiae that cycle between arthropod and vertebrate hosts cause human diseases with a spectrum of severity, primarily by targeting microvascular endothelial cells, resulting in endothelial dysfunction. Endothelial cells and mononuclear phagocytes have important roles in the intracellular killing of rickettsiae upon activation by the effector molecules of innate and adaptive immunity. In overwhelming infection, immunosuppressive effects contribute to the severity of illness. Rickettsia-host cell interactions involve host cell receptors for rickettsial ligands that mediate cell adhesion and, in some instances, trigger induced phagocytosis. Rickettsiae interact with host cell actin to effect both cellular entry and intracellular actin-based mobility. The interaction of rickettsiae with the host cell also involves rickettsial evasion of host defense mechanisms and exploitation of the intracellular environment. Signal transduction events exemplify these effects. An intriguing frontier is the array of rickettsial noncoding RNA molecules and their potential effects on the pathogenesis and transmission of rickettsial diseases.
Collapse
Affiliation(s)
- Abha Sahni
- The University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0609, USA; , , ,
| | - Rong Fang
- The University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0609, USA; , , ,
| | - Sanjeev K Sahni
- The University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0609, USA; , , ,
| | - David H Walker
- The University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0609, USA; , , ,
| |
Collapse
|
27
|
Li L, Cheng Y, Emrich S, Schorey J. Activation of endothelial cells by extracellular vesicles derived from Mycobacterium tuberculosis infected macrophages or mice. PLoS One 2018; 13:e0198337. [PMID: 29851993 PMCID: PMC5979010 DOI: 10.1371/journal.pone.0198337] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/17/2018] [Indexed: 12/12/2022] Open
Abstract
Endothelial cells play an essential role in regulating an immune response through promoting leukocyte adhesion and cell migration and production of cytokines such as TNFα. Regulation of endothelial cell immune function is tightly regulated and recent studies suggest that extracellular vesicles (EVs) are prominently involved in this process. However, the importance of EVs in regulating endothelial activation in the context of a bacterial infection is poorly understood. To begin addressing this knowledge gap we characterized the endothelial cell response to EVs released from Mycobacterium tuberculosis (Mtb) infected macrophages. Our result showed increased macrophage migration through the monolayer when endothelial cells were pretreated with EVs isolated from Mtb-infected macrophages. Transcriptome analysis showed a significant upregulation of genes involved in cell adhesion and the inflammatory process in endothelial cells treated with EVs. These results were validated by quantitative PCR and flow cytometry. Pathway analysis of these differentially expressed genes indicated that several immune response-related pathways were up-regulated. Endothelial cells were also treated with EVs isolated from the serum of Mtb-infected mice. Interestingly, EVs isolated 14 days but not 7 or 21 days post-infection showed a similar ability to induce endothelial cell activation suggesting a change in EV function during the course of an Mtb infection. Immunofluorescence microscopy result indicated that NF-κB and the Type 1 interferon pathways were involved in endothelial activation by EVs. In summary, our data suggest that EVs can activate endothelial cells and thus may play an important role in modulating host immune responses during an Mtb infection.
Collapse
Affiliation(s)
- Li Li
- Department of Biological Sciences, Eck Institute for Global Health, Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN, United States of America
| | - Yong Cheng
- Department of Biological Sciences, Eck Institute for Global Health, Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN, United States of America
| | - Scott Emrich
- Department of Electrical Engineering and Computer Science, University of Tennessee, Knoxville, TN, United States of America
| | - Jeffrey Schorey
- Department of Biological Sciences, Eck Institute for Global Health, Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN, United States of America
- * E-mail:
| |
Collapse
|
28
|
Gonzalez-Quintial R, Baccala R. Murine Models for Viral Hemorrhagic Fever. Methods Mol Biol 2018; 1604:257-267. [PMID: 28986841 DOI: 10.1007/978-1-4939-6981-4_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Hemorrhagic fever (HF) viruses, such as Lassa, Ebola, and dengue viruses, represent major human health risks due to their highly contagious nature, the severity of the clinical manifestations induced, the lack of vaccines, and the very limited therapeutic options currently available. Appropriate animal models are obviously critical to study disease pathogenesis and develop efficient therapies. We recently reported that the clone 13 (Cl13) variant of the lymphocytic choriomeningitis virus (LCMV-Cl13), a prototype arenavirus closely related to Lassa virus, causes in some mouse strains endothelial damage, vascular leakage, platelet loss, and death, mimicking pathological aspects typically observed in Lassa and other HF syndromes. This model has the advantage that the mice used are fully immunocompetent, allowing studies on the contribution of the immune response to disease progression. Moreover, LCMV is very well characterized and exhibits limited pathogenicity in humans, allowing handling in convenient BSL-2 facilities. In this chapter we outline protocols for the induction and analysis of arenavirus-mediated pathogenesis in the NZB/LCMV model, including mouse infection, virus titer determination, platelet counting, phenotypic analysis of virus-specific T cells, and assessment of vascular permeability.
Collapse
Affiliation(s)
- Rosana Gonzalez-Quintial
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Roberto Baccala
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
29
|
Sahni A, Patel J, Narra HP, Schroeder CLC, Walker DH, Sahni SK. Fibroblast growth factor receptor-1 mediates internalization of pathogenic spotted fever rickettsiae into host endothelium. PLoS One 2017; 12:e0183181. [PMID: 28806774 PMCID: PMC5555671 DOI: 10.1371/journal.pone.0183181] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/31/2017] [Indexed: 11/25/2022] Open
Abstract
Rickettsial infections continue to cause serious morbidity and mortality in severe human cases around the world. Host cell adhesion and invasion is an essential requisite for intracellular growth, replication, and subsequent dissemination of pathogenic rickettsiae. Heparan sulfate proteoglycans [HSPGs] facilitate the interactions between fibroblast growth factor(s) and their tyrosine kinase receptors resulting in receptor dimerization/activation and have been implicated in bacterial adhesion to target host cells. In the present study, we have investigated the contributions of fibroblast growth factor receptors [FGFRs] in rickettsial entry into the host cells. Inhibition of HSPGs by heparinase and FGFRs by AZD4547 (a selective small-molecule inhibitor) results in significant reduction in rickettsial internalization into cultured human microvascular endothelial cells (ECs), which represent the primary targets of pathogenic rickettsiae during human infections. Administration of AZD4547 during R. conorii infection in a murine model of endothelial-target spotted fever rickettsiosis also diminishes pulmonary rickettsial burden in comparison to mock-treated controls. Silencing of FGFR1 expression using a small interfering RNA also leads to similar inhibition of R. rickettsii invasion into ECs. Consistent with these findings, R. rickettsii infection of ECs also results in phosphorylation of tyrosine 653/654, suggesting activation of FGFR1. Using isobaric tag for relative and absolute quantitation [iTRAQ]-based proteomics approach, we further demonstrate association of β-peptide of rickettsial outer membrane protein OmpA with FGFR1. Mechanistically, FGFR1 binds to caveolin-1 and mediates bacterial entry via caveolin-1 dependent endocytosis. Together, these results identify host cell FGFR1 and rickettsial OmpA as another novel receptor-ligand pair contributing to the internalization of pathogenic rickettsiae into host endothelial cells and the potential application of FGFR-inhibitor drugs as adjunct therapeutics against spotted fever rickettsioses.
Collapse
Affiliation(s)
- Abha Sahni
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail: (AS); (SKS)
| | - Jignesh Patel
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Hema P. Narra
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Casey L. C. Schroeder
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - David H. Walker
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Sanjeev K. Sahni
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail: (AS); (SKS)
| |
Collapse
|
30
|
Zhang LC, Jin X, Huang Z, Yan ZN, Li PB, Duan RF, Feng H, Jiang JH, Peng H, Liu W. Protective effects of choline against hypoxia-induced injuries of vessels and endothelial cells. Exp Ther Med 2017; 13:2316-2324. [PMID: 28565844 PMCID: PMC5443310 DOI: 10.3892/etm.2017.4276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 01/06/2017] [Indexed: 01/08/2023] Open
Abstract
The current study aimed to lay a theoretical foundation for further development of choline as an anti-hypoxia damage drug. Wild-type, 3- to 5-month-old male Sprague-Dawley rats, weighing 180-220 g, were used in this study. The rats were randomly divided into a normoxic control group (n=16) and a chronic intermittent hypoxia (CIH) group (n=16). The effects of CIH on acetylcholine (ACh)-mediated endothelium-dependent vasodilatation in the rat cerebral basilar arterioles and mesenteric arterioles, as well as the protective effects of choline on the arterioles damaged by hypoxia were observed. Moreover, the effects of choline on endothelial cell proliferation during hypoxia were observed, and choline's functional mechanism further explored. The ACh-mediated vasodilatation of rat cerebral basilar and mesenteric arterioles significantly reduced during hypoxia (P<0.01). Choline significantly increased dilation in the rat cerebral basilar (P<0.01) and mesenteric arterioles (P<0.05) damaged by CIH compared with those in the control group. In addition, under hypoxic conditions, choline significantly promoted the proliferation of rat aortic endothelial cells (P<0.05) and significantly reduced lactate dehydrogenase activity in the cell culture supernatant in vitro (P<0.05). Furthermore, the effect of choline could be related to its ability to significantly increase the secretion of vascular endothelial growth factor (P<0.01) and activation of α7 non-neuronal nicotinic acetylcholine receptors under hypoxia (P<0.01). This study demonstrated that choline could have protective effects against hypoxic injuries.
Collapse
Affiliation(s)
- Lian-Cheng Zhang
- Department of Environment and Pharmacy, Tianjin Institute of Health and Environmental Medicine, Tianjin 300050, P.R. China
| | - Xin Jin
- Department of Environment and Pharmacy, Tianjin Institute of Health and Environmental Medicine, Tianjin 300050, P.R. China
| | - Zhao Huang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, P.R. China
| | - Zhen-Nan Yan
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, P.R. China
| | - Pei-Bing Li
- Department of Environment and Pharmacy, Tianjin Institute of Health and Environmental Medicine, Tianjin 300050, P.R. China
| | - Rui-Feng Duan
- Department of Environment and Pharmacy, Tianjin Institute of Health and Environmental Medicine, Tianjin 300050, P.R. China
| | - Hong Feng
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, P.R. China
| | - Jian-Hua Jiang
- Department of Environment and Pharmacy, Tianjin Institute of Health and Environmental Medicine, Tianjin 300050, P.R. China
| | - Hui Peng
- Department of Environment and Pharmacy, Tianjin Institute of Health and Environmental Medicine, Tianjin 300050, P.R. China
| | - Wei Liu
- Department of Environment and Pharmacy, Tianjin Institute of Health and Environmental Medicine, Tianjin 300050, P.R. China
| |
Collapse
|
31
|
Siragam V, Qiu X. How can Ebola virus infection lead to endothelial dysfunction and coagulopathy? Future Virol 2017. [DOI: 10.2217/fvl-2016-0143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Vinayakumar Siragam
- Special Pathogens Program, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xiangguo Qiu
- Special Pathogens Program, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
32
|
Ermonval M, Baychelier F, Tordo N. What Do We Know about How Hantaviruses Interact with Their Different Hosts? Viruses 2016; 8:v8080223. [PMID: 27529272 PMCID: PMC4997585 DOI: 10.3390/v8080223] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/27/2016] [Accepted: 08/05/2016] [Indexed: 11/26/2022] Open
Abstract
Hantaviruses, like other members of the Bunyaviridae family, are emerging viruses that are able to cause hemorrhagic fevers. Occasional transmission to humans is due to inhalation of contaminated aerosolized excreta from infected rodents. Hantaviruses are asymptomatic in their rodent or insectivore natural hosts with which they have co-evolved for millions of years. In contrast, hantaviruses cause different pathologies in humans with varying mortality rates, depending on the hantavirus species and its geographic origin. Cases of hemorrhagic fever with renal syndrome (HFRS) have been reported in Europe and Asia, while hantavirus cardiopulmonary syndromes (HCPS) are observed in the Americas. In some cases, diseases caused by Old World hantaviruses exhibit HCPS-like symptoms. Although the etiologic agents of HFRS were identified in the early 1980s, the way hantaviruses interact with their different hosts still remains elusive. What are the entry receptors? How do hantaviruses propagate in the organism and how do they cope with the immune system? This review summarizes recent data documenting interactions established by pathogenic and nonpathogenic hantaviruses with their natural or human hosts that could highlight their different outcomes.
Collapse
Affiliation(s)
- Myriam Ermonval
- Unité des Stratégies Antivirales, Département de Virologie, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France.
| | - Florence Baychelier
- Unité des Stratégies Antivirales, Département de Virologie, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France.
| | - Noël Tordo
- Unité des Stratégies Antivirales, Département de Virologie, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France.
| |
Collapse
|
33
|
Rengarajan M, Hayer A, Theriot JA. Endothelial Cells Use a Formin-Dependent Phagocytosis-Like Process to Internalize the Bacterium Listeria monocytogenes. PLoS Pathog 2016; 12:e1005603. [PMID: 27152864 PMCID: PMC4859537 DOI: 10.1371/journal.ppat.1005603] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 04/06/2016] [Indexed: 01/11/2023] Open
Abstract
Vascular endothelial cells act as gatekeepers that protect underlying tissue from blood-borne toxins and pathogens. Nevertheless, endothelial cells are able to internalize large fibrin clots and apoptotic debris from the bloodstream, although the precise mechanism of such phagocytosis-like uptake is unknown. We show that cultured primary human endothelial cells (HUVEC) internalize both pathogenic and non-pathogenic Listeria bacteria comparably, in a phagocytosis-like process. In contrast with previously studied host cell types, including intestinal epithelial cells and hepatocytes, we find that endothelial internalization of Listeria is independent of all known pathogenic bacterial surface proteins. Consequently, we exploited the internalization and intracellular replication of L. monocytogenes to identify distinct host cell factors that regulate phagocytosis-like uptake in HUVEC. Using siRNA screening and subsequent genetic and pharmacologic perturbations, we determined that endothelial infectivity was modulated by cytoskeletal proteins that normally modulate global architectural changes, including phosphoinositide-3-kinase, focal adhesions, and the small GTPase Rho. We found that Rho kinase (ROCK) is acutely necessary for adhesion of Listeria to endothelial cells, whereas the actin-nucleating formins FHOD1 and FMNL3 specifically regulate internalization of bacteria as well as inert beads, demonstrating that formins regulate endothelial phagocytosis-like uptake independent of the specific cargo. Finally, we found that neither ROCK nor formins were required for macrophage phagocytosis of L. monocytogenes, suggesting that endothelial cells have distinct requirements for bacterial internalization from those of classical professional phagocytes. Our results identify a novel pathway for L. monocytogenes uptake by human host cells, indicating that this wily pathogen can invade a variety of tissues by using a surprisingly diverse suite of distinct uptake mechanisms that operate differentially in different host cell types.
Collapse
Affiliation(s)
- Michelle Rengarajan
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
| | - Arnold Hayer
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Julie A. Theriot
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
Toscana virus infects dendritic and endothelial cells opening the way for the central nervous system. J Neurovirol 2015; 22:307-15. [PMID: 26510872 DOI: 10.1007/s13365-015-0395-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/06/2015] [Accepted: 10/12/2015] [Indexed: 02/06/2023]
Abstract
Toscana virus (TOSV) is a Phlebovirus responsible for human neurological infections in endemic Mediterranean areas. The main viral target is the central nervous system, with viremia as a way of dissemination throughout the host. This study was aimed at understanding the spread of TOSV in the host by identifying the cell population infected by the virus and the vehicle to the organs. In vivo studies provided evidence that endothelial cells are infected by TOSV, indicating their potential role in the diffusion of the virus following viremic spread. These results were further confirmed in vitro. Human peripheral mononuclear blood cells were infected with TOSV; only monocyte-derived dendritic cells were identified as susceptible to TOSV infection. Productive viral replication was then observed in human monocyte-derived dendritic cells (moDCs) and in human endothelial cells by recovery of the virus from a cell supernatant. Interleukin-6 was produced by both cell types upon TOSV infection, mostly by endothelial cells, while moDCs particularly expressed TNF-α, which is known to induce a long-lasting decrease in endothelial cell barrier function. These cells could therefore be implicated in the spread of the virus in the host and in the infection of tissues that are affected by the disease, such as the central nervous system. The identification of in vitro and in vivo TOSV cell targets is an important tool for understanding the pathogenesis of the infection, providing new insight into virus-cell interaction for improved knowledge and control of this viral disease.
Collapse
|
35
|
Wang W, Mu X, Zhao L, Wang J, Chu Y, Feng X, Feng B, Wang X, Zhang J, Qiao J. Transcriptional response of human umbilical vein endothelial cell to H9N2 influenza virus infection. Virology 2015; 482:117-27. [PMID: 25863179 DOI: 10.1016/j.virol.2015.03.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 03/13/2015] [Accepted: 03/18/2015] [Indexed: 11/25/2022]
Abstract
Endothelial cells are believed to play an important role in response to virus infection. Here, we used a microarray technology to study the gene expression profile in human umbilical vein endothelial cells at 24h postinfection with H9N2 viruses or inactivated H9N2 viral particles. The results showed that H9N2 virus infection induced an abundance of differential expressed genes, exhibiting a transcriptional signature of viral infection. High levels of chemokine gene expressions were detected following treatment. Surprisingly, the most significantly up-regulated genes were mainly interferon-stimulated genes (ISGs), although there was no change in interferon gene expression and interferon protein level. We also found that viral particles were more potent than viruses in inducing ISGs expression. These results suggest that induction of expression of ISGs is mainly dependent on the interaction between viral particles and endothelial cells. Our data offer further insight into the interaction between endothelial cells and H9N2 influenza viruses.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pathophysiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People׳s Republic of China
| | - Xiang Mu
- Department of Animal Science and Technology, Beijing Agricultural College, Beijing 102206, People׳s Republic of China
| | - Lihong Zhao
- Department of Pathophysiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People׳s Republic of China
| | - Jianfang Wang
- Department of Animal Science and Technology, Beijing Agricultural College, Beijing 102206, People׳s Republic of China
| | - Yaocheng Chu
- Department of Veterinary Medicine, College of Animal Science, Hebei North University, Zhangjiakou 075131, Hebei Province, People׳s Republic of China
| | - Xuejian Feng
- Department of Pathophysiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People׳s Republic of China
| | - Bo Feng
- Department of Pathophysiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People׳s Republic of China
| | - Xiaohong Wang
- Department of Pathophysiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People׳s Republic of China
| | - Jianjun Zhang
- Department of Animal Science and Technology, Beijing Agricultural College, Beijing 102206, People׳s Republic of China
| | - Jian Qiao
- Department of Pathophysiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People׳s Republic of China.
| |
Collapse
|
36
|
Hepojoki J, Vaheri A, Strandin T. The fundamental role of endothelial cells in hantavirus pathogenesis. Front Microbiol 2014; 5:727. [PMID: 25566236 PMCID: PMC4273638 DOI: 10.3389/fmicb.2014.00727] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 12/03/2014] [Indexed: 01/17/2023] Open
Abstract
Hantavirus, a genus of rodent- and insectivore-borne viruses in the family Bunyaviridae, is a group of emerging zoonotic pathogens. Hantaviruses cause hemorrhagic fever with renal syndrome and hantavirus cardiopulmonary syndrome in man, often with severe consequences. Vascular leakage is evident in severe hantavirus infections, and increased permeability contributes to the pathogenesis. This review summarizes the current knowledge on hantavirus interactions with hematopoietic and endothelial cells, and their effects on the increased vascular permeability.
Collapse
Affiliation(s)
- Jussi Hepojoki
- Department of Virology, Haartman Institute, University of Helsinki Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Haartman Institute, University of Helsinki Helsinki, Finland
| | - Tomas Strandin
- Department of Virology, Haartman Institute, University of Helsinki Helsinki, Finland
| |
Collapse
|
37
|
Cellular visualization of macrophage pyroptosis and interleukin-1β release in a viral hemorrhagic infection in zebrafish larvae. J Virol 2014; 88:12026-40. [PMID: 25100833 DOI: 10.1128/jvi.02056-14] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Hemorrhagic viral diseases are distributed worldwide with important pathogens, such as dengue virus or hantaviruses. The lack of adequate in vivo infection models has limited the research on viral pathogenesis and the current understanding of the underlying infection mechanisms. Although hemorrhages have been associated with the infection of endothelial cells, other cellular types could be the main targets for hemorrhagic viruses. Our objective was to take advantage of the use of zebrafish larvae in the study of viral hemorrhagic diseases, focusing on the interaction between viruses and host cells. Cellular processes, such as transendothelial migration of leukocytes, virus-induced pyroptosis of macrophages. and interleukin-1β (Il-1β) release, could be observed in individual cells, providing a deeper knowledge of the immune mechanisms implicated in the disease. Furthermore, the application of these techniques to other pathogens will improve the current knowledge of host-pathogen interactions and increase the potential for the discovery of new therapeutic targets. Importance: Pathogenic mechanisms of hemorrhagic viruses are diverse, and most of the research regarding interactions between viruses and host cells has been performed in cell lines that might not be major targets during natural infections. Thus, viral pathogenesis research has been limited because of the lack of adequate in vivo infection models. The understanding of the relative pathogenic roles of the viral agent and the host response to the infection is crucial. This will be facilitated by the establishment of in vivo infection models using organisms such as zebrafish, which allows the study of the diseases in the context of a complete individual. The use of this animal model with other pathogens could improve the current knowledge on host-pathogen interactions and increase the potential for the discovery of new therapeutic targets against diverse viral diseases.
Collapse
|
38
|
Dalrymple NA, Mackow ER. Virus interactions with endothelial cell receptors: implications for viral pathogenesis. Curr Opin Virol 2014; 7:134-40. [PMID: 25063986 PMCID: PMC4206553 DOI: 10.1016/j.coviro.2014.06.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 06/10/2014] [Accepted: 06/27/2014] [Indexed: 01/14/2023]
Abstract
The endothelial lining of the vasculature performs a vital role in maintaining fluid barrier functions despite balancing nutrient and fluid content of tissues, repairing localized damage, coordinating responses of a plethora of factors, immune cells and platelets through a multitude of endothelial cell surface receptors. Viruses that nonlytically cause lethal hemorrhagic or edematous diseases engage receptors on vascular and lymphatic endothelial cells, altering normal cellular responses that control capillary leakage and fluid clearance functions with lethal consequences. Recent studies indicate that receptors directing dengue virus and hantavirus infection of the endothelium contribute to the dysregulation of normal endothelial cell signaling responses that control capillary permeability and immune responses that contribute to pathogenesis. Here we present recent studies of virally altered endothelial functions that provide new insight into targeting barrier functions of the endothelium as a potential therapeutic approach.
Collapse
|
39
|
Beaufort N, Corvazier E, Mlanaoindrou S, de Bentzmann S, Pidard D. Disruption of the endothelial barrier by proteases from the bacterial pathogen Pseudomonas aeruginosa: implication of matrilysis and receptor cleavage. PLoS One 2013; 8:e75708. [PMID: 24069438 PMCID: PMC3777978 DOI: 10.1371/journal.pone.0075708] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 08/19/2013] [Indexed: 12/19/2022] Open
Abstract
Within the vasculature, uncontrolled pericellular proteolysis can lead to disruption of cell-to-cell and cell-to-matrix interactions and subsequent detachment-induced cell apoptosis, or anoikis, contributing to inflammatory vascular diseases, with the endothelium as the major target. Most studies so far have focused on endogenous proteinases. However, during bloodstream infections, bacterial proteinases may also trigger endothelial anoikis. We thus investigated the potential apoptotic activity of the proteinases secreted by the haematotropic opportunistic pathogen, Pseudomonas aeruginosa, and particularly its predominant metalloproteinase, LasB. For this, we used the secretome of the LasB-expressing pseudomonal strain, PAO1, and compared it with that from the isogenic, LasB-deficient strain (PAO1∆lasB), as well as with purified LasB. Secretomes were tested for apoptotic activity on cultured human endothelial cells derived from the umbilical vein or from the cerebral microvasculature. We found that the PAO1 secretome readily induced endothelial cell anoikis, as did secretomes of LasB-positive clinical pseudomonal isolates, while the PAO1∆lasB secretome had only a limited impact on endothelial adherence and viability. Notably, purified LasB reproduced most of the effects of the LasB-containing secretomes, and these were drastically reduced in the presence of the LasB-selective inhibitor, phosphoramidon. A precocious and extensive LasB-dependent degradation of several proteins associated with the endothelial extracellular matrix, fibronectin and von Willebrand factor, was observed by immunofluorescence and/or immunoblotting analysis of cell cultures. Moreover, the PAO1 secretome, but not that from PAO1∆lasB, specifically induced rapid endoproteolysis of two major interendothelial junction components, VE-cadherin and occludin, as well as of the anti-anoikis, integrin-associated urokinase receptor, uPAR. Taken as a prototype for exogenous haemorrhagic proteinases, pseudomonal LasB thus appears to induce endothelial anoikis not only via matrilysis, as observed for many pro-apoptotic proteinases, but also via cleavage of some essential cell-to-cell and cell-to-matrix adhesion receptors implicated in the maintenance of the endothelial barrier.
Collapse
Affiliation(s)
- Nathalie Beaufort
- Inserm, U698, Paris, France
- Université Denis Diderot, UMR-S698, Paris, France
| | - Elisabeth Corvazier
- Inserm, U698, Paris, France
- Université Denis Diderot, UMR-S698, Paris, France
| | - Saouda Mlanaoindrou
- Inserm, U698, Paris, France
- Université Denis Diderot, UMR-S698, Paris, France
| | - Sophie de Bentzmann
- CNRS, UMR 7255-LISM, Marseille, France
- Aix-Marseille University, Marseille, France
| | - Dominique Pidard
- Inserm, U698, Paris, France
- Université Denis Diderot, UMR-S698, Paris, France
- * E-mail:
| |
Collapse
|
40
|
Mackow ER, Gorbunova EE, Dalrymple NA, Gavrilovskaya IN. Role of vascular and lymphatic endothelial cells in hantavirus pulmonary syndrome suggests targeted therapeutic approaches. Lymphat Res Biol 2013; 11:128-35. [PMID: 24024573 DOI: 10.1089/lrb.2013.0006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Hantaviruses in the Americas cause a highly lethal acute pulmonary edema termed hantavirus pulmonary syndrome (HPS). Hantaviruses nonlytically infect microvascular and lymphatic endothelial cells and cause dramatic changes in barrier functions without disrupting the endothelium. Hantaviruses cause changes in the function of infected endothelial cells that normally regulate fluid barrier functions. The endothelium of arteries, veins, and lymphatic vessels are unique and central to the function of vast pulmonary capillary beds that regulate pulmonary fluid accumulation. RESULTS We have found that HPS-causing hantaviruses alter vascular barrier functions of microvascular and lymphatic endothelial cells by altering receptor and signaling pathway responses that serve to permit fluid tissue influx and clear tissue edema. Infection of the endothelium provides several mechanisms for hantaviruses to cause acute pulmonary edema, as well as potential therapeutic targets for reducing the severity of HPS disease. CONCLUSIONS Here we discuss interactions of HPS-causing hantaviruses with the endothelium, roles for unique lymphatic endothelial responses in HPS, and therapeutic targeting of the endothelium as a means of reducing the severity of HPS disease.
Collapse
Affiliation(s)
- Erich R Mackow
- Department of Molecular Genetics and Microbiology, Stony Brook University , Stony Brook, New York
| | | | | | | |
Collapse
|
41
|
Gong B, Lee YS, Lee I, Shelite TR, Kunkeaw N, Xu G, Lee K, Jeon SH, Johnson BH, Chang Q, Ha T, Mendell NL, Cheng X, Bouyer DH, Boor PJ, Ksiazek TG, Walker DH. Compartmentalized, functional role of angiogenin during spotted fever group rickettsia-induced endothelial barrier dysfunction: evidence of possible mediation by host tRNA-derived small noncoding RNAs. BMC Infect Dis 2013; 13:285. [PMID: 23800282 PMCID: PMC3699377 DOI: 10.1186/1471-2334-13-285] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/12/2013] [Indexed: 01/19/2023] Open
Abstract
Background Microvascular endothelial barrier dysfunction is the central enigma in spotted fever group (SFG) rickettsioses. Angiogenin (ANG) is one of the earliest identified angiogenic factors, of which some are relevant to the phosphorylation of VE-cadherins that serve as endothelial adherens proteins. Although exogenous ANG is known to translocate into the nucleus of growing endothelial cells (ECs) where it plays a functional role, nuclear ANG is not detected in quiescent ECs. Besides its nuclear role, ANG is thought to play a cytoplasmic role, owing to its RNase activity that cleaves tRNA to produce small RNAs. Recently, such tRNA-derived RNA fragments (tRFs) have been shown to be induced under stress conditions. All these observations raise an intriguing hypothesis about a novel cytoplasmic role of ANG, which is induced upon infection with Rickettsia and generates tRFs that may play roles in SFG rickettsioses. Methods C3H/HeN mice were infected intravenously with a sublethal dose of R. conorii. At days 1, 3, and 5 post infection (p.i.), liver, lung and brain were collected for immunofluorescence (IF) studies of R. conorii and angiogenin (ANG). Human umbilical vein endothelial cells (HUVECs) were infected with R. conorii for 24, 48, and 72 hrs before incubation with 1μg/ml recombinant human ANG (rANG) in normal medium for 2 hrs. HUVEC samples were subjected to IF, exogenous ANG translocation, endothelial permeability, and immunoprecipitation phosphorylation assays. To identify small non-coding RNAs (sncRNAs) upon rickettsial infection, RNAs from pulverized mouse lung tissues and HUVECs were subjected to library preparation and deep sequencing analysis using an Illumina 2000 instrument. Identified sncRNAs were confirmed by Northern hybridization, and their target mRNAs were predicted in silico using BLAST and RNA hybrid programs. Results In the present study, we have demonstrated endothelial up-regulation of ANG, co-localized with SFG rickettsial infection in vivo. We also have provided direct evidence that rickettsial infection sensitizes human ECs to the translocation of exogenous ANG in a compartmentalized pattern at different times post-infection. Typically, exogenous ANG translocates into the nucleus at 24 hrs and to the cytoplasm at 72 hrs post-infection. The ANG cytoplasmic translocation enhances phosphorylation and destabilization of VE-cadherin and attenuates endothelial barrier function. Of note, deep sequencing analysis detected tRFs, mostly derived from the 5'-halves of host tRNAs, that are induced by ANG. Northern hybridization validates the two most abundantly cloned tRFs derived from tRNA-ValGTG and tRNA-GlyGCC, in both mouse tissues and human cells. Bioinformatics analysis predicted that these tRFs may interact with transcripts associated with the endothelial barrier, the host cell inflammatory response, and autophagy. Conclusions Our data provide new insight into the role of compartmentalized ANG during SFG rickettsioses, and highlight its possible mediation through tRFs.
Collapse
Affiliation(s)
- Bin Gong
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Aamelfot M, Weli SC, Dale OB, Koppang EO, Falk K. Characterisation of a monoclonal antibody detecting Atlantic salmon endothelial and red blood cells, and its association with the infectious salmon anaemia virus cell receptor. J Anat 2013; 222:547-57. [PMID: 23439106 PMCID: PMC3633344 DOI: 10.1111/joa.12033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2013] [Indexed: 01/29/2023] Open
Abstract
Endothelial cells (ECs) line the luminal surfaces of the cardiovascular system and play an important role in cardiovascular functions such as regulation of haemostasis and vasomotor tone. A number of fish and mammalian viruses target these cells in the course of their infection. Infectious salmon anaemia virus (ISAV) attacks ECs and red blood cells (RBCs) of farmed Atlantic salmon (Salmo salar L.), producing the severe disease of infectious salmon anaemia (ISA). The investigation of ISA has up to now been hampered by the lack of a functional marker for ECs in Atlantic salmon in situ. In this study, we report the characterisation and use of a novel monoclonal antibody (MAb) detecting Atlantic salmon ECs (e.g. vessel endothelium, endocardial cells and scavenger ECs) and RBCs. The antibody can be used with immunohistochemistry, IFAT and on Western blots. It appears that the epitope recognised by the antibody is associated with the ISAV cellular receptor. Besides being a tool to identify ECs in situ, it could be useful in further studies of the pathogenicity of ISA. Finally, the detection of an epitope shared by ECs and RBCs agrees with recent findings that these cells share a common origin, thus the MAb can potentially be used to study the ontogeny of these cells in Atlantic salmon.
Collapse
Affiliation(s)
| | | | - Ole B Dale
- Norwegian Veterinary InstituteOslo, Norway
| | | | - Knut Falk
- Norwegian Veterinary InstituteOslo, Norway
| |
Collapse
|
43
|
Valbuena G, Walker DH. Approaches to vaccines against Orientia tsutsugamushi. Front Cell Infect Microbiol 2013; 2:170. [PMID: 23316486 PMCID: PMC3539663 DOI: 10.3389/fcimb.2012.00170] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 12/14/2012] [Indexed: 12/02/2022] Open
Abstract
Scrub typhus is a severe mite-borne infection caused by Orientia tsutsugamushi, an obligately intracellular bacterium closely related to Rickettsia. The disease explains a substantial proportion of acute undifferentiated febrile cases that require hospitalization in rural areas of Asia, the North of Australia, and many islands of the Pacific Ocean. Delayed antibiotic treatment is common due to the lack of effective commercially available diagnostic tests and the lack of specificity of the early clinical presentation. The systemic infection of endothelial cells that line the vasculature with Orientia can lead to many complications and fatalities. In survivors, immunity does not last long, and is poorly cross-reactive among numerous strains. In addition, chronic infections are established in an unknown number of patients. All those characteristics justify the pursuit of a prophylactic vaccine against O. tsutsugamushi; however, despite continuous efforts to develop such a vaccine since World War II, the objective has not been attained. In this review, we discuss the history of vaccine development against Orientia to provide a clear picture of the challenges that we continue to face from the perspective of animal models and the immunological challenges posed by an intracellular bacterium that normally triggers a short-lived immune response. We finish with a proposal for development of an effective and safe vaccine for scrub typhus through a new approach with a strong focus on T cell-mediated immunity, empirical testing of the immunogenicity of proteins encoded by conserved genes, and assessment of protection in relevant animal models that truly mimic human scrub typhus.
Collapse
Affiliation(s)
- Gustavo Valbuena
- Department of Pathology, University of Texas Medical Branch Galveston, TX, USA.
| | | |
Collapse
|
44
|
Sustained high level of serum VEGF at convalescent stage contributes to the renal recovery after HTNV infection in patients with hemorrhagic fever with renal syndrome. Clin Dev Immunol 2012; 2012:812386. [PMID: 23097674 PMCID: PMC3477746 DOI: 10.1155/2012/812386] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 09/15/2012] [Accepted: 09/16/2012] [Indexed: 12/07/2022]
Abstract
To investigate the role of vascular endothelial growth factor (VEGF) in the increased permeability of vascular endothelial cells after Hantaan virus (HTNV) infection in humans, the concentration of VEGF in serum from HTNV infected patients was quantified with sandwich ELISA. Generally, the level of serum VEGF in patients was elevated to 607.0 (542.2-671.9) pg/mL, which was dramatically higher compared with healthy controls (P < 0.001). There was a rapid increase of the serum VEGF level in all patients from the fever onset to oliguric stage, at which the serum creatinine reached the peak level of the disease, indicating that VEGF may be involved in the pathogenesis of renal hyper-permeability. Moreover, the serum VEGF level at convalescent stage was positively correlated with the degree of the disease severity. The sustained high level of serum VEGF at convalescence was observed in critical HFRS patients, suggesting that VEGF would probably contribute to the renal recovery after the virus clearance. Taken together, our results suggested that the VEGF would be involved in the pathogenesis of renal dysfunction at the oliguric stage after HTNV infection, but may function as a recovery factor during the convalescence to help the body self-repair of the renal injury.
Collapse
|
45
|
Roles for endothelial cells in dengue virus infection. Adv Virol 2012; 2012:840654. [PMID: 22952474 PMCID: PMC3431041 DOI: 10.1155/2012/840654] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 07/19/2012] [Indexed: 02/06/2023] Open
Abstract
Dengue viruses cause two severe diseases that alter vascular fluid barrier functions, dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). The endothelium is the primary fluid barrier of the vasculature and ultimately the effects of dengue virus infection that cause capillary leakage impact endothelial cell (EC) barrier functions. The ability of dengue virus to infect the endothelium provides a direct means for dengue to alter capillary permeability, permit virus replication, and induce responses that recruit immune cells to the endothelium. Recent studies focused on dengue virus infection of primary ECs have demonstrated that ECs are efficiently infected, rapidly produce viral progeny, and elicit immune enhancing cytokine responses that may contribute to pathogenesis. Furthermore, infected ECs have also been implicated in enhancing viremia and immunopathogenesis within murine dengue disease models. Thus dengue-infected ECs have the potential to directly contribute to immune enhancement, capillary permeability, viremia, and immune targeting of the endothelium. These effects implicate responses of the infected endothelium in dengue pathogenesis and rationalize therapeutic targeting of the endothelium and EC responses as a means of reducing the severity of dengue virus disease.
Collapse
|
46
|
The Role of the Endothelium in HPS Pathogenesis and Potential Therapeutic Approaches. Adv Virol 2012; 2012:467059. [PMID: 22811711 PMCID: PMC3395186 DOI: 10.1155/2012/467059] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 05/16/2012] [Accepted: 05/18/2012] [Indexed: 02/07/2023] Open
Abstract
American hantaviruses cause a highly lethal acute pulmonary edema termed hantavirus pulmonary syndrome (HPS). Hantaviruses nonlytically infect endothelial cells and cause dramatic changes in barrier functions of the endothelium without disrupting the endothelium. Instead hantaviruses cause changes in the function of infected endothelial cells that normally regulate fluid barrier functions of capillaries. The endothelium of arteries, veins, and lymphatic vessels is unique and central to the function of vast pulmonary capillary beds, which regulate pulmonary fluid accumulation. The endothelium maintains vascular barrier functions through a complex series of redundant receptors and signaling pathways that serve to both permit fluid and immune cell efflux into tissues and restrict tissue edema. Infection of the endothelium provides several mechanisms for hantaviruses to alter capillary permeability but also defines potential therapeutic targets for regulating acute pulmonary edema and HPS disease. Here we discuss interactions of HPS causing hantaviruses with the endothelium, potential endothelial cell-directed permeability mechanisms, and therapeutic targeting of the endothelium as a means of reducing the severity of HPS disease.
Collapse
|
47
|
Gong B, Ma L, Liu Y, Gong Q, Shelite T, Bouyer D, Boor PJ, Lee YS, Oberhauser A. Rickettsiae induce microvascular hyperpermeability via phosphorylation of VE-cadherins: evidence from atomic force microscopy and biochemical studies. PLoS Negl Trop Dis 2012; 6:e1699. [PMID: 22720111 PMCID: PMC3373609 DOI: 10.1371/journal.pntd.0001699] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 05/02/2012] [Indexed: 11/18/2022] Open
Abstract
The most prominent pathophysiological effect of spotted fever group (SFG) rickettsial infection of microvascular endothelial cells (ECs) is an enhanced vascular permeability, promoting vasogenic cerebral edema and non-cardiogenic pulmonary edema, which are responsible for most of the morbidity and mortality in severe cases. To date, the cellular and molecular mechanisms by which SFG Rickettsia increase EC permeability are largely unknown. In the present study we used atomic force microscopy (AFM) to study the interactive forces between vascular endothelial (VE)-cadherin and human cerebral microvascular EC infected with R. montanensis, which is genetically similar to R. rickettsii and R. conorii, and displays a similar ability to invade cells, but is non-pathogenic and can be experimentally manipulated under Biosafety Level 2 (BSL2) conditions. We found that infected ECs show a significant decrease in VE-cadherin-EC interactions. In addition, we applied immunofluorescent staining, immunoprecipitation phosphorylation assay, and an in vitro endothelial permeability assay to study the biochemical mechanisms that may participate in the enhanced vascular permeability as an underlying pathologic alteration of SFG rickettsial infection. A major finding is that infection of R. montanensis significantly activated tyrosine phosphorylation of VE-cadherin beginning at 48 hr and reaching a peak at 72 hr p.i. In vitro permeability assay showed an enhanced microvascular permeability at 72 hr p.i. On the other hand, AFM experiments showed a dramatic reduction in VE-cadherin-EC interactive forces at 48 hr p.i. We conclude that upon infection by SFG rickettsiae, phosphorylation of VE-cadherin directly attenuates homophilic protein-protein interactions at the endothelial adherens junctions, and may lead to endothelial paracellular barrier dysfunction causing microvascular hyperpermeability. These new approaches should prove useful in characterizing the antigenically related SFG rickettsiae R. conorii and R. rickettsii in a BSL3 environment. Future studies may lead to the development of new therapeutic strategies to inhibit the VE-cadherin-associated microvascular hyperpermeability in SFG rickettsioses.
Collapse
Affiliation(s)
- Bin Gong
- Department of Pathology, University of Texas Medical Branch at Galveston, TX, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Dengue viruses cause two severe diseases that alter vascular fluid barrier functions, dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Preexisting antibodies to dengue virus disposes patients to immune-enhanced edema (DSS) or hemorrhagic (DHF) disease following infection by a discrete dengue virus serotype. Although the endothelium is the primary vascular fluid barrier, direct effects of dengue virus on endothelial cells (ECs) have not been considered primary factors in pathogenesis. Here, we show that dengue virus infection of human ECs elicits immune-enhancing EC responses. Our results suggest that rapid early dengue virus proliferation within ECs is permitted by dengue virus regulation of early, but not late, beta interferon (IFN-β) responses. The analysis of EC responses following synchronous dengue virus infection revealed the high-level induction and secretion of immune cells (T cells, B cells, and mast cells) as well as activating and recruiting cytokines BAFF (119-fold), IL-6/8 (4- to 7-fold), CXCL9/10/11 (45- to 338-fold), RANTES (724-fold), and interleukin-7 (IL-7; 128-fold). Moreover, we found that properdin factor B, an alternative pathway complement activator that directs chemotactic anaphylatoxin C3a and C5a production, was induced 34-fold. Thus, dengue virus-infected ECs evoke key inflammatory responses observed in dengue virus patients which are linked to DHF and DSS. Our findings suggest that dengue virus-infected ECs directly contribute to immune enhancement, capillary permeability, viremia, and immune targeting of the endothelium. These data implicate EC responses in dengue virus pathogenesis and further rationalize therapeutic targeting of the endothelium as a means of reducing the severity of dengue virus disease.
Collapse
|
49
|
Wang W, Zhang Y, Li Y, Pan L, Bai L, Zhuang Y, Huang CX, Wang JP, Yu HT, Wei X, Jiang W, Nan YY, Yang DQ, Su WJ, Wang PZ, Bai XF. Dysregulation of the β3 integrin-VEGFR2 complex in Hantaan virus-directed hyperpermeability upon treatment with VEGF. Arch Virol 2012; 157:1051-61. [PMID: 22407444 DOI: 10.1007/s00705-012-1245-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 12/27/2011] [Indexed: 01/13/2023]
Abstract
Hantaviruses infect human endothelial cells (ECs) and are known to cause vascular-permeability-based diseases, including hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). The αvβ3 integrins, which are highly expressed on the surface of ECs, serve as hantavirus receptors. Specifically, the β3 integrin and vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) form a functional complex and interact with each other. Signaling through this complex causes cytoskeletal reorganization, which is one of the most important mechanisms underlying hyperpermeability. In this study, we show that VEGF dramatically enhances Hantaan virus (HTNV)-directed permeability and increases the reorganization of the cytoskeleton and the disruption of junctional organizations in an EC monolayer at 3 days postinfection. HTNV infection reduced the effect of VEGF on adhesion, migration, and the upregulation of β3 expression, but the infection alone upregulated the expression of β3 and VEGFR2. These results indicate that in addition to its role in blocking β3 integrin activation as reported previously, HTNV blocks the function of the complex of VEGFR2 and β3 integrin, and the dysfunction of the complex may contribute to cytoskeletal reorganization in an HTNV-directed hyperpermeability response to VEGF.
Collapse
Affiliation(s)
- Wei Wang
- Center of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Shear stress levels generated by circulating blood have a strong impact on biological processes taking place in the vasculature. It is therefore important to take them into account when studying infectious agents targeting the endothelium. Here we describe a protocol using disposable laminar-flow chambers and video microcopy to study bacterial infections in an environment that mimics the bloodstream. We initially focused on the interaction of Neisseria meningitidis with human endothelial cells and determined that shear stress is an important factor for the pathogen's initial adhesion and for the formation of micro-colonies. The experimental set-up can be used to investigate other pathogens that interact with the endothelium as well as with other sites where shear stress is present.
Collapse
|