1
|
Rosen H, Roberts E. The discovery and development of the sphingosine 1-phosphate receptor modulator ozanimod in ulcerative colitis. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:183-202. [PMID: 39521600 DOI: 10.1016/bs.apha.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Sphingosine-1 phosphate (S1P) modulators have received recent FDA-approval for the treatment of moderate-to-severe ulcerative colitis, including agents ozanimod, approved in 2021, and etrasimod, approved in 2023. These oral drugs are uniquely efficacious in UC as they have multimodal mechanisms contributing to their beneficial immunomodulatory effects, while preserving host response to pathogens and attenuating toxicities observed with less specific agents. In this review, the discovery and development of the first approved S1P modulator, ozanimod, is described in detail: from design of initial screens to discover unique binding agents, to extensive chemical modifications to improve pharmacokinetic and safety profiles, and through preclinical and clinical studies validating mechanism and establishing safety and efficacy. Ultimately, this review will not only inform the reader of the unique path to development of a clinical S1P modulator for UC, but will also highlight advances made and gaps remaining to individualize therapeutic approaches for inflammatory bowel disease.
Collapse
Affiliation(s)
- Hugh Rosen
- Department of Cellular and Molecular Biology, Scripps Research, La Jolla, CA, United States.
| | - Edward Roberts
- Department of Chemistry, Scripps Research, La Jolla, CA, United States
| |
Collapse
|
2
|
Riyaz Tramboo S, Elkhalifa AM, Quibtiya S, Ali SI, Nazir Shah N, Taifa S, Rakhshan R, Hussain Shah I, Ahmad Mir M, Malik M, Ramzan Z, Bashir N, Ahad S, Khursheed I, Bazie EA, Mohamed Ahmed E, Elderdery AY, Alenazy FO, Alanazi A, Alzahrani B, Alruwaili M, Manni E, E. Hussein S, Abdalhabib EK, Nabi SU. The critical impacts of cytokine storms in respiratory disorders. Heliyon 2024; 10:e29769. [PMID: 38694122 PMCID: PMC11058722 DOI: 10.1016/j.heliyon.2024.e29769] [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: 01/16/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/03/2024] Open
Abstract
Cytokine storm (CS) refers to the spontaneous dysregulated and hyper-activated inflammatory reaction occurring in various clinical conditions, ranging from microbial infection to end-stage organ failure. Recently the novel coronavirus involved in COVID-19 (Coronavirus disease-19) caused by SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) has been associated with the pathological phenomenon of CS in critically ill patients. Furthermore, critically ill patients suffering from CS are likely to have a grave prognosis and a higher case fatality rate. Pathologically CS is manifested as hyper-immune activation and is clinically manifested as multiple organ failure. An in-depth understanding of the etiology of CS will enable the discovery of not just disease risk factors of CS but also therapeutic approaches to modulate the immune response and improve outcomes in patients with respiratory diseases having CS in the pathogenic pathway. Owing to the grave consequences of CS in various diseases, this phenomenon has attracted the attention of researchers and clinicians throughout the globe. So in the present manuscript, we have attempted to discuss CS and its ramifications in COVID-19 and other respiratory diseases, as well as prospective treatment approaches and biomarkers of the cytokine storm. Furthermore, we have attempted to provide in-depth insight into CS from both a prophylactic and therapeutic point of view. In addition, we have included recent findings of CS in respiratory diseases reported from different parts of the world, which are based on expert opinion, clinical case-control research, experimental research, and a case-controlled cohort approach.
Collapse
Affiliation(s)
- Shahana Riyaz Tramboo
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, J&K, 190006, India
| | - Ahmed M.E. Elkhalifa
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh, 11673, Saudi Arabia
- Department of Haematology, Faculty of Medical Laboratory Sciences, University of El Imam El Mahdi, Kosti, 1158, Sudan
| | - Syed Quibtiya
- Department of General Surgery, Sher-I-Kashmir Institute of Medical Sciences, Medical College, Srinagar, 190011, Jammu & Kashmir, India
| | - Sofi Imtiyaz Ali
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, J&K, 190006, India
| | - Naveed Nazir Shah
- Department of Chest Medicine, Govt. Medical College, Srinagar, 191202, Jammu & Kashmir, India
| | - Syed Taifa
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, J&K, 190006, India
| | - Rabia Rakhshan
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu & Kashmir, 190006, India
| | - Iqra Hussain Shah
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, J&K, 190006, India
| | - Muzafar Ahmad Mir
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, J&K, 190006, India
| | - Masood Malik
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, J&K, 190006, India
| | - Zahid Ramzan
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, J&K, 190006, India
| | - Nusrat Bashir
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, J&K, 190006, India
| | - Shubeena Ahad
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, J&K, 190006, India
| | - Ibraq Khursheed
- Department of Zoology, Central University of Kashmir, 191201, Nunar, Ganderbal, Jammu & Kashmir, India
| | - Elsharif A. Bazie
- Pediatric Department, Faculty of Medicine, University of El Imam El Mahdi, Kosti, 1158, Sudan
| | - Elsadig Mohamed Ahmed
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, 61922, Saudi Arabia
- Department of Clinical Chemistry, Faculty of Medical Laboratory Sciences, University of El Imam El Mahdi, Kosti, 1158, Sudan
| | - Abozer Y. Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al-Qurayyat, Saudi Arabia
| | - Fawaz O. Alenazy
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al-Qurayyat, Saudi Arabia
| | - Awadh Alanazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al-Qurayyat, Saudi Arabia
| | - Badr Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al-Qurayyat, Saudi Arabia
| | - Muharib Alruwaili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al-Qurayyat, Saudi Arabia
| | - Emad Manni
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al-Qurayyat, Saudi Arabia
| | - Sanaa E. Hussein
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al-Qurayyat, Saudi Arabia
| | - Ezeldine K. Abdalhabib
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al-Qurayyat, Saudi Arabia
| | - Showkat Ul Nabi
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, J&K, 190006, India
| |
Collapse
|
3
|
Modak A, Mishra SR, Awasthi M, Aravind A, Singh S, Sreekumar E. Fingolimod (FTY720), an FDA-approved sphingosine 1-phosphate (S1P) receptor agonist, restores endothelial hyperpermeability in cellular and animal models of dengue virus serotype 2 infection. IUBMB Life 2024; 76:267-285. [PMID: 38031996 DOI: 10.1002/iub.2795] [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: 04/07/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023]
Abstract
Extensive vascular leakage and shock is a major cause of dengue-associated mortality. At present, there are no specific treatments available. Sphingolipid pathway is a key player in the endothelial barrier integrity; and is mediated through the five sphingosine-1-phosphate receptors (S1PR1-S1PR5). Signaling through S1PR2 promotes barrier disruption; and in Dengue virus (DENV)-infection, there is overexpression of this receptor. Fingolimod (FTY720) is a specific agonist that targets the remaining barrier-protective S1P receptors, without targeting S1PR2. In the present study, we explored whether FTY720 treatment can alleviate DENV-induced endothelial hyperpermeability. In functional assays, in both in vitro systems and in AG129 animal models, FTY720 treatment was found effective. Upon treatment, there was complete restoration of the monolayer integrity in DENV serotype 2-infected human microvascular endothelial cells (HMEC-1). At the molecular level, the treatment reversed activation of the S1P pathway. It significantly reduced the phosphorylation of the key molecules such as PTEN, RhoA, and VE-Cadherin; and also, the expression levels of S1PR2. In DENV2-infected AG129 mice treated with FTY720, there was significant improvement in weight gain, in overall clinical symptoms, and in survival. Whereas 100% of the DENV2-infected, untreated animals died by day-10 post-infection, 70% of the FTY720-treated animals were alive; and at the end of the 15-day post-infection observation period, 30% of them were still surviving. There was a significant reduction in the Evan's-blue dye permeability in the organs of FTY720-treated, DENV-2 infected animals; and also improvement in the hemogram, with complete restoration of thrombocytopenia and hepatic function. Our results show that the FDA-approved molecule Fingolimod (FTY720) is a promising therapeutic intervention in severe dengue.
Collapse
Affiliation(s)
- Ayan Modak
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad, Haryana (NCR Delhi), India
| | - Srishti Rajkumar Mishra
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad, Haryana (NCR Delhi), India
| | - Mansi Awasthi
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad, Haryana (NCR Delhi), India
| | - Arya Aravind
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Sneha Singh
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Easwaran Sreekumar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Molecular Bioassay Laboratory, Institute of Advanced Virology (IAV), Thiruvananthapuram, Kerala, India
| |
Collapse
|
4
|
Qi J, Lv C, Guo J, Li Y, Sima M, Luo R, Xiang H, Xia X, Zhou Y, Wang T. Schisandra chinensis (Turcz.) Baill. polysaccharide inhibits influenza A virus in vitro and in vivo. FEBS Open Bio 2023; 13:1831-1843. [PMID: 37544014 PMCID: PMC10549229 DOI: 10.1002/2211-5463.13690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/08/2023] Open
Abstract
Influenza virus is prone to seasonal spread and widespread outbreaks, which pose important challenges to public health security. Therefore, it is important to effectively prevent and treat influenza virus infection. Schisandra polysaccharide (SPJ) is a polysaccharide derived from the fruit of Schisandra chinensis (Turcz.) Baill. In this study, we evaluated the antiviral activity of SPJ in vitro and in vivo, especially against influenza A virus (IAV) infection. By analyzing SPJ structure and monosaccharide composition, the molecular weight of SPJ was determined to be 115.5 KD, and it is composed of galacturonic acid (89.4%), rhamnose (0.8%), galactose (4.4%), arabinose (3.8%), and glucose (1.7%). Immunofluorescence analysis showed that SPJ treatment reduced the positive rate of viral nucleoproteins in cells, indicating that the compound had an inhibitory effect on influenza virus replication. Furthermore, SPJ therapy improved the survival of infected mice. Lung virus titer assays indicated that SPJ treatment significantly reduced viral loading in the lung tissue of infected mice and alleviated the pathological damage caused by influenza virus infection. Moreover, SPJ reduced cytokine expression during influenza virus challenge. In conclusion, SPJ has anti-influenza virus effects and may have potential as an anti-influenza drug candidate in further clinical studies.
Collapse
Affiliation(s)
- Jing Qi
- College of Life SciencesNortheast Normal UniversityChangchunChina
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChina
| | - Chaoxiang Lv
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChina
- The Research Center for Preclinical MedicineSouthwest Medical UniversityLuzhouChina
| | - Jin Guo
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChina
- College of Life SciencesShandong Normal UniversityJinanChina
| | - Yuanguo Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChina
| | - Mingwei Sima
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChina
- College of Basic MedicineChangchun University of Chinese MedicineChina
| | - Rongbo Luo
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChina
| | - Haiyang Xiang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChina
| | - Xianzhu Xia
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChina
| | - Yifa Zhou
- College of Life SciencesNortheast Normal UniversityChangchunChina
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChina
| |
Collapse
|
5
|
Teymouri S, Pourbayram Kaleybar S, Hejazian SS, Hejazian SM, Ansarin K, Ardalan M, Zununi Vahed S. The effect of Fingolimod on patients with moderate to severe COVID-19. Pharmacol Res Perspect 2023; 11:e01039. [PMID: 36567519 PMCID: PMC9791159 DOI: 10.1002/prp2.1039] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 12/09/2022] [Indexed: 12/27/2022] Open
Abstract
Hyper-inflammation, cytokine storm, and recruitment of immune cells lead to uncontrollable endothelial cell damage in patients with coronavirus disease 2019 (COVID-19). Sphingosine 1-phosphate (S1P) signaling is needed for endothelial integrity and its decreased serum level is a predictor of clinical severity in COVID-19. In this clinical trial, the effect of Fingolimod, an agonist of S1P, was evaluated on patients with COVID-19. Forty patients with moderate to severe COVID-19 were enrolled and divided into two groups including (1) the control group (n = 21) receiving the national standard regimen for COVID-19 patients and (2) the intervention group (n = 19) that prescribed daily Fingolimod (0.5 mg) for 3 days besides receiving the standard national regimen for COVID-19. The hospitalization period, re-admission rate, intensive care unit (ICU) administration, need for mechanical ventilation, and mortality rate were assessed as primary outcomes in both groups. The results showed that re-admission was significantly decreased in COVID-19 patients who received Fingolimod compared to the controls (p = .04). In addition, the hemoglobin levels of the COVID-19 patients in the intervention group were increased compared to the controls (p = .018). However, no significant differences were found regarding the intubation or mortality rate between the groups (p > .05). Fingolimod could significantly reduce the re-admission rate after hospitalization with COVID-19. Fingolimod may not enhance patients' outcomes with moderate COVID-19. It is necessary to examine these findings in a larger cohort of patients with severe to critical COVID-19.
Collapse
Affiliation(s)
- Soheil Teymouri
- Tuberculosis and Lung Disease Research CenterTabriz University of Medical SciencesTabrizIran
| | - Siamak Pourbayram Kaleybar
- Kidney Research CenterFaculty of MedicineTabriz University of Medical SciencesTabrizIran
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | | | | | - Khalil Ansarin
- Tuberculosis and Lung Disease Research CenterTabriz University of Medical SciencesTabrizIran
| | - Mohammadreza Ardalan
- Kidney Research CenterFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| | - Sepideh Zununi Vahed
- Kidney Research CenterFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
6
|
Hach T, Shakeri-Nejad K, Bigaud M, Dahlke F, de Micco M, Petricoul O, Graham G, Piani-Meier D, Turrini R, Brinkmann V, Nicoletti F. Rationale for Use of Sphingosine-1-Phosphate Receptor Modulators in COVID-19 Patients: Overview of Scientific Evidence. J Interferon Cytokine Res 2022. [DOI: 10.1089/jir.2022.0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Thomas Hach
- Patient Engagement, Novartis Pharma AG, Basel, Switzerland
| | - Kasra Shakeri-Nejad
- Department of Clinical Pharmacology; Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Marc Bigaud
- Department of Autoimmunity, Transplantation & Inflammation; Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Frank Dahlke
- Patient Engagement, Novartis Pharma AG, Basel, Switzerland
| | | | - Olivier Petricoul
- Department of Neuroscience; Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Gordon Graham
- Patient Engagement, Novartis Pharma AG, Basel, Switzerland
| | | | - Renato Turrini
- Department of Autoimmunity, Transplantation & Inflammation; Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University Sapienza of Rome, Rome, Italy
- Department of Molecular Neuropharmacology, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
7
|
Feng T, Tong H, Ming Z, Deng L, Liu J, Wu J, Chen Z, Yan Y, Dai J. Matrix metalloproteinase 3 restricts viral infection by enhancing host antiviral immunity. Antiviral Res 2022; 206:105388. [PMID: 35940311 DOI: 10.1016/j.antiviral.2022.105388] [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: 05/12/2022] [Revised: 07/14/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022]
Abstract
Viral pandemics pose great threats to human health and the economy. The host evolved a complex immune response against viral infection. Matrix metalloproteinase 3 (MMP3), also known as stromelysin-1, has an emerging role in immune regulation during pathogen infection. Using in vitro and in vivo infection models, we showed that MMP3 exhibits broad-spectrum antiviral activities against vesicular stomatitis virus (VSV), influenza A virus (H1N1) and human herpes virus 1 (HSV-1). MMP3 deficient mice are susceptible to viral infection and display a compromised antiviral immune response. Correspondingly, the mice with MMP3 overexpression are resistant to viral infection. The mechanistic study suggested that MMP3 is translocated from the cytoplasm into the cell nucleus upon virus infection and influence NF-κB activities, thus amplifying antiviral immune responses. This study suggested a novel function of MMP3 in viral infection and provided new ideas for developing antiviral drugs based on modulating MMP activity.
Collapse
Affiliation(s)
- Tingting Feng
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Soochow University, Suzhou, China; Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| | - Hao Tong
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| | - Zhihao Ming
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| | - Lei Deng
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| | - Jiayan Liu
- Department of Radiology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China.
| | - Jiahui Wu
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| | - Zhengrong Chen
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Soochow University, Suzhou, China.
| | - Yongdong Yan
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Soochow University, Suzhou, China.
| | - Jianfeng Dai
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| |
Collapse
|
8
|
Jiang Y, Zhao T, Zhou X, Xiang Y, Gutierrez‐Castrellon P, Ma X. Inflammatory pathways in COVID-19: Mechanism and therapeutic interventions. MedComm (Beijing) 2022; 3:e154. [PMID: 35923762 PMCID: PMC9340488 DOI: 10.1002/mco2.154] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 02/05/2023] Open
Abstract
The 2019 coronavirus disease (COVID-19) pandemic has become a global crisis. In the immunopathogenesis of COVID-19, SARS-CoV-2 infection induces an excessive inflammatory response in patients, causing an inflammatory cytokine storm in severe cases. Cytokine storm leads to acute respiratory distress syndrome, pulmonary and other multiorgan failure, which is an important cause of COVID-19 progression and even death. Among them, activation of inflammatory pathways is a major factor in generating cytokine storms and causing dysregulated immune responses, which is closely related to the severity of viral infection. Therefore, elucidation of the inflammatory signaling pathway of SARS-CoV-2 is important in providing otential therapeutic targets and treatment strategies against COVID-19. Here, we discuss the major inflammatory pathways in the pathogenesis of COVID-19, including induction, function, and downstream signaling, as well as existing and potential interventions targeting these cytokines or related signaling pathways. We believe that a comprehensive understanding of the regulatory pathways of COVID-19 immune dysregulation and inflammation will help develop better clinical therapy strategies to effectively control inflammatory diseases, such as COVID-19.
Collapse
Affiliation(s)
- Yujie Jiang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduPR China
| | - Tingmei Zhao
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduPR China
| | - Xueyan Zhou
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduPR China
| | - Yu Xiang
- Department of BiotherapyState Key Laboratory of Biotherapy Cancer CenterWest China HospitalSichuan UniversityChengduPR China
| | - Pedro Gutierrez‐Castrellon
- Center for Translational Research on Health Science Hospital General Dr. Manuel Gea GonzalezMinistry of HealthMexico CityMexico
| | - Xuelei Ma
- Department of BiotherapyState Key Laboratory of Biotherapy Cancer CenterWest China HospitalSichuan UniversityChengduPR China
| |
Collapse
|
9
|
Pasharawipas T. Perspectives Concerning Various Symptoms of SARS-CoV-2 Detected Individuals. Open Microbiol J 2021. [DOI: 10.2174/1874285802115010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
After exposure to SARS-CoV-2, varying symptoms of COVID-19 ranging from asymptomatic symptoms to morbidity and mortality have been exhibited in each individual. SARS-CoV-2 requires various cellular molecules for penetration into a target host cell. Angiotensin-converting enzyme2 (ACE2) acts as the viral receptor molecule. After attachment, SARS-CoV-2 also requires the transmembrane protease serine-2 (TMPRSS-2) and furin molecules, which serve as co-receptors for penetration into the target cell and for subsequent replication. In the meantime, a major histocompatibility complex (MHC) is required for the induction of adaptive immune cells, especially cytotoxic T cells and helper T cells, to clear the virally infected cells. This perspective review article proposes different aspects to explain the varying symptoms of the individuals who have been exposed to SARS-CoV-2, which relates to the polymorphisms of these involved molecules.
Collapse
|
10
|
Pandemics of the 21st Century: The Risk Factor for Obese People. Viruses 2021; 14:v14010025. [PMID: 35062229 PMCID: PMC8779521 DOI: 10.3390/v14010025] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023] Open
Abstract
The number of obese adults and children is increasing worldwide, with obesity now being a global epidemic. Around 2.8 million people die annually from clinical overweight or obesity. Obesity is associated with numerous comorbid conditions including hypertension, cardiovascular disease, type 2 diabetes, hypercholesterolemia, hypertriglyceridemia, nonalcoholic fatty liver disease, and cancer, and even the development of severe disease after infection with viruses. Over the past twenty years, a number of new viruses has emerged and entered the human population. Moreover, influenza (H1N1)pdm09 virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have caused pandemics. During pandemics, the number of obese patients presents challenging and complex issues in medical and surgical intensive care units. Morbidity amongst obese individuals is directly proportional to body mass index. In this review, we describe the impact of obesity on the immune system, adult mortality, and immune response after infection with pandemic influenza virus and SARS-CoV-2. Finally, we address the effect of obesity on vaccination.
Collapse
|
11
|
Jiang H, Gu J, Zhao H, Joshi S, Perlmutter JS, Gropler RJ, Klein RS, Benzinger TLS, Tu Z. PET Study of Sphingosine-1-phosphate Receptor 1 Expression in Response to S. aureus Infection. Mol Imaging 2021; 2021:9982020. [PMID: 34934406 PMCID: PMC8654346 DOI: 10.1155/2021/9982020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/02/2021] [Indexed: 11/22/2022] Open
Abstract
Sphingosine-1-phosphate receptor 1 (S1PR1) plays a crucial role in infectious diseases. Targeting S1PR1 provides protection against pathogens, such as influenza viruses. This study is aimed at investigating S1PR1 in response to bacterial infection by assessing S1PR1 expression in S. aureus-infected mice. A rodent local muscle bacterial infection model was developed by injecting S. aureus to the lower hind limb of Balb/c mice. The changes of S1PR1 expression in response to bacterial infection and blocking treatment were assessed using ex vivo biodistribution and in vivo positron emission tomography (PET) after intravenous injection of an S1PR1-specific radiotracer [18F]TZ4877. The specificity of [18F]TZ4877 was assessed using S1PR1-specific antagonist, NIBR-0213, and S1PR1-specific DsiRNA pretreated the animals. Immunohistochemical studies were performed to confirm the increase of S1PR1 expression in response to infection. Ex vivo biodistribution data showed that the uptake of [18F]TZ4877 was increased 30.6%, 54.3%, 74.3%, and 115.3% in the liver, kidney, pancreas, and thymus of the infected mice, respectively, compared to that in normal control mice, indicating that S1PR1 is involved in the early immune response to bacterial infection. NIBR-0213 or S1PR1-specific DsiRNA pretreatment reduced the tissue uptake of [18F]TZ4877, suggesting that uptake of [18F]TZ4877 is specific. Our PET/CT study data also confirmed that infected mice have increased [18F]TZ4877 uptake in several organs comparing to that in normal control mice. Particularly, compared to control mice, a 39% increase of [18F]TZ4877 uptake was observed in the infected muscle of S. aureus mice, indicating that S1PR1 expression was directly involved in the inflammatory response to infection. Overall, our study suggested that S1PR1 plays an important role in the early immune response to bacterial infection. The uptake of [18F]TZ4877 is tightly correlated with the S1R1 expression in response to S. aureus infection. PET with S1PR1-specific radiotracer [18F]TZ4877 could provide a noninvasive tool for detecting the early S1PR1 immune response to infectious diseases.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jiwei Gu
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Haiyang Zhao
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Sumit Joshi
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Joel S. Perlmutter
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, USA
- Department of Neuroscience, Neurology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Robert J. Gropler
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Robyn S. Klein
- Department of Neuroscience, Neurology, Washington University School of Medicine, St Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Tammie L. S. Benzinger
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, USA
- Department of Neurological Surgery, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
12
|
Abstract
Influenza viruses are one of the leading causes of respiratory tract infections in humans and their newly emerging and re-emerging virus strains are responsible for seasonal epidemics and occasional pandemics, leading to a serious threat to global public health systems. The poor clinical outcome and pathogenesis during influenza virus infection in humans and animal models are often associated with elevated proinflammatory cytokines and chemokines production, which is also known as hypercytokinemia or "cytokine storm", that precedes acute respiratory distress syndrome (ARDS) and often leads to death. Although we still do not fully understand the complex nature of cytokine storms, the use of immunomodulatory drugs is a promising approach for treating hypercytokinemia induced by an acute viral infection, including highly pathogenic avian influenza virus infection and Coronavirus Disease 2019 (COVID-19). This review aims to discuss the immune responses and cytokine storm pathology induced by influenza virus infection and also summarize alternative experimental strategies for treating hypercytokinemia caused by influenza virus.
Collapse
Affiliation(s)
- Fanhua Wei
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China.,College of Agriculture, Ningxia University, Yinchuan, China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Yujiong Wang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China.,College of Life Science, Ningxia University, Yinchuan, China
| |
Collapse
|
13
|
Morris G, Bortolasci CC, Puri BK, Marx W, O'Neil A, Athan E, Walder K, Berk M, Olive L, Carvalho AF, Maes M. The cytokine storms of COVID-19, H1N1 influenza, CRS and MAS compared. Can one sized treatment fit all? Cytokine 2021; 144:155593. [PMID: 34074585 PMCID: PMC8149193 DOI: 10.1016/j.cyto.2021.155593] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/03/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023]
Abstract
An analysis of published data appertaining to the cytokine storms of COVID-19, H1N1 influenza, cytokine release syndrome (CRS), and macrophage activation syndrome (MAS) reveals many common immunological and biochemical abnormalities. These include evidence of a hyperactive coagulation system with elevated D-dimer and ferritin levels, disseminated intravascular coagulopathy (DIC) and microthrombi coupled with an activated and highly permeable vascular endothelium. Common immune abnormalities include progressive hypercytokinemia with elevated levels of TNF-α, interleukin (IL)-6, and IL-1β, proinflammatory chemokines, activated macrophages and increased levels of nuclear factor kappa beta (NFκB). Inflammasome activation and release of damage associated molecular patterns (DAMPs) is common to COVID-19, H1N1, and MAS but does not appear to be a feature of CRS. Elevated levels of IL-18 are detected in patients with COVID-19 and MAS but have not been reported in patients with H1N1 influenza and CRS. Elevated interferon-γ is common to H1N1, MAS, and CRS but levels of this molecule appear to be depressed in patients with COVID-19. CD4+ T, CD8+ and NK lymphocytes are involved in the pathophysiology of CRS, MAS, and possibly H1N1 but are reduced in number and dysfunctional in COVID-19. Additional elements underpinning the pathophysiology of cytokine storms include Inflammasome activity and DAMPs. Treatment with anakinra may theoretically offer an avenue to positively manipulate the range of biochemical and immune abnormalities reported in COVID-19 and thought to underpin the pathophysiology of cytokine storms beyond those manipulated via the use of, canakinumab, Jak inhibitors or tocilizumab. Thus, despite the relative success of tocilizumab in reducing mortality in COVID-19 patients already on dexamethasone and promising results with Baricitinib, the combination of anakinra in combination with dexamethasone offers the theoretical prospect of further improvements in patient survival. However, there is currently an absence of trial of evidence in favour or contravening this proposition. Accordingly, a large well powered blinded prospective randomised controlled trial (RCT) to test this hypothesis is recommended.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | | | - Wolfgang Marx
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Melbourne School of Population and Global Health, Melbourne, Australi
| | - Eugene Athan
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, Australia
| | - Lisa Olive
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, School of Psychology, Geelong, Australia
| | - Andre F Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, University of Toronto, Toronto, Canada, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
| |
Collapse
|
14
|
Fouladseresht H, Doroudchi M, Rokhtabnak N, Abdolrahimzadehfard H, Roudgari A, Sabetian G, Paydar S. Predictive monitoring and therapeutic immune biomarkers in the management of clinical complications of COVID-19. Cytokine Growth Factor Rev 2021; 58:32-48. [PMID: 33199179 PMCID: PMC7544568 DOI: 10.1016/j.cytogfr.2020.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022]
Abstract
The coronavirus disease-2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), appears with a wide spectrum of mild-to-critical clinical complications. Many clinical and experimental findings suggest the role of inflammatory mechanisms in the immunopathology of COVID-19. Hence, cellular and molecular mediators of the immune system can be potential targets for predicting, monitoring, and treating the progressive complications of COVID-19. In this review, we assess the latest cellular and molecular data on the immunopathology of COVID-19 according to the pathological evidence (e.g., mucus and surfactants), dysregulations of pro- and anti-inflammatory mediators (e.g., cytokines and chemokines), and impairments of innate and acquired immune system functions (e.g., mononuclear cells, neutrophils and antibodies). Furthermore, we determine the significance of immune biomarkers for predicting, monitoring, and treating the progressive complications of COVID-19. We also discuss the clinical importance of recent immune biomarkers in COVID-19, and at the end of each section, recent clinical trials in immune biomarkers for COVID-19 are mentioned.
Collapse
Affiliation(s)
- Hamed Fouladseresht
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Rokhtabnak
- Department of Biology, Faculty of Science, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Hossein Abdolrahimzadehfard
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Roudgari
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Golnar Sabetian
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahram Paydar
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
15
|
McGowan EM, Haddadi N, Nassif NT, Lin Y. Targeting the SphK-S1P-SIPR Pathway as a Potential Therapeutic Approach for COVID-19. Int J Mol Sci 2020; 21:ijms21197189. [PMID: 33003377 PMCID: PMC7583882 DOI: 10.3390/ijms21197189] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
The world is currently experiencing the worst health pandemic since the Spanish flu in 1918-the COVID-19 pandemic-caused by the coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This pandemic is the world's third wake-up call this century. In 2003 and 2012, the world experienced two major coronavirus outbreaks, SARS-CoV-1 and Middle East Respiratory syndrome coronavirus (MERS-CoV), causing major respiratory tract infections. At present, there is neither a vaccine nor a cure for COVID-19. The severe COVID-19 symptoms of hyperinflammation, catastrophic damage to the vascular endothelium, thrombotic complications, septic shock, brain damage, acute disseminated encephalomyelitis (ADEM), and acute neurological and psychiatric complications are unprecedented. Many COVID-19 deaths result from the aftermath of hyperinflammatory complications, also referred to as the "cytokine storm syndrome", endotheliitus and blood clotting, all with the potential to cause multiorgan dysfunction. The sphingolipid rheostat plays integral roles in viral replication, activation/modulation of the immune response, and importantly in maintaining vasculature integrity, with sphingosine 1 phosphate (S1P) and its cognate receptors (SIPRs: G-protein-coupled receptors) being key factors in vascular protection against endotheliitus. Hence, modulation of sphingosine kinase (SphK), S1P, and the S1P receptor pathway may provide significant beneficial effects towards counteracting the life-threatening, acute, and chronic complications associated with SARS-CoV-2 infection. This review provides a comprehensive overview of SARS-CoV-2 infection and disease, prospective vaccines, and current treatments. We then discuss the evidence supporting the targeting of SphK/S1P and S1P receptors in the repertoire of COVID-19 therapies to control viral replication and alleviate the known and emerging acute and chronic symptoms of COVID-19. Three clinical trials using FDA-approved sphingolipid-based drugs being repurposed and evaluated to help in alleviating COVID-19 symptoms are discussed.
Collapse
Affiliation(s)
- Eileen M McGowan
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precise Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China;
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
- Correspondence: ; Tel.: +61-405814048
| | - Nahal Haddadi
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
| | - Najah T. Nassif
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
| | - Yiguang Lin
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precise Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China;
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
| |
Collapse
|
16
|
Ao DS, Gao LY, Gu JH, Qiao JH, Wang H, Liu YF, Song H. Study on Adenovirus Infection in vitro with Nanoself-Assembling Peptide as Scaffolds for 3D Culture. Int J Nanomedicine 2020; 15:6327-6338. [PMID: 32922004 PMCID: PMC7457861 DOI: 10.2147/ijn.s239395] [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: 04/08/2020] [Accepted: 07/30/2020] [Indexed: 01/04/2023] Open
Abstract
Purpose To construct a three-dimensional (3D) culture model of adenovirus in vitro using the nanoself-assembling peptide RADA16-I as a 3D cell culture scaffold combined with virology experimental technology to provide a novel research method for virus isolation and culture, pathogenesis research, antiviral drug screening and vaccine preparation. Methods The nanoself-assembling peptide RADA16-I was used as a 3D scaffold material for 293T cell culture, and adenovirus was cultured in the cells. The growth, morphological characteristics and pathological effects of 3D-cultured 293T cells after adenovirus infection were observed with an inverted microscope and MTS. The proliferation of adenovirus in 293T cells was observed by TEM and detected by qPCR. The levels of TNF-α and IL-8 secreted by adenovirus-infected 293T cells in the RADA16-I 3D culture system were detected by ELISA. Results The 293T cells grew well in the RADA16-I 3D culture system for a prolonged period of time. The adenovirus infection persisted for a long time with multiple proliferation peaks, which closely resembled those of in vivo infections. The adenovirus virions amplified in the 3D system remained infectious. There were multiple secretion peaks of TNF-α and IL-8 secretion levels in adenovirus-infected 293T cells cultured in 3D culture systems. Conclusion The nanoself-assembling peptide RADA16-I can be used as a 3D scaffold for adenovirus isolation, culture and research. The 3D culture system shows more realistic in vivo effects than two-dimensional (2D) culture.
Collapse
Affiliation(s)
- Di-Shu Ao
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Lu-Yao Gao
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Jing-Han Gu
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Jun-Hua Qiao
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Huan Wang
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Yan-Fei Liu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Hong Song
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| |
Collapse
|
17
|
Immunopathology of SARS-CoV-2 Infection: Immune Cells and Mediators, Prognostic Factors, and Immune-Therapeutic Implications. Int J Mol Sci 2020; 21:ijms21134782. [PMID: 32640747 PMCID: PMC7370171 DOI: 10.3390/ijms21134782] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
The present is a comprehensive review of the immunopathology of Covid-19. The immune reaction to SARS-CoV-2 infection is characterized by differentiation and proliferation of a variety of immune cells with immune mediator production and release, and activation of other pathogen resistance mechanisms. We fully address the humoral and cellular immune changes induced by the virus, with particular emphasis on the role of the “cytokine storm” in the evolution of the disease. Moreover, we also propose some immune alterations (i.e., inflammatory parameters, cytokines, leukocytes and lymphocyte subpopulations) as prognostic markers of the disease. Furthermore, we discuss how immune modifying drugs, such as tocilizumab, chloroquine, glucocorticoids and immunoglobulins, and blood purification therapy, can constitute a fundamental moment in the therapy of the infection. Finally, we made a critical analysis of a number of substances, not yet utilized, but potentially useful in SARS-CoV-2 patients, such as IFN lambda, TNF blockers, ulinastatin, siponimod, tacrolimus, mesenchymal stem cells, inhibitors of mononuclear macrophage recruitment, IL-1 family antagonists, JAK-2 or STAT-3 inhibitors.
Collapse
|
18
|
Ye Q, Wang B, Mao J. The pathogenesis and treatment of the `Cytokine Storm' in COVID-19. J Infect 2020; 80:607-613. [PMID: 32283152 PMCID: PMC7194613 DOI: 10.1016/j.jinf.2020.03.037] [Citation(s) in RCA: 1947] [Impact Index Per Article: 389.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 02/06/2023]
Abstract
Cytokine storm is an excessive immune response to external stimuli. The pathogenesis of the cytokine storm is complex. The disease progresses rapidly, and the mortality is high. Certain evidence shows that, during the coronavirus disease 2019 (COVID-19) epidemic, the severe deterioration of some patients has been closely related to the cytokine storm in their bodies. This article reviews the occurrence mechanism and treatment strategies of the COVID-19 virus-induced inflammatory storm in attempt to provide valuable medication guidance for clinical treatment.
Collapse
Affiliation(s)
- Qing Ye
- National Clinical Research Center for Child Health, National Children's Regional Medical Center, the Children's Hospital, Zhejiang University School of Medicine, No 3333, Binsheng Road, Hangzhou 310052, China
| | - Bili Wang
- National Clinical Research Center for Child Health, National Children's Regional Medical Center, the Children's Hospital, Zhejiang University School of Medicine, No 3333, Binsheng Road, Hangzhou 310052, China
| | - Jianhua Mao
- National Clinical Research Center for Child Health, National Children's Regional Medical Center, the Children's Hospital, Zhejiang University School of Medicine, No 3333, Binsheng Road, Hangzhou 310052, China.
| |
Collapse
|
19
|
Turianová L, Lachová V, Svetlíkova D, Kostrábová A, Betáková T. Comparison of cytokine profiles induced by nonlethal and lethal doses of influenza A virus in mice. Exp Ther Med 2019; 18:4397-4405. [PMID: 31777543 PMCID: PMC6862669 DOI: 10.3892/etm.2019.8096] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023] Open
Abstract
Influenza viruses are among the most common human pathogens and are responsible for causing extensive seasonal morbidity and mortality. To investigate the immunological factors associated with severe influenza infection, the immune responses in mice infected with nonlethal (LD0) doses of A/PR/8/34 (H1N1) influenza virus were compared with those of mice infected with a lethal dose (LD100) of the virus. The virus titer and activation of retinoic acid-inducible gene (RIG)-I-like receptor signaling pathways were similar in the mice infected with LD0 and LD100 at 2 days post-infection; however, mice infected with LD100 exhibited a greater abundance of cytokines and a more diverse cytokine profile. Infection with LD100 induced the expression of the following factors: Interleukins (ILs), IL-4, IL-7, IL-10, IL-11, IL-12p40, IL-13 and IL-15; inflammatory chemokines, C-C motif chemokine ligand (CCL)2, CCL3/4, CCL12, CCL17, CCL19; and lung injury-associated cytokines, leptin, leukaemia inhibitory factor, macrophage colony stimulating factor, pentraxin (PTX)2 and PTX3, WNT1-inducible-signaling pathway protein 1, matrix metallopeptidase (MMP)-2, MMP-3, proprotein convertase subtilisin/kexin type 9, and T cell immunoglobulin and mucin domain. Switching in macrophage polarization from M1 to M2 was evidenced by the increase in M2 markers, including arginase-1 (Arg1) and early growth response protein 2 (Egr2), in the lungs of mice infected with LD100. Since IL-12 and interferon-γ are the major T helper (Th)1 cytokines, increased expression of interferon regulatory factor 4, IL-4, IL-10 and IL-13 promoted the differentiation of naïve CD4+ T cells into Th2 cells. In conclusion, the present study identified key cytokines involved in the pathogenicity of influenza infection, and demonstrated that lethal influenza virus infection induces a mixed Th1/Th2 response.
Collapse
Affiliation(s)
- Lucia Turianová
- Biomedical Research Center-Slovak Academy of Sciences, Institute of Virology, 84505 Bratislava, Slovak Republic
| | - Veronika Lachová
- Biomedical Research Center-Slovak Academy of Sciences, Institute of Virology, 84505 Bratislava, Slovak Republic
| | - Darina Svetlíkova
- Biomedical Research Center-Slovak Academy of Sciences, Institute of Virology, 84505 Bratislava, Slovak Republic
| | - Anna Kostrábová
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 84215 Bratislava, Slovak Republic
| | - Tatiana Betáková
- Biomedical Research Center-Slovak Academy of Sciences, Institute of Virology, 84505 Bratislava, Slovak Republic.,Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 84215 Bratislava, Slovak Republic
| |
Collapse
|
20
|
Zhao J, Zhu M, Jiang H, Shen S, Su X, Shi Y. Combination of sphingosine-1-phosphate receptor 1 (S1PR1) agonist and antiviral drug: a potential therapy against pathogenic influenza virus. Sci Rep 2019; 9:5272. [PMID: 30918324 PMCID: PMC6437142 DOI: 10.1038/s41598-019-41760-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/13/2019] [Indexed: 01/13/2023] Open
Abstract
The pandemic 2009 influenza A H1N1 virus is associated with significant mortality. Targeting S1PR1, which is known to modulate the immune response, provides protection against pathogenic influenza virus. The functional role and molecular mechanism of S1PR1 were analysed by generating inducible endothelial cell-specific S1PR1 knockout mice and assessing the therapeutic efficacy of the selective S1PR1 agonist CYM5442 against acute lung injury (ALI) induced by the 2009 influenza A H1N1 virus. Immune-mediated pulmonary injury is aggravated by the absence of endothelial S1PR1 and alleviated by treatment with CYM-5442, suggesting a protective function of S1PR1 signaling during H1N1 infection. S1PR1 signaling does not affect viral clearance in mice infected with influenza. Mechanistically, the MAPK and NF-kB signaling pathways are involved in the ALI mediated by S1PR1 in infected mice. Combined administration of the S1PR1 agonist CYM-5442 and the antiviral drug oseltamivir provides maximum protection from ALI. Our current study provides insight into the molecular mechanism of S1PR1 mediating the ALI induced by H1N1 infection and indicates that the combination of S1PR1 agonist with antiviral drug could potentially be used as a therapeutic remedy for future H1N1 virus pandemics.
Collapse
Affiliation(s)
- Jiangnan Zhao
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Meiying Zhu
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Hao Jiang
- Department of Emergency Medicine, the Second Affiliated Hospital, Southeast University, Nanjing, 210002, China
| | - Simen Shen
- Department of Respiratory Medicine, the First People's Hospital of Nantong, Nantong, 226000, China
| | - Xin Su
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Yi Shi
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China.
| |
Collapse
|
21
|
Abstract
Sepsis in children is typically presumed to be bacterial in origin until proven otherwise, but frequently bacterial cultures ultimately return negative. Although viruses may be important causative agents of culture-negative sepsis worldwide, the incidence, disease burden and mortality of viral-induced sepsis is poorly elucidated. Consideration of viral sepsis is critical as its recognition carries implications on appropriate use of antibacterial agents, infection control measures, and, in some cases, specific, time-sensitive antiviral therapies. This review outlines our current understanding of viral sepsis in children and addresses its epidemiology and pathophysiology, including pathogen-host interaction during active infection. Clinical manifestation, diagnostic testing, and management options unique to viral infections will be outlined.
Collapse
Affiliation(s)
- Neha Gupta
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Robert Richter
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stephen Robert
- Division of Pediatric Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Michele Kong
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
22
|
A Systems and Treatment Perspective of Models of Influenza Virus-Induced Host Responses. Processes (Basel) 2018. [DOI: 10.3390/pr6090138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Severe influenza infections are often characterized as having unique host responses (e.g., early, severe hypercytokinemia). Neuraminidase inhibitors can be effective in controlling the severe symptoms of influenza but are often not administered until late in the infection. Several studies suggest that immune modulation may offer protection to high risk groups. Here, we review the current state of mathematical models of influenza-induced host responses. Selecting three models with conserved immune response components, we determine if the immune system components which most affect virus replication when perturbed are conserved across the models. We also test each model’s response to a pre-induction of interferon before the virus is administered. We find that each model emphasizes the importance of controlling the infected cell population to control viral replication. Moreover, our work shows that the structure of current models does not allow for significant responses to increased interferon concentrations. These results suggest that the current library of available published models of influenza infection does not adequately represent the complex interactions of the virus, interferon, and other aspects of the immune response. Specifically, the method used to model virus-resistant cells may need to be adapted in future work to more realistically represent the immune response to viral infection.
Collapse
|
23
|
Ranadheera C, Coombs KM, Kobasa D. Comprehending a Killer: The Akt/mTOR Signaling Pathways Are Temporally High-Jacked by the Highly Pathogenic 1918 Influenza Virus. EBioMedicine 2018; 32:142-163. [PMID: 29866590 PMCID: PMC6021456 DOI: 10.1016/j.ebiom.2018.05.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/08/2018] [Accepted: 05/21/2018] [Indexed: 02/06/2023] Open
Abstract
Previous transcriptomic analyses suggested that the 1918 influenza A virus (IAV1918), one of the most devastating pandemic viruses of the 20th century, induces a dysfunctional cytokine storm and affects other innate immune response patterns. Because all viruses are obligate parasites that require host cells for replication, we globally assessed how IAV1918 induces host protein dysregulation. We performed quantitative mass spectrometry of IAV1918-infected cells to measure host protein dysregulation. Selected proteins were validated by immunoblotting and phosphorylation levels of members of the PI3K/AKT/mTOR pathway were assessed. Compared to mock-infected controls, >170 proteins in the IAV1918-infected cells were dysregulated. Proteins mapped to amino sugar metabolism, purine metabolism, steroid biosynthesis, transmembrane receptors, phosphatases and transcription regulation. Immunoblotting demonstrated that IAV1918 induced a slight up-regulation of the lamin B receptor whereas all other tested virus strains induced a significant down-regulation. IAV1918 also strongly induced Rab5b expression whereas all other tested viruses induced minor up-regulation or down-regulation. IAV1918 showed early reduced phosphorylation of PI3K/AKT/mTOR pathway members and was especially sensitive to rapamycin. These results suggest the 1918 strain requires mTORC1 activity in early replication events, and may explain the unique pathogenicity of this virus. Proteomic analyses of influenza 1918 virus-infected cells identified >170 dysregulated host proteins. Dysregulated proteins mapped to numerous important cellular pathways. 1918 virus infection showed prominent early reduced phosphorylation of PI3K/Akt/mTOR.
The 1918 influenza pandemic was one of the most devastating infectious disease events of the 20th century, resulting in 20–100 million deaths. Gene-based assays showed severe dysregulation of the host's cytokine responses, but little was known about global protein responses to virus infection. This work identifies unique and temporal alterations in phosphorylation of the PI3K/AKT/mTOR signaling pathway, which is important in determining cell death. This work paves the way for further research on how this pathway influences host mechanisms responsible for aiding virus replication and in determining levels and severity of influenza virus-induced patho
Collapse
Affiliation(s)
- Charlene Ranadheera
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J6, Canada; Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
| | - Kevin M Coombs
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J6, Canada; Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Avenue, Winnipeg, Manitoba R3E 3P4, Canada; Manitoba Institute of Child Health, John Buhler Research Centre, Room 513, 715 McDermot Avenue, Winnipeg, Manitoba R3E 3P4, Canada.
| | - Darwyn Kobasa
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J6, Canada; Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada.
| |
Collapse
|
24
|
Chiricozzi E, Loberto N, Schiumarini D, Samarani M, Mancini G, Tamanini A, Lippi G, Dechecchi MC, Bassi R, Giussani P, Aureli M. Sphingolipids role in the regulation of inflammatory response: From leukocyte biology to bacterial infection. J Leukoc Biol 2018; 103:445-456. [PMID: 29345379 DOI: 10.1002/jlb.3mr0717-269r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 10/13/2017] [Accepted: 11/07/2017] [Indexed: 12/14/2022] Open
Abstract
Sphingolipids (SLs) are amphiphilic molecules mainly associated with the external leaflet of eukaryotic plasma membrane, and are structural membrane components with key signaling properties. Since the beginning of the last century, a large number of papers described the involvement of these molecules in several aspects of cell physiology and pathology. Several lines of evidence support the critical role of SLs in inflammatory diseases, by acting as anti- or pro-inflammatory mediators. They are involved in control of leukocyte activation and migration, and are recognized as essential players in host response to pathogenic infection. We propose here a critical overview of current knowledge on involvement of different classes of SLs in inflammation, focusing on the role of simple and complex SLs in pathogen-mediated inflammatory response.
Collapse
Affiliation(s)
- Elena Chiricozzi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Nicoletta Loberto
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Domitilla Schiumarini
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Maura Samarani
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Giulia Mancini
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Anna Tamanini
- Laboratorio di Patologia Molecolare-Laboratorio Analisi, Dipartimento di Patologia e Diagnostica, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Giuseppe Lippi
- Sezione di Biochimica Clinica, Università degli Studi di Verona, Verona, Italy
| | - Maria Cristina Dechecchi
- Laboratorio di Patologia Molecolare-Laboratorio Analisi, Dipartimento di Patologia e Diagnostica, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Rosaria Bassi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Paola Giussani
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Massimo Aureli
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
25
|
Jie C, Luo Z, Chen H, Wang M, Yan C, Mao ZF, Xiao GK, Kurihara H, Li YF, He RR. Indirubin, a bisindole alkaloid from Isatis indigotica, reduces H1N1 susceptibility in stressed mice by regulating MAVS signaling. Oncotarget 2017; 8:105615-105629. [PMID: 29285277 PMCID: PMC5739664 DOI: 10.18632/oncotarget.22350] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 07/18/2017] [Indexed: 12/22/2022] Open
Abstract
Isatis indigotica has a long history in treating virus infection and related symptoms in China. Nevertheless, its antivirus evidence in animal studies is not satisfactory, which might be due to the lack of appropriate animal model. Previously, we had utilized restraint stress to establish mouse H1N1 susceptibility model which was helpful in evaluating the anti-virus effect of medicines targeting host factors, such as type I interferon production. In this study, this model was employed to investigate the effect and mechanism of indirubin, a natural bisindole alkaloid from Isatis indigotica, on influenza A virus susceptibility. In the in vitro study, the stress hormone corticosterone was used to simulate restraint stress. Our results demonstrated that indirubin decreased the susceptibility to influenza virus with lowered mortality and alleviated lung damage in restraint-stressed mice model. Moreover, indirubin promoted the expression of interferon-β and interferon inducible transmembrane 3. In addition, indirubin maintained the morphology and function of mitochondria following influenza A virus infection. Further study revealed that indirubin promoted interferon-β production through promoting mitochondrial antiviral signaling pathway. Our study indicated that indirubin could be a candidate for the therapy of influenza.
Collapse
Affiliation(s)
- Chong Jie
- Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China.,Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, China
| | - Zhuo Luo
- Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China.,Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, China
| | - Huan Chen
- Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China.,Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, China
| | - Min Wang
- Department of Pharmacy, Hainan General Hospital, Hainan 570311, China
| | - Chang Yan
- Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China.,Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, China
| | - Zhong-Fu Mao
- Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China.,Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, China
| | - Gao-Keng Xiao
- Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, China
| | - Hiroshi Kurihara
- Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China.,Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, China
| | - Yi-Fang Li
- Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China.,Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, China
| | - Rong-Rong He
- Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou 510632, China.,Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, China
| |
Collapse
|
26
|
Smee DF, Dagley A, Tarbet EB. Combinations of L-N G-monomethyl-arginine and oseltamivir against pandemic influenza A virus infections in mice. Antivir Chem Chemother 2017; 25:11-17. [PMID: 28417640 DOI: 10.1177/2040206617691885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
L-NG-monomethyl-arginine (L-NMMA) is an experimental compound that suppresses nitric oxide production in animals. The compound was combined with oseltamivir to treat lethal influenza A/California/04/2009 (H1N1) pandemic virus infections in mice. Treatments were given twice a day for five days starting 4 h (oseltamivir, by oral gavage) or three days (L-NMMA, by intraperitoneal route; corresponding to the time previously reported for nitric oxide induction in the animals) after infection. Low doses of oseltamivir were used in order to demonstrate synergy or antagonism. Oseltamivir monotherapy protected 70% of mice from death at 1 mg/kg/day. L-NMMA (40 and 80 mg/kg/day) was ineffective alone in preventing mortality. Compared to oseltamivir treatment alone, L-NMMA combined with oseltamivir was synergistically effective (as evaluated by three-dimensional MacSynergy analysis), resulting in survival increases from 20 to 70% when 40 or 80 mg/kg/day of L-NMMA was combined with 0.3 mg/kg/day of oseltamivir, and from 70 to 100% survival increases when these doses were combined with 1 mg/kg/day of oseltamivir. These data demonstrate that a nitric oxide inhibitor such as L-NMMA has the potential to be beneficial when combined with oseltamivir in treating influenza virus infections.
Collapse
Affiliation(s)
- Donald F Smee
- Department of Animal, Dairy and Veterinary Sciences, Institute for Antiviral Research, Utah State University, Logan, UT, USA
| | - Ashley Dagley
- Department of Animal, Dairy and Veterinary Sciences, Institute for Antiviral Research, Utah State University, Logan, UT, USA
| | - E B Tarbet
- Department of Animal, Dairy and Veterinary Sciences, Institute for Antiviral Research, Utah State University, Logan, UT, USA
| |
Collapse
|
27
|
Wang Y, Cheng T, Bryant SH. PubChem BioAssay: A Decade's Development toward Open High-Throughput Screening Data Sharing. SLAS DISCOVERY 2017; 22:655-666. [PMID: 28346087 PMCID: PMC5480605 DOI: 10.1177/2472555216685069] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
High-throughput screening (HTS) is now routinely conducted for drug discovery by both pharmaceutical companies and screening centers at academic institutions and universities. Rapid advance in assay development, robot automation, and computer technology has led to the generation of terabytes of data in screening laboratories. Despite the technology development toward HTS productivity, fewer efforts were devoted to HTS data integration and sharing. As a result, the huge amount of HTS data was rarely made available to the public. To fill this gap, the PubChem BioAssay database (https://www.ncbi.nlm.nih.gov/pcassay/) was set up in 2004 to provide open access to the screening results tested on chemicals and RNAi reagents. With more than 10 years’ development and contributions from the community, PubChem has now become the largest public repository for chemical structures and biological data, which provides an information platform to worldwide researchers supporting drug development, medicinal chemistry study, and chemical biology research. This work presents a review of the HTS data content in the PubChem BioAssay database and the progress of data deposition to stimulate knowledge discovery and data sharing. It also provides a description of the database’s data standard and basic utilities facilitating information access and use for new users.
Collapse
Affiliation(s)
- Yanli Wang
- 1 National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Tiejun Cheng
- 1 National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Stephen H Bryant
- 1 National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
28
|
14-Deoxy-11,12-didehydroandrographolide attenuates excessive inflammatory responses and protects mice lethally challenged with highly pathogenic A(H5N1) influenza viruses. Antiviral Res 2016; 133:95-105. [DOI: 10.1016/j.antiviral.2016.07.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 07/18/2016] [Accepted: 07/26/2016] [Indexed: 11/19/2022]
|
29
|
Zhu L, Yuan C, Ding X, Xu S, Yang J, Liang Y, Zhu Q. PLC-γ1 is involved in the inflammatory response induced by influenza A virus H1N1 infection. Virology 2016; 496:131-137. [PMID: 27310357 DOI: 10.1016/j.virol.2016.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/05/2016] [Accepted: 06/06/2016] [Indexed: 12/20/2022]
Abstract
We have previously reported that phosphoinositide-specific phospholipase γ1 (PLC-γ1) signaling is activated by influenza virus H1N1 infection and mediates efficient viral entry in human epithelial cells. In this study, we show that H1N1 also activates PLCγ-1 signaling in human promonocytic cell line -derived macrophages. Surprisingly, the activated PLCγ-1 signaling is not important for viral replication in macrophages, but is involved in the virus-induced inflammatory responses. PLC-γ1-specific inhibitor U73122 strongly inhibits the H1N1 virus-induced NF-κB signaling, blocking the up-regulation of TNF-α, IL-6, MIP-1α, and reactive oxidative species. In a positive feedback loop, IL-1β and TNF-α activate the PLCγ-1 signaling in both epithelial and macrophage cell lines. In summary, we have shown for the first time that the PLCγ-1 signaling plays an important role in the H1N1-induced inflammatory responses. Our study suggests that targeting the PLCγ-1 signaling is a potential antiviral therapy against H1N1 by inhibiting both viral replication and excessive inflammation.
Collapse
Affiliation(s)
- Liqian Zhu
- College of Veterinary Medicine, Yangzhou University and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 48 Wenhui East Road, Yangzhou 225009, China
| | - Chen Yuan
- College of Veterinary Medicine, Yangzhou University and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 48 Wenhui East Road, Yangzhou 225009, China
| | - Xiuyan Ding
- College of Veterinary Medicine, Yangzhou University and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 48 Wenhui East Road, Yangzhou 225009, China
| | - Shuai Xu
- The State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 Xujiaping, Chengguan District, Lanzhou 730046, Gansu, China
| | - Jiayun Yang
- The State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 Xujiaping, Chengguan District, Lanzhou 730046, Gansu, China
| | - Yuying Liang
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, MN 55108, USA.
| | - Qiyun Zhu
- The State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 Xujiaping, Chengguan District, Lanzhou 730046, Gansu, China.
| |
Collapse
|
30
|
S1PR1-mediated IFNAR1 degradation modulates plasmacytoid dendritic cell interferon-α autoamplification. Proc Natl Acad Sci U S A 2016; 113:1351-6. [PMID: 26787880 DOI: 10.1073/pnas.1525356113] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Blunting immunopathology without abolishing host defense is the foundation for safe and effective modulation of infectious and autoimmune diseases. Sphingosine 1-phosphate receptor 1 (S1PR1) agonists are effective in treating infectious and multiple autoimmune pathologies; however, mechanisms underlying their clinical efficacy are yet to be fully elucidated. Here, we uncover an unexpected mechanism of convergence between S1PR1 and interferon alpha receptor 1 (IFNAR1) signaling pathways. Activation of S1PR1 signaling by pharmacological tools or endogenous ligand sphingosine-1 phosphate (S1P) inhibits type 1 IFN responses that exacerbate numerous pathogenic conditions. Mechanistically, S1PR1 selectively suppresses the type I IFN autoamplification loop in plasmacytoid dendritic cells (pDCs), a specialized DC subset, for robust type I IFN release. S1PR1 agonist suppression is pertussis toxin-resistant, but inhibited by an S1PR1 C-terminal-derived transactivating transcriptional activator (Tat)-fusion peptide that blocks receptor internalization. S1PR1 agonist treatment accelerates turnover of IFNAR1, suppresses signal transducer and activator of transcription 1 (STAT1) phosphorylation, and down-modulates total STAT1 levels, thereby inactivating the autoamplification loop. Inhibition of S1P-S1PR1 signaling in vivo using the selective antagonist Ex26 significantly elevates IFN-α production in response to CpG-A. Thus, multiple lines of evidence demonstrate that S1PR1 signaling sets the sensitivity of pDC amplification of IFN responses, thereby blunting pathogenic immune responses. These data illustrate a lipid G-protein coupled receptor (GPCR)-IFNAR1 regulatory loop that balances effective and detrimental immune responses and elevated endogenous S1PR1 signaling. This mechanism will likely be advantageous in individuals subject to a range of inflammatory conditions.
Collapse
|
31
|
Blanc F, Furio L, Moisy D, Yen HL, Chignard M, Letavernier E, Naffakh N, Mok CKP, Si-Tahar M. Targeting host calpain proteases decreases influenza A virus infection. Am J Physiol Lung Cell Mol Physiol 2016; 310:L689-99. [PMID: 26747784 DOI: 10.1152/ajplung.00314.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/07/2016] [Indexed: 11/22/2022] Open
Abstract
Influenza A viruses (IAV) trigger contagious acute respiratory diseases. A better understanding of the molecular mechanisms of IAV pathogenesis and host immune responses is required for the development of more efficient treatments of severe influenza. Calpains are intracellular proteases that participate in diverse cellular responses, including inflammation. Here, we used in vitro and in vivo approaches to investigate the role of calpain signaling in IAV pathogenesis. Calpain expression and activity were found altered in IAV-infected bronchial epithelial cells. With the use of small-interfering RNA (siRNA) gene silencing, specific synthetic inhibitors of calpains, and mice overexpressing calpastatin, we found that calpain inhibition dampens IAV replication and IAV-triggered secretion of proinflammatory mediators and leukocyte infiltration. Remarkably, calpain inhibition has a protective impact in IAV infection, since it significantly reduced mortality of mice challenged not only by seasonal H3N2- but also by hypervirulent H5N1 IAV strains. Hence, our study suggests that calpains are promising therapeutic targets for treating IAV acute pneumonia.
Collapse
Affiliation(s)
- Fany Blanc
- Institut Pasteur, Unité de Défense Innée et Inflammation, Paris, France; Institut National de la Santé et de la Recherche Médicale U874, Paris, France
| | - Laetitia Furio
- Institut Pasteur, Unité de Défense Innée et Inflammation, Paris, France; Institut National de la Santé et de la Recherche Médicale U874, Paris, France
| | - Dorothée Moisy
- Institut Pasteur, Unité de Génétique Moléculaire des Virus ARN, Paris, France
| | - Hui-Ling Yen
- School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Michel Chignard
- Institut Pasteur, Unité de Défense Innée et Inflammation, Paris, France; Institut National de la Santé et de la Recherche Médicale U874, Paris, France
| | - Emmanuel Letavernier
- Institut National de la Santé et de la Recherche Médicale UMR-S1155, Paris, France
| | - Nadia Naffakh
- Institut Pasteur, Unité de Génétique Moléculaire des Virus ARN, Paris, France
| | - Chris Ka Pun Mok
- The HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Mustapha Si-Tahar
- Institut Pasteur, Unité de Défense Innée et Inflammation, Paris, France; Institut National de la Santé et de la Recherche Médicale U874, Paris, France; Institut National de la Santé et de la Recherche Médicale U1100, Centre d'Etude des Pathologies Respiratoires, Tours, France; and Université François Rabelais, Tours, France
| |
Collapse
|
32
|
McClain MT, Henao R, Williams J, Nicholson B, Veldman T, Hudson L, Tsalik EL, Lambkin-Williams R, Gilbert A, Mann A, Ginsburg GS, Woods CW. Differential evolution of peripheral cytokine levels in symptomatic and asymptomatic responses to experimental influenza virus challenge. Clin Exp Immunol 2015; 183:441-51. [PMID: 26506932 DOI: 10.1111/cei.12736] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 10/04/2015] [Accepted: 10/13/2015] [Indexed: 02/02/2023] Open
Abstract
Exposure to influenza virus triggers a complex cascade of events in the human host. In order to understand more clearly the evolution of this intricate response over time, human volunteers were inoculated with influenza A/Wisconsin/67/2005 (H3N2), and then had serial peripheral blood samples drawn and tested for the presence of 25 major human cytokines. Nine of 17 (53%) inoculated subjects developed symptomatic influenza infection. Individuals who will go on to become symptomatic demonstrate increased circulating levels of interleukin (IL)-6, IL-8, IL-15, monocyte chemotactic protein (MCP)-1 and interferon (IFN) gamma-induced protein (IP)-10 as early as 12-29 h post-inoculation (during the presymptomatic phase), whereas challenged patients who remain asymptomatic do not. Overall, the immunological pathways of leucocyte recruitment, Toll-like receptor (TLR)-signalling, innate anti-viral immunity and fever production are all over-represented in symptomatic individuals very early in disease, but are also dynamic and evolve continuously over time. Comparison with simultaneous peripheral blood genomics demonstrates that some inflammatory mediators (MCP-1, IP-10, IL-15) are being expressed actively in circulating cells, while others (IL-6, IL-8, IFN-α and IFN-γ) are probable effectors produced locally at the site of infection. Interestingly, asymptomatic exposed subjects are not quiescent either immunologically or genomically, but instead exhibit early and persistent down-regulation of important inflammatory mediators in the periphery. The host inflammatory response to influenza infection is variable but robust, and evolves over time. These results offer critical insight into pathways driving influenza-related symptomatology and offer the potential to contribute to early detection and differentiation of infected hosts.
Collapse
Affiliation(s)
- M T McClain
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.,Durham Veterans Affairs Medical Center, Durham, NC, USA.,Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | - R Henao
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.,Department of Electrical Engineering, Duke University, Durham, NC, USA
| | - J Williams
- Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - B Nicholson
- Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - T Veldman
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
| | - L Hudson
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
| | - E L Tsalik
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.,Durham Veterans Affairs Medical Center, Durham, NC, USA.,Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | | | | | | | - G S Ginsburg
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
| | - C W Woods
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.,Durham Veterans Affairs Medical Center, Durham, NC, USA.,Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
33
|
Scanlon KM, Skerry C, Carbonetti NH. Novel therapies for the treatment of pertussis disease. Pathog Dis 2015; 73:ftv074. [PMID: 26394802 PMCID: PMC4626598 DOI: 10.1093/femspd/ftv074] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/09/2015] [Accepted: 09/16/2015] [Indexed: 12/13/2022] Open
Abstract
Whooping cough, or pertussis, incidence has reached levels not seen since the 1950s. Previous studies have shown that antibiotics fail to improve the course of disease unless diagnosed early. Early diagnosis is complicated by the non-diagnostic presentation of disease early in infection. This review focuses on previous attempts at developing novel host-directed therapies for the treatment of pertussis. In addition, two novel approaches from our group are discussed. Manipulation of the signaling pathway of sphingosine-1-phosphate, a lipid involved in many immune processes, has shown great promise, but is in its infancy. Pendrin, a host epithelial anion exchanger upregulated in the airways with B. pertussis infection, appears to drive mucus production and dysregulation of airway surface liquid pH and salinity. In addition to detailing these potential new therapeutic targets, the need for greater focus on the neonatal model of disease is highlighted.
Collapse
Affiliation(s)
- Karen M Scanlon
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD 21201, USA
| | - Ciaran Skerry
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD 21201, USA
| | - Nicholas H Carbonetti
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD 21201, USA
| |
Collapse
|
34
|
Ramos I, Fernandez-Sesma A. Modulating the Innate Immune Response to Influenza A Virus: Potential Therapeutic Use of Anti-Inflammatory Drugs. Front Immunol 2015; 6:361. [PMID: 26257731 PMCID: PMC4507467 DOI: 10.3389/fimmu.2015.00361] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/04/2015] [Indexed: 12/27/2022] Open
Abstract
Infection by influenza A viruses (IAV) is frequently characterized by robust inflammation that is usually more pronounced in the case of avian influenza. It is becoming clearer that the morbidity and pathogenesis caused by IAV are consequences of this inflammatory response, with several components of the innate immune system acting as the main players. It has been postulated that using a therapeutic approach to limit the innate immune response in combination with antiviral drugs has the potential to diminish symptoms and tissue damage caused by IAV infection. Indeed, some anti-inflammatory agents have been shown to be effective in animal models in reducing IAV pathology as a proof of principle. The main challenge in developing such therapies is to selectively modulate signaling pathways that contribute to lung injury while maintaining the ability of the host cells to mount an antiviral response to control virus replication. However, the dissection of those pathways is very complex given the numerous components regulated by the same factors (i.e., NF kappa B transcription factors) and the large number of players involved in this regulation, some of which may be undescribed or unknown. This article provides a comprehensive review of the current knowledge regarding the innate immune responses associated with tissue damage by IAV infection, the understanding of which is essential for the development of effective immunomodulatory drugs. Furthermore, we summarize the recent advances on the development and evaluation of such drugs as well as the lessons learned from those studies.
Collapse
Affiliation(s)
- Irene Ramos
- Department of Microbiology, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| | - Ana Fernandez-Sesma
- Department of Microbiology, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| |
Collapse
|
35
|
Schreiber SL, Kotz JD, Li M, Aubé J, Austin CP, Reed JC, Rosen H, White EL, Sklar LA, Lindsley CW, Alexander BR, Bittker JA, Clemons PA, de Souza A, Foley MA, Palmer M, Shamji AF, Wawer MJ, McManus O, Wu M, Zou B, Yu H, Golden JE, Schoenen FJ, Simeonov A, Jadhav A, Jackson MR, Pinkerton AB, Chung TDY, Griffin PR, Cravatt BF, Hodder PS, Roush WR, Roberts E, Chung DH, Jonsson CB, Noah JW, Severson WE, Ananthan S, Edwards B, Oprea TI, Conn PJ, Hopkins CR, Wood MR, Stauffer SR, Emmitte KA. Advancing Biological Understanding and Therapeutics Discovery with Small-Molecule Probes. Cell 2015; 161:1252-65. [PMID: 26046436 PMCID: PMC4564295 DOI: 10.1016/j.cell.2015.05.023] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Indexed: 02/06/2023]
Abstract
Small-molecule probes can illuminate biological processes and aid in the assessment of emerging therapeutic targets by perturbing biological systems in a manner distinct from other experimental approaches. Despite the tremendous promise of chemical tools for investigating biology and disease, small-molecule probes were unavailable for most targets and pathways as recently as a decade ago. In 2005, the NIH launched the decade-long Molecular Libraries Program with the intent of innovating in and broadening access to small-molecule science. This Perspective describes how novel small-molecule probes identified through the program are enabling the exploration of biological pathways and therapeutic hypotheses not otherwise testable. These experiences illustrate how small-molecule probes can help bridge the chasm between biological research and the development of medicines but also highlight the need to innovate the science of therapeutic discovery.
Collapse
Affiliation(s)
- Stuart L Schreiber
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Joanne D Kotz
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Min Li
- Johns Hopkins School of Medicine Ion Channel Center, Baltimore, MD 21205, USA
| | - Jeffrey Aubé
- University of Kansas Specialized Chemistry Center, Lawrence, KS 66045, USA; Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, 66045, USA
| | - Christopher P Austin
- NIH Chemical Genomics Center, National Institutes of Health, Rockville, MD 20850, USA; National Center for Advancing Translational Sciences, Bethesda, MD 20892, USA
| | - John C Reed
- Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, and Lake Nona, FL 32827, USA
| | - Hugh Rosen
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA; Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - E Lucile White
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - Larry A Sklar
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM 87131, USA; Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Craig W Lindsley
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Benjamin R Alexander
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Joshua A Bittker
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Development of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Paul A Clemons
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Andrea de Souza
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Michael A Foley
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Michelle Palmer
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Alykhan F Shamji
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Mathias J Wawer
- Probe Development Center, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Owen McManus
- Johns Hopkins School of Medicine Ion Channel Center, Baltimore, MD 21205, USA
| | - Meng Wu
- Johns Hopkins School of Medicine Ion Channel Center, Baltimore, MD 21205, USA
| | - Beiyan Zou
- Johns Hopkins School of Medicine Ion Channel Center, Baltimore, MD 21205, USA
| | - Haibo Yu
- Johns Hopkins School of Medicine Ion Channel Center, Baltimore, MD 21205, USA
| | - Jennifer E Golden
- University of Kansas Specialized Chemistry Center, Lawrence, KS 66045, USA
| | - Frank J Schoenen
- University of Kansas Specialized Chemistry Center, Lawrence, KS 66045, USA
| | - Anton Simeonov
- NIH Chemical Genomics Center, National Institutes of Health, Rockville, MD 20850, USA; National Center for Advancing Translational Sciences, Bethesda, MD 20892, USA
| | - Ajit Jadhav
- NIH Chemical Genomics Center, National Institutes of Health, Rockville, MD 20850, USA; National Center for Advancing Translational Sciences, Bethesda, MD 20892, USA
| | - Michael R Jackson
- Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, and Lake Nona, FL 32827, USA
| | - Anthony B Pinkerton
- Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, and Lake Nona, FL 32827, USA
| | - Thomas D Y Chung
- Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, and Lake Nona, FL 32827, USA
| | - Patrick R Griffin
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA; Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Benjamin F Cravatt
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA; Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Peter S Hodder
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA
| | - William R Roush
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA; Department of Chemistry, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Edward Roberts
- Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, and Jupiter, FL 33458, USA
| | - Dong-Hoon Chung
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - Colleen B Jonsson
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - James W Noah
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - William E Severson
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - Subramaniam Ananthan
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, AL 35205, USA
| | - Bruce Edwards
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM 87131, USA; Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Tudor I Oprea
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM 87131, USA; Department of Internal Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - P Jeffrey Conn
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Corey R Hopkins
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Michael R Wood
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Shaun R Stauffer
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kyle A Emmitte
- The Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| |
Collapse
|
36
|
Gonzalez-Cabrera PJ, Brown S, Studer SM, Rosen H. S1P signaling: new therapies and opportunities. F1000PRIME REPORTS 2014; 6:109. [PMID: 25580263 PMCID: PMC4251414 DOI: 10.12703/p6-109] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Development of sphingosine-1-phosphate receptor 1 (S1P1) modulators to dampen inflammation and its sequelae is becoming increasingly promising for treating medical conditions characterized by significant immunopathology. As shown by the non-selective S1P receptor modulator FTY720 (fingolimod [Gilenya(®)]) in the treatment of relapsing-remitting multiple sclerosis (MS), the ability to use S1P1 modulation to precisely block immune cell traffic-immunomodulation-while maintaining immunosurveillance, has opened therapeutic opportunities in various other immune-derived chronic pathologies, including inflammatory bowel disease (IBD), lupus, psoriasis, as well as, potentially, in early acute viral respiratory infection. Proof-of-concept studies across validated animal models with S1P receptor modulators highly selective for S1P1, such as BAF-312 (Siponimod), KRP-203, ONO-4641 (Ceralifimod), ponesimod and RPC-1063, and emerging clinical trials for safety and efficacy in humans, particularly in MS, ulcerative colitis (UC) and psoriasis, have set the stage for us to consider additional testing in various other autoimmune diseases.
Collapse
|
37
|
Gomes L, Fernando S, Fernando RH, Wickramasinghe N, Shyamali NLA, Ogg GS, Malavige GN. Sphingosine 1-phosphate in acute dengue infection. PLoS One 2014; 9:e113394. [PMID: 25409037 PMCID: PMC4237441 DOI: 10.1371/journal.pone.0113394] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/23/2014] [Indexed: 12/26/2022] Open
Abstract
Background Vascular leak is the hallmark of severe dengue infections and leads to complications such as shock and multi-organ failure. Although many mediators have been implicated in the vascular leak in dengue, the role of sphingosine 1-phosphate (S1P) has not been investigated. Metholodology/Principal findings As S1P has been shown to be important in barrier integrity, we assessed the S1P levels in 28 patients with acute dengue and 12 healthy individuals. The S1P levels were significantly lower in patients with acute dengue (p = 0.002) and the levels in patients with grade IV dengue haemorrhagic fever (DHF) were significantly lower than those with dengue fever (p = 0.005). We then investigated the kinetics of S1P levels throughout the course of the illness in another 32 patients in serum samples obtained twice a day. We found that S1P levels were low throughout the course of illness and S1P levels were <0.5 µM in 12/23 patients with DHF when compared to 1/9 with DF. Conclusions/Significance As S1P has shown to be important in the endothelial barrier integrity and increases transendothelial resistance, low levels of S1P in acute dengue infection are likely to contribute to increased vascular permeability.
Collapse
Affiliation(s)
- Laksiri Gomes
- Centre for Dengue Research, Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayawardanapura, Gangodawila, Sri Lanka
| | - Samitha Fernando
- Centre for Dengue Research, Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayawardanapura, Gangodawila, Sri Lanka
| | - Randika Heshan Fernando
- Centre for Dengue Research, Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayawardanapura, Gangodawila, Sri Lanka
| | - Nilanka Wickramasinghe
- Centre for Dengue Research, Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayawardanapura, Gangodawila, Sri Lanka
| | - Narangoda Liyanage Ajantha Shyamali
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford NIHR Biomedical Research Centre and University of Oxford, Oxford, United Kingdom
| | - Graham S Ogg
- Department of Dermatology, Churchill Hospital, Oxford, United Kingdom
| | - Gathsaurie Neelika Malavige
- Centre for Dengue Research, Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayawardanapura, Gangodawila, Sri Lanka; Department of Dermatology, Churchill Hospital, Oxford, United Kingdom
| |
Collapse
|
38
|
Animal model of respiratory syncytial virus: CD8+ T cells cause a cytokine storm that is chemically tractable by sphingosine-1-phosphate 1 receptor agonist therapy. J Virol 2014; 88:6281-93. [PMID: 24672024 DOI: 10.1128/jvi.00464-14] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED The cytokine storm is an intensified, dysregulated, tissue-injurious inflammatory response driven by cytokine and immune cell components. The cytokine storm during influenza virus infection, whereby the amplified innate immune response is primarily responsible for pulmonary damage, has been well characterized. Now we describe a novel event where virus-specific T cells induce a cytokine storm. The paramyxovirus pneumonia virus of mice (PVM) is a model of human respiratory syncytial virus (hRSV). Unexpectedly, when C57BL/6 mice were infected with PVM, the innate inflammatory response was undetectable until day 5 postinfection, at which time CD8(+) T cells infiltrated into the lung, initiating a cytokine storm by their production of gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α). Administration of an immunomodulatory sphingosine-1-phosphate (S1P) receptor 1 (S1P1R) agonist significantly inhibited PVM-elicited cytokine storm by blunting the PVM-specific CD8(+) T cell response, resulting in diminished pulmonary disease and enhanced survival. IMPORTANCE A dysregulated overly exuberant immune response, termed a "cytokine storm," accompanies virus-induced acute respiratory diseases (VARV), is primarily responsible for the accompanying high morbidity and mortality, and can be controlled therapeutically in influenza virus infection of mice and ferrets by administration of sphingosine-1-phosphate 1 receptor (S1P1R) agonists. Here, two novel findings are recorded. First, in contrast to influenza infection, where the cytokine storm is initiated early by the innate immune system, for pneumonia virus of mice (PVM), a model of RSV, the cytokine storm is initiated late in infection by the adaptive immune response: specifically, by virus-specific CD8 T cells via their release of IFN-γ and TNF-α. Blockading these cytokines with neutralizing antibodies blunts the cytokine storm and protects the host. Second, PVM infection is controlled by administration of an S1P1R agonist.
Collapse
|
39
|
Kash JC, Xiao Y, Davis AS, Walters KA, Chertow DS, Easterbrook JD, Dunfee RL, Sandouk A, Jagger BW, Schwartzman LM, Kuestner RE, Wehr NB, Huffman K, Rosenthal RA, Ozinsky A, Levine RL, Doctrow SR, Taubenberger JK. Treatment with the reactive oxygen species scavenger EUK-207 reduces lung damage and increases survival during 1918 influenza virus infection in mice. Free Radic Biol Med 2014; 67:235-47. [PMID: 24140866 PMCID: PMC3927540 DOI: 10.1016/j.freeradbiomed.2013.10.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 10/07/2013] [Accepted: 10/08/2013] [Indexed: 12/16/2022]
Abstract
The 1918 influenza pandemic caused over 40 million deaths worldwide, with 675,000 deaths in the United States alone. Studies in several experimental animal models showed that 1918 influenza virus infection resulted in severe lung pathology associated with dysregulated immune and cell death responses. To determine if reactive oxygen species produced by host inflammatory responses play a central role in promoting severity of lung pathology, we treated 1918 influenza virus-infected mice with the catalytic catalase/superoxide dismutase mimetic, salen-manganese complex EUK-207 beginning 3 days postinfection. Postexposure treatment of mice infected with a lethal dose of the 1918 influenza virus with EUK-207 resulted in significantly increased survival and reduced lung pathology without a reduction in viral titers. In vitro studies also showed that EUK-207 treatment did not affect 1918 influenza viral replication. Immunohistochemical analysis showed a reduction in the detection of the apoptosis marker cleaved caspase-3 and the oxidative stress marker 8-oxo-2'-deoxyguanosine in lungs of EUK-207-treated animals compared to vehicle controls. High-throughput sequencing and RNA expression microarray analysis revealed that treatment resulted in decreased expression of inflammatory response genes and increased lung metabolic and repair responses. These results directly demonstrate that 1918 influenza virus infection leads to an immunopathogenic immune response with excessive inflammatory and cell death responses that can be limited by treatment with the catalytic antioxidant EUK-207.
Collapse
Affiliation(s)
- John C Kash
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Yongli Xiao
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - A Sally Davis
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Daniel S Chertow
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Judith D Easterbrook
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rebecca L Dunfee
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aline Sandouk
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brett W Jagger
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Louis M Schwartzman
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Nancy B Wehr
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Karl Huffman
- Pulmonary Center, Department of Medicine, Boston University Medical School, Boston, MA 02118, USA
| | - Rosalind A Rosenthal
- Pulmonary Center, Department of Medicine, Boston University Medical School, Boston, MA 02118, USA
| | | | - Rodney L Levine
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Susan R Doctrow
- Pulmonary Center, Department of Medicine, Boston University Medical School, Boston, MA 02118, USA
| | - Jeffery K Taubenberger
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
40
|
Vijayan M, Hahm B. Influenza viral manipulation of sphingolipid metabolism and signaling to modulate host defense system. SCIENTIFICA 2014; 2014:793815. [PMID: 24672735 PMCID: PMC3920843 DOI: 10.1155/2014/793815] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 12/24/2013] [Indexed: 06/03/2023]
Abstract
Viruses attempt to create a distinctive cellular environment to favor viral replication and spread. Recent studies uncovered new functions of the sphingolipid signaling/metabolism during pathogenic virus infections. While sphingolipids such as sphingomyelin and ceramide were reported to influence the entry step of several viruses, sphingolipid-metabolizing enzymes could directly alter viral replication processes. Influenza virus was shown to increase the level of sphingosine kinase (SK) 1 to promote virus propagation. The mechanism involves regulation of intracellular signaling pathways, leading to the amplification of influenza viral RNA synthesis and nuclear export of viral ribonucleoprotein (RNP) complex. However, bovine viral diarrhea virus inhibits SK1 to enhance the efficacy of virus replication, demonstrating the presence of virus-specific strategies for modulation of the sphingolipid system. Therefore, investigating the sphingolipid metabolism and signaling in the context of virus replication could help us design innovative therapeutic approaches to improve human health.
Collapse
Affiliation(s)
- Madhuvanthi Vijayan
- Departments of Surgery and Molecular Microbiology & Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Bumsuk Hahm
- Departments of Surgery and Molecular Microbiology & Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
- University of Missouri-Columbia, One Hospital Drive, Medical Sciences Building, NW301C, Columbia, MO 65212, USA
| |
Collapse
|
41
|
Abstract
The understanding of the role of the sphingosine 1-phosphate signaling system in immunology and host defense has deepened exponentially over the past 12 years since the discovery that lymphocyte egress was reversibly modulated by sphingosine 1-phosphate receptors, and with the development of fingolimod, a prodrug of a nonselective S1P receptor agonist, for therapeutic use in the treatment of relapsing, remitting multiple sclerosis. Innovative genetic and chemical approaches, together with structural biology, now provide a more detailed molecular understanding of a regulated lysophospholipid ligand with a variety of autocrine, paracrine, and systemic effects in physiology and pathology, based upon selective interactions with a high affinity and selective evolutionary cluster of G-protein-coupled receptors.
Collapse
|
42
|
Tong S, Tian J, Wang H, Huang Z, Yu M, Sun L, Liu R, Liao M, Ning Z. H9N2 avian influenza infection altered expression pattern of sphiogosine-1-phosphate receptor 1 in BALB/c mice. Virol J 2013; 10:296. [PMID: 24073762 PMCID: PMC3849581 DOI: 10.1186/1743-422x-10-296] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 09/24/2013] [Indexed: 01/30/2023] Open
Abstract
Background The pathological damage inflicted by virulent AIV strains is often caused by inducing a positive feedback loop of cytokines in immune cells that cause excessive inflammation. Previous research has shown that a G protein-coupled receptor, sphingosine-1-phosphate receptor 1 (S1PR1), plays a crucial role in the development of excessive inflammation in influenza virus infection (Cell 146:861–862, 2011; Cell 146:980–991, 2011). BALB/c mice are common laboratory animals used in research of influenza virus; however the effects of influenza infections on expression patterns of S1PR1 in mice are unknown. Methods We investigated the expression patterns of S1PR1 in normal BALB/c mice and those infected by two distinct H9N2 AIV strains, one (A/chicken/Guangdong/V/2008,V) highly pathogenic, and the other (A/chicken/Guangdong/Ts/2004,Ts), non-pathogenic in mice, using quantitative PCR and immunohistochemistry (IHC) to detect S1PR1 mRNA and protein, respectively. Results S1PR1 mRNA was ubiquitously expressed in all the tissues examined, and significant differences were seen in mRNA expression between infected Ts, V and control mice in detected tissues, heart, liver, spleen, kidney and brain. S1PR1 protein was expressed in the cytoplasm and also demonstrated quantitative changes in expression in the various tissues between mice infected with the two strains of AIV. Conclusions Our results provided the first look at differences in S1PR1 expression patterns in BALB/c mice infected by non-pathogenic and highly pathogenic H9N2 influenza viruses. This information will not only be helpful in designing experiments to better understand the role of S1PR1 in virus-host interactions but also in developing novel anti-influenza agents to minimize the mortality and morbidity associated with highly virulent strains in avian and human populations.
Collapse
Affiliation(s)
- Shuang Tong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, People's Republic of China.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Armstrong SM, Darwish I, Lee WL. Endothelial activation and dysfunction in the pathogenesis of influenza A virus infection. Virulence 2013; 4:537-42. [PMID: 23863601 PMCID: PMC5359731 DOI: 10.4161/viru.25779] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The development of severe influenza has been attributed, in part, to a heightened innate immune response. Recent evidence suggests that endothelial activation, loss of barrier function, and consequent microvascular leak may also serve important mechanistic roles in the pathogenesis of severe influenza. The aim of this review is to summarize the current evidence in support of endothelial activation and dysfunction as a central feature preceding the development of severe influenza. We also discuss the effect of influenza on platelet–endothelial interactions.
Collapse
|
44
|
Darwish I, Liles WC. Emerging therapeutic strategies to prevent infection-related microvascular endothelial activation and dysfunction. Virulence 2013; 4:572-82. [PMID: 23863603 PMCID: PMC5359747 DOI: 10.4161/viru.25740] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Recent evidence suggests that loss of endothelial barrier function and resulting microvascular leak play important mechanistic roles in the pathogenesis of infection-related end-organ dysfunction and failure. Several distinct therapeutic strategies, designed to prevent or limit infection-related microvascular endothelial activation and permeability, thereby mitigating end-organ injury/dysfunction, have recently been investigated in pre-clinical models. In this review, these potential therapeutic strategies, namely, VEGFR2/Src antagonists, sphingosine-1-phosphate agonists, fibrinopeptide Bβ15–42, slit2N, secinH3, angiopoietin-1/tie-2 agonists, angiopoietin-2 antagonists, statins, atrial natriuretic peptide, and mesenchymal stromal (stem) cells, are discussed in terms of their translational potential for the management of clinical infectious diseases.
Collapse
Affiliation(s)
- Ilyse Darwish
- University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | | |
Collapse
|
45
|
Boonnak K, Vogel L, Orandle M, Zimmerman D, Talor E, Subbarao K. Antigen-activated dendritic cells ameliorate influenza A infections. J Clin Invest 2013; 123:2850-61. [PMID: 23934125 DOI: 10.1172/jci67550] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 04/11/2013] [Indexed: 02/06/2023] Open
Abstract
Influenza A viruses cause significant morbidity and mortality worldwide. There is a need for alternative or adjunct therapies, as resistance to currently used antiviral drugs is emerging rapidly. We tested ligand epitope antigen presentation system (LEAPS) technology as a new immune-based treatment for influenza virus infection in a mouse model. Influenza-J-LEAPS peptides were synthesized by conjugating the binding ligand derived from the β2-microglobulin chain of the human MHC class I molecule (J-LEAPS) with 15 to 30 amino acid-long peptides derived from influenza virus NP, M, or HA proteins. DCs were stimulated with influenza-J-LEAPS peptides (influenza-J-LEAPS) and injected intravenously into infected mice. Antigen-specific LEAPS-stimulated DCs were effective in reducing influenza virus replication in the lungs and enhancing survival of infected animals. Additionally, they augmented influenza-specific T cell responses in the lungs and reduced the severity of disease by limiting excessive cytokine responses, which are known to contribute to morbidity and mortality following influenza virus infection. Our data demonstrate that influenza-J-LEAPS-pulsed DCs reduce virus replication in the lungs, enhance survival, and modulate the protective immune responses that eliminate the virus while preventing excessive cytokines that could injure the host. This approach shows promise as an adjunct to antiviral treatment of influenza virus infections.
Collapse
Affiliation(s)
- Kobporn Boonnak
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892-3203, USA
| | | | | | | | | | | |
Collapse
|
46
|
Relationships between IL-17(+) subsets, Tregs and pDCs that distinguish among SIV infected elite controllers, low, medium and high viral load rhesus macaques. PLoS One 2013; 8:e61264. [PMID: 23620737 PMCID: PMC3631185 DOI: 10.1371/journal.pone.0061264] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/07/2013] [Indexed: 01/13/2023] Open
Abstract
Comprehensive studies of the frequencies and absolute numbers of the various cell lineages that synthesize IL-17 in the blood and corresponding gastrointestinal (GI) tissues, their correlation with CD4(+) Tregs, CD8(+) Tregs, total and IFN-α synthesizing plasmacytoid dendritic cells (pDC) relative to plasma viral load in SIV infection has been lacking. The unique availability of SIV infected rhesus macaques (RM) classified as Elite Controllers (EC), and those with Low, Intermediate and High Viral Loads (HVL) provided a unique opportunity to address this issue. Results of these studies showed that EC demonstrated a remarkable ability to reverse changes that are induced acutely by SIV in the various cell lineages. Highlights of the differences between EC and HVL RM within Gastro-intestinal tissues (GIT) was the maintenance and/or increases in the levels of IL-17 synthesizing CD4, CD8, and NK cells and pDCs associated with slight decreases in the levels of CD4(+) Tregs and IFN-α synthesizing pDCs in EC as compared with decreases in the levels of IL-17 synthesizing CD4, CD8 and NK cells associated with increases in pDCs and IFN-α synthesizing pDCs in HVL monkeys. A previously underappreciated role for CD8(+) Tregs was also noted with a moderate increase in ECs but further increases of CD8(+) Tregs with increasing VL in viremic monkeys. Positive correlations between plasma VL and decreases in the levels of Th17, Tc17, NK-17, CD4(+) Tregs and increases in the levels of CD8(+) Tregs, total and IFN-α synthesizing pDCs were also noted. This study also identified 2 additional IL-17(+) subsets in GIT as CD3(-/)CD8(+)/NKG2a(-) and CD3(+)/CD8(+)/NKG2a(+) subsets. Studies also suggest a limited role for IFN-α synthesizing pDCs in chronic immune activation despite persistent up-regulation of ISGs. Finally, elevated persistent innate immune responses appear associated with poor prognosis. These findings provide an initial foundation for markers important to follow for vaccine design.
Collapse
|
47
|
Nicholls JM. The battle between influenza and the innate immune response in the human respiratory tract. Infect Chemother 2013; 45:11-21. [PMID: 24265946 PMCID: PMC3780943 DOI: 10.3947/ic.2013.45.1.11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Indexed: 12/23/2022] Open
Abstract
Influenza is a viral infection of the respiratory tract. Infection is normally confined to the upper respiratory tract but certain viral strains have evolved the ability to infect the lower respiratory tract, including the alveoli, leading to inflammation and a disease pattern of diffuse alveolar damage. Factors leading to this sequence of events are novel influenza strains, or strains that have viral proteins, in particular the NS1 protein that allow it to escape the innate immune system. There are three main barriers that prevent infection of pneumocytes - mucin, host defence lectins and cells such as macrophages. Viruses have developed strategies such as neuraminidase and glycosylation patterns that allow this evasion. Though there has been much investment in antiviral drugs, it is proposed that more attention should be directed towards developing or utilizing compounds that enhance the ability of the innate immune system to combat viral infection.
Collapse
Affiliation(s)
- John M Nicholls
- Department of Pathology, Hong Kong University, Hong Kong, Hong Kong
| |
Collapse
|
48
|
Matheu MP, Teijaro JR, Walsh KB, Greenberg ML, Marsolais D, Parker I, Rosen H, Oldstone MBA, Cahalan MD. Three phases of CD8 T cell response in the lung following H1N1 influenza infection and sphingosine 1 phosphate agonist therapy. PLoS One 2013; 8:e58033. [PMID: 23533579 PMCID: PMC3606384 DOI: 10.1371/journal.pone.0058033] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/29/2013] [Indexed: 12/13/2022] Open
Abstract
Influenza-induced lung edema and inflammation are exacerbated by a positive feedback loop of cytokine and chemokine production termed a 'cytokine storm', a hallmark of increased influenza-related morbidity and mortality. Upon infection, an immune response is rapidly initiated in the lungs and draining lymph node, leading to expansion of virus-specific effector cells. Using two-photon microscopy, we imaged the dynamics of dendritic cells (DC) and virus-specific eGFP(+)CD8(+) T cells in the lungs and draining mediastinal lymph nodes during the first two weeks following influenza infection. Three distinct phases of T cell and CD11c(+) DC behavior were revealed: 1) Priming, facilitated by the arrival of lung DCs in the lymph node and characterized by antigen recognition and expansion of antigen-specific CD8(+) T cells; asymmetric T cell division in contact with DCs was frequently observed. 2) Clearance, during which DCs re-populate the lung and T cells leave the draining lymph node and re-enter the lung tissue where enlarged, motile T cells come into contact with DCs and form long-lived interactions. 3) Maintenance, characterized by T-cell scanning of the lung tissue and dissociation from local antigen presenting cells; the T cells spend less time in association with DCs and migrate rapidly on collagen. A single dose of a sphingosine-1-phosphate receptor agonist, AAL-R, sufficient to suppress influenza-induced cytokine-storm, altered T cell and DC behavior during influenza clearance, delaying T cell division, cellular infiltration in the lung, and suppressing T-DC interactions in the lung. Our results provide a detailed description of T cell and DC choreography and dynamics in the lymph node and the lung during influenza infection. In addition, we suggest that phase lags in T cell and DC dynamics induced by targeting S1P receptors in vivo may attenuate the intensity of the immune response and can be manipulated for therapeutic benefit.
Collapse
Affiliation(s)
- Melanie P. Matheu
- Department of Physiology and Biophysics and the Center for Immunology, University of California Irvine, Irvine, California, United States of America
| | - John R. Teijaro
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Kevin B. Walsh
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Milton L. Greenberg
- Department of Physiology and Biophysics and the Center for Immunology, University of California Irvine, Irvine, California, United States of America
| | - David Marsolais
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
- Department of Medicine, Faculty of Medicine, Laval University; IUCPQ Research Center, Québec, Québec, Canada
| | - Ian Parker
- Department of Physiology and Biophysics and the Center for Immunology, University of California Irvine, Irvine, California, United States of America
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, United States of America
| | - Hugh Rosen
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Michael B A. Oldstone
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Michael D. Cahalan
- Department of Physiology and Biophysics and the Center for Immunology, University of California Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
49
|
Rosen H, Stevens RC, Hanson M, Roberts E, Oldstone MBA. Sphingosine-1-phosphate and its receptors: structure, signaling, and influence. Annu Rev Biochem 2013; 82:637-62. [PMID: 23527695 DOI: 10.1146/annurev-biochem-062411-130916] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The sphingosine-1-phosphate (S1P) receptor signaling system has biological and medical importance and is the first lipid G protein-coupled receptor (GPCR) structure to be solved to 2.8-Å resolution. S1P binds to five high-affinity GPCRs generating multiple downstream signals that play essential roles in vascular development and endothelial integrity, control of cardiac rhythm, and routine oral treatment of multiple sclerosis. Genetics, chemistry, and now structural biology have advanced this integrated biochemical system. The S1P receptors have a novel N-terminal fold that occludes access to the binding pocket from the extracellular environment as well as orthosteric and bitopic ligands with very different physicochemical properties. S1P receptors and metabolizing enzymes have been deleted, inducibly deleted, and knocked in as tagged or altered receptors in mice. An array of genetic models allows analysis of integrated receptor function in vivo. We can now directly understand causal relationships among protein expression, signal, and control points in physiology and pathology.
Collapse
Affiliation(s)
- Hugh Rosen
- Department of Chemical Physiology and Immunology, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | | | | | |
Collapse
|
50
|
Abstract
The substantial increase in the worldwide prevalence of asthma and atopy has been attributed to lifestyle changes that reduce exposure to bacteria. A recent insight is that the largely bacterial microbiome maintains a state of basal immune homoeostasis, which modulates immune responses to microbial pathogens. However, some respiratory viral infections cause bronchiolitis of infancy and childhood wheeze, and can exacerbate established asthma; whereas allergens can partly mimic infectious agents. New insights into the host’s innate sensing systems, combined with recently developed methods that characterise commensal and pathogenic microbial exposure, now allow a unified theory for how microbes cause mucosal inflammation in asthma. The respiratory mucosa provides a key microbial interface where epithelial and dendritic cells interact with a range of functionally distinct lymphocytes. Lymphoid cells then control a range of pathways, both innate and specific, which organise the host mucosal immune response. Fundamental to innate immune responses to microbes are the interactions between pathogen-associated molecular patterns and pattern recognition receptors, which are associated with production of type I interferons, proinflammatory cytokines, and the T-helper-2 cell pathway in predisposed people. These coordinated, dynamic immune responses underlie the differing asthma phenotypes, which we delineate in terms of Seven Ages of Asthma. An understanding of the role of microbes in the atopic march towards asthma, and in causing exacerbations of established asthma, provides the rationale for new specific treatments that can be assessed in clinical trials. On the basis of these new ideas, specific host biomarkers might then allow personalised treatment to become a reality for patients with asthma.
Collapse
Affiliation(s)
- Trevor T Hansel
- National Heart and Lung Institute, Centre for Respiratory Infection, MRC, London, UK.
| | | | | |
Collapse
|