1
|
Gerber-Tichet E, Blanchet FP, Majzoub K, Kremer EJ. Toll-like receptor 4 - a multifunctional virus recognition receptor. Trends Microbiol 2024:S0966-842X(24)00171-9. [PMID: 39179422 DOI: 10.1016/j.tim.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/30/2024] [Accepted: 07/02/2024] [Indexed: 08/26/2024]
Abstract
Since the initial description of Toll receptors in Drosophila and their mammalian counterparts Toll-like receptors (TLRs), numerous fundamental and applied studies have explored their crucial role as sensors of pathogen-associated molecular patterns (PAMPs). Among the ten human TLRs, TLR4 is particularly well known for its ability to detect lipopolysaccharides (LPS), a component of the Gram-negative bacterial cell wall. In addition to its archetypal functions, TLR4 is also a versatile virus sensor. This review provides a background on the discovery of TLR4 and how this knowledge laid a foundation for characterization of its diverse roles in antiviral responses, examined through genetic, biochemical, structural, and immunological approaches. These advances have led to a deeper understanding of the molecular functions that enable TLR4 to orchestrate multi-nodal control by professional antigen-presenting cells (APCs) to initiate appropriate and regulated antiviral immune responses.
Collapse
Affiliation(s)
- Elina Gerber-Tichet
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS UMR 5535, 34090 Montpellier, France
| | - Fabien P Blanchet
- Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, CNRS UMR 9004, 34090 Montpellier, France
| | - Karim Majzoub
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS UMR 5535, 34090 Montpellier, France
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS UMR 5535, 34090 Montpellier, France.
| |
Collapse
|
2
|
Zou Y, Wang X, Chen P, Zheng Z, Li X, Chen Z, Guo M, Zhou Y, Sun C, Wang R, Zhu W, Zheng P, Cho WJ, Cho YC, Liang G, Tang Q. Fragment-Based Anti-inflammatory Agent Design and Target Identification: Discovery of AF-45 as an IRAK4 Inhibitor to Treat Ulcerative Colitis and Acute Lung Injury. J Med Chem 2024; 67:10687-10709. [PMID: 38913701 DOI: 10.1021/acs.jmedchem.4c00202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
UC and ALI are inflammatory diseases with limited treatment in the clinic. Herein, fragment-based anti-inflammatory agent designs were carried out deriving from cyclohexylamine/cyclobutylamine and several fragments from anti-inflammatory agents in our lab. AF-45 (IC50 = 0.53/0.60 μM on IL-6/TNF-α in THP-1 macrophages) was identified as the optimal molecule using ELISA and MTT assays from the 33 synthesized compounds. Through mechanistic studies and a systematic target search process, AF-45 was found to block the NF-κB/MAPK pathway and target IRAK4, a promising target for inflammation and autoimmune diseases. The selectivity of AF-45 targeting IRAK4 was validated by comparing its effects on other kinase/nonkinase proteins. In vivo, AF-45 exhibited a good therapeutic effect on UC and ALI, and favorable PK proprieties. Since there are currently no clinical or preclinical trials for IRAK4 inhibitors to treat UC and ALI, AF-45 provides a new lead compound or candidate targeting IRAK4 for the treatment of these diseases.
Collapse
Affiliation(s)
- Yu Zou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, China
| | - Xiemin Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, China
| | - Pan Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, China
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Zhiwei Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, China
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea
| | - Xiaobo Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, China
| | - Zhichao Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, China
| | - Mi Guo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, China
| | - Ying Zhou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, China
| | - Chenhui Sun
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, China
| | - Ran Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Pengwu Zheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Won-Jea Cho
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea
| | - Young-Chang Cho
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- School of Pharmacy, Hangzhou Medical College, Hangzhou 311399, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, China
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Qidong Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, China
| |
Collapse
|
3
|
Toya S, Struyf S, Huerta L, Morris P, Gavioli E, Minnella EM, Cesta MC, Allegretti M, Proost P. A narrative review of chemokine receptors CXCR1 and CXCR2 and their role in acute respiratory distress syndrome. Eur Respir Rev 2024; 33:230172. [PMID: 39048127 PMCID: PMC11267298 DOI: 10.1183/16000617.0172-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 05/15/2024] [Indexed: 07/27/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe form of acute respiratory failure characterised by extensive inflammatory injury to the alveolocapillary barrier leading to alveolar oedema, impaired gas exchange and, ultimately, hypoxaemia necessitating the use of supplemental oxygen combined with some degree of positive airway pressure. Although much heterogeneity exists regarding the aetiology, localisation and endotypic characterisation of ARDS, what remains largely undisputed is the role of the innate immune system, and in particular of neutrophils, in precipitating and propagating lung injury. Activated neutrophils, recruited to the lung through chemokine gradients, promote injury by releasing oxidants, proteases and neutrophil extracellular traps, which ultimately cause platelet aggregation, microvascular thrombosis and cellular death. Among various neutrophilic chemoattractants, interleukin-8/C-X-C motif ligand 8 and related chemokines, collectively called ELR+ chemokines, acting on neutrophils through the G protein-coupled receptors CXCR1 and CXCR2, are pivotal in orchestrating the neutrophil activation status and chemotaxis in the inflamed lung. This allows efficient elimination of infectious agents while at the same time minimising collateral damage to host tissue. Therefore, understanding how CXCR1 and CXCR2 receptors are regulated is important if we hope to effectively target them for therapeutic use in ARDS. In the following narrative review, we provide an overview of the role of ELR+ chemokines in acute lung injury (ALI) and ARDS, we summarise the relevant regulatory pathways of their cognisant receptors CXCR1/2 and highlight current preclinical and clinical evidence on the therapeutic role of CXCR1 and CXCR2 inhibition in animal models of ALI, as well as in ARDS patients.
Collapse
Affiliation(s)
| | - Sofie Struyf
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Luis Huerta
- Keck School of Medicine of USC, Department of Medicine, Pulmonary and Critical Care Medicine, Los Angeles, CA, USA
| | - Peter Morris
- The University of Alabama at Birmingham, Department of Medicine, Pulmonary, Allergy, and Critical Care Medicine, Birmingham, AL, USA
| | | | | | | | | | - Paul Proost
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| |
Collapse
|
4
|
Zhang C, Simón M, Harder JM, Lim H, Montgomery C, Wang Q, John SW. TLR4 deficiency does not alter glaucomatous progression in a mouse model of chronic glaucoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597951. [PMID: 38895321 PMCID: PMC11185798 DOI: 10.1101/2024.06.07.597951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Glaucoma is a leading cause of irreversible blindness worldwide. Toll-like receptor 4 (TLR4) is a pattern-recognition transmembrane receptor that induces neuroinflammatory processes in response to injury. Tlr4 is highly expressed in ocular tissues and is known to modulate inflammatory processes in both anterior and posterior segment tissues. TLR4 activation can lead to mitochondrial dysfunction and metabolic deficits in inflammatory disorders. Due to its effects on inflammation and metabolism, TLR4 is a candidate to participate in glaucoma pathogenesis. It has been suggested as a therapeutic target based on studies using acute models, such as experimentally raising IOP to ischemia-inducing levels. Nevertheless, its role in chronic glaucoma needs further evaluation. In the current study, we investigated the role of TLR4 in an inherited mouse model of chronic glaucoma, DBA/2J. To do this, we analyzed the effect of Tlr4 knockout (Tlr4 -/-) on glaucoma-associated phenotypes in DBA/2J mice. Our studies found no significant differences in intraocular pressure, iris disease, or glaucomatous progression in Tlr4 -/- compared to Tlr4 +/+ DBA/2J mice. These data do not identify a role for TLR4 in this chronic glaucoma, but further research is warranted to understand its role in other glaucoma models and different genetic contexts.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY
| | - Marina Simón
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY
| | | | - Haeyn Lim
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY
| | - Christa Montgomery
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY
| | - Qing Wang
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY
| | - Simon W.M. John
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY
- The Jackson Laboratory, Bar Harbor, ME
| |
Collapse
|
5
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Recent Insights into the Molecular Mechanisms of the Toll-like Receptor Response to Influenza Virus Infection. Int J Mol Sci 2024; 25:5909. [PMID: 38892096 PMCID: PMC11172706 DOI: 10.3390/ijms25115909] [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: 04/21/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Influenza A viruses (IAVs) pose a significant global threat to human health. A tightly controlled host immune response is critical to avoid any detrimental effects of IAV infection. It is critical to investigate the association between the response of Toll-like receptors (TLRs) and influenza virus. Because TLRs may act as a double-edged sword, a balanced TLR response is critical for the overall benefit of the host. Consequently, a thorough understanding of the TLR response is essential for targeting TLRs as a novel therapeutic and prophylactic intervention. To date, a limited number of studies have assessed TLR and IAV interactions. Therefore, further research on TLR interactions in IAV infection should be conducted to determine their role in host-virus interactions in disease causation or clearance of the virus. Although influenza virus vaccines are available, they have limited efficacy, which should be enhanced to improve their efficacy. In this study, we discuss the current status of our understanding of the TLR response in IAV infection and the strategies adopted by IAVs to avoid TLR-mediated immune surveillance, which may help in devising new therapeutic or preventive strategies. Furthermore, recent advances in the use of TLR agonists as vaccine adjuvants to enhance influenza vaccine efficacy are discussed.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal 8210, Bangladesh
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-0065, Japan
| |
Collapse
|
6
|
Maler MD, Zwick S, Kallfass C, Engelhard P, Shi H, Hellig L, Zhengyang P, Hardt A, Zissel G, Ruzsics Z, Jahnen-Dechent W, Martin SF, Nielsen PJ, Stolz D, Lopatecka J, Bastyans S, Beutler B, Schamel WW, Fejer G, Freudenberg MA. Type I Interferon, Induced by Adenovirus or Adenoviral Vector Infection, Regulates the Cytokine Response to Lipopolysaccharide in a Macrophage Type-Specific Manner. J Innate Immun 2024; 16:226-247. [PMID: 38527452 PMCID: PMC11023693 DOI: 10.1159/000538282] [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: 02/17/2023] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
INTRODUCTION While TLR ligands derived from microbial flora and pathogens are important activators of the innate immune system, a variety of factors such as intracellular bacteria, viruses, and parasites can induce a state of hyperreactivity, causing a dysregulated and potentially life-threatening cytokine over-response upon TLR ligand exposure. Type I interferon (IFN-αβ) is a central mediator in the induction of hypersensitivity and is strongly expressed in splenic conventional dendritic cells (cDC) and marginal zone macrophages (MZM) when mice are infected with adenovirus. This study investigates the ability of adenoviral infection to influence the activation state of the immune system and underlines the importance of considering this state when planning the treatment of patients. METHODS Infection with adenovirus-based vectors (Ad) or pretreatment with recombinant IFN-β was used as a model to study hypersensitivity to lipopolysaccharide (LPS) in mice, murine macrophages, and human blood samples. The TNF-α, IL-6, IFN-αβ, and IL-10 responses induced by LPS after pretreatment were measured. Mouse knockout models for MARCO, IFN-αβR, CD14, IRF3, and IRF7 were used to probe the mechanisms of the hypersensitive reaction. RESULTS We show that, similar to TNF-α and IL-6 but not IL-10, the induction of IFN-αβ by LPS increases strongly after Ad infection. This is true both in mice and in human blood samples ex vivo, suggesting that the regulatory mechanisms seen in the mouse are also present in humans. In mice, the scavenger receptor MARCO on IFN-αβ-producing cDC and splenic marginal zone macrophages is important for Ad uptake and subsequent cytokine overproduction by LPS. Interestingly, not all IFN-αβ-pretreated macrophage types exposed to LPS exhibit an enhanced TNF-α and IL-6 response. Pretreated alveolar macrophages and alveolar macrophage-like murine cell lines (MPI cells) show enhanced responses, while bone marrow-derived and peritoneal macrophages show a weaker response. This correlates with the respective absence or presence of the anti-inflammatory IL-10 response in these different macrophage types. In contrast, Ad or IFN-β pretreatment enhances the subsequent induction of IFN-αβ in all macrophage types. IRF3 is dispensable for the LPS-induced IFN-αβ overproduction in infected MPI cells and partly dispensable in infected mice, while IRF7 is required. The expression of the LPS co-receptor CD14 is important but not absolutely required for the elicitation of a TNF-α over-response to LPS in Ad-infected mice. CONCLUSION Viral infections or application of virus-based vaccines induces type I interferon and can tip the balance of the innate immune system in the direction of hyperreactivity to a subsequent exposure to TLR ligands. The adenoviral model presented here is one example of how multiple factors, both environmental and genetic, affect the physiological responses to pathogens. Being able to measure the current reactivity state of the immune system would have important benefits for infection-specific therapies and for the prevention of vaccination-elicited adverse effects.
Collapse
Affiliation(s)
- Mareike D. Maler
- Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Allergy Research Group, Department of Dermatology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Sophie Zwick
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Carsten Kallfass
- Institute of Virology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Peggy Engelhard
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Hexin Shi
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Laura Hellig
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Pang Zhengyang
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Annika Hardt
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Gernot Zissel
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Zsolt Ruzsics
- Institute of Virology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Willi Jahnen-Dechent
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Stefan F. Martin
- Allergy Research Group, Department of Dermatology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Peter Jess Nielsen
- Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Daiana Stolz
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Justyna Lopatecka
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Faculty of Medicine, Aachen, Germany
| | - Sarah Bastyans
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Faculty of Medicine, Aachen, Germany
| | - Bruce Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wolfgang W. Schamel
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Plymouth, UK
| | - György Fejer
- Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Faculty of Medicine, Aachen, Germany
| | - Marina Alexandra Freudenberg
- Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| |
Collapse
|
7
|
Yuan HX, Chen YT, Li YQ, Wang YS, Ou ZJ, Li Y, Gao JJ, Deng MJ, Song YK, Fu L, Ci HB, Chang FJ, Cao Y, Jian YP, Kang BA, Mo ZW, Ning DS, Peng YM, Liu ZL, Liu XJ, Xu YQ, Xu J, Ou JS. Endothelial extracellular vesicles induce acute lung injury via follistatin-like protein 1. SCIENCE CHINA. LIFE SCIENCES 2024; 67:475-487. [PMID: 37219765 PMCID: PMC10202752 DOI: 10.1007/s11427-022-2328-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 03/06/2023] [Indexed: 05/24/2023]
Abstract
Cardiopulmonary bypass has been speculated to elicit systemic inflammation to initiate acute lung injury (ALI), including acute respiratory distress syndrome (ARDS), in patients after cardiac surgery. We previously found that post-operative patients showed an increase in endothelial cell-derived extracellular vesicles (eEVs) with components of coagulation and acute inflammatory responses. However, the mechanism underlying the onset of ALI owing to the release of eEVs after cardiopulmonary bypass, remains unclear. Plasma plasminogen-activated inhibitor-1 (PAI-1) and eEV levels were measured in patients with cardiopulmonary bypass. Endothelial cells and mice (C57BL/6, Toll-like receptor 4 knockout (TLR4-/-) and inducible nitric oxide synthase knockout (iNOS-/-)) were challenged with eEVs isolated from PAI-1-stimulated endothelial cells. Plasma PAI-1 and eEVs were remarkably enhanced after cardiopulmonary bypass. Plasma PAI-1 elevation was positively correlated with the increase in eEVs. The increase in plasma PAI-1 and eEV levels was associated with post-operative ARDS. The eEVs derived from PAI-1-stimulated endothelial cells could recognize TLR4 to stimulate a downstream signaling cascade identified as the Janus kinase 2/3 (JAK2/3)-signal transducer and activator of transcription 3 (STAT3)-interferon regulatory factor 1 (IRF-1) pathway, along with iNOS induction, and cytokine/chemokine production in vascular endothelial cells and C57BL/6 mice, ultimately contributing to ALI. ALI could be attenuated by JAK2/3 or STAT3 inhibitors (AG490 or S3I-201, respectively), and was relieved in TLR4-/- and iNOS-/- mice. eEVs activate the TLR4/JAK3/STAT3/IRF-1 signaling pathway to induce ALI/ARDS by delivering follistatin-like protein 1 (FSTL1), and FSTL1 knockdown in eEVs alleviates eEV-induced ALI/ARDS. Our data thus demonstrate that cardiopulmonary bypass may increase plasma PAI-1 levels to induce FSTL1-enriched eEVs, which target the TLR4-mediated JAK2/3/STAT3/IRF-1 signaling cascade and form a positive feedback loop, leading to ALI/ARDS after cardiac surgery. Our findings provide new insight into the molecular mechanisms and therapeutic targets for ALI/ARDS after cardiac surgery.
Collapse
Affiliation(s)
- Hao-Xiang Yuan
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Ya-Ting Chen
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Yu-Quan Li
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Yan-Sheng Wang
- State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Zhi-Jun Ou
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
- Division of Hypertension and Vascular Diseases, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yan Li
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Jian-Jun Gao
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Meng-Jie Deng
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Yuan-Kai Song
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Li Fu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Hong-Bo Ci
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Feng-Jun Chang
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Yang Cao
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Yu-Peng Jian
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Bi-Ang Kang
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Zhi-Wei Mo
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Da-Sheng Ning
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Yue-Ming Peng
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Ze-Long Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Xiao-Jun Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Ying-Qi Xu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Jun Xu
- State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China.
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China.
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Jing-Song Ou
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, 510080, China.
| |
Collapse
|
8
|
Xiao S, Zhang P, Zhang G, Li W, Lin H, Hu X. Inhibition of toll-like receptor 4 activation by apigenin and chrysin via competition for sites and conformational changes. Int J Biol Macromol 2023; 252:126415. [PMID: 37598817 DOI: 10.1016/j.ijbiomac.2023.126415] [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: 07/10/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
The activation of toll-like receptor 4 (TLR4) signaling is crucial for initiating and coordinating the immune response against infections, and is proved as a vital target for inflammatory diseases. Herein, TLR4 with sufficient amount and functional activity was generated by heterologous expression and used to investigate the mechanism of apigenin (Api)/chrysin (Chr) inhibition of TLR4 activation. The results demonstrated that Api/Chr exhibited a strong fluorescence quenching effect through a static quenching and a high binding affinity (Ka > 105 L·mol-1) with TLR4, indicating the potential of Api/Chr as a TLR4 inhibitor. Additionally, the binding of Api/Chr induced a loose and unstable conformation of TLR4 with evidence like the decreased hydrophobicity of the tryptophan microenvironment, decreased α-helix content and increased free sulfhydryl content, resulting in reduced stability of the TLR4. The computer simulations revealed that Api/Chr occupied the myeloid differentiation factor 2 (MD-2) binding region, preventing MD-2 from binding to TLR4. Furthermore, the accuracy of the binding site between Api/Chr and TLR4 was confirmed through genetic mutations. Overall, the mechanism by which Api/Chr inhibited TLR4 activation was elucidated at the macroscopic and molecular levels, providing the worthful information concerning the future therapeutic application of Api/Chr as a natural TLR4 inhibitor.
Collapse
Affiliation(s)
- Shuang Xiao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Peng Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Guowen Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Wenwen Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Haowen Lin
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Xing Hu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
9
|
Numata M, Kandasamy P, Voelker DR. The anti-inflammatory and antiviral properties of anionic pulmonary surfactant phospholipids. Immunol Rev 2023; 317:166-186. [PMID: 37144896 PMCID: PMC10524216 DOI: 10.1111/imr.13207] [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: 03/15/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/06/2023]
Abstract
The pulmonary surfactant system of the lung is a lipid and protein complex, which regulates the biophysical properties of the alveoli to prevent lung collapse and the innate immune system in the lung. Pulmonary surfactant is a lipoprotein complex consisting of 90% phospholipids and 10% protein, by weight. Two minor components of pulmonary surfactant phospholipids, phosphatidylglycerol (PG) and phosphatidylinositol (PI), exist at very high concentrations in the extracellular alveolar compartments. We have reported that one of the most dominant molecular species of PG, palmitoyl-oleoyl-phosphatidylglycerol (POPG) and PI inhibit inflammatory responses induced by multiple toll-like receptors (TLR2/1, TLR3, TLR4, and TLR2/6) by interacting with subsets of multiprotein receptor components. These lipids also exert potent antiviral effects against RSV and influenza A, in vitro, by inhibiting virus binding to host cells. POPG and PI inhibit these viral infections in vivo, in multiple animal models. Especially noteworthy, these lipids markedly attenuate SARS-CoV-2 infection including its variants. These lipids are natural compounds that already exist in the lung and, thus, are less likely to cause adverse immune responses by hosts. Collectively, these data demonstrate that POPG and PI have strong potential as novel therapeutics for applications as anti-inflammatory compounds and preventatives, as treatments for broad ranges of RNA respiratory viruses.
Collapse
Affiliation(s)
- Mari Numata
- Department of Medicine, National Jewish Health, Denver, CO 80206
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO 80206
| | - Pitchaimani Kandasamy
- Department of Medicine, National Jewish Health, Denver, CO 80206
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO 80206
| | - Dennis R. Voelker
- Department of Medicine, National Jewish Health, Denver, CO 80206
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO 80206
| |
Collapse
|
10
|
Yu H, Deng W, Chen S, Qin B, Yao Y, Zhou C, Guo M. Strongylocentrotus nudus egg polysaccharide (SEP) suppresses HBV replication via activation of TLR4-induced immune pathway. Int J Biol Macromol 2023:125539. [PMID: 37355064 DOI: 10.1016/j.ijbiomac.2023.125539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023]
Abstract
Chronic hepatitis B virus (HBV) infection is a worldwide public health problem that causes significant liver-related morbidity and mortality. In our previous study, Strongylocentrotus nudus eggs polysaccharide (SEP), extracted from sea urchins, had immunomodulatory and antitumor effects. Whether SEP has anti-HBV activity is still obscure. This study demonstrated that SEP decreased the secretion of hepatitis B surface antigen (HBsAg) and e antigen (HBeAg), as well as the replication and transcription of HBV both in vitro and in vivo. Immunofluorescence and immunohistochemistry results showed that the level of HBV core antigen (HBcAg) was clearly reduced by SEP treatment. Mechanistically, RT-qPCR, western blot, and confocal microscopy analysis showed that SEP significantly increased the expression of toll-like receptor 4 (TLR4) and co-localization with TLR4. The downstream molecules of TLR4, including NF-κb and IRF3, were activated and the expression of IFN-β, TNF-α, IL-6, OAS, and MxA were also increased, which could suppress HBV replication. Moreover, SEP inhibited other genotypes of HBV and hepatitis C virus (HCV) replication in vitro. In summary, SEP could be investigated as a potential anti-HBV drug capable of modulating the innate immune.
Collapse
Affiliation(s)
- Haifei Yu
- State Key Laboratory of Natural Medicines, School of Life Science & Technolgy, China Pharmaceutical University, Nanjing 211198, Jiangsu province, China
| | - Wanyu Deng
- College of life science, Shangrao Normal University, Shangrao 334001, Jiangxi province, China
| | - Shuo Chen
- State Key Laboratory of Natural Medicines, School of Life Science & Technolgy, China Pharmaceutical University, Nanjing 211198, Jiangsu province, China
| | - Bo Qin
- Shaoxing Women and Children's Hospital, Shaoxing 312000, Zhejiang, China
| | - Yongxuan Yao
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children Medical Center, Guangzhou 510623, China.
| | - Changlin Zhou
- State Key Laboratory of Natural Medicines, School of Life Science & Technolgy, China Pharmaceutical University, Nanjing 211198, Jiangsu province, China.
| | - Min Guo
- State Key Laboratory of Natural Medicines, School of Life Science & Technolgy, China Pharmaceutical University, Nanjing 211198, Jiangsu province, China.
| |
Collapse
|
11
|
Liu PY, Chen CY, Lin YL, Lin CM, Tsai WC, Tsai YL, Lin GJ, Chen YG, Wang SY, Sun RN, Huang YC, Chang H, Chen YC. RNF128 regulates neutrophil infiltration and myeloperoxidase functions to prevent acute lung injury. Cell Death Dis 2023; 14:369. [PMID: 37344492 DOI: 10.1038/s41419-023-05890-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/04/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023]
Abstract
Acute lung injury (ALI) is characterised by severe pulmonary inflammation, alveolar-capillary barrier disruption, and pulmonary oedema. Therefore, establishing effective therapeutic targets for ALI prevention is crucial. The present study reports a novel function of RNF128 in regulating LPS-induced ALI. Severe lung damage and increased immune cell infiltration were detected in RNF128-deficient mice. In vitro experiments revealed that RNF128 inhibits neutrophil activation by binding to myeloperoxidase (MPO) and reducing its levels and activity. Moreover, RNF128 regulates alveolar macrophage activation and neutrophil infiltration by interacting with TLR4, targeting it for degradation, and inhibiting NF-κB activation, hence decreasing pro-inflammatory cytokines. Our results demonstrate for the first time that RNF128 is a negative regulator of MPO and TLR4 in neutrophils and alveolar macrophages, respectively. However, AAV9-mediated RNF128 overexpression alleviated lung tissue damage and reduced inflammatory cell infiltration. Thus, RNF128 is a promising therapeutic candidate for pharmacological interventions in ALI.
Collapse
Affiliation(s)
- Pei-Yao Liu
- Department of Physiology & Biophysics, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Chih-Yuan Chen
- Department of Physiology & Biophysics, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
- Department of Thoracic surgery, Tri-Service General Hospital Taipei, National Defense Medical Center, Taiwan, Republic of China
| | - Yu-Lung Lin
- The Ph.D. Program for Translational Medicine, College for Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
| | - Chien-Ming Lin
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Yu-Ling Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Gu-Jiun Lin
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Yu-Guang Chen
- Division of Hematology/Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
- Cancer Institute, University College London, London, UK
| | - Shih-Yun Wang
- Department of Physiology & Biophysics, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Rui-Nong Sun
- Department of Physiology & Biophysics, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Yu-Chuan Huang
- School of Pharmacy & Institute Pharmacy, National Defense Medical Center, Taipei, Republic of China
- Department of Research and Development, National Defense Medical Center, Taipei, Republic of China
| | - Hung Chang
- Department of Physiology & Biophysics, National Defense Medical Center, Taipei 114, Taiwan, Republic of China.
- Department of Thoracic surgery, Tri-Service General Hospital Taipei, National Defense Medical Center, Taiwan, Republic of China.
| | - Ying-Chuan Chen
- Department of Physiology & Biophysics, National Defense Medical Center, Taipei 114, Taiwan, Republic of China.
| |
Collapse
|
12
|
Žilienė E, Inčiūra A, Ugenskienė R, Juozaitytė E. Pathomorphological Manifestations and the Course of the Cervical Cancer Disease Determined by Variations in the TLR4 Gene. Diagnostics (Basel) 2023; 13:1999. [PMID: 37370894 DOI: 10.3390/diagnostics13121999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Cervical cancer (CC) is often associated with human papillomavirus (HPV). Chronic inflammation has been described as one of the triggers of cancer. The immune system fights diseases, including cancer. The genetic polymorphism of pathogen recognition receptors potentially influences the infectious process, development, and disease progression. Many candidate genes SNPs have been contradictory demonstrated to be associated with cervical cancer by association studies, GWAS. TLR4 gene activation can promote antitumor immunity. It can also result in immunosuppression and tumor growth. Our study aimed to investigate eight selected polymorphisms of the TLR4 gene (rs10759932, rs1927906, rs11536898, rs11536865, rs10983755, rs4986790, rs4986791, rs11536897) and to determine the impact of polymorphisms in genotypes and alleles on the pathomorphological characteristics and progression in a group of 172 cervical cancer subjects with stage I-IV. Genotyping was performed by RT-PCR assay. We detected that the CA genotype and A allele of rs11536898 were significantly more frequent in patients with metastases (p = 0.026; p = 0.008). The multivariate logistic regression analysis confirmed this link to be significant. The effect of rs10759932 and rs11536898 on progression-free survival (PFS) and overall survival (OS) has been identified as important. In univariate and multivariate Cox analyses, AA genotype of rs11536898 was a negative prognostic factor for PFS (p = 0.024; p = 0.057, respectively) and OS (p = 0.008; p = 0.042, respectively). Rs11536898 C allele predisposed for longer PFS (univariate and multivariate: p = 0.025; p = 0.048, respectively) and for better OS (univariate and multivariate: p = 0.010; p = 0.043). The worse prognostic factor of rs10759932 in a univariate and multivariate Cox analysis for survival was CC genotype: shorter PFS (p = 0.032) and increased risk of death (p = 0.048; p = 0.015, respectively). The T allele of rs10759932 increased longer PFS (univariate and multivariate: p = 0.048; p = 0.019, respectively) and longer OS (univariate and multivariate: p = 0.037; p = 0.009, respectively). Our study suggests that SNPs rs10759932 and rs11536898 may have the potential to be markers contributing to the assessment of the cervical cancer prognosis. Further studies, preferably with larger groups of different ethnic backgrounds, are needed to confirm the results of the current study.
Collapse
Affiliation(s)
- Eglė Žilienė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Arturas Inčiūra
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Rasa Ugenskienė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Elona Juozaitytė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| |
Collapse
|
13
|
Zhou B, Wang L, Yang S, Liang Y, Zhang Y, Pan X, Li J. Diosmetin alleviates benzo[ a]pyrene-exacerbated H1N1 influenza virus-induced acute lung injury and dysregulation of inflammation through modulation of the PPAR-γ-NF-κB/P38 MAPK signaling axis. Food Funct 2023; 14:3357-3378. [PMID: 36942763 DOI: 10.1039/d2fo02590f] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
The severity of a viral respiratory illness was greatly exacerbated after exposure to a contaminant containing benzo[a]pyrene (B[a]P). Flavonoid-rich fruit intake has gained intense interest due to their health-promoting benefits for viral respiratory diseases, including influenza viruses. In our study, diosmetin (3',5,7-trihydroxy-4'-methoxyflavone), a naturally occurring hydroxylated methoxyflavone that is abundant in Citrus fruits, was explored for its effects on B[a]P-exacerbated H1N1 influenza virus-mediated inflammation and lung injury. Initially, in vivo results demonstrated that diosmetin protected against H1N1 virus-elicited acute lung injury. Simultaneously, H1N1 virus or B[a]P-stimulated A549 cells treated with diosmetin inhibited NF-κB and P-P38 activation, resulting in suppression of pro-inflammatory cytokines and apoptosis. Interestingly, diosmetin obviously promoted the expression of PPAR-γ as well as nuclear translocation of PPAR-γ, whereas, PPAR-γ inhibition by GW9662 weakened the inhibitory effects of diosmetin on H1N1 virus or B[a]P-mediated activation of NF-κB and P-P38, elevated expression of pro-inflammatory mediators as well as apoptosis. Furthermore, it was surprising to discover that mice exposed to both B[a]P and H1N1 viruses contributed to exacerbated acute lung injury, which were significantly ameliorated by diosmetin administration. In vitro studies showed that A549 cells with the combination of B[a]P and H1N1 virus augmented NF-κB and P-P38 activation, accompanied by higher levels of pro-inflammatory mediators and apoptosis, all of which were also significantly reduced by diosmetin treatment. Repressing PPAR-γ abrogated the inhibitory effects of diosmetin on B[a]P-exacerbated H1N1 virus-mediated NF-κB and P-P38 activation, inflammation, and apoptosis in A549 cells. Our findings suggest that diosmetin protected against B[a]P-exacerbated H1N1 virus-mediated lung injury by suppressing the exacerbation of NF-κB and P38 kinase activation in a PPAR-γ-dependent manner, suggesting potential benefits for B[a]P-exacerbated influenza-related illness therapeutics.
Collapse
Affiliation(s)
- Beixian Zhou
- The People's Hospital of Gaozhou, Gaozhou 525200, China
| | | | - Sushan Yang
- The People's Hospital of Gaozhou, Gaozhou 525200, China
| | - Yueyun Liang
- The People's Hospital of Gaozhou, Gaozhou 525200, China
| | - Yuehan Zhang
- The People's Hospital of Gaozhou, Gaozhou 525200, China
| | | | - Jing Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
- Institute of Chinese Integrative Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Li H, Hu W, Lin Y, Xu T, Zhang X, Wang C. MicroRNA-9-5p Is Involved in Lipopolysaccharide-Induced Acute Lung Injury Via the Regulation of Macrophage Polarization. Int J Toxicol 2023; 42:156-164. [PMID: 36537157 DOI: 10.1177/10915818221146446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
MicroRNA (miR)-9-5 p has been shown to affect lung cancer progression and lung fibrosis, but the efficacy of miR-9-5 p in acute lung injury (ALI) remained indefinite. The study was performed to probe the modulating mechanism of miR-9-5 p in ALI via regulating macrophage polarization. The ALI mouse model was established and blood samples of ALI patients were obtained. MiR-9-5 p levels in ALI mice and ALI patients were detected. Mouse pulmonary macrophages were extracted from bronchoalveolar lavage fluid and polarized into M1 and M2 macrophages. Intervention of miR-9-5 p expression was performed to observe the effects on M1 polarization and M2 polarization in lung macrophages, inflammatory factors in BALF, wet/dry weight ratio (W/D) in lung tissues, myeloperoxidase (MPO) activity in lung tissues, and lung tissue lesion condition. MiR-9-5 p levels were elevated in the lung tissues of ALI mice and ALI patients. MiR-9-5 p silencing could repress lung macrophages in ALI mice polarized toward the M1 phenotype and promoted the polarization toward the M2 phenotype, reduced the lung lesions, the lung water content, and the secretion levels of the pro-inflammatory factors TNF-α, IL-6, and IL-1β in BALF, increased the secretion of the anti-inflammatory factor IL-10, as well as impeded the MPO activity in the lung tissues of ALI mice. MiR-9-5 p deletion ameliorates LPS-induced inflammatory infiltration in lung tissues via inhibiting the polarization of mouse lung macrophages to the M1 phenotype and promoting the polarization to the M2 phenotype.
Collapse
Affiliation(s)
- Hao Li
- Department of Emergency Medicine, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Weimi Hu
- Department of Emergency Medicine, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Yueyue Lin
- Department of Gastroscope Room, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Tengxiao Xu
- Department of Emergency Medicine, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Xianjing Zhang
- Department of Emergency Medicine, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Chen Wang
- Department of Respiratory and Critical Care Medicine, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| |
Collapse
|
15
|
Wang H, Su Y, Chen D, Li Q, Shi S, Huang X, Fang M, Yang M. Advances in the mechanisms and applications of inhibitory oligodeoxynucleotides against immune-mediated inflammatory diseases. Front Pharmacol 2023; 14:1119431. [PMID: 36825156 PMCID: PMC9941346 DOI: 10.3389/fphar.2023.1119431] [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: 12/09/2022] [Accepted: 01/26/2023] [Indexed: 02/09/2023] Open
Abstract
Inhibitory oligodeoxynucleotides (ODNs) are short single-stranded DNA, which capable of folding into complex structures, enabling them to bind to a large variety of targets. With appropriate modifications, the inhibitory oligodeoxynucleotides exhibited many features of long half-life time, simple production, low toxicity and immunogenicity. In recent years, inhibitory oligodeoxynucleotides have received considerable attention for their potential therapeutic applications in immune-mediated inflammatory diseases (IMIDs). Inhibitory oligodeoxynucleotides could be divided into three categories according to its mechanisms and targets, including antisense ODNs (AS-ODNs), DNA aptamers and immunosuppressive ODNs (iSup ODNs). As a synthetic tool with immunomodulatory activity, it can target RNAs or proteins in a specific way, resulting in the reduction, increase or recovery of protein expression, and then regulate the state of immune activation. More importantly, inhibitory oligodeoxynucleotides have been used to treat immune-mediated inflammatory diseases, including inflammatory disorders and autoimmune diseases. Several inhibitory oligodeoxynucleotide drugs have been developed and approved on the market already. These drugs vary in their chemical structures, action mechanisms and cellular targets, but all of them could be capable of inhibiting excessive inflammatory responses. This review summarized their chemical modifications, action mechanisms and applications of the three kinds of inhibitory oligodeoxynucleotidesin the precise treatment of immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Hongrui Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Yingying Su
- Department of Anatomy, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Duoduo Chen
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Qi Li
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Shuyou Shi
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Xin Huang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Mingli Fang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China,*Correspondence: Mingli Fang, ; Ming Yang,
| | - Ming Yang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China,*Correspondence: Mingli Fang, ; Ming Yang,
| |
Collapse
|
16
|
Resiliac J, Rohlfing M, Santoro J, Hussain SRA, Grayson MH. Low-Dose Lipopolysaccharide Protects from Lethal Paramyxovirus Infection in a Macrophage- and TLR4-Dependent Process. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:348-355. [PMID: 36480273 PMCID: PMC9851983 DOI: 10.4049/jimmunol.2200604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/14/2022] [Indexed: 01/03/2023]
Abstract
Respiratory diseases are a major public health burden and a leading cause of death and disability in the world. Understanding antiviral immune responses is crucial to alleviate morbidity and mortality associated with these respiratory viral infections. Previous data from human and animal studies suggested that pre-existing atopy may provide some protection against severe disease from a respiratory viral infection. However, the mechanism(s) of protection is not understood. Low-dose LPS has been shown to drive an atopic phenotype in mice. In addition, LPS has been shown in vitro to have an antiviral effect. We examined the effect of LPS treatment on mortality to the murine parainfluenza virus Sendai virus. Low-dose LPS treatment 24 h before inoculation with a normally lethal dose of Sendai virus greatly reduced death. This protection was associated with a reduced viral titer and reduced inflammatory cytokine production in the airways. The administration of LPS was associated with a marked increase in lung neutrophils and macrophages. Depletion of neutrophils failed to reverse the protective effect of LPS; however, depletion of macrophages reversed the protective effect of LPS. Further, we demonstrate that the protective effect of LPS depends on type I IFN and TLR4-MyD88 signaling. Together, these studies demonstrate pretreatment with low-dose LPS provides a survival advantage against a severe respiratory viral infection through a macrophage-, TLR4-, and MyD88-dependent pathway.
Collapse
Affiliation(s)
- Jenny Resiliac
- The Ohio State University College of Medicine, Biomedical Sciences Graduate Program, Columbus, Ohio
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Michelle Rohlfing
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Jennifer Santoro
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Syed-Rehan A. Hussain
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Mitchell H. Grayson
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children’s Hospital and The Ohio State University, Columbus, OH
| |
Collapse
|
17
|
Sun H, Chan JFW, Yuan S. Cellular Sensors and Viral Countermeasures: A Molecular Arms Race between Host and SARS-CoV-2. Viruses 2023; 15:352. [PMID: 36851564 PMCID: PMC9962416 DOI: 10.3390/v15020352] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the coronavirus disease 2019 (COVID-19) pandemic that has caused disastrous effects on the society and human health globally. SARS-CoV-2 is a sarbecovirus in the Coronaviridae family with a positive-sense single-stranded RNA genome. It mainly replicates in the cytoplasm and viral components including RNAs and proteins can be sensed by pattern recognition receptors including toll-like receptors (TLRs), RIG-I-like receptors (RLRs), and NOD-like receptors (NLRs) that regulate the host innate and adaptive immune responses. On the other hand, the SARS-CoV-2 genome encodes multiple proteins that can antagonize the host immune response to facilitate viral replication. In this review, we discuss the current knowledge on host sensors and viral countermeasures against host innate immune response to provide insights on virus-host interactions and novel approaches to modulate host inflammation and antiviral responses.
Collapse
Affiliation(s)
- Haoran Sun
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518009, China
| | - Jasper Fuk-Woo Chan
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518009, China
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Shuofeng Yuan
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518009, China
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| |
Collapse
|
18
|
Borrmann M, Brandes F, Kirchner B, Klein M, Billaud JN, Reithmair M, Rehm M, Schelling G, Pfaffl MW, Meidert AS. Extensive blood transcriptome analysis reveals cellular signaling networks activated by circulating glycocalyx components reflecting vascular injury in COVID-19. Front Immunol 2023; 14:1129766. [PMID: 36776845 PMCID: PMC9909741 DOI: 10.3389/fimmu.2023.1129766] [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: 12/22/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
Background Degradation of the endothelial protective glycocalyx layer during COVID-19 infection leads to shedding of major glycocalyx components. These circulating proteins and their degradation products may feedback on immune and endothelial cells and activate molecular signaling cascades in COVID-19 associated microvascular injury. To test this hypothesis, we measured plasma glycocalyx components in patients with SARS-CoV-2 infection of variable disease severity and identified molecular signaling networks activated by glycocalyx components in immune and endothelial cells. Methods We studied patients with RT-PCR confirmed COVID-19 pneumonia, patients with COVID-19 Acute Respiratory Distress Syndrome (ARDS) and healthy controls (wildtype, n=20 in each group) and measured syndecan-1, heparan sulfate and hyaluronic acid. The in-silico construction of signaling networks was based on RNA sequencing (RNAseq) of mRNA transcripts derived from blood cells and of miRNAs isolated from extracellular vesicles from the identical cohort. Differentially regulated RNAs between groups were identified by gene expression analysis. Both RNAseq data sets were used for network construction of circulating glycosaminoglycans focusing on immune and endothelial cells. Results Plasma concentrations of glycocalyx components were highest in COVID-19 ARDS. Hyaluronic acid plasma levels in patients admitted with COVID-19 pneumonia who later developed ARDS during hospital treatment (n=8) were significantly higher at hospital admission than in patients with an early recovery. RNAseq identified hyaluronic acid as an upregulator of TLR4 in pneumonia and ARDS. In COVID-19 ARDS, syndecan-1 increased IL-6, which was significantly higher than in pneumonia. In ARDS, hyaluronic acid activated NRP1, a co-receptor of activated VEGFA, which is associated with pulmonary vascular hyperpermeability and interacted with VCAN (upregulated), a proteoglycan important for chemokine communication. Conclusions Circulating glycocalyx components in COVID-19 have distinct biologic feedback effects on immune and endothelial cells and result in upregulation of key regulatory transcripts leading to further immune activation and more severe systemic inflammation. These consequences are most pronounced during the early hospital phase of COVID-19 before pulmonary failure develops. Elevated levels of circulating glycocalyx components may early identify patients at risk for microvascular injury and ARDS. The timely inhibition of glycocalyx degradation could provide a novel therapeutic approach to prevent the development of ARDS in COVID-19.
Collapse
Affiliation(s)
- Melanie Borrmann
- Department of Anesthesiology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Florian Brandes
- Department of Anesthesiology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Benedikt Kirchner
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Matthias Klein
- Department of Neurology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | | | - Marlene Reithmair
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Markus Rehm
- Department of Anesthesiology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany,Department of Anesthesiology and intensive Care Medicine, Hospital Agatharied, Hausham, Germany
| | - Gustav Schelling
- Department of Anesthesiology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany,*Correspondence: Gustav Schelling,
| | - Michael W. Pfaffl
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Agnes S. Meidert
- Department of Anesthesiology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
19
|
Liu Z, Deng P, Liu S, Bian Y, Xu Y, Zhang Q, Wang H, Pi J. Is Nuclear Factor Erythroid 2-Related Factor 2 a Target for the Intervention of Cytokine Storms? Antioxidants (Basel) 2023; 12:antiox12010172. [PMID: 36671034 PMCID: PMC9855012 DOI: 10.3390/antiox12010172] [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: 12/10/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
The term "cytokine storm" describes an acute pathophysiologic state of the immune system characterized by a burst of cytokine release, systemic inflammatory response, and multiple organ failure, which are crucial determinants of many disease outcomes. In light of the complexity of cytokine storms, specific strategies are needed to prevent and alleviate their occurrence and deterioration. Nuclear factor erythroid 2-related factor 2 (NRF2) is a CNC-basic region-leucine zipper protein that serves as a master transcription factor in maintaining cellular redox homeostasis by orchestrating the expression of many antioxidant and phase II detoxification enzymes. Given that inflammatory response is intertwined with oxidative stress, it is reasonable to assume that NRF2 activation limits inflammation and thus cytokine storms. As NRF2 can mitigate inflammation at many levels, it has emerged as a potential target to prevent and treat cytokine storms. In this review, we summarized the cytokine storms caused by different etiologies and the rationale of interventions, focusing mainly on NRF2 as a potential therapeutic target.
Collapse
Affiliation(s)
- Zihang Liu
- The First Department of Clinical Medicine, China Medical University, Shenyang 110122, China
| | - Panpan Deng
- The First Department of Clinical Medicine, China Medical University, Shenyang 110122, China
| | - Shengnan Liu
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang 110122, China
| | - Yiying Bian
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang 110122, China
| | - Yuanyuan Xu
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, Shenyang 110122, China
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Huihui Wang
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, Shenyang 110122, China
- Correspondence: (H.W.); or (J.P.)
| | - Jingbo Pi
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang 110122, China
- Correspondence: (H.W.); or (J.P.)
| |
Collapse
|
20
|
Heine H, Zamyatina A. Therapeutic Targeting of TLR4 for Inflammation, Infection, and Cancer: A Perspective for Disaccharide Lipid A Mimetics. Pharmaceuticals (Basel) 2022; 16:23. [PMID: 36678520 PMCID: PMC9864529 DOI: 10.3390/ph16010023] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
The Toll-like receptor 4 (TLR4) signaling pathway plays a central role in the prompt defense against infectious challenge and provides immediate response to Gram-negative bacterial infection. The TLR4/MD-2 complex can sense and respond to various pathogen-associated molecular patterns (PAMPs) with bacterial lipopolysaccharide (LPS) being the most potent and the most frequently occurring activator of the TLR4-mediated inflammation. TLR4 is believed to be both a friend and foe since improperly regulated TLR4 signaling can result in the overactivation of immune responses leading to sepsis, acute lung injury, or pathologic chronic inflammation involved in cancer and autoimmune disease. TLR4 is also considered a legitimate target for vaccine adjuvant development since its activation can boost the adaptive immune responses. The dual action of the TLR4 complex justifies the efforts in the development of both TLR4 antagonists as antisepsis drug candidates or remedies for chronic inflammatory diseases and TLR4 agonists as vaccine adjuvants or immunotherapeutics. In this review, we provide a brief overview of the biochemical evidences for possible pharmacologic applications of TLR4 ligands as therapeutics and report our systematic studies on the design, synthesis, and immunobiological evaluation of carbohydrate-based TLR4 antagonists with nanomolar affinity for MD-2 as well as disaccharide-based TLR4 agonists with picomolar affinity for the TLR4/MD-2 complex.
Collapse
Affiliation(s)
- Holger Heine
- Research Group Innate Immunity, Research Center Borstel—Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Parkallee 22, 23845 Borstel, Germany
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
21
|
Vogel SN, Richard K, Shirey KA, Sylla FY, Boukhvalova MS, Blanco JC. Evidence for Interplay Between the Renin-Angiotensin System and Toll-Like Receptor 4 Signaling Pathways in the Induction of Virus-Induced Acute Lung Injury. J Interferon Cytokine Res 2022; 42:618-623. [PMID: 36206057 PMCID: PMC9805881 DOI: 10.1089/jir.2022.0081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/24/2022] [Indexed: 01/13/2023] Open
Abstract
Dedication: This article is dedicated to Howard Young, an exceptional scientist who has provided outstanding mentorship to many postbaccalaureates, graduate students, and postdoctoral fellows during his career. Howard has been a colleague to many and was never tired of learning new things. He has brought "thinking out of the box" to the level of an art form and has always provided thoughtful and constructive suggestions to those who have sought his counsel. I am personally greatly indebted to Howard for his guidance in molecular biology over the past 30 years, and hope that we will continue to share a passion for learning and mentoring others for years to come. Thank you, Howard! -Stephanie N. Vogel The SARS-CoV-2 pandemic has led to an unprecedented explosion in studies that have sought to identify key mechanisms that underlie the ravaging aspects of this disease on individuals. SARS-CoV-2 virus gains access to cells by (1) binding of the viral spike (S) protein to cell-associated angiotensin-converting enzyme 2 (ACE2), a key receptor in the renin-angiotensin system (RAS), followed by (2) cleavage of S protein by a cellular serine protease ("S protein priming") to facilitate viral entry. Dysregulation of the RAS system has been implicated in the spectrum of clinical symptoms associated with SARS-CoV-2, including hypercytokinemia, elevated markers of endothelial injury and thrombosis, and both localized and systemic inflammation. However, the underlying mechanisms have yet to be fully delineated.
Collapse
Affiliation(s)
- Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Katharina Richard
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Kari Ann Shirey
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
22
|
Kaplan M, Şahutoğlu AS, Sarıtaş S, Duman H, Arslan A, Pekdemir B, Karav S. Role of milk glycome in prevention, treatment, and recovery of COVID-19. Front Nutr 2022; 9:1033779. [PMID: 36424926 PMCID: PMC9680090 DOI: 10.3389/fnut.2022.1033779] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/10/2022] [Indexed: 08/23/2023] Open
Abstract
Milk contains all essential macro and micro-nutrients for the development of the newborn. Its high therapeutic and antimicrobial content provides an important function for the prevention, treatment, and recovery of certain diseases throughout life. The bioactive components found in milk are mostly decorated with glycans, which provide proper formation and modulate the biological functions of glycosylated compounds. The glycome of milk consists of free glycans, glycolipids, and N- and O- glycosylated proteins. Recent studies have shown that both free glycans and glycan-containing molecules have antiviral characteristics based on different mechanisms such as signaling, microbiome modulation, natural decoy strategy, and immunomodulatory action. In this review, we discuss the recent clinical studies and potential mechanisms of free and conjugated glycans' role in the prevention, treatment, and recovery of COVID-19.
Collapse
Affiliation(s)
- Merve Kaplan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | | | - Sümeyye Sarıtaş
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Ayşenur Arslan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Burcu Pekdemir
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
23
|
Stephens M. The emerging potential of Aptamers as therapeutic agents in infection and inflammation. Pharmacol Ther 2022; 238:108173. [DOI: 10.1016/j.pharmthera.2022.108173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 10/18/2022]
|
24
|
Halajian EA, LeBlanc EV, Gee K, Colpitts CC. Activation of TLR4 by viral glycoproteins: A double-edged sword? Front Microbiol 2022; 13:1007081. [PMID: 36246240 PMCID: PMC9557975 DOI: 10.3389/fmicb.2022.1007081] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/15/2022] [Indexed: 11/25/2022] Open
Abstract
Recognition of viral infection by pattern recognition receptors is paramount for a successful immune response to viral infection. However, an unbalanced proinflammatory response can be detrimental to the host. Recently, multiple studies have identified that the SARS-CoV-2 spike protein activates Toll-like receptor 4 (TLR4), resulting in the induction of proinflammatory cytokine expression. Activation of TLR4 by viral glycoproteins has also been observed in the context of other viral infection models, including respiratory syncytial virus (RSV), dengue virus (DENV) and Ebola virus (EBOV). However, the mechanisms involved in virus-TLR4 interactions have remained unclear. Here, we review viral glycoproteins that act as pathogen-associated molecular patterns to induce an immune response via TLR4. We explore the current understanding of the mechanisms underlying how viral glycoproteins are recognized by TLR4 and discuss the contribution of TLR4 activation to viral pathogenesis. We identify contentious findings and research gaps that highlight the importance of understanding viral glycoprotein-mediated TLR4 activation for potential therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Che C. Colpitts
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| |
Collapse
|
25
|
Lidocaine reinforces the anti-inflammatory action of dexamethasone on myeloid and epithelial cells activated by inflammatory cytokines or SARS-CoV-2 infection. Biomed J 2022; 46:81-92. [PMID: 35948250 PMCID: PMC9357286 DOI: 10.1016/j.bj.2022.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/18/2022] [Accepted: 07/30/2022] [Indexed: 12/15/2022] Open
Abstract
Background Severe cases of Coronavirus Disease 2019 (COVID-19) that require admission to the Intensive Care Unit (ICU) and mechanical ventilation assistance show a high mortality rate with currently few therapeutic options available. Severe COVID-19 is characterized by a systemic inflammatory condition, also called “cytokine storm”, which can lead to various multi-organ complications and ultimately death. Lidocaine, a safe local anesthetic that given intravenously is used to treat arrhythmias, has long been reported to have an anti-inflammatory and pro-homeostatic activity. Methods We studied the capacity of lidocaine to modulate cytokine secretion of mouse and human myeloid cell lines activated by different cytokines or Toll Like Receptor (TLR) ligands (flagellin (FliC), Lipopolysaccharide (LPS), Polyinosinic:polycytidylic acid (Poly I:C) and N-Palmitoyl-S- [2,3-bis(palmitoyloxy)-(2RS)-propyl]-(R)-cysteinyl-(S)-seryl-(S)-lysyl-(S)-lysyl-(S)-lysyl-(S)-lysine x 3HCl (Pam3Cys-SKKKK)) or by Severe acute respiratory syndromecoronavirus 2 (SARS-CoV-2) infection to epithelial cells. Reporter cell lines were used to study modulation of lidocaine of specific signaling pathways. Results Lidocaine used in combination with dexamethasone, had an additive effect in the modulation of cellular inflammatory response triggered by Tumoral Necrosis Factor alpha (TNFα), Interleukin 1 beta (IL-1β) as well as different TLR ligands. We also found that lidocaine in combination with dexamethasone modulates the Nuclear factor kappa B (NF-κB) pathway, inflammasome activation as well as interferon gamma receptor (IFNγR) signaling without affecting the type I interferons (Type I IFNs) pathway. Furthermore, we showed that lidocaine and dexamethasone treatment of epithelial cells infected with SARS-CoV-2 modulated the expression of chemokines that contribute to pro-inflammatory effects in severe COVID. Conclusions We reported for the first time in vitro anti-inflammatory capacity of lidocaine on SARS-CoV-2 triggered immune pathways. These results indicated the potential of lidocaine to treat COVID-19 patients and add tools to the therapeutic options available for these concerning cases.
Collapse
|
26
|
Girkin JLN, Maltby S, Bartlett NW. Toll-like receptor-agonist-based therapies for respiratory viral diseases: thinking outside the cell. Eur Respir Rev 2022; 31:31/164/210274. [PMID: 35508333 PMCID: PMC9488969 DOI: 10.1183/16000617.0274-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 11/24/2022] Open
Abstract
Respiratory virus infections initiate in the upper respiratory tract (URT). Innate immunity is critical for initial control of infection at this site, particularly in the absence of mucosal virus-neutralising antibodies. If the innate immune response is inadequate, infection can spread to the lower respiratory tract (LRT) causing community-acquired pneumonia (as exemplified by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)/coronavirus disease 2019). Vaccines for respiratory viruses (influenza and SARS-CoV-2) leverage systemic adaptive immunity to protect from severe lung disease. However, the URT remains vulnerable to infection, enabling viral transmission and posing an ongoing risk of severe disease in populations that lack effective adaptive immunity. Innate immunity is triggered by host cell recognition of viral pathogen-associated molecular patterns via molecular sensors such as Toll-like receptors (TLRs). Here we review the role of TLRs in respiratory viral infections and the potential of TLR-targeted treatments to enhance airway antiviral immunity to limit progression to severe LRT disease and reduce person-to-person viral transmission. By considering cellular localisation and antiviral mechanisms of action and treatment route/timing, we propose that cell surface TLR agonist therapies are a viable strategy for preventing respiratory viral diseases by providing immediate, durable pan-viral protection within the URT. Respiratory virus infections are a significant disease burden and new treatment options are required. Treatments that stimulate innate immunity in the upper respiratory tract by targeting Toll-like receptors may provide rapid, pan-viral protection.https://bit.ly/3BNH2Em
Collapse
Affiliation(s)
- Jason L N Girkin
- Viral Immunology and Respiratory Disease Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Steven Maltby
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Nathan W Bartlett
- Viral Immunology and Respiratory Disease Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia .,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| |
Collapse
|
27
|
Wang Z, Li F, Liu J, Luo Y, Guo H, Yang Q, Xu C, Ma S, Chen H. Intestinal Microbiota - An Unmissable Bridge to Severe Acute Pancreatitis-Associated Acute Lung Injury. Front Immunol 2022; 13:913178. [PMID: 35774796 PMCID: PMC9237221 DOI: 10.3389/fimmu.2022.913178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022] Open
Abstract
Severe acute pancreatitis (SAP), one of the most serious abdominal emergencies in general surgery, is characterized by acute and rapid onset as well as high mortality, which often leads to multiple organ failure (MOF). Acute lung injury (ALI), the earliest accompanied organ dysfunction, is the most common cause of death in patients following the SAP onset. The exact pathogenesis of ALI during SAP, however, remains unclear. In recent years, advances in the microbiota-gut-lung axis have led to a better understanding of SAP-associated lung injury (PALI). In addition, the bidirectional communications between intestinal microbes and the lung are becoming more apparent. This paper aims to review the mechanisms of an imbalanced intestinal microbiota contributing to the development of PALI, which is mediated by the disruption of physical, chemical, and immune barriers in the intestine, promotes bacterial translocation, and results in the activation of abnormal immune responses in severe pancreatitis. The pathogen-associated molecular patterns (PAMPs) mediated immunol mechanisms in the occurrence of PALI via binding with pattern recognition receptors (PRRs) through the microbiota-gut-lung axis are focused in this study. Moreover, the potential therapeutic strategies for alleviating PALI by regulating the composition or the function of the intestinal microbiota are discussed in this review. The aim of this study is to provide new ideas and therapeutic tools for PALI patients.
Collapse
Affiliation(s)
- Zhengjian Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fan Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jin Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yalan Luo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Haoya Guo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qi Yang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Caiming Xu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Shurong Ma
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- *Correspondence: Shurong Ma, ; Hailong Chen,
| | - Hailong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- *Correspondence: Shurong Ma, ; Hailong Chen,
| |
Collapse
|
28
|
Zhang Y, Liang X, Bao X, Xiao W, Chen G. Toll-like receptor 4 (TLR4) inhibitors: Current research and prospective. Eur J Med Chem 2022; 235:114291. [DOI: 10.1016/j.ejmech.2022.114291] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 01/10/2023]
|
29
|
López-Cortés A, Guerrero S, Ortiz-Prado E, Yumiceba V, Vera-Guapi A, León Cáceres Á, Simbaña-Rivera K, Gómez-Jaramillo AM, Echeverría-Garcés G, García-Cárdenas JM, Guevara-Ramírez P, Cabrera-Andrade A, Puig San Andrés L, Cevallos-Robalino D, Bautista J, Armendáriz-Castillo I, Pérez-Villa A, Abad-Sojos A, Ramos-Medina MJ, León-Sosa A, Abarca E, Pérez-Meza ÁA, Nieto-Jaramillo K, Jácome AV, Morillo A, Arias-Erazo F, Fuenmayor-González L, Quiñones LA, Kyriakidis NC. Pulmonary Inflammatory Response in Lethal COVID-19 Reveals Potential Therapeutic Targets and Drugs in Phases III/IV Clinical Trials. Front Pharmacol 2022; 13:833174. [PMID: 35422702 PMCID: PMC9002106 DOI: 10.3389/fphar.2022.833174] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/07/2022] [Indexed: 12/26/2022] Open
Abstract
Background: It is imperative to identify drugs that allow treating symptoms of severe COVID-19. Respiratory failure is the main cause of death in severe COVID-19 patients, and the host inflammatory response at the lungs remains poorly understood. Methods: Therefore, we retrieved data from post-mortem lungs from COVID-19 patients and performed in-depth in silico analyses of single-nucleus RNA sequencing data, inflammatory protein interactome network, and shortest pathways to physiological phenotypes to reveal potential therapeutic targets and drugs in advanced-stage COVID-19 clinical trials. Results: Herein, we analyzed transcriptomics data of 719 inflammatory response genes across 19 cell types (116,313 nuclei) from lung autopsies. The functional enrichment analysis of the 233 significantly expressed genes showed that the most relevant biological annotations were inflammatory response, innate immune response, cytokine production, interferon production, macrophage activation, blood coagulation, NLRP3 inflammasome complex, and the TLR, JAK-STAT, NF-κB, TNF, oncostatin M signaling pathways. Subsequently, we identified 34 essential inflammatory proteins with both high-confidence protein interactions and shortest pathways to inflammation, cell death, glycolysis, and angiogenesis. Conclusion: We propose three small molecules (baricitinib, eritoran, and montelukast) that can be considered for treating severe COVID-19 symptoms after being thoroughly evaluated in COVID-19 clinical trials.
Collapse
Affiliation(s)
- Andrés López-Cortés
- Programa de Investigación en Salud Global, Facultad de Ciencias de la Salud, Universidad Internacional SEK, Quito, Ecuador.,Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | - Santiago Guerrero
- Escuela de Medicina, Facultad de Ciencias Médicas de la Salud y de la Vida, Universidad Internacional del Ecuador, Quito, Ecuador
| | - Esteban Ortiz-Prado
- One Health Research Group, Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Verónica Yumiceba
- Institut für Humangenetik Lübeck, Universität zu Lübeck, Lübeck, Germany
| | - Antonella Vera-Guapi
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Ángela León Cáceres
- Heidelberg Institute of Global Health, Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | - Katherine Simbaña-Rivera
- One Health Research Group, Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador.,Latin American Network for Cancer Research (LAN-CANCER), Lima, Peru
| | - Ana María Gómez-Jaramillo
- Centro de Investigación para la Salud en América Latina (CISeAL), Pontificia Universidad Católica del Ecuador, Quito, Ecuador
| | - Gabriela Echeverría-Garcés
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | - Jennyfer M García-Cárdenas
- Escuela de Medicina, Facultad de Ciencias Médicas de la Salud y de la Vida, Universidad Internacional del Ecuador, Quito, Ecuador
| | - Patricia Guevara-Ramírez
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | | | | | | | | | - Isaac Armendáriz-Castillo
- Facultade de Ciencias, Universidade da Coruña, A Coruña, Spain.,Instituto Nacional de Investigación en Salud Pública, Quito, Ecuador
| | - Andy Pérez-Villa
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | | | | | | | | | - Álvaro A Pérez-Meza
- Biotechnology Engineering Career, Faculty of Life Sciences, Universidad Regional Amazónica Ikiam, Tena, Ecuador
| | | | - Andrea V Jácome
- Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | | | | | | | - Luis Abel Quiñones
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain.,Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Nikolaos C Kyriakidis
- One Health Research Group, Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| |
Collapse
|
30
|
Yue M, Hu M, Fu F, Ruan H, Wu C. Emerging Roles of Platelets in Allergic Asthma. Front Immunol 2022; 13:846055. [PMID: 35432313 PMCID: PMC9010873 DOI: 10.3389/fimmu.2022.846055] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 01/21/2023] Open
Abstract
Allergic asthma is a complex chronic inflammatory disease of the airways, driven by Th2 immune responses and characterized by eosinophilic pulmonary inflammation, airway hyperresponsiveness, excessive mucus production, and airway remodeling. Overwhelming evidence from studies in animal models and allergic asthmatic patients suggests that platelets are aberrantly activated and recruited to the lungs. It has been established that platelets can interact with other immune cells and secrete various biochemical mediators to promote allergic sensitization and airway inflammatory response, and platelet deficiency may alleviate the pathological features and symptoms of allergic asthma. However, the comprehensive roles of platelets in allergic asthma have not been fully clarified, leaving attempts to treat allergic asthma with antiplatelet agents questionable. In this review, we summarize the role of platelet activation and pulmonary accumulation in allergic asthma; emphasis is placed on the different interactions between platelets with crucial immune cell types and the contribution of platelet-derived mediators in this context. Furthermore, clinical antiplatelet approaches to treat allergic asthma are discussed. This review provides a clearer understanding of the roles of platelets in the pathogenesis of allergic asthma and could be informative in the development of novel strategies for the treatment of allergic asthma.
Collapse
Affiliation(s)
- Ming Yue
- Department of Physiology, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengjiao Hu
- Department of Immunology and Microbiology, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangda Fu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongfeng Ruan
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Hongfeng Ruan,
| | - Chengliang Wu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
31
|
Chutipongtanate S, Morrow AL, Newburg DS. Human Milk Oligosaccharides: Potential Applications in COVID-19. Biomedicines 2022; 10:biomedicines10020346. [PMID: 35203555 PMCID: PMC8961778 DOI: 10.3390/biomedicines10020346] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/25/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) has become a global health crisis with more than four million deaths worldwide. A substantial number of COVID-19 survivors continue suffering from long-COVID syndrome, a long-term complication exhibiting chronic inflammation and gut dysbiosis. Much effort is being expended to improve therapeutic outcomes. Human milk oligosaccharides (hMOS) are non-digestible carbohydrates known to exert health benefits in breastfed infants by preventing infection, maintaining immune homeostasis and nurturing healthy gut microbiota. These beneficial effects suggest the hypothesis that hMOS might have applications in COVID-19 as receptor decoys, immunomodulators, mucosal signaling agents, and prebiotics. This review summarizes hMOS biogenesis and classification, describes the possible mechanisms of action of hMOS upon different phases of SARS-CoV-2 infection, and discusses the challenges and opportunities of hMOS research for clinical applications in COVID-19.
Collapse
Affiliation(s)
- Somchai Chutipongtanate
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
- Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Mahidol University, Samut Prakan 10540, Thailand
- Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Ardythe L. Morrow
- Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children′s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - David S. Newburg
- Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
- Correspondence: or
| |
Collapse
|
32
|
Abstract
Toll-like receptors were discovered as proteins playing a crucial role in the dorsoventral patterning during embryonic development in the Drosophila melanogaster (D. melanogaster) almost 40 years ago. Subsequently, further research also showed a role of the Toll protein or Toll receptor in the recognition of Gram-positive bacterial and fungal pathogens infecting D. melanogaster. In 1997, the human homolog was reported and the receptor was named the Toll-like receptor 4 (TLR4) that recognizes lipopolysaccharide (LPS) of the Gram-negative bacteria as a pathogen-associated molecular pattern (PAMP). Identification of TLR4 in humans filled the long existing gap in the field of infection and immunity, addressing the mystery surrounding the recognition of foreign pathogens/microbes by the immune system. It is now known that mammals (mice and humans) express 13 different TLRs that are expressed on the outer cell membrane or intracellularly, and which recognize different PAMPs or microbe-associated molecular patterns (MAMPs) and death/damage-associated molecular patterns (DAMPs) to initiate the protective immune response. However, their dysregulation generates profound and prolonged pro-inflammatory immune responses responsible for different inflammatory and immune-mediated diseases. This chapter provides an overview of TLRs in the control of the immune response, their association with different diseases, including TLR single nucleotide polymorphisms (SNPs), interactions with microRNAs (miRs), use in drug development and vaccine design, and expansion in neurosciences to include pain, addiction, metabolism, reproduction, and wound healing.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - James E Barrett
- Drexel University College of Medicine, Philadelphia, PA, USA.
- Department of Neural Sciences, Centre for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
33
|
Alves HR, Lomba GSB, Gonçalves-de-Albuquerque CF, Burth P. Irisin, Exercise, and COVID-19. Front Endocrinol (Lausanne) 2022; 13:879066. [PMID: 35784579 PMCID: PMC9248970 DOI: 10.3389/fendo.2022.879066] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
Muscle and adipose tissue produce irisin during exercise. Irisin is thermogenic adipomyokine, improves glucose and lipid metabolism, and ameliorates the effects of obesity-driven inflammation, metabolic syndrome, and diabetes. In addition, exercise-induced irisin activates anti-inflammatory pathways and may play an essential role in improving the outcomes of inflammatory conditions, such as coronavirus disease (COVID-19). COVID-19 infection can activate different intracellular receptors and modulate various pathways during the course of the disease. The cytokine release storm (CRS) produced is significant because it promotes the context for systemic inflammation, which increases the risk of mortality in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV2). In addition, viral infection and the resulting organ damage may stimulate the mitogen-activated protein kinase(MAPK) and toll-like receptor 4 (TLR4)/toll interleukin receptor (TIR)-domain-containing adaptor (MyD88) pathways while negatively modulating the AMP-activated protein kinase (AMPK) pathway, leading to increased inflammatory cytokine production. Exercise-induced irisin may counteract this inflammatory modulation by decreasing cytokine production. Consequently, increased irisin levels, as found in healthy patients, may favor a better prognosis in patients with SARS-CoV2. This review aims to explore the molecular mechanisms underlying the anti-inflammatory properties of irisin in mitigating CRS and preventing severe outcomes due to infection with SARS-CoV2.
Collapse
Affiliation(s)
- Hugo Rodrigues Alves
- Department of Cell and Molecular Biology, Fluminense Federal University, Niterói, Brazil
| | | | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratory of Immunopharmacology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate Program in Biotechnology, Fluminense Federal University, Rio de Janeiro, Brazil
- *Correspondence: Patricia Burth, ; Cassiano Felippe Gonçalves-de-Albuquerque,
| | - Patricia Burth
- Department of Cell and Molecular Biology, Fluminense Federal University, Niterói, Brazil
- Postgraduate Program in Biotechnology, Fluminense Federal University, Rio de Janeiro, Brazil
- *Correspondence: Patricia Burth, ; Cassiano Felippe Gonçalves-de-Albuquerque,
| |
Collapse
|