1
|
Lv F, Zhou W, Li K. The roles of collectins in renal diseases and transplantation. Immunol Lett 2025; 271:106945. [PMID: 39542045 DOI: 10.1016/j.imlet.2024.106945] [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: 08/30/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
The collectins are soluble C-type lectins and a group of proteins characterized with common structural features: a collagen-like domain and a carbohydrate-binding domain. These proteins are essential components of the innate immune system, pivotal for recognizing and eliminating pathogens to protect against infections. Over recent decades, research has significantly advanced our understanding of collectins. Beyond their fundamental role in host defense, collectins have been emerged as multifunctional proteins involved in modulating inflammatory and immune responses, facilitating the clearance of cellular debris, and even stimulating cell proliferation. These diverse roles are critical for maintaining physiological balance and hold substantial implications in various disease processes, particularly in renal diseases and transplantation. Here, we review the roles of collectins in renal diseases and transplantation focusing on four prominent members of the collectin family: mannose-binding lectin (MBL), surfactant proteins (SP-A and SP-D), and collectin-11 (CL-11). These proteins have gained considerable attention in current research due to their roles in renal diseases and transplantation, shedding light on their impact beyond traditional immune defense mechanisms. Understanding their involvement in these contexts is crucial for exploring potential therapeutic avenues and interventions aimed at mitigating renal pathology and improving outcomes in transplantation settings.
Collapse
Affiliation(s)
- Fu Lv
- Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wuding Zhou
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK
| | - Ke Li
- Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Core Research Laboratory, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
2
|
Niri P, Saha A, Polopalli S, Kumar M, Das S, Saha B, Goyary D, Bhutia YD, Karmakar S, Kishor S, Rahaman S, Chattopadhyay P. β-Caryophyllene attenuates oxidative stress and inflammatory response in LPS induced acute lung injury by targeting ACE2/MasR and Nrf2/HO-1/NF-κB axis. Biochem Biophys Res Commun 2025; 746:151286. [PMID: 39756207 DOI: 10.1016/j.bbrc.2024.151286] [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: 07/15/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), is a clinical syndrome that can cause pulmonary edema, inflammation, oxidative stress, and immunological dysregulations. β-Caryophyllene (BCP), a natural bicyclic sesquiterpene, possesses a variety of pharmacological properties and has the potential to be a therapeutic agent. This study aimed to comprehend the effect of BCP on Nrf2/HO-1/NF-κB and ACE2/MasR axis in a rat model of ALI by lipopolysaccharide (LPS) and the underlying mechanisms during this process. The study also examined pulmonary edema, BALF, and cytokine production to investigate inflammation and oxidative stress. In the LPS group, Western blot analysis showed decreased Nrf2/HO-1 and ACE2/MasR, including increased lung edema, elevated vascular permeability, neutrophil infiltration in BALF, increased cytokine levels, and histological changes. In comparison to the LPS group, BCP dramatically reduced lung edema, vascular permeability, and histological changes. Additionally, by lowering malondialdehyde and myeloperoxidase activity in lung tissues, it also reduced oxidative stress. BCP boosted IL-10 production and decreased the levels of pro-inflammatory cytokines and neutrophil infiltration. BCP administration decreased VEGF-A and SP-D expression, subsequently lowering NF-κB activation and cytokine production. Further, BCP altered ACE2 expression, indicating its involvement by activating the ACE2/Angiotensin (1-7)/MasR axis. In addition, BCP could stimulate the Nrf2/HO-1 anti-oxidant axis to suppress NF-κB and reduce inflammation. BCP modulation of the ACE2/MasR and Nrf2/HO-1/NF-κB axis impedes the course of ALI by influencing immunological response including but not limited to oxidative stress, the influx of neutrophils, and cytokine production. Hence, BCP may act as a potential candidate for management of ALI.
Collapse
Affiliation(s)
- Pakter Niri
- Division of Pharmaceutical Technology, Defence Research Laboratory, DRDO, Tezpur, Assam, 784001, India; Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Subramanyam Polopalli
- Division of Pharmaceutical Technology, Defence Research Laboratory, DRDO, Tezpur, Assam, 784001, India; Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Mohit Kumar
- Division of Pharmaceutical Technology, Defence Research Laboratory, DRDO, Tezpur, Assam, 784001, India
| | - Sanghita Das
- Division of Pharmaceutical Technology, Defence Research Laboratory, DRDO, Tezpur, Assam, 784001, India; Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Bidisha Saha
- Division of Pharmaceutical Technology, Defence Research Laboratory, DRDO, Tezpur, Assam, 784001, India
| | - Danswrang Goyary
- Division of Pharmaceutical Technology, Defence Research Laboratory, DRDO, Tezpur, Assam, 784001, India
| | - Yangchen Doma Bhutia
- Division of Pharmaceutical Technology, Defence Research Laboratory, DRDO, Tezpur, Assam, 784001, India
| | - Sanjeev Karmakar
- Division of Pharmaceutical Technology, Defence Research Laboratory, DRDO, Tezpur, Assam, 784001, India
| | - Sumit Kishor
- Division of Pharmaceutical Technology, Defence Research Laboratory, DRDO, Tezpur, Assam, 784001, India
| | - Saidur Rahaman
- Division of Pharmaceutical Technology, Defence Research Laboratory, DRDO, Tezpur, Assam, 784001, India
| | - Pronobesh Chattopadhyay
- Division of Pharmaceutical Technology, Defence Research Laboratory, DRDO, Tezpur, Assam, 784001, India.
| |
Collapse
|
3
|
Shamim A, Abdul Aziz M, Saeed F, Kumari R, Mary Joseph A, Ponnachan P, Kishore U, Masmoudi K. Revisiting surfactant protein D: an immune surveillance molecule bridging innate and adaptive immunity. Front Immunol 2024; 15:1491175. [PMID: 39742280 PMCID: PMC11685232 DOI: 10.3389/fimmu.2024.1491175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/12/2024] [Indexed: 01/03/2025] Open
Abstract
Surfactant protein D (SP-D) is a C-type lectin that was originally discovered as a lung surfactant associated phospholipid recognising protein. It was originally shown to be of great importance in surfactant turnover and homeostasis in conjunction with another hydrophilic surfactant protein i.e. SP-A. In addition, it was found to agglutinate bacteria in suspension and likely a key defence molecule in the lungs. Since its early days of characterization in 1990s, SP-D has turned out to be a central player in the mucosal immunity as pulmonary as well as extrapulmonary innate immune molecule. The most exciting development has been characterization of its C-type lectin or carbohydrate recognition domain (CRDs) that exists in a homotrimeric form in native as well as recombinant versions. SP-D has a range of strategies to recognise pathogen-associated molecular patterns (PAMPs) and thus act as a soluble PAMP-recognizing receptor (PRR), and subsequent destruction of the pathogens directly, or indirectly via phagocytic cells. SP-D also recognizes a range of allergens, competes out with specific IgE antibodies, and downregulates histamine release by basophils and mast cells. These anti-microbial and anti-allergic properties of SP-D have been validated by in vivo murine models of infection and allergy. The SP-D gene deficient mice exhibit remarkable phenotypes where lungs are leaky, showing features of fibrosis and emphysema. One of the seminal discoveries in the field has been the observation that activated eosinophils (and other immune cells) can be induced into apoptotic pathways by SP-D. This raised the possibility that SP-D can be an innate immune surveillance molecule. Studies have revealed the ability of a recombinant fragment of human SP-D containing homotrimeric neck and CRD region to induce apoptosis via intrinsic as well as extrinsic pathways; in addition, it also seems capable of interfering with epithelial-to-mesenchymal transition. These studies have opened up enormous possibilities for setting up pre-clinical and clinical trials.
Collapse
Affiliation(s)
- Azra Shamim
- Department Integrative Agriculture, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mughair Abdul Aziz
- Department Integrative Agriculture, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Faryal Saeed
- Department Integrative Agriculture, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rekha Kumari
- Department of Zoology, A.N College, Patliputra University, Patna, Bihar, India
| | - Ann Mary Joseph
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Pretty Ponnachan
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Uday Kishore
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Khaled Masmoudi
- Department Integrative Agriculture, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
4
|
Zhao Q, Zhang R, Wang Y, Li T, Xue J, Chen Z. FOXQ1, deubiquitinated by USP10, alleviates sepsis-induced acute kidney injury by targeting the CREB5/NF-κB signaling axis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167331. [PMID: 38960057 DOI: 10.1016/j.bbadis.2024.167331] [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: 11/22/2023] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Sepsis-induced acute kidney injury (S-AKI) is a severe and frequent complication that occurs during sepsis. This study aimed to understand the role of FOXQ1 in S-AKI and its potential upstream and downstream regulatory mechanisms. A cecal ligation and puncture induced S-AKI mouse model in vivo and an LPS-induced HK-2 cell model in vitro were used. FOXQ1 was significantly upregulated in CLP mice and downregulated in the LPS-induced HK-2 cells. Upregulation of FOXQ1 improved kidney injury and dysfunction in CLP mice. Overexpression of FOXQ1 remarkably suppressed the apoptosis and inflammatory response via down-regulating oxidative stress indicators and pro-inflammatory factors (IL-1β, IL-6, and TNF-α), both in vivo and in vitro. From online analysis, the CREB5/NF-κB axis was identified as the downstream target of FOXQ1. FOXQ1 transcriptionally activated CREB5, upregulating its expression. Overexpression of FOXQ1 suppressed the phosphorylation level and nucleus transport of p65. Rescue experiments showed that CREB5 mediates the protective role of FOXQ1 on S-AKI. Furthermore, FOXQ1 was identified as a substrate of USP10, a deubiquitinating enzyme. Ectopic expression of USP10 reduced the ubiquitination of FOXQ1, promoting its protein stability. USP10 upregulation alleviated LPS-induced cell apoptosis and inflammatory response, while suppression of FOXQ1 augmented these trends. Collectively, our results suggest that FOXQ1, deubiquitinated by USP10, plays a protective role in S-AKI induced inflammation and apoptosis by targeting CREB5/NF-κB axis.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ran Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yu Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Zhiguang Chen
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
5
|
Jacob IB, Lawal AO, Mahmoud SS, Kopsack EM, Reynolds ES, Meng Q, Fan H, Massa PT, Thangamani S, Jia H, Wang G. Differential Immunoregulation by Human Surfactant Protein A Variants Determines Severity of SARS-CoV-2-induced Lung Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612497. [PMID: 39314485 PMCID: PMC11418998 DOI: 10.1101/2024.09.11.612497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
COVID-19 remains a significant threat to public health globally. Infection in some susceptible individuals causes life-threatening acute lung injury (ALI/ARDS) and/or death. Human surfactant protein A (SP-A) is a C-type lectin expressed in the lung and other mucosal tissues, and it plays a critical role in host defense against various pathogens. The human SP-A genes ( SFTPA1 and SFTPA2 ) are highly polymorphic and comprise several common genetic variants, i.e., SP-A1 (variants 6A 2 , 6A 4 ) and SP-A2 (variants 1A 0 , 1A 3 ). Here, we elucidated the differential antiviral and immunoregulatory roles of SP-A variants in response to SARS-CoV-2 infection in vivo . Six genetically-modified mouse lines, expressing both hACE2 (SARS-CoV-2 receptor) and individual SP-A variants: (hACE2/6A 2 (6A 2 ), hACE2/6A 4 (6A 4 ), hACE2/1A 0 (1A 0 ), and hACE2/1A 3 (1A 3 ), one SP-A knockout (hACE2/SP-A KO (KO) and one hACE2/mouse SP-A (K18) mice, were challenged intranasally with 10 3 PFU SARS-CoV-2 or saline (Sham). Infected KO and 1A 0 mice had more weight loss and mortality compared to other mouse lines. Relative to other infected mouse lines, a more severe ALI was observed in KO, 1A 0 , and 6A 2 mice. Reduced viral titers were generally observed in the lungs of infected SP-A mice relative to KO mice. Transcriptomic analysis revealed an upregulation in genes that play central roles in immune responses such as MyD88 , Stat3 , IL-18 , and Jak2 in the lungs of KO and 1A 0 mice. However, Mapk1 was significantly downregulated in 6A 2 versus 1A 0 mice. Analysis of biological pathways identified those involved in lung host defense and innate immunity, including pathogen-induced cytokine, NOD1/2, and Trem1 signaling pathways. Consistent with the transcriptomic data, levels of cytokines and chemokines such as G-CSF, IL-6 and IL-1β were comparatively higher in the lungs and sera of KO and 1A 0 mice with the highest mortality rate. These findings demonstrate that human SP-A variants differentially modulate SARS-CoV-2-induced lung injury and disease severity by differentially inhibiting viral infectivity and regulating immune-related gene expressions.
Collapse
|
6
|
Chen LL, Weng H. Clinical significance of lower respiratory tract culture within 48 h of admission in patients with viral pneumonia: an observational study. BMC Pulm Med 2024; 24:372. [PMID: 39085852 PMCID: PMC11293002 DOI: 10.1186/s12890-024-03162-y] [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: 04/12/2023] [Accepted: 07/11/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND The aim of this retrospective study was to examine the risk factors of positive lower respiratory tract cultures and to investigate whether nosocomial infections are common in patients with positive lower respiratory tract cultures. METHODS We enrolled 86 patients diagnosed with influenza A-related critical illness who were treated at Fuzhou Pulmonary Hospital of Fujian in China between 1st October 2013 and 31st March 2019. The of admission were used to divide the enrolled patients into two groups. Sputum and bronchoalveolar lavage fluid specimens were collected within 48 h after admission for culture. All samples were cultured immediately after sampling. Nosocomial infections are defined as any symptom or sign of pulmonary infiltration, confirmed by X-ray, after 5 days of admission and positive results from one or more cultures. RESULTS The average age of this cohort was (54.13 ± 16.52) years. Based on the culture results, Staphylococcus aureus and Candida albicans had the highest positive rates (3.40% (3/86) and 20.90% (18/86), respectively). In patients with positive lower respiratory tract cultures, the incidence of nosocomial infection was 73.30% (22/30) five days after admission. However, the incidence of nosocomial infection was lower (42.80%, 24/56) in patients with negative lower respiratory tract cultures. Hemoptysis, systolic pressure at admission, and blood urea nitrogen level at admission were all independent risk factors for positive lower respiratory tract cultures within 48 h of admission. CONCLUSION Our data showed that a significant proportion of patients with pneumonia exhibited co-infections with bacteria or fungi within five days of hospital admission. Hemoptysis, systolic pressure, and blood urea nitrogen levels at admission emerged as the key risk factors. These findings underscore the necessity of closely monitoring patients with influenza infection, particularly for positive bacterial or fungal cultures within the initial 48 h of admission.
Collapse
Affiliation(s)
- Lu-Lu Chen
- Department of Respiratory Diseases, People' Hospital Affiliated to Fujian University of Traditional Chinese Medicine, No 602, 817 Middle Road, Taijiang District, Fuzhou, 350009, China
| | - Heng Weng
- Department of Respiratory Diseases, People' Hospital Affiliated to Fujian University of Traditional Chinese Medicine, No 602, 817 Middle Road, Taijiang District, Fuzhou, 350009, China.
| |
Collapse
|
7
|
Guo J, Chen X, Wang C, Ruan F, Xiong Y, Wang L, Abdel-Razek O, Meng Q, Shahbazov R, Cooney RN, Wang G. LIRAGLUTIDE ALLEVIATES ACUTE LUNG INJURY AND MORTALITY IN PNEUMONIA-INDUCED SEPSIS THROUGH REGULATING SURFACTANT PROTEIN EXPRESSION AND SECRETION. Shock 2024; 61:601-610. [PMID: 38150354 PMCID: PMC11009087 DOI: 10.1097/shk.0000000000002285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
ABSTRACT Glucagon-like peptide 1 (GLP-1) analogs are used to treat type 2 diabetes, and they can regulate insulin secretion, energy homeostasis, inflammation, and immune cell function. This study sought to determine whether the GLP-1 analog liraglutide exerts a beneficial action in an acute lung injury model of pneumonia-induced sepsis. Methods: Wild-type FVB/NJ mice (n = 114) were infected by intratracheal injection with Pseudomonas aeruginosa Xen5 (4 × 10 4 CFU/mouse) or an equal volume (50 μL) of saline (control) with or without a subcutaneous injection of liraglutide (2 mg/kg, 30 min after infection). Mice were killed 24 h after infection. Lung tissues and BALF were analyzed. In separate experiments, the dynamic growth of bacteria and animal mortality was monitored using in vivo imaging system within 48 h after infection. In addition, primary lung alveolar type II cells isolated from mice were used to study the mechanism of liraglutide action. Result: Liraglutide improved survival ( P < 0.05), decreased bacterial loads in vivo , and reduced lung injury scores ( P < 0.01) in septic mice. Liraglutide-treated mice showed decreased levels of inflammatory cells ( P < 0.01) and proinflammatory cytokines (TNF-α and IL-6) ( P < 0.01) in the lung compared with septic controls. Liraglutide significantly increased pulmonary surfactant proteins (SP-A and SP-B) expression/secretion ( P < 0.01) and phospholipid secretion ( P < 0.01) in vivo . Primary alveolar type II cells pretreated with liraglutide improved SP-A and SP-B expression after LPS exposure ( P < 0.01). Conclusion: Liraglutide attenuates mortality and lung inflammation/injury in pneumonia-induced sepsis. The increased surfactant expression/secretion and anti-inflammatory effects of liraglutide represent potential mechanisms by GLP-1 agonists potentiate host defense and maintain alveolar respiratory function in acute lung injury.
Collapse
Affiliation(s)
- Junping Guo
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Rainbowfish Rehabilitation & Nursing School, Hangzhou Vocational & Technical College, Hangzhou 310018, China
| | - Xinghua Chen
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Nephrology, Wuhan University, Renmin Hospital, Wuhan 430060, China
| | - Cole Wang
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Feng Ruan
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Yunhe Xiong
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Lijun Wang
- Department of Endocrinology, Zhejiang Provincial People’s Hospital, Hangzhou 310014, China
| | - Osama Abdel-Razek
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Qinghe Meng
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Rauf Shahbazov
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Robert N Cooney
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Guirong Wang
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
8
|
Jacob IB, Gemmiti A, Xiong W, Reynolds E, Nicholas B, Thangamani S, Jia H, Wang G. Human surfactant protein A inhibits SARS-CoV-2 infectivity and alleviates lung injury in a mouse infection model. Front Immunol 2024; 15:1370511. [PMID: 38596675 PMCID: PMC11002091 DOI: 10.3389/fimmu.2024.1370511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
Introduction SARS coronavirus 2 (SARS-CoV-2) infects human angiotensin-converting enzyme 2 (hACE2)-expressing lung epithelial cells through its spike (S) protein. The S protein is highly glycosylated and could be a target for lectins. Surfactant protein A (SP-A) is a collagen-containing C-type lectin, expressed by mucosal epithelial cells and mediates its antiviral activities by binding to viral glycoproteins. Objective This study examined the mechanistic role of human SP-A in SARS-CoV-2 infectivity and lung injury in vitro and in vivo. Results Human SP-A can bind both SARS-CoV-2 S protein and hACE2 in a dose-dependent manner (p<0.01). Pre-incubation of SARS-CoV-2 (Delta) with human SP-A inhibited virus binding and entry and reduced viral load in human lung epithelial cells, evidenced by the dose-dependent decrease in viral RNA, nucleocapsid protein (NP), and titer (p<0.01). We observed significant weight loss, increased viral burden, and mortality rate, and more severe lung injury in SARS-CoV-2 infected hACE2/SP-A KO mice (SP-A deficient mice with hACE2 transgene) compared to infected hACE2/mSP-A (K18) and hACE2/hSP-A1 (6A2) mice (with both hACE2 and human SP-A1 transgenes) 6 Days Post-infection (DPI). Furthermore, increased SP-A level was observed in the saliva of COVID-19 patients compared to healthy controls (p<0.05), but severe COVID-19 patients had relatively lower SP-A levels than moderate COVID-19 patients (p<0.05). Discussion Collectively, human SP-A attenuates SARS-CoV-2-induced acute lung injury (ALI) by directly binding to the S protein and hACE2, and inhibiting its infectivity; and SP-A level in the saliva of COVID-19 patients might serve as a biomarker for COVID-19 severity.
Collapse
Affiliation(s)
- Ikechukwu B. Jacob
- Department of Surgery, the State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Amanda Gemmiti
- Department of Otolaryngology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Weichuan Xiong
- Department of Surgery, the State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Erin Reynolds
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Brian Nicholas
- Department of Otolaryngology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Saravanan Thangamani
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Hongpeng Jia
- Department of Surgery, Johns-Hopkins University, Baltimore, MD, United States
| | - Guirong Wang
- Department of Surgery, the State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
9
|
Liu T, Han R, Yan Y. Preliminary study on molecular mechanism of COVID-19 intervention by Polygonum cuspidatum through computer bioinformatics. Medicine (Baltimore) 2024; 103:e36918. [PMID: 38215091 PMCID: PMC10783314 DOI: 10.1097/md.0000000000036918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 12/19/2023] [Indexed: 01/14/2024] Open
Abstract
To explore the mechanism of action of Polygonum cuspidatum in intervening in coronavirus disease 2019 using a network pharmacology approach and to preliminarily elucidate its mechanism. The active ingredients and action targets of P cuspidatum were classified and summarized using computer virtual technology and molecular informatics methods. The active ingredients and relevant target information of P cuspidatum were identified using the TCM Systematic Pharmacology Database and Analysis Platform, the TCM Integrated Pharmacology Research Platform v2.0, and the SwissTarget database. The GENECARDS database was used to search for COVID-19 targets. The STRING database was analyzed and combined with Cytoscape 3.7.1 software to construct a protein interaction network map to screen the core targets. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis was then performed. The core compound, polydatin, was selected and the core targets were analyzed by computer virtual docking using software such as discovery studio autodock tool. In vitro cell models were constructed to experimentally validate the activity of the core compound, polydatin. By computer screening, we identified 9 active ingredients and their corresponding 286 targets from P cuspidatum. A search of the GENECARDS database for COVID-19 yielded 303 core targets. By mapping the active ingredient targets to the disease targets, 27 overlapping targets could be extracted as potential targets for the treatment of COVID-19 with P cuspidatum. In addition, the enrichment analysis of Kyoto Encyclopedia of Genes and Genomes pathway on core targets showed that the coronavirus disease, MAPK signaling pathway, NF kappa B signaling pathway, and other signaling pathways were highly enriched. Combined with the degree-high target analysis in the protein interaction network, it was found to be mainly concentrated in the NF-kappaB (NF-κB) signaling pathway, indicating that the NF-κB signaling pathway may be an important pathway for P cuspidatum intervention. In vitro assays showed no effect of 0.1 to 10 μM polydatin on cell viability, but an inhibitory effect on the transcriptional activity of NF-κB-RE. Molecular docking showed stable covalent bonding of polydatin molecules with Il-1β protein at residue leu-26, TNF protein ser-60, residue gly-121, and residue ile-258 of ICAM-1 protein, indicating a stable docking result. The treatment of COVID-19 with P cuspidatum is characterized by multi-component, multi-target, and multi-pathway, which can exert a complex network of regulatory effects through the interaction between different targets, providing a new idea and basis for further exploration of the mechanism of action of P cuspidatum in the treatment of COVID-19.
Collapse
Affiliation(s)
- Tao Liu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Han
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiqi Yan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
10
|
Hu Y, He S, Xu X, Cui X, Wei Y, Zhao C, Ye H, Zhao J, Liu Q. Shenhuangdan decoction alleviates sepsis-induced lung injury through inhibition of GSDMD-mediated pyroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117047. [PMID: 37586442 DOI: 10.1016/j.jep.2023.117047] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/29/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sepsis-induced lung injury is closely associated with it remarkable morbidity and mortality. Shenhuangdan (SHD) decoction, a traditional Chinese medicine prescription, has been clinically proven to be an effective treatment for sepsis. However, the mechanism of SHD decoction in treating sepsis remains unclear. AIM OF THE STUDY This study aimed to evaluate the therapeutic effect of SHD decoction on sepsis-induced lung injury and its underlying mechanism. MATERIALS AND METHODS In this study, we established a mouse model of sepsis by cecum ligation and puncture (CLP) surgery. Firstly, seven-day survival analysis and histological staining of lung tissue were used to evaluate the therapeutic effect of SHD decoction on lung injury in septic mice. Multifactor microarray was used to detect cytokine expression changes in serum and bronchoalveolar lavage fluid (BALF). Subsequently, the main components in medicated serum of SHD decoction were inspected by Ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). The material basis of SHD decoction and potential interaction mechanisms were analysed by systemic pharmacology. To confirm the reliability of network pharmacology for predicting outcomes, we used molecular docking techniques to identify interactions between the core components and targets of SHD. Finally, TUNEL staining, immunofluorescence and western blotting were used to explore the mechanism of SHD decoction through the inhibition of GSDMD-mediated pyroptosis in septic mice. RESULTS SHD was found to be effective in reducing the mortality and alleviating lung pathological damage in septic mice. Multifactor microarray results showed that SHD can reduce the expression of inflammation factors (IL-18, IL-1β, IL-5, IL-6 and TNF-α) in serum and BALF of septic mice. There were 22 major blood-entry components detected by UPLC-MS/MS. Then, combined with the network pharmacological analysis, it is evident that the main components of SHD for sepsis are Renshen-ginsenoside Rh2, Danshen-tanshinone IIA and Dahuang-rhein. The main targets were IL-1β and caspase-1, which were related to GSDMD-mediated pyroptosis signalling pathway. Molecular docking exhibited that Renshen-ginsenoside Rh2, Danshen-tanshinone IIA and Dahuang-rhein can closely bind to GSDMD, IL-1β and caspase-1. In addition, TUNEL staining and immunohistochemistry demonstrated that SHD alleviated the expression of GSDMD protein. The western blotting showed that SHD significantly inhibited the protein expression levels of NLRP3, GSDMD, GSDMD-N, cleved caspase-1, caspase-1 and ASC in lung tissue. CONCLUSIONS Our study revealed that SHD improves CLP-induced lung injury by blocking the GSDMD-mediated pyroptosis signalling pathway in septic mice. This study provides evidence to support that SHD had a potential therapeutic effect on sepsis.
Collapse
Affiliation(s)
- Yahui Hu
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China; Beijing Institute of Chinese Medicine, Beijing 100010, China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China.
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China; Beijing Institute of Chinese Medicine, Beijing 100010, China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China.
| | - Xiaolong Xu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China; Beijing Institute of Chinese Medicine, Beijing 100010, China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China.
| | - Xuran Cui
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China; Beijing Institute of Chinese Medicine, Beijing 100010, China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China.
| | - Yiming Wei
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China; Beijing Institute of Chinese Medicine, Beijing 100010, China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China; Beijing University of Chinese Medicine, Beijing 100010, China.
| | - Chunxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China; Beijing Institute of Chinese Medicine, Beijing 100010, China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China; Capital Medical University, Beijing 100069, China.
| | - Haoran Ye
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China; Beijing Institute of Chinese Medicine, Beijing 100010, China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China; Capital Medical University, Beijing 100069, China.
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China; Beijing Institute of Chinese Medicine, Beijing 100010, China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China.
| | - Qingquan Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China; Beijing Institute of Chinese Medicine, Beijing 100010, China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China.
| |
Collapse
|
11
|
Elmore A, Almuntashiri A, Wang X, Almuntashiri S, Zhang D. Circulating Surfactant Protein D: A Biomarker for Acute Lung Injury? Biomedicines 2023; 11:2517. [PMID: 37760958 PMCID: PMC10525947 DOI: 10.3390/biomedicines11092517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening lung diseases in critically ill patients. The lack of prognostic biomarkers has halted detection methods and effective therapy development. Quantitative biomarker-based approaches in the systemic circulation have been proposed as a means of enhancing diagnostic strategies as well as pharmacotherapy in a patient-specific manner. Pulmonary surfactants are complex mixtures made up of lipids and proteins, which are secreted into the alveolar space by epithelial type II cells under normal and pathological conditions. In this review, we summarize the current knowledge of SP-D in lung injury from both preclinical and clinical studies. Among surfactant proteins, surfactant protein-D (SP-D) has been more widely studied in ALI and ARDS. Recent studies have reported that SP-D has a superior discriminatory ability compared to other lung epithelial proteins for the diagnosis of ARDS, which could reflect the severity of lung injury. Furthermore, we shed light on recombinant SP-D treatment and its benefits as a potential drug for ALI, and we encourage further studies to translate SP-D into clinical use for diagnosis and treatment.
Collapse
Affiliation(s)
- Alyssa Elmore
- College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | - Ali Almuntashiri
- Department of Dentistry, Security Forces Hospital, Dammam 32314, Saudi Arabia
- Department of Preventive Dentistry, College of Dentistry, Qassim University, Ar Rass 52571, Saudi Arabia
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA (D.Z.)
| | - Sultan Almuntashiri
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA (D.Z.)
- Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail 55473, Saudi Arabia
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA (D.Z.)
| |
Collapse
|
12
|
Emin MT, Lee MJ, Bhattacharya J, Hough RF. Mitochondria of lung venular capillaries mediate lung-liver cross talk in pneumonia. Am J Physiol Lung Cell Mol Physiol 2023; 325:L277-L287. [PMID: 37431588 PMCID: PMC10625830 DOI: 10.1152/ajplung.00209.2022] [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: 06/29/2022] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/12/2023] Open
Abstract
Failure of the lung's endothelial barrier underlies lung injury, which causes the high mortality acute respiratory distress syndrome (ARDS). Multiple organ failure predisposes to the mortality, but mechanisms are poorly understood. Here, we show that mitochondrial uncoupling protein 2 (UCP2), a component of the mitochondrial inner membrane, plays a role in the barrier failure. Subsequent lung-liver cross talk mediated by neutrophil activation causes liver congestion. We intranasally instilled lipopolysaccharide (LPS). Then, we viewed the lung endothelium by real-time confocal imaging of the isolated, blood-perfused mouse lung. LPS caused alveolar-capillary transfer of reactive oxygen species and mitochondrial depolarization in lung venular capillaries. The mitochondrial depolarization was inhibited by transfection of alveolar Catalase and vascular knockdown of UCP2. LPS instillation caused lung injury as indicated by increases in bronchoalveolar lavage (BAL) protein content and extravascular lung water. LPS or Pseudomonas aeruginosa instillation also caused liver congestion, quantified by liver hemoglobin and plasma aspartate aminotransferase (AST) increases. Genetic inhibition of vascular UCP2 prevented both lung injury and liver congestion. Antibody-mediated neutrophil depletion blocked the liver responses, but not lung injury. Knockdown of lung vascular UCP2 mitigated P. aeruginosa-induced mortality. Together, these data suggest a mechanism in which bacterial pneumonia induces oxidative signaling to lung venular capillaries, known sites of inflammatory signaling in the lung microvasculature, depolarizing venular mitochondria. Successive activation of neutrophils induces liver congestion. We conclude that oxidant-induced UCP2 expression in lung venular capillaries causes a mechanistic sequence leading to liver congestion and mortality. Lung vascular UCP2 may present a therapeutic target in ARDS.NEW & NOTEWORTHY We report that mitochondrial injury in lung venular capillaries underlies barrier failure in pneumonia, and venular capillary uncoupling protein 2 (UCP2) causes neutrophil-mediated liver congestion. Using in situ imaging, we found that epithelial-endothelial transfer of H2O2 activates UCP2, depolarizing mitochondria in venular capillaries. The conceptual advance from our findings is that mitochondrial depolarization in lung capillaries mediates liver cross talk through circulating neutrophils. Pharmacologic blockade of UCP2 could be a therapeutic strategy for lung injury.
Collapse
Affiliation(s)
- Memet T Emin
- Department of Pediatrics, Pediatric Critical Care and Hospital Medicine, Columbia University Irving Medical Center, New York, New York, United States
| | - Michael J Lee
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States
| | - Jahar Bhattacharya
- Lung Biology Laboratory, Pulmonary Division, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, United States
| | - Rebecca F Hough
- Department of Pediatrics, Pediatric Critical Care and Hospital Medicine, Columbia University Irving Medical Center, New York, New York, United States
- Lung Biology Laboratory, Pulmonary Division, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, United States
| |
Collapse
|
13
|
Chen X, Guo J, Mahmoud S, Vanga G, Liu T, Xu W, Xiong Y, Xiong W, Abdel-Razek O, Wang G. Regulatory roles of SP-A and exosomes in pneumonia-induced acute lung and kidney injuries. Front Immunol 2023; 14:1188023. [PMID: 37256132 PMCID: PMC10225506 DOI: 10.3389/fimmu.2023.1188023] [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: 03/16/2023] [Accepted: 05/02/2023] [Indexed: 06/01/2023] Open
Abstract
Introduction Pneumonia-induced sepsis can cause multiple organ dysfunction including acute lung and kidney injury (ALI and AKI). Surfactant protein A (SP-A), a critical innate immune molecule, is expressed in the lung and kidney. Extracellular vesicles like exosomes are involved in the processes of pathophysiology. Here we tested one hypothesis that SP-A regulates pneumonia-induced AKI through the modulation of exosomes and cell death. Methods Wild-type (WT), SP-A knockout (KO), and humanized SP-A transgenic (hTG, lung-specific SP-A expression) mice were used in this study. Results After intratracheal infection with Pseudomonas aeruginosa, KO mice showed increased mortality, higher injury scores, more severe inflammation in the lung and kidney, and increased serum TNF-α, IL-1β, and IL-6 levels compared to WT and hTG mice. Infected hTG mice exhibited similar lung injury but more severe kidney injury than infected WT mice. Increased renal tubular apoptosis and pyroptosis in the kidney of KO mice were found when compared with WT and hTG mice. We found that serum exosomes from septic mice cause ALI and AKI through mediating apoptosis and proptosis when mice were injected intravenously. Furthermore, primary proximal tubular epithelial cells isolated from KO mice showed more sensitivity than those from WT mice after exposure to septic serum exosomes. Discussion Collectively, SP-A attenuates pneumonia-induced ALI and AKI by regulating inflammation, apoptosis and pyroptosis; serum exosomes are important mediators in the pathogenesis of AKI.
Collapse
Affiliation(s)
- Xinghua Chen
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
- Department of Nephrology, Wuhan University, Renmin Hospital, Wuhan, Hubei, China
| | - Junping Guo
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Salma Mahmoud
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Gautam Vanga
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Tianyi Liu
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Wanwen Xu
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Yunhe Xiong
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Weichuan Xiong
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Osama Abdel-Razek
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Guirong Wang
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
14
|
Ding F, Zhu J, Hu Y. Circular RNA protein tyrosine kinase 2 aggravates pyroptosis and inflammation in septic lung tissue by promoting microRNA-766/eukaryotic initiation factor 5A axis-mediated ATP efflux. Acta Cir Bras 2023; 38:e380323. [PMID: 36888755 PMCID: PMC10037555 DOI: 10.1590/acb380323] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/06/2023] [Indexed: 03/08/2023] Open
Abstract
PURPOSE Sepsis is characterized by an acute inflammatory response to infection, often with multiple organ failures, especially severe lung injury. This study was implemented to probe circular RNA (circRNA) protein tyrosine kinase 2 (circPTK2)-associated regulatory mechanisms in septic acute lung injury (ALI). METHODS A cecal ligation and puncture-based mouse model and an lipopolysaccharides (LPS)-based alveolar type II cell (RLE-6TN) model were generated to mimic sepsis. In the two models, inflammation- and pyroptosis-related genes were measured. RESULTS The degree of lung injury in mice was analyzed by hematoxylin and eosin (H&E) staining and the apoptosis was by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining. In addition, pyroptosis and toxicity were detected in cells. Finally, the binding relationship between circPTK2, miR-766, and eukaryotic initiation factor 5A (eIF5A) was detected. Data indicated that circPTK2 and eIF5A were up-regulated and miR-766 was down-regulated in LPS-treated RLE-6TN cells and lung tissue of septic mice. Lung injury in septic mice was ameliorated after inhibition of circPTK2. CONCLUSIONS It was confirmed in the cell model that knockdown of circPTK2 effectively ameliorated LPS-induced ATP efflux, pyroptosis, and inflammation. Mechanistically, circPTK2 mediated eIF5A expression by competitively adsorbing miR-766. Taken together, circPTK2/miR-766/eIF5A axis ameliorates septic ALI, developing a novel therapeutic target for the disease.
Collapse
Affiliation(s)
- FuYan Ding
- Zhengzhou University - Central China Fuwai Hospital - Department of Adult Cardiovascular Surgical Intensive Care Unit - Zhengzhou (Henan), China
| | - JiaLu Zhu
- Zhengzhou University - Central China Fuwai Hospital - Department of Adult Cardiovascular Surgical Intensive Care Unit - Zhengzhou (Henan), China
| | - YanLei Hu
- Zhengzhou University - Central China Fuwai Hospital - Department of Adult Cardiovascular Surgical Intensive Care Unit - Zhengzhou (Henan), China
| |
Collapse
|
15
|
Jian Y, Yang Y, Cheng L, Yang X, Liu H, Li W, Wan Y, Yang D. Sirt3 mitigates LPS-induced mitochondrial damage in renal tubular epithelial cells by deacetylating YME1L1. Cell Prolif 2022; 56:e13362. [PMID: 36433732 PMCID: PMC9890524 DOI: 10.1111/cpr.13362] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/21/2022] [Accepted: 11/01/2022] [Indexed: 11/28/2022] Open
Abstract
Acute kidney injury (AKI) is often secondary to sepsis. Increasing evidence suggests that mitochondrial dysfunction contributes to the pathological process of AKI. In this study, we aimed to examine the regulatory roles of Sirt3 in Lipopolysaccharide (LPS)-induced mitochondrial damage in renal tubular epithelial cells (TECs). Sirt3 knockout mice were intraperitoneally injected with LPS, and cultured TECs were stimulated with LPS to evaluate the effects of Sirt3 on mitochondrial structure and function in TECs. Electron microscopy was used to assess mitochondrial morphology. Immunofluorescence staining was performed to detect protein expression and examine mitochondrial morphology. Western blotting was used to quantify protein expression. We observed that LPS increased apoptosis, induced disturbances in mitochondrial function and dynamics, and downregulated Sirt3 expression in a sepsis-induced AKI mouse model and human proximal tubular (HK-2) cells in vitro. Sirt3 deficiency further exacerbated LPS-induced renal pathological damage, apoptosis and disturbances in mitochondrial function and dynamics. On the contrary, Sirt3 overexpression in HK-2 cells alleviated these lesions. Functional studies revealed that Sirt3 overexpression alleviated LPS-induced mitochondrial damage and apoptosis in TECs by promoting OPA1-mediated mitochondrial fusion through the deacetylation of i-AAA protease (YME1L1), an upstream regulatory molecule of OPA1. Our study has identified Sirt3 as a vital factor that protects against LPS-induced mitochondrial damage and apoptosis in TECs via the YME1L1-OPA1 signaling pathway.
Collapse
Affiliation(s)
- Yonghong Jian
- Department of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina,Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| | - Yifei Yang
- Department of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina,Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| | - Lingli Cheng
- Department of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina,Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| | - Xueyan Yang
- Department of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina,Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| | - Hongyan Liu
- Department of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina,Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| | - Wei Li
- Department of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina,Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| | - Yuhan Wan
- Department of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina,Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| | - Dingping Yang
- Department of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina,Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| |
Collapse
|
16
|
Gregor C. Generation of bright autobioluminescent bacteria by chromosomal integration of the improved lux operon ilux2. Sci Rep 2022; 12:19039. [PMID: 36351939 PMCID: PMC9646698 DOI: 10.1038/s41598-022-22068-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/10/2022] [Indexed: 11/10/2022] Open
Abstract
The bacterial bioluminescence system enables the generation of light by living cells without the requirement of an external luciferin. Due to the relatively low light emission, many applications of bioluminescence imaging would benefit from an increase in brightness of this system. In this report, a new approach of mutagenesis and screening of the involved proteins is described that is based on the identification of mutants with improved properties under rate-limiting reaction conditions. Multiple rounds of screening in Escherichia coli resulted in the operon ilux2 that contains 26 new mutations in the fatty acid reductase complex which provides the aldehyde substrate for the bioluminescence reaction. Chromosomal integration of ilux2 yielded an autonomously bioluminescent E. coli strain with sixfold increased brightness compared to the previously described ilux operon. The ilux2 strain produces sufficient signal for the robust detection of individual cells and enables highly sensitive long-term imaging of bacterial propagation without a selection marker.
Collapse
Affiliation(s)
- Carola Gregor
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Department of Optical Nanoscopy, Institut für Nanophotonik Göttingen e.V., Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
17
|
Ruan F, Chen J, Yang J, Wang G. MILD TRAUMATIC BRAIN INJURY ATTENUATES PNEUMONIA-INDUCED LUNG INJURY BY MODULATIONS OF ALVEOLAR MACROPHAGE BACTERICIDAL ACTIVITY AND M1 POLARIZATION. Shock 2022; 58:400-407. [PMID: 36166827 PMCID: PMC9712263 DOI: 10.1097/shk.0000000000001989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
ABSTRACT Traumatic brain injury is one of the main causes of death and disability worldwide, and results in multisystem complications. However, the mechanism of mild traumatic brain injury (MTBI) on lung injury remains unclear. In this study, we used a murine model of MTBI and pneumonia ( Pseudomonas aeruginosa ;) to explore the relationship between these conditions and the underlying mechanism. Methods: Mice (n = 104) were divided into control, MTBI, pneumonia, and MTBI + pneumonia groups. MTBI was induced by the weight-drop method. Pneumonia was induced by intratracheal injection with P. aeruginosa Xen5 strain. Animals were killed 24 h after bacterial challenging. Histological, cellular, and molecular indices of brain and lung injury were assessed using various methods. Results: Mice in both the MTBI and pneumonia groups had more Fluoro-Jade C-positive neurons than did the controls ( P < 0.01), but mice in the MTBI + pneumonia group had fewer Fluoro-Jade C-positive cells than did the pneumonia group ( P < 0.01). The MTBI + pneumonia mice showed decreased bacterial load ( P < 0.05), reduced lung injury score and pulmonary permeability ( P < 0.01), less inflammatory cells, and lower levels of proinflammatory cytokines (TNF-α and IL-1β; P < 0.01) when compared with the pneumonia group. Molecular analysis indicated lower levels of phosphorylated nuclear factor-κB in the lung of MTBI + pneumonia mice compared with the pneumonia group ( P < 0.01). Furthermore, alveolar macrophages from MTBI mice exhibited enhanced bactericidal capacity compared with those from controls ( P < 0.01). Moreover, MTBI + pneumonia mice exhibited less CD86-positive M1 macrophages compared with the pneumonia group ( P < 0.01). Conclusions: MTBI attenuates pneumonia-induced acute lung injury through the modulation of alveolar macrophage bactericidal capacity and M1 polarization in bacterial pneumonia model.
Collapse
Affiliation(s)
- Feng Ruan
- Department of Emergency Medicine, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Department of Surgery, SUNY Upstate Medical University, Syracuse, New York, 13210, USA
| | - Jing Chen
- Department of Ophthalmology, Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, P.R. China
| | - Jianxin Yang
- Department of Emergency Medicine, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Guirong Wang
- Department of Surgery, SUNY Upstate Medical University, Syracuse, New York, 13210, USA
| |
Collapse
|
18
|
Xiong W, Perna A, Jacob IB, Lundgren BR, Wang G. The Enhancer-Binding Protein MifR, an Essential Regulator of α-Ketoglutarate Transport, Is Required for Full Virulence of Pseudomonas aeruginosa PAO1 in a Mouse Model of Pneumonia. Infect Immun 2022; 90:e0013622. [PMID: 36125307 PMCID: PMC9584295 DOI: 10.1128/iai.00136-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The opportunistic human pathogen Pseudomonas aeruginosa PAO1 has an extensive metabolism, enabling it to utilize a wide range of structurally diverse compounds to meet its nutritional and energy needs. Interestingly, the utilization of some of the more unusual compounds often associated with a eukaryotic-host environment is regulated via enhancer-binding proteins (EBPs) in P. aeruginosa. Whether the utilization of such compounds and the EBPs involved contribute to the pathogenesis of P. aeruginosa remains to be fully understood. To narrow this gap, we investigated the roles of the EBPs EatR (regulator of ethanolamine catabolism), DdaR (regulator of methylarginine catabolism), and MifR (regulator of α-ketoglutarate or α-KG transport) in the virulence of P. aeruginosa PAO1 in a pneumonia-induced septic mouse model. Deletion of genes encoding EatR and DdaR had no significant effect on the mortality of P. aeruginosa PAO1-infected mice compared to wide-type (WT) PAO1-infected mice. In contrast, infected mice with ΔmifR mutant exhibited a significant reduction (~50%) in the mortality rate compared with WT PAO1 (P < 0.05). Infected mice with ΔmifR PAO1 had lower lung injury scores, fewer inflammatory cells, decreased proinflammatory cytokines, and decreased apoptosis and cell death compared to mice infected with WT PAO1 (P < 0.05). Furthermore, molecular analysis revealed decreased NLRP3 inflammasome activation in infected mice with ΔmifR PAO1 compared to WT PAO1 (P < 0.05). These results suggested that the utilization of α-KG was a contributing factor in P. aeruginosa-mediated pneumonia and sepsis and that MifR-associated regulation may be a potential therapeutic target for P. aeruginosa infectious disease.
Collapse
Affiliation(s)
- Weichuan Xiong
- Department of Surgery, State University of New York (SUNY) Upstate Medical University, Syracuse, New York, USA
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Alexander Perna
- Department of Surgery, State University of New York (SUNY) Upstate Medical University, Syracuse, New York, USA
| | - Ikechukwu B. Jacob
- Department of Surgery, State University of New York (SUNY) Upstate Medical University, Syracuse, New York, USA
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York, USA
| | | | - Guirong Wang
- Department of Surgery, State University of New York (SUNY) Upstate Medical University, Syracuse, New York, USA
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
19
|
Wang R, Gao D, Yu F, Han J, Yuan H, Hu F. Phospholipase A 2 inhibitor varespladib prevents wasp sting-induced nephrotoxicity in rats. Toxicon 2022; 215:69-76. [PMID: 35724947 DOI: 10.1016/j.toxicon.2022.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 02/07/2023]
Abstract
This study aimed to clarify whether varespladib, a phospholipase A2 (PLA2) inhibitor, can be used as a therapeutic agent for wasp sting-induced acute kidney injury (AKI). Rats were divided into control, AKI, and AKI + varespladib groups. The AKI model was established by subcutaneously injecting wasp venom at five different sites in rats. Varespladib treatment showed a significant inhibitory effect on wasp venom PLA2in vitro and in vivo. Moreover, we observed that varespladib decreased the levels of rhabdomyolysis and hemolysis markers compared with that in the AKI group. Histopathological changes in the kidney decreased significantly, and rat serum creatinine levels were reduced after varespladib administration. The significantly regulated genes in the kidney of the AKI group were mostly involved in inflammatory response pathway, and the administration of varespladib remarkably attenuated the expression of these genes. Therefore, varespladib inhibited wasp sting-induced functional and pathological damage to the kidneys. We propose that the PLA2 inhibitor varespladib protects the kidney tissue in a wasp sting-induced AKI model by inhibiting PLA2 activity.
Collapse
Affiliation(s)
- Rui Wang
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China; Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China
| | - Dan Gao
- Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China
| | - Fanglin Yu
- Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China
| | - Jiamin Han
- Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China
| | - Hai Yuan
- Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China.
| | - Fengqi Hu
- Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China.
| |
Collapse
|
20
|
Surfactant Protein D Influences Mortality During Abdominal Sepsis by Facilitating Escherichia coli Colonization in the Gut. Crit Care Explor 2022; 4:e0699. [PMID: 35620769 PMCID: PMC9119639 DOI: 10.1097/cce.0000000000000699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
21
|
Srinath BS, Shastry RP, Kumar SB. Role of gut-lung microbiome crosstalk in COVID-19. RESEARCH ON BIOMEDICAL ENGINEERING 2022. [PMCID: PMC7685301 DOI: 10.1007/s42600-020-00113-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
22
|
Escobar‐Salom M, Torrens G, Jordana‐Lluch E, Oliver A, Juan C. Mammals' humoral immune proteins and peptides targeting the bacterial envelope: from natural protection to therapeutic applications against multidrug‐resistant
Gram
‐negatives. Biol Rev Camb Philos Soc 2022; 97:1005-1037. [PMID: 35043558 PMCID: PMC9304279 DOI: 10.1111/brv.12830] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
Mammalian innate immunity employs several humoral ‘weapons’ that target the bacterial envelope. The threats posed by the multidrug‐resistant ‘ESKAPE’ Gram‐negative pathogens (Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.) are forcing researchers to explore new therapeutic options, including the use of these immune elements. Here we review bacterial envelope‐targeting (peptidoglycan and/or membrane‐targeting) proteins/peptides of the mammalian immune system that are most likely to have therapeutic applications. Firstly we discuss their general features and protective activity against ESKAPE Gram‐negatives in the host. We then gather, integrate, and discuss recent research on experimental therapeutics harnessing their bactericidal power, based on their exogenous administration and also on the discovery of bacterial and/or host targets that improve the performance of this endogenous immunity, as a novel therapeutic concept. We identify weak points and knowledge gaps in current research in this field and suggest areas for future work to obtain successful envelope‐targeting therapeutic options to tackle the challenge of antimicrobial resistance.
Collapse
Affiliation(s)
- María Escobar‐Salom
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Gabriel Torrens
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Elena Jordana‐Lluch
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Antonio Oliver
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Carlos Juan
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| |
Collapse
|
23
|
Liu W, Zeng Y, Li Y, Li N, Peng M, Cheng J, Tian B, Chen M. Exploring the Potential Targets and Mechanisms of Huang Lian Jie Du Decoction in the Treatment of Coronavirus Disease 2019 Based on Network Pharmacology. Int J Gen Med 2021; 14:9873-9885. [PMID: 34938107 PMCID: PMC8687521 DOI: 10.2147/ijgm.s337025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022] Open
Abstract
Background In December 2019, coronavirus disease 2019 (COVID-19) caused by a novel coronavirus (severe acute respiratory syndrome coronavirus 2, SARS-CoV-2; previously known as 2019-nCoV) emerged in Wuhan, China, and caused many infections and deaths. At present, there are no specific drugs for the etiology and treatment of COVID-19. A combination of traditional Chinese and western medicine is proposed to treat COVID-19, in which Huang Lian Jie Du decoction (HLJDD) is recommended for the treatment of COVID-19 in many provinces in China and has been widely used in the clinic. This study explored the potential targets of HLJDD in the treatment of COVID-19 based on network pharmacology. Methods First, the chemical composition and targets of HLJDD and COVID-19-related targets were obtained through the TCMSP, UniProt, GeneCards and OMIM databases. Second, HLJDD target and HLJDD-COVID-19 target networks were constructed via the STRING database and Cytoscape software. Finally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of the HLJDD-COVID-19 targets was applied via the DAVID database. Results Our study identified a total of 67 active ingredients of HLJDD and 204 targets of HLJDD. A total of 502 COVID-19-related targets were obtained, of which 47 were intersecting targets of HLJDD and COVID-19. A total of 179 GO terms and 77 KEGG terms, including the TNF signaling pathway, NF-κB signaling pathway and HIF-1 signaling pathway, were identified. Conclusion The present study explored the potential targets and signaling pathways of HLJDD during the treatment of COVID-19, which may provide a basis for the research and development of drugs for the treatment of COVID-19.
Collapse
Affiliation(s)
- Wang Liu
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| | - Yu Zeng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| | - Yanda Li
- Department of Internal Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, People's Republic of China
| | - Nanhong Li
- Department of Pathology and Pathophysiology, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Min Peng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| | - Junfen Cheng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| | - Binbin Tian
- Department of Critical Care Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, People's Republic of China
| | - Mingdi Chen
- Department of Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| |
Collapse
|
24
|
Release of HMGB1 in Podocytes Exacerbates Lipopolysaccharide-Induced Acute Kidney Injury. Mediators Inflamm 2021; 2021:5220226. [PMID: 34616232 PMCID: PMC8490059 DOI: 10.1155/2021/5220226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/31/2021] [Indexed: 12/29/2022] Open
Abstract
Objective Acute kidney injury (AKI) usually occurs during sepsis. Inflammation factors, such as high-mobility group box 1 (HMGB1), are dramatically upregulated under septic conditions. In our current work, the functions of HMGB1 in AKI were explored. Methods An AKI model was induced by the lipopolysaccharide (LPS) challenge in C57 mice. Podocytes were challenged by LPS for different durations. Subsequently, podocytes transfected with HMGB1 siRNA were exposed to LPS for 24 h. The expressions of supernatant HMGB1 and cellular active caspase-3 were examined by Western blotting analysis. To explore the effect of HMGB1 on tubular epithelial cells (TECs), HK-2 cells were exposed to HMGB1 at various concentrations for 24 h. Epithelial-mesenchymal transition (EMT) of HK-2 cells was evaluated by Western blotting analysis. Mitochondrial division and apoptosis of HK-2 cells were assessed by MitoTracker Red and Western blotting analysis, respectively. Results Compared with the sham control group, the expression of HMGB1 was increased in the kidney of AKI mice. Moreover, the expression of supernatant HMGB1 was increased in LPS-challenged podocytes compared with the control group. Knockdown of HMGB1 attenuated LPS-induced podocyte injury. Besides, EMT in TECs was triggered by HMGB1. Mitochondrial damage and apoptosis of HK-2 cells exposed to HMGB1 were markedly elevated compared with the control group. Conclusions Collectively, HMGB1 release in podocytes was induced by LPS, subsequently leading to exacerbated AKI.
Collapse
|
25
|
Tong M, Xiong Y, Zhu C, Xu H, Zheng Q, Jiang Y, Zou L, Xiao X, Chen F, Yan X, Hu C, Zhu Y. Serum surfactant protein D in COVID-19 is elevated and correlated with disease severity. BMC Infect Dis 2021; 21:737. [PMID: 34344306 PMCID: PMC8329621 DOI: 10.1186/s12879-021-06447-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 07/20/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The serum surfactant protein D (SP-D) level is suggested to be a useful biomarker for acute lung injuries and acute respiratory distress syndrome. Whether the serum SP-D level could identify the severity of coronavirus disease 2019 (COVID-19) in the early stage has not been elucidated. METHODS We performed an observational study on 39 laboratory-confirmed COVID-19 patients from The Fourth People's Hospital of Yiyang, Hunan, China. Receiver operating characteristic (ROC) curve analysis, correlation analysis, and multivariate logistic regression model analysis were performed. RESULTS In the acute phase, the serum levels of SP-D were elevated significantly in severe COVID-19 patients than in mild cases (mean value ± standard deviation (SD), 449.7 ± 125.8 vs 245.9 ± 90.0 ng/mL, P<0.001), while the serum levels of SP-D in the recovery period were decreased dramatically than that in the acute phase (mean value ± SD, 129.5 ± 51.7 vs 292.9 ± 130.7 ng/ml, P<0.001), and so were for the stratified patients. The chest CT imaging scores were considerably higher in the severe group compared with those in the mild group (median value, 10.0 vs 9.0, P = 0.011), while markedly lower in the recovery period than those in the acute phase (median value, 2.0 vs 9.0, P<0.001), and so were for the stratified patients. ROC curve analysis revealed that areas under the curve of lymphocyte counts (LYM), C-reaction protein (CRP), erythrocyte sedimentation rate (ESR), interleukin-6 (IL-6), and SP-D for severe COVID-19 were 0.719, 0.833, 0.817, 0.837, and 0.922, respectively. Correlation analysis showed that the SP-D levels were negatively correlated with LYM (r = - 0.320, P = 0.047), while positively correlated with CRP (r = 0.658, P<0.001), IL-6 (r = 0.471, P = 0.002), the duration of nucleic acid of throat swab turning negative (r = 0.668, P<0.001), chest CT imaging score on admission (r = 0.695, P<0.001) and length of stay (r = 0.420, P = 0.008). Multivariate logistic regression model analysis showed that age (P = 0.041, OR = 1.093) and SP-D (P = 0.008, OR = 1.018) were risk factors for severe COVID-19. CONCLUSIONS Elevated serum SP-D level was a potential biomarker for the severity of COVID-19; this may be useful in identifying patients whose condition worsens at an early stage.
Collapse
Affiliation(s)
- Ming Tong
- Department of Infectious Diseases, Hunan Provincial People's Hospital (The First-affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital (The First-affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Ying Xiong
- The Fourth People's Hospital of Yiyang, Yiyang, Hunan, China
| | - Chen Zhu
- Department of Pediatrics, Yiyang Central Hospital, Yiyang, Hunan, China
| | - Hong Xu
- The Fourth People's Hospital of Yiyang, Yiyang, Hunan, China
| | - Qing Zheng
- Department of Geriatrics, Hunan Provincial People's Hospital (The First-affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Yu Jiang
- Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital (The First-affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Lianhong Zou
- Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital (The First-affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Xiaolin Xiao
- The Fourth People's Hospital of Yiyang, Yiyang, Hunan, China
| | - Fang Chen
- Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital (The First-affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Xiquan Yan
- Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital (The First-affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Changping Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China.
| | - Yimin Zhu
- Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital (The First-affiliated Hospital of Hunan Normal University), Changsha, Hunan, China.
- School of Life Sciences, Hunan Normal University, Changsha, Hunan, China.
| |
Collapse
|
26
|
Regulatory Roles of Human Surfactant Protein B Variants on Genetic Susceptibility to Pseudomonas Aeruginosa Pneumonia-Induced Sepsis. Shock 2021; 54:507-519. [PMID: 31851120 DOI: 10.1097/shk.0000000000001494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Surfactant protein B (SP-B) is essential for life and plays critical roles in host defense and lowering alveolar surface tension. A single-nucleotide polymorphism (SNP rs1130866) of human SP-B (hSP-B) alters the N-linked glycosylation, thus presumably affecting SP-B function. This study has investigated the regulatory roles of hSP-B genetic variants on lung injury in pneumonia-induced sepsis. METHODS Wild-type (WT) FVB/NJ and humanized transgenic SP-B-T and SP-B-C mice (expressing either hSP-B C or T allele without mouse SP-B gene) were infected intratracheally with 50 μL (4 × 10 colony-forming units [CFUs]/mouse) Pseudomonas aeruginosa Xen5 or saline, and then killed 24 or 48 h after infection. Bacterial dynamic growths were monitored from 0 to 48 h postinfection by in vivo imaging. Histopathological, cellular, and molecular changes of lung tissues and bronchoalveolar lavage fluid (BALF) were analyzed. Surface tension of surfactants was determined with constrained drop surfactometry. RESULTS SP-B-C mice showed higher bioluminescence and CFUs, increased inflammation and mortality, the higher score of lung injury, and reduced numbers of lamellar bodies in type II cells compared with SP-B-T or WT (P < 0.05). Minimum surface tension increased dramatically in infected mice (P < 0.01) with the order of SP-B-C > SP-B-T > WT. Levels of multiple cytokines in the lung of infected SP-B-C were higher than those of SP-B-T and WT (P < 0.01). Furthermore, compared with SP-B-T or WT, SP-B-C exhibited lower SP-B, higher NF-κB and NLRP3 inflammasome activation, and higher activated caspase-3. CONCLUSIONS hSP-B variants differentially regulate susceptibility through modulating the surface activity of surfactant, cell death, and inflammatory signaling in sepsis.
Collapse
|
27
|
Logette E, Lorin C, Favreau C, Oshurko E, Coggan JS, Casalegno F, Sy MF, Monney C, Bertschy M, Delattre E, Fonta PA, Krepl J, Schmidt S, Keller D, Kerrien S, Scantamburlo E, Kaufmann AK, Markram H. A Machine-Generated View of the Role of Blood Glucose Levels in the Severity of COVID-19. Front Public Health 2021; 9:695139. [PMID: 34395368 PMCID: PMC8356061 DOI: 10.3389/fpubh.2021.695139] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/30/2021] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 started spreading toward the end of 2019 causing COVID-19, a disease that reached pandemic proportions among the human population within months. The reasons for the spectrum of differences in the severity of the disease across the population, and in particular why the disease affects more severely the aging population and those with specific preconditions are unclear. We developed machine learning models to mine 240,000 scientific articles openly accessible in the CORD-19 database, and constructed knowledge graphs to synthesize the extracted information and navigate the collective knowledge in an attempt to search for a potential common underlying reason for disease severity. The machine-driven framework we developed repeatedly pointed to elevated blood glucose as a key facilitator in the progression of COVID-19. Indeed, when we systematically retraced the steps of the SARS-CoV-2 infection, we found evidence linking elevated glucose to each major step of the life-cycle of the virus, progression of the disease, and presentation of symptoms. Specifically, elevations of glucose provide ideal conditions for the virus to evade and weaken the first level of the immune defense system in the lungs, gain access to deep alveolar cells, bind to the ACE2 receptor and enter the pulmonary cells, accelerate replication of the virus within cells increasing cell death and inducing an pulmonary inflammatory response, which overwhelms an already weakened innate immune system to trigger an avalanche of systemic infections, inflammation and cell damage, a cytokine storm and thrombotic events. We tested the feasibility of the hypothesis by manually reviewing the literature referenced by the machine-generated synthesis, reconstructing atomistically the virus at the surface of the pulmonary airways, and performing quantitative computational modeling of the effects of glucose levels on the infection process. We conclude that elevation in glucose levels can facilitate the progression of the disease through multiple mechanisms and can explain much of the differences in disease severity seen across the population. The study provides diagnostic considerations, new areas of research and potential treatments, and cautions on treatment strategies and critical care conditions that induce elevations in blood glucose levels.
Collapse
Affiliation(s)
- Emmanuelle Logette
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Henry Markram
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| |
Collapse
|
28
|
Arroyo R, Kingma PS. Surfactant protein D and bronchopulmonary dysplasia: a new way to approach an old problem. Respir Res 2021; 22:141. [PMID: 33964929 PMCID: PMC8105703 DOI: 10.1186/s12931-021-01738-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Surfactant protein D (SP-D) is a collectin protein synthesized by alveolar type II cells in the lungs. SP-D participates in the innate immune defense of the lungs by helping to clear infectious pathogens and modulating the immune response. SP-D has shown an anti-inflammatory role by down-regulating the release of pro-inflammatory mediators in different signaling pathways such as the TLR4, decreasing the recruitment of inflammatory cells to the lung, and modulating the oxidative metabolism in the lungs. Recombinant human SP-D (rhSP-D) has been successfully produced mimicking the structure and functions of native SP-D. Several in vitro and in vivo experiments using different animal models have shown that treatment with rhSP-D reduces the lung inflammation originated by different insults, and that rhSP-D could be a potential treatment for bronchopulmonary dysplasia (BPD), a rare disease for which there is no effective therapy up to date. BPD is a complex disease in preterm infants whose incidence increases with decreasing gestational age at birth. Lung inflammation, which is caused by different prenatal and postnatal factors like infections, lung hyperoxia and mechanical ventilation, among others, is the key player in BPD. Exacerbated inflammation causes lung tissue injury that results in a deficient gas exchange in the lungs of preterm infants and frequently leads to long-term chronic lung dysfunction during childhood and adulthood. In addition, low SP-D levels and activity in the first days of life in preterm infants have been correlated with a worse pulmonary outcome in BPD. Thus, SP-D mediated functions in the innate immune response could be critical aspects of the pathogenesis in BPD and SP-D could inhibit lung tissue injury in this preterm population. Therefore, administration of rhSP-D has been proposed as promising therapy that could prevent BPD.
Collapse
Affiliation(s)
- Raquel Arroyo
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. ML7029, Cincinnati, OH, 45229-3039, USA
| | - Paul S Kingma
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. ML7029, Cincinnati, OH, 45229-3039, USA. .,Airway Therapeutics Inc, Cincinnati, OH, 45249, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| |
Collapse
|
29
|
Yamamoto R, Fujishima S, Sasaki J, Gando S, Saitoh D, Shiraishi A, Kushimoto S, Ogura H, Abe T, Mayumi T, Kotani J, Nakada TA, Shiino Y, Tarui T, Okamoto K, Sakamoto Y, Shiraishi SI, Takuma K, Tsuruta R, Masuno T, Takeyama N, Yamashita N, Ikeda H, Ueyama M, Hifumi T, Yamakawa K, Hagiwara A, Otomo Y. Hyperoxemia during resuscitation of trauma patients and increased intensive care unit length of stay: inverse probability of treatment weighting analysis. World J Emerg Surg 2021; 16:19. [PMID: 33926507 PMCID: PMC8082221 DOI: 10.1186/s13017-021-00363-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/16/2021] [Indexed: 12/28/2022] Open
Abstract
Background Information on hyperoxemia among patients with trauma has been limited, other than traumatic brain injuries. This study aimed to elucidate whether hyperoxemia during resuscitation of patients with trauma was associated with unfavorable outcomes. Methods A post hoc analysis of a prospective observational study was carried out at 39 tertiary hospitals in 2016–2018 in adult patients with trauma and injury severity score (ISS) of > 15. Hyperoxemia during resuscitation was defined as PaO2 of ≥ 300 mmHg on hospital arrival and/or 3 h after arrival. Intensive care unit (ICU)-free days were compared between patients with and without hyperoxemia. An inverse probability of treatment weighting (IPW) analysis was conducted to adjust patient characteristics including age, injury mechanism, comorbidities, vital signs on presentation, chest injury severity, and ISS. Analyses were stratified with intubation status at the emergency department (ED). The association between biomarkers and ICU length of stay were then analyzed with multivariate models. Results Among 295 severely injured trauma patients registered, 240 were eligible for analysis. Patients in the hyperoxemia group (n = 58) had shorter ICU-free days than those in the non-hyperoxemia group [17 (10–21) vs 23 (16–26), p < 0.001]. IPW analysis revealed the association between hyperoxemia and prolonged ICU stay among patients not intubated at the ED [ICU-free days = 16 (12–22) vs 23 (19–26), p = 0.004], but not among those intubated at the ED [18 (9–20) vs 15 (8–23), p = 0.777]. In the hyperoxemia group, high inflammatory markers such as soluble RAGE and HMGB-1, as well as low lung-protective proteins such as surfactant protein D and Clara cell secretory protein, were associated with prolonged ICU stay. Conclusions Hyperoxemia until 3 h after hospital arrival was associated with prolonged ICU stay among severely injured trauma patients not intubated at the ED. Trial registration UMIN-CTR, UMIN000019588. Registered on November 15, 2015. Supplementary Information The online version contains supplementary material available at 10.1186/s13017-021-00363-2.
Collapse
Affiliation(s)
- Ryo Yamamoto
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Seitaro Fujishima
- Center for General Medicine Education, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan.
| | - Junichi Sasaki
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Gando
- Department of Acute and Critical Care Medicine, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan.,Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Daizoh Saitoh
- Division of Traumatology, Research Institute, National Defense Medical College, Tokorozawa, Japan
| | | | - Shigeki Kushimoto
- Division of Emergency and Critical Care Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Toshikazu Abe
- Department of General Medicine, Juntendo University, Tokyo, Japan.,Health Services Research and Development Center, University of Tsukuba, Tsukuba, Japan
| | - Toshihiko Mayumi
- Department of Emergency Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Joji Kotani
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Taka-Aki Nakada
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yasukazu Shiino
- Department of Acute Medicine, Kawasaki Medical School, Kurashiki, Japan
| | - Takehiko Tarui
- Department of Trauma and Critical Care Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Kohji Okamoto
- Department of Surgery, Center for Gastroenterology and Liver Disease, Kitakyushu City Yahata Hospital, Kitakyushu, Japan
| | - Yuichiro Sakamoto
- Emergency and Critical Care Medicine, Saga University Hospital, Saga, Japan
| | - Shin-Ichiro Shiraishi
- Department of Emergency and Critical Care Medicine, Aizu Chuo Hospital, Aizuwakamatsu, Japan
| | - Kiyotsugu Takuma
- Emergency & Critical Care Center, Kawasaki Municipal Kawasaki Hospital, Kawasaki, Japan
| | - Ryosuke Tsuruta
- Advanced Medical Emergency & Critical Care Center, Yamaguchi University Hospital, Ube, Japan
| | - Tomohiko Masuno
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Naoshi Takeyama
- Advanced Critical Care Center, Aichi Medical University Hospital, Nagakute, Japan
| | - Norio Yamashita
- Advanced Emergency Medical Service Center, Kurume University Hospital, Kurume, Japan
| | - Hiroto Ikeda
- Department of Emergency Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Masashi Ueyama
- Department of Trauma, Critical Care Medicine, and Burn Center, Japan Community Healthcare Organization, Chukyo Hospital, Nagoya, Japan
| | - Toru Hifumi
- Department of Emergency and Critical Care Medicine, St. Luke's International Hospital, Tokyo, Japan
| | - Kazuma Yamakawa
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| | - Akiyoshi Hagiwara
- Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Yasuhiro Otomo
- Trauma and Acute Critical Care Center, Medical Hospital, Tokyo Medical and Dental University, Tokyo, Japan
| | | |
Collapse
|
30
|
Watson A, Madsen J, Clark HW. SP-A and SP-D: Dual Functioning Immune Molecules With Antiviral and Immunomodulatory Properties. Front Immunol 2021; 11:622598. [PMID: 33542724 PMCID: PMC7851053 DOI: 10.3389/fimmu.2020.622598] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/14/2020] [Indexed: 01/08/2023] Open
Abstract
Surfactant proteins A (SP-A) and D (SP-D) are soluble innate immune molecules which maintain lung homeostasis through their dual roles as anti-infectious and immunomodulatory agents. SP-A and SP-D bind numerous viruses including influenza A virus, respiratory syncytial virus (RSV) and human immunodeficiency virus (HIV), enhancing their clearance from mucosal points of entry and modulating the inflammatory response. They also have diverse roles in mediating innate and adaptive cell functions and in clearing apoptotic cells, allergens and other noxious particles. Here, we review how the properties of these first line defense molecules modulate inflammatory responses, as well as host-mediated immunopathology in response to viral infections. Since SP-A and SP-D are known to offer protection from viral and other infections, if their levels are decreased in some disease states as they are in severe asthma and chronic obstructive pulmonary disease (COPD), this may confer an increased risk of viral infection and exacerbations of disease. Recombinant molecules of SP-A and SP-D could be useful in both blocking respiratory viral infection while also modulating the immune system to prevent excessive inflammatory responses seen in, for example, RSV or coronavirus disease 2019 (COVID-19). Recombinant SP-A and SP-D could have therapeutic potential in neutralizing both current and future strains of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus as well as modulating the inflammation-mediated pathology associated with COVID-19. A recombinant fragment of human (rfh)SP-D has recently been shown to neutralize SARS-CoV-2. Further work investigating the potential therapeutic role of SP-A and SP-D in COVID-19 and other infectious and inflammatory diseases is indicated.
Collapse
Affiliation(s)
- Alastair Watson
- Clinical and Experimental Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, United Kingdom
- Birmingham Medical School, University of Birmingham, Birmingham, United Kingdom
| | - Jens Madsen
- Neonatology, EGA Institute for Women’s Health, Faculty of Population Health Sciences, University College London, London, United Kingdom
| | - Howard William Clark
- Neonatology, EGA Institute for Women’s Health, Faculty of Population Health Sciences, University College London, London, United Kingdom
- NIHR Biomedical Research Centre, University College London Hospital (UCLH), University College London (UCL), London, United Kingdom
| |
Collapse
|
31
|
Yuan H, Gao Z, Lu X, Hu F. Role of collectin-11 in innate defence against uropathogenic Escherichia coli infection. Innate Immun 2020; 27:50-60. [PMID: 33241978 PMCID: PMC7780352 DOI: 10.1177/1753425920974766] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Classical collectins (surfactant protein A and D) play a significant role in innate immunity and host defence in uropathogenic Escherichia coli (UPEC)-induced urinary tract infection (UTI). However, the functions of collectin-11 (CL-11) with respect to UPEC and UTI remain largely unexplored. This study aimed to investigate the effect of CL-11 on UPEC and its role in UTI. We further examined its modulatory effect on inflammatory reactions in proximal tubular epithelial cells (PTECs). The present study provides evidence for the effect of CL-11 on the growth, agglutination, binding, epithelial adhesion and invasion of UPEC. We found increased basal levels of phosphorylated p38 MAPK and human cytokine homologue (keratinocyte-derived chemokine) expression in CL-11 knockdown PTECs. Furthermore, signal regulatory protein α blockade reversed the increased basal levels of inflammation associated with CL-11 knockdown in PTECs. Additionally, CL-11 knockdown effectively inhibited UPEC-induced p38 MAPK phosphorylation and cytokine production in PTECs. These were further inhibited by CD91 blockade. We conclude that CL-11 functions as a mediator of innate immunity via direct antibacterial roles as well as dual modulatory roles in UPEC-induced inflammatory responses during UTI. Thus, the study findings suggest a possible function for CL-11 in defence against UTI.
Collapse
Affiliation(s)
- Hai Yuan
- Department of Nephrology, 74731Xiangyang Central Hospital, Affiliated Hospital of 118302Hubei University of Arts and Science, PR China
| | - Zhao Gao
- Department of Nephrology, 74731Xiangyang Central Hospital, Affiliated Hospital of 118302Hubei University of Arts and Science, PR China
| | - Xiaohan Lu
- Department of Nephrology, 74731Xiangyang Central Hospital, Affiliated Hospital of 118302Hubei University of Arts and Science, PR China
| | - Fengqi Hu
- Department of Nephrology, 74731Xiangyang Central Hospital, Affiliated Hospital of 118302Hubei University of Arts and Science, PR China
| |
Collapse
|
32
|
Wang XX, Sha XL, Li YL, Li CL, Chen SH, Wang JJ, Xia Z. Lung injury induced by short-term mechanical ventilation with hyperoxia and its mitigation by deferoxamine in rats. BMC Anesthesiol 2020; 20:188. [PMID: 32738874 PMCID: PMC7395352 DOI: 10.1186/s12871-020-01089-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/09/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Long-term mechanical ventilation with hyperoxia can induce lung injury. General anesthesia is associated with a very high incidence of hyperoxaemia, despite it usually lasts for a relatively short period of time. It remains unclear whether short-term mechanical ventilation with hyperoxia has an adverse impact on or cause injury to the lungs. The present study aimed to assess whether short-term mechanical ventilation with hyperoxia may cause lung injury in rats and whether deferoxamine (DFO), a ferrous ion chelator, could mitigate such injury to the lungs and explore the possible mechanism. METHODS Twenty-four SD rats were randomly divided into 3 groups (n = 8/group): mechanical ventilated with normoxia group (MV group, FiO2 = 21%), with hyperoxia group (HMV group, FiO2 = 90%), or with hyperoxia + DFO group (HMV + DFO group, FiO2 = 90%). Mechanical ventilation under different oxygen concentrations was given for 4 h, and ECG was monitored. The HMV + DFO group received continuous intravenous infusion of DFO at 50 mg•kg- 1•h- 1, while the MV and HMV groups received an equal volume of normal saline. Carotid artery cannulation was carried out to monitor the blood gas parameters under mechanical ventilation for 2 and 4 h, respectively, and the PaO2/FiO2 ratio was calculated. After 4 h ventilation, the right anterior lobe of the lung and bronchoalveolar lavage fluid from the right lung was sampled for pathological and biochemical assays. RESULTS PaO2 in the HMV and HMV + DFO groups were significantly higher, but the PaO2/FiO2 ratio were significantly lower than those of the MV group (all p < 0.01), while PaO2 and PaO2/FiO2 ratio between HMV + DFO and HMV groups did not differ significantly. The lung pathological scores and the wet-to-dry weight ratio (W/D) in the HMV and HMV + DFO groups were significantly higher than those of the MV group, but the lung pathological score and the W/D ratio were reduced by DFO (p < 0.05, HMV + DFO vs. HMV). Biochemically, HMV resulted in significant reductions in Surfactant protein C (SP-C), Surfactant protein D (SP-D), and Glutathion reductase (GR) levels and elevation of xanthine oxidase (XOD) in both the Bronchoalveolar lavage fluid and the lung tissue homogenate, and all these changes were prevented or significantly reverted by DFO. CONCLUSIONS Mechanical ventilation with hyperoxia for 4 h induced oxidative injury of the lungs, accompanied by a dramatic reduction in the concentrations of SP-C and SP-D. DFO could mitigate such injury by lowering XOD activity and elevating GR activity.
Collapse
Affiliation(s)
- Xiao-Xia Wang
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Xiao-Lan Sha
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Yu-Lan Li
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.
| | - Chun-Lan Li
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Su-Heng Chen
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Jing-Jing Wang
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Zhengyuan Xia
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, 999077, People's Republic of China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, People's Republic of China
| |
Collapse
|
33
|
Lipid-Protein and Protein-Protein Interactions in the Pulmonary Surfactant System and Their Role in Lung Homeostasis. Int J Mol Sci 2020; 21:ijms21103708. [PMID: 32466119 PMCID: PMC7279303 DOI: 10.3390/ijms21103708] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary surfactant is a lipid/protein complex synthesized by the alveolar epithelium and secreted into the airspaces, where it coats and protects the large respiratory air–liquid interface. Surfactant, assembled as a complex network of membranous structures, integrates elements in charge of reducing surface tension to a minimum along the breathing cycle, thus maintaining a large surface open to gas exchange and also protecting the lung and the body from the entrance of a myriad of potentially pathogenic entities. Different molecules in the surfactant establish a multivalent crosstalk with the epithelium, the immune system and the lung microbiota, constituting a crucial platform to sustain homeostasis, under health and disease. This review summarizes some of the most important molecules and interactions within lung surfactant and how multiple lipid–protein and protein–protein interactions contribute to the proper maintenance of an operative respiratory surface.
Collapse
|
34
|
A novel designer drug, 25N-NBOMe, exhibits abuse potential via the dopaminergic system in rodents. Brain Res Bull 2019; 152:19-26. [DOI: 10.1016/j.brainresbull.2019.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/02/2019] [Accepted: 07/02/2019] [Indexed: 12/29/2022]
|
35
|
Surfactant Protein D Is Altered in Experimental Malaria-Associated Acute Lung Injury/Acute Respiratory Distress Syndrome. J Trop Med 2019; 2019:9281605. [PMID: 31467567 PMCID: PMC6699403 DOI: 10.1155/2019/9281605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/07/2019] [Indexed: 11/17/2022] Open
Abstract
Surfactant protein D (SP-D) is in the collectin family of C-type lectins and plays an important role in the regulation of inflammation and the innate immune defense against pathogens. This protein has been proposed as a biomarker for acute lung injury. However, the expression of SP-D in the lung and the circulating levels of SP-D during malaria infection have received limited attention. Therefore, the aim of this study was to determine the location and expression of the SP-D protein in lung tissue and to measure the plasma level of SP-D in experimental malaria-associated acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Malaria-infected mice induced by Plasmodium berghei ANKA were classified into two groups, namely, the ALI/ARDS and non-ALI/ARDS groups, according to lung histopathology. The lungs of uninfected mice were used as a control group. The location and expression of SP-D in the lung tissues were investigated by immunohistochemical staining and Western blot analysis. In addition, the level of SP-D in plasma and lung homogenate was measured by an enzyme-linked immunosorbent assay. Immunohistochemical staining of SP-D was significantly increased in the lung tissues of the malaria-infected mice in the ALI/ARDS group compared with that in the malaria-infected mice in the non-ALI/ARDS group and the mice in the control group (p < 0.05). The levels of SP-D in the plasma and lung homogenate were significantly increased in the malaria-infected mice in the ALI/ARDS group compared with those in the malaria-infected mice in the non-ALI/ARDS group and the mice in the control group (p < 0.05). There was a significant positive correlation between SP-D in the plasma and SP-D in the lung homogenate (r s = 0.900, p = 0.037). In conclusion, this study demonstrated increased expression levels of SP-D in the lung tissue and high levels of plasma SP-D in the malaria-infected mice with ALI/ARDS compared with those in the mice in the other groups. The current study supports that the elevation of the plasma SP-D level may provide useful biological confirmation of the diagnosis of ALI/ARDS during malaria infection.
Collapse
|