1
|
Li L, Liu Z, Tian L, Yao S, Feng L, Lai F, Wang K, Zhang Y, Li Y, Wang J, Ren W. Single-cell proteomics delineates murine systemic immune response to blast lung injury. Commun Biol 2024; 7:1429. [PMID: 39489806 DOI: 10.1038/s42003-024-07151-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024] Open
Abstract
Victims of explosive events frequently suffer from blast lung injuries. Immune system has been implicated in the pathogenesis of this disease. However, systemic immune responses underlying the progression and recovery of injury repair remain poorly understood. Here, we depict the systemic landscape of immune dysregulation during blast lung injury and uncover immune recovery patterns. Single-cell analyses reveal dramatic changes in neutrophils, macrophages, monocytes, dendritic cells, and eosinophils after a gas explosion, along with early involvement of CD4 T, CD8 T, and Th17 cells. We demonstrate that myeloid cells primarily exert functions during the acute phase, while the spleen serves as an alternative source of granulocytes. Granulopoiesis is initiated in the bone marrow at a later stage during blast lung injury recovery, rather than at the acute stage. These findings contribute to a better understanding of the pathogenesis and provide valuable insights for potential immune interventions in blast lung injury.
Collapse
Affiliation(s)
- Long Li
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, China
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Zhongrui Liu
- The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Linqiang Tian
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, China
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Sanqiao Yao
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Lili Feng
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Feng Lai
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Kunxi Wang
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yue Zhang
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yanyan Li
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jinheng Wang
- The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.
| | - Wenjie Ren
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, China.
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.
- Clinical Medical Centre of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
2
|
Cong P, Tong C, Mao S, Shi L, Hou M, Liu Y. DDAH1 deficiency exacerbates cerebral vascular endothelial dysfunction by aggravating BBB disruption and oxidative stress in thoracic blast-induced brain injury. Exp Neurol 2024; 383:114994. [PMID: 39424041 DOI: 10.1016/j.expneurol.2024.114994] [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: 05/24/2024] [Revised: 09/07/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
As terrorist incidents and underground explosion events have become more frequent around the world, brain injury caused by thoracic blast exposure has been more highlighted due to its injured organ, subsequent social and economic burden. It has been reported dimethylarginine dimethylaminohydrolase 1 (DDAH1) plays important roles in regulating vascular endothelial injury repair and angiogenesis, but its role in thoracic blast-induced brain injury remains to be explained. This study seeks to investigate the mechanism of DDAH1 on thoracic blast-induced brain injury. 40 C57BL/6 wild type mice and 40 DDAH1 knockout mice were randomly and equally divided into control group and blast group, respectively. The integrity of blood-brain barrier (BBB) was detected by Evans blue test. The serum inflammatory factors, nitric oxide (NO) contents, and asymmetric dimethylarginine (ADMA) levels were determined through ELISA. HE staining and reactive oxygen species (ROS) detection were performed for histopathological changes. Western blot was used to detect the proteins related to oxidative stress, tight junction, focal adhesion, vascular endothelial injury, and the DDAH1/ADMA/eNOS signaling pathway. DDAH1 deficiency aggravated thoracic blast-induced BBB leakage, inflammatory response, and the increased levels of inflammatory-related factors. Additionally, DDAH1 deficiency also increased ROS generation, MDA and IRE-α expression. Regarding cerebral vascular endothelial dysfunction, DDAH1 deficiency increased the expression of MCAM, FN1, LIMK1, VEGF, MMP9, Vimentin and N-cadherin, while lowering the expression of FMR1, Occludin, claudin-3, claudin-5, Lyn, LIMA1, Glrb, Sez6, Dystrophin, and phosphorylation of VASP. Also, DDAH1 deficiency exacerbated explosion-induced increase of ADMA and decrease of eNOS activity and NO contents. Thus, we conclude that DDAH1 could prevent cerebral vascular endothelial dysfunction and related injury by inhibiting ADMA signaling and increasing eNOS activity in thoracic blast induced brain injury.
Collapse
Affiliation(s)
- Peifang Cong
- Shenyang Medical College, No.146, Huanghe North Street, Yuhong District, Shenyang, Liaoning Province 110034, China
| | - Changci Tong
- Shenyang Medical College, No.146, Huanghe North Street, Yuhong District, Shenyang, Liaoning Province 110034, China
| | - Shun Mao
- Shenyang Medical College, No.146, Huanghe North Street, Yuhong District, Shenyang, Liaoning Province 110034, China
| | - Lin Shi
- Shenyang Medical College, No.146, Huanghe North Street, Yuhong District, Shenyang, Liaoning Province 110034, China
| | - Mingxiao Hou
- Shenyang Medical College, No.146, Huanghe North Street, Yuhong District, Shenyang, Liaoning Province 110034, China
| | - Yunen Liu
- Shenyang Medical College, No.146, Huanghe North Street, Yuhong District, Shenyang, Liaoning Province 110034, China.
| |
Collapse
|
3
|
Bar-Kochba E, Iwaskiw AS, Dunn JM, Ott KA, Harrigan TP, Demetropoulos CK. The dynamic response of human lungs due to underwater shock wave exposure. PLoS One 2024; 19:e0303325. [PMID: 38748668 PMCID: PMC11095682 DOI: 10.1371/journal.pone.0303325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/23/2024] [Indexed: 05/19/2024] Open
Abstract
Since the 19th century, underwater explosions have posed a significant threat to service members. While there have been attempts to establish injury criteria for the most vulnerable organs, namely the lungs, existing criteria are highly variable due to insufficient human data and the corresponding inability to understand the underlying injury mechanisms. This study presents an experimental characterization of isolated human lung dynamics during simulated exposure to underwater shock waves. We found that the large acoustic impedance at the surface of the lung severely attenuated transmission of the shock wave into the lungs. However, the shock wave initiated large bulk pressure-volume cycles that are distinct from the response of the solid organs under similar loading. These pressure-volume cycles are due to compression of the contained gas, which we modeled with the Rayleigh-Plesset equation. The extent of these lung dynamics was dependent on physical confinement, which in real underwater blast conditions is influenced by factors such as rib cage properties and donned equipment. Findings demonstrate a potential causal mechanism for implosion injuries, which has significant implications for the understanding of primary blast lung injury due to underwater blast exposures.
Collapse
Affiliation(s)
- Eyal Bar-Kochba
- Research and Exploratory Development Department, Johns Hopkins University Applied Physics Laboratory, Laurel, MD, United States of America
| | - Alexander S. Iwaskiw
- Research and Exploratory Development Department, Johns Hopkins University Applied Physics Laboratory, Laurel, MD, United States of America
| | - Jenna M. Dunn
- Research and Exploratory Development Department, Johns Hopkins University Applied Physics Laboratory, Laurel, MD, United States of America
| | - Kyle A. Ott
- Research and Exploratory Development Department, Johns Hopkins University Applied Physics Laboratory, Laurel, MD, United States of America
| | - Timothy P. Harrigan
- Research and Exploratory Development Department, Johns Hopkins University Applied Physics Laboratory, Laurel, MD, United States of America
| | - Constantine K. Demetropoulos
- Research and Exploratory Development Department, Johns Hopkins University Applied Physics Laboratory, Laurel, MD, United States of America
| |
Collapse
|
4
|
Shao S, Wu Z, Wang Y, Wang Y, Wang Z, Ye H, Zhao H. Esophageal pressure monitoring and its clinical significance in severe blast lung injury. Front Bioeng Biotechnol 2024; 12:1280679. [PMID: 38784763 PMCID: PMC11112033 DOI: 10.3389/fbioe.2024.1280679] [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: 08/21/2023] [Accepted: 03/22/2024] [Indexed: 05/25/2024] Open
Abstract
Background The incidence of blast lung injury (BLI) has been escalating annually due to military conflicts and industrial accidents. Currently, research into these injuries predominantly uses animal models. Despite the availability of various models, there remains a scarcity of studies focused on monitoring respiratory mechanics post-BLI. Consequently, our objective was to develop a model for monitoring esophageal pressure (Pes) following BLI using a biological shock tube (BST), aimed at providing immediate and precise monitoring of respiratory mechanics parameters post-injury. Methods Six pigs were subjected to BLI using a BST, during which Pes was monitored. We assessed vital signs; conducted blood gas analysis, hemodynamics evaluations, and lung ultrasound; and measured respiratory mechanics before and after the inflicted injury. Furthermore, the gross anatomy of the lungs 3 h post-injury was examined, and hematoxylin and eosin staining was conducted on the injured lung tissues for further analysis. Results The pressure in the experimental section of the BST reached 402.52 ± 17.95 KPa, with a peak pressure duration of 53.22 ± 1.69 ms. All six pigs exhibited an anatomical lung injury score ≥3, and pathology revealed classic signs of severe BLI. Post-injury vital signs showed an increase in HR and SI, along with a decrease in MAP (p < 0.05). Blood gas analyses indicated elevated levels of Lac, CO2-GAP, A-aDO2, HB, and HCT and reduced levels of DO2, OI, SaO2, and OER (p < 0.05). Hemodynamics and lung ultrasonography findings showed increased ELWI, PVPI, SVRI, and lung ultrasonography scores and decreased CI, SVI, GEDI, and ITBI (p < 0.05). Analysis of respiratory mechanics revealed increased Ppeak, Pplat, Driving P, MAP, PEF, Ri, lung elastance, MP, Ptp, Ppeak - Pplat, and ΔPes, while Cdyn, Cstat, and time constant were reduced (p < 0.05). Conclusion We have successfully developed a novel respiratory mechanics monitoring model for severe BLI. This model is reliable, repeatable, stable, effective, and user-friendly. Pes monitoring offers a non-invasive and straightforward alternative to blood gas analysis, facilitating early clinical decision-making. Our animal study lays the groundwork for the early diagnosis and management of severe BLI in clinical settings.
Collapse
Affiliation(s)
- Shifeng Shao
- Department of ICU, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhengbin Wu
- Department of ICU, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi Wang
- The Fifth Outpatient Clinic, Western Theater General Hospital, Chengdu, China
| | - Yaoli Wang
- Department of ICU, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhen Wang
- Department of ICU, Daping Hospital, Army Medical University, Chongqing, China
| | - Huan Ye
- Department of Rehabilitation, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Hui Zhao
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
5
|
Fu H, Liang X, Tan W, Hu X. Unraveling the protective mechanisms of Chuanfangyihao against acute lung injury: Insights from experimental validation. Exp Ther Med 2023; 26:535. [PMID: 37869635 PMCID: PMC10587870 DOI: 10.3892/etm.2023.12234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 08/21/2023] [Indexed: 10/24/2023] Open
Abstract
Chuanfangyihao (CFYH) is an effective treatment for acute lung injury (ALI) in clinical practice; however, its underlying mechanism of action remains unclear. Therefore, the aim of the present study was to elucidate the pharmacological mechanism of action of CFYH in ALI through experimental validation. First, a rat model of ALI was established using lipopolysaccharide (LPS). Next, the pathological changes in the lungs of the rats and the pathological damage were scored. The wet/dry weight ratios were measured, and ROS content was detected using flow cytometry. ELISA was used to examine IL-6, TNF-α, IL-1β, IL-18, and LDH levels. Immunohistochemistry was used to detect Beclin-1 and NLRP3 expression. Western blotting was performed to analyze the expression of HMGB1, RAGE, TLR4, NF-κB p65, AMPK, p-AMPK, mTOR, p-mTOR, Beclin-1, LC3-II/I, p62, Bcl-2, Bax, Caspase-3, Caspase-1, and GSDMD-NT. The mRNA levels of HMGB1, RAGE, AMPK, mTOR, and HIF-1α were determined using reverse transcription quantitative PCR. CFYH alleviated pulmonary edema and decreased the expression of IL-6, TNF-α, TLR4, NF-κB p65, HMGB1/RAGE, ROS, and HIF-1α. In addition, pretreatment with CFYH reversed ALI-induced programmed cell death. In conclusion, CFYH alleviates LPS-induced ALI, and these findings provide a preliminary clarification of the predominant mechanism of action of CFYH in ALI.
Collapse
Affiliation(s)
- Hongfang Fu
- Infectious Disease Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Xiao Liang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Wanying Tan
- Infectious Disease Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Xiaoyu Hu
- Infectious Disease Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
6
|
Bishani A, Makarova DM, Shmendel EV, Maslov MA, Sen‘kova AV, Savin IA, Gladkikh DV, Zenkova MA, Chernolovskaya EL. Influence of the Composition of Cationic Liposomes on the Performance of Cargo Immunostimulatory RNA. Pharmaceutics 2023; 15:2184. [PMID: 37765155 PMCID: PMC10535620 DOI: 10.3390/pharmaceutics15092184] [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: 08/08/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
In this study, the impact of different delivery systems on the cytokine-inducing, antiproliferative, and antitumor activities of short immunostimulatory double-stranded RNA (isRNA) was investigated. The delivery systems, consisting of the polycationic amphiphile 1,26-bis(cholest-5-en-3-yloxycarbonylamino)-7,11,16,20 tetraazahexacosan tetrahydrochloride (2X3), and the lipid-helper dioleoylphosphatidylethanolamine (DOPE), were equipped with polyethylene glycol lipoconjugates differing in molecular weight and structure. The main findings of this work are as follows: (i) significant activation of MCP-1 and INF-α, β, and γ production in CBA mice occurs under the action of isRNA complexes with liposomes containing lipoconjugates with long PEG chains, while activation of MCP-1 and INF-γ, but not INF-α or β, was observed under the action of isRNA lipoplexes containing lipoconjugates with short PEG chains; (ii) a pronounced antiproliferative effect on B16 melanoma cells in vitro, as well as an antitumor and hepatoprotective effect in vivo, was induced by isRNA pre-complexes with non-pegylated liposomes, while complexes containing lipoconjugates with long-chain liposomes were inactive; (iii) the antitumor activity of isRNA correlated with the efficiency of its accumulation in the cells and did not explicitly depend on the activation of cytokine and interferon production. Thus, the structure of the delivery system plays a vital role in determining the response to isRNA and allows for the choice of a delivery system depending on the desired effect.
Collapse
Affiliation(s)
- Ali Bishani
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia; (A.B.); (A.V.S.); (I.A.S.); (D.V.G.); (M.A.Z.)
| | - Darya M. Makarova
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadsky Ave. 86, 119571 Moscow, Russia; (D.M.M.); (E.V.S.); (M.A.M.)
| | - Elena V. Shmendel
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadsky Ave. 86, 119571 Moscow, Russia; (D.M.M.); (E.V.S.); (M.A.M.)
| | - Mikhail A. Maslov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadsky Ave. 86, 119571 Moscow, Russia; (D.M.M.); (E.V.S.); (M.A.M.)
| | - Aleksandra V. Sen‘kova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia; (A.B.); (A.V.S.); (I.A.S.); (D.V.G.); (M.A.Z.)
| | - Innokenty A. Savin
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia; (A.B.); (A.V.S.); (I.A.S.); (D.V.G.); (M.A.Z.)
| | - Daniil V. Gladkikh
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia; (A.B.); (A.V.S.); (I.A.S.); (D.V.G.); (M.A.Z.)
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia; (A.B.); (A.V.S.); (I.A.S.); (D.V.G.); (M.A.Z.)
| | - Elena L. Chernolovskaya
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia; (A.B.); (A.V.S.); (I.A.S.); (D.V.G.); (M.A.Z.)
| |
Collapse
|
7
|
Geng C, Wang X, Chen J, Sun N, Wang Y, Li Z, Han L, Hou S, Fan H, Li N, Gong Y. Repetitive Low-Level Blast Exposure via Akt/NF-κB Signaling Pathway Mediates the M1 Polarization of Mouse Alveolar Macrophage MH-S Cells. Int J Mol Sci 2023; 24:10596. [PMID: 37445774 DOI: 10.3390/ijms241310596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 07/15/2023] Open
Abstract
Repetitive low-level blast (rLLB) exposure is a potential risk factor for the health of soldiers or workers who are exposed to it as an occupational characteristic. Alveolar macrophages (AMs) are susceptible to external blast waves and produce pro-inflammatory or anti-inflammatory effects. However, the effect of rLLB exposure on AMs is still unclear. Here, we generated rLLB waves through a miniature manual Reddy-tube and explored their effects on MH-S cell morphology, phenotype transformation, oxidative stress status, and apoptosis by immunofluorescence, real-time quantitative PCR (qPCR), western blotting (WB) and flow cytometry. Ipatasertib (GDC-0068) or PDTC was used to verify the role of the Akt/NF-κB signaling pathway in these processes. Results showed that rLLB treatment could cause morphological irregularities and cytoskeletal disorders in MH-S cells and promote their polarization to the M1 phenotype by increasing iNOS, CD86 and IL-6 expression. The molecular mechanism is through the Akt/NF-κB signaling pathway. Moreover, we found reactive oxygen species (ROS) burst, Ca2+ accumulation, mitochondrial membrane potential reduction, and early apoptosis of MH-S cells. Taken together, our findings suggest rLLB exposure may cause M1 polarization and early apoptosis of AMs. Fortunately, it is blocked by specific inhibitors GDC-0068 or PDTC. This study provides a new treatment strategy for preventing and alleviating health damage in the occupational population caused by rLLB exposure.
Collapse
Affiliation(s)
- Chenhao Geng
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Xinyue Wang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Jiale Chen
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Na Sun
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yuru Wang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Zizheng Li
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Lu Han
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Ning Li
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yanhua Gong
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| |
Collapse
|
8
|
Li Y, Yan B, Wu Y, Peng Q, Wei Y, Chen Y, Zhang Y, Ma N, Yang X, Ma P. Ferroptosis participates in dibutyl phthalate-aggravated allergic asthma in ovalbumin-sensitized mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 256:114848. [PMID: 37018853 DOI: 10.1016/j.ecoenv.2023.114848] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 06/19/2023]
Abstract
Dibutyl phthalate (DBP), used as a plasticizer, is of wide concern as an environmental pollutant since it has certain immunotoxicity. Although there is growing evidence supporting a link between DBP exposure and allergic airway inflammation, there is less information concerned with whether the ferroptosis pathway is involved in DBP-aggravated allergic asthma in ovalbumin (OVA)-sensitized mice. This study aimed to investigate the role and underlying mechanisms of ferroptosis in DBP-exposed allergic asthmatic mice. Balb/c mice were orally exposed to 40 mg/kg-1 DBP for 28 days, followed by sensitization with OVA and seven consecutive challenges with nebulized OVA. We analyzed airway hyperresponsiveness (AHR), immunoglobulins, inflammation and pulmonary histopathology, to investigate whether DBP exacerbates allergic asthma in OVA-induced mice. We also measured the biomarkers of ferroptosis (Fe2+, GPX4, PTGS2), proteins related to the ferroptosis pathway (VEGF, IL-33, HMGB1, SLC7A11, ALOX15, PEBP1), and indices of lipid peroxidation (ROS, Lipid ROS, GSH, MDA, 4-HNE), to explore the role of ferroptosis in DBP+OVA mice. Finally, we used ferrostatin-1 (Fer-1) as an antagonist against the harmful effects of DBP. The results showed that, DBP+OVA mice had a significant increase in AHR, airway wall remodeling and airway inflammation. Further, we showed that DBP aggravated allergic asthma via ferroptosis and lipid peroxidation, and that Fer-1 inhibited ferroptosis and alleviated the pulmonary toxicity of DBP. These results suggest that ferroptosis participates in the exacerbation of allergic asthma resulting from oral exposure to DBP, highlighting a novel pathway for the connection between DBP and allergic asthma.
Collapse
Affiliation(s)
- Yan Li
- Xianning Engineering Research Center for Healthy Environment, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Biao Yan
- Xianning Engineering Research Center for Healthy Environment, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Yang Wu
- Xianning Engineering Research Center for Healthy Environment, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Qi Peng
- Xianning Engineering Research Center for Healthy Environment, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Yaolu Wei
- Xianning Engineering Research Center for Healthy Environment, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Yenan Chen
- Xianning Engineering Research Center for Healthy Environment, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Yuping Zhang
- Xianning Engineering Research Center for Healthy Environment, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Ning Ma
- Xianning Engineering Research Center for Healthy Environment, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Xu Yang
- Xianning Engineering Research Center for Healthy Environment, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China; Section of Environmental Biomedicine, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan 430079, PR China
| | - Ping Ma
- Xianning Engineering Research Center for Healthy Environment, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China.
| |
Collapse
|
9
|
Díaz-García E, García-Sánchez A, Sánz-Rubio D, Alfaro E, López-Fernández C, Casitas R, Mañas Baena E, Cano-Pumarega I, Cubero P, Marin-Oto M, López-Collazo E, Marin JM, García-Río F, Cubillos-Zapata C. SMAD4 Expression in Monocytes as a Potential Biomarker for Atherosclerosis Risk in Patients with Obstructive Sleep Apnea. Int J Mol Sci 2023; 24:ijms24097900. [PMID: 37175608 PMCID: PMC10178665 DOI: 10.3390/ijms24097900] [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: 03/13/2023] [Revised: 04/14/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Obstructive sleep apnea (OSA) patients are at special risk of suffering atherosclerosis, leading to major cardiovascular diseases. Notably, the transforming growth factor (TGF-β) plays a crucial role in the development and progression of atherosclerosis. In this context, the central regulator of TGF-β pathway, SMAD4 (small mother against decapentaplegic homolog 4), has been previously reported to be augmented in OSA patients, which levels were even higher in patients with concomitant cardiometabolic diseases. Here, we analyzed soluble and intracellular SMAD4 levels in plasma and monocytes from OSA patients and non-apneic subjects, with or without early subclinical atherosclerosis (eSA). In addition, we used in vitro and ex vivo models to explore the mechanisms underlying SMAD4 upregulation and release. Our study confirmed elevated sSMAD4 levels in OSA patients and identified that its levels were even higher in those OSA patients with eSA. Moreover, we demonstrated that SMAD4 is overexpressed in OSA monocytes and that intermittent hypoxia contributes to SMAD4 upregulation and release in a process mediated by NLRP3. In conclusion, this study highlights the potential role of sSMAD4 as a biomarker for atherosclerosis risk in OSA patients and provides new insights into the mechanisms underlying its upregulation and release to the extracellular space.
Collapse
Affiliation(s)
- Elena Díaz-García
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| | - Aldara García-Sánchez
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Servicio de Neumología, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - David Sánz-Rubio
- Precision Medicine in Respiratory Diseases Group, Miguel Servet University Hospital-IIS Aragon, 50009 Zaragoza, Spain
| | - Enrique Alfaro
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| | - Cristina López-Fernández
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| | - Raquel Casitas
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| | - Eva Mañas Baena
- Servicio de Neumología, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Irene Cano-Pumarega
- Servicio de Neumología, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Pablo Cubero
- Precision Medicine in Respiratory Diseases Group, Miguel Servet University Hospital-IIS Aragon, 50009 Zaragoza, Spain
| | - Marta Marin-Oto
- Precision Medicine in Respiratory Diseases Group, Miguel Servet University Hospital-IIS Aragon, 50009 Zaragoza, Spain
| | - Eduardo López-Collazo
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- The Innate Immune Response Group, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| | - José María Marin
- Precision Medicine in Respiratory Diseases Group, Miguel Servet University Hospital-IIS Aragon, 50009 Zaragoza, Spain
- Department of Medicine, University of Zaragoza School of Medicine, 50009 Zaragoza, Spain
| | - Francisco García-Río
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
- Faculty of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Carolina Cubillos-Zapata
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| |
Collapse
|
10
|
Yiang GT, Wu CC, Lu CL, Hu WC, Tsai YJ, Huang YM, Su WL, Lu KC. Endoplasmic Reticulum Stress in Elderly Patients with COVID-19: Potential of Melatonin Treatment. Viruses 2023; 15:156. [PMID: 36680196 PMCID: PMC9863214 DOI: 10.3390/v15010156] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Aging processes, including immunosenescence, inflammation, inflammasome formation, genomic instability, telomeric attrition, and altered autophagy, are involved in viral infections and they may contribute to increased pathophysiological responses to the SARS-CoV-2 infection in the elderly; this poses additional risks of accelerated aging, which could be found even after recovery. Aging is associated with oxidative damage. Moreover, SARS-CoV-2 infections may increase the production of reactive oxygen species and such infections will disturb the Ca++ balance via an endoplasmic reticulum (ER) stress-mediated unfolded protein response. Although vaccine development and anti-inflammation therapy lower the severity of COVID-19, the prevalence and mortality rates are still alarming in some countries worldwide. In this review, we describe the involvement of viral proteins in activating ER stress transducers and their downstream signals and in inducing inflammation and inflammasome formation. Furthermore, we propose the potential of melatonin as an ER stress modulator, owing to its antioxidant, anti-inflammatory, and immunoregulatory effects in viral infections. Considering its strong safety profile, we suggest that additive melatonin supplementation in the elderly could be beneficial in treating COVID-19.
Collapse
Affiliation(s)
- Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
- School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Chia-Chao Wu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan
| | - Chien-Lin Lu
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei 24352, Taiwan
| | - Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| | - Yi-Ju Tsai
- Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei 243, Taiwan
| | - Yiao-Mien Huang
- Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| | - Wen-Lin Su
- School of Medicine, Tzu Chi University, Hualien 970, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei 24352, Taiwan
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| |
Collapse
|
11
|
Zhang Y, Xi Y, Yang C, Gong W, Wang C, Wu L, Wang D. Short-Chain Fatty Acids Attenuate 5-Fluorouracil-Induced THP-1 Cell Inflammation through Inhibiting NF-κB/NLRP3 Signaling via Glycerolphospholipid and Sphingolipid Metabolism. Molecules 2023; 28:molecules28020494. [PMID: 36677551 PMCID: PMC9864921 DOI: 10.3390/molecules28020494] [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: 12/14/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
5-Fluorouracil (5-FU) is a common anti-tumor drug, but there is no effective treatment for its side effect, intestinal mucositis. The inflammatory reaction of macrophages in intestinal mucosa induced by 5-FU is an important cause of intestinal mucositis. In this study, we investigated the anti-inflammatory effects of the three important short-chain fatty acids (SCFAs), including sodium acetate (NaAc), sodium propionate (NaPc), and sodium butyrate (NaB), on human mononuclear macrophage-derived THP-1 cells induced by 5-FU. The expressions of intracellular ROS, pro-inflammatory/anti-inflammatory cytokines, as well as the nuclear factor-κB/NLR family and pyrin domain-containing protein 3 (NF-κB/NLRP3) signaling pathway proteins were determined. Furthermore, the cell metabolites were analyzed by untargeted metabolomics techniques. Our results revealed that the three SCFAs inhibited pro-inflammatory factor expressions, including IL-1β and IL-6, when treated with 5-FU (p < 0.05). The ROS expression and NF-κB activity of 5-FU-treated THP-1 cells were inhibited by the three SCFAs pre-incubated (p < 0.05). Moreover, NLRP3 knockdown abolished 5-FU-induced IL-1β expression (p < 0.05). Further experiments showed that the three SCFAs affected 20 kinds of metabolites that belong to amino acid and phosphatidylcholine metabolism in THP-1 cells. These significantly altered metabolites were involved in amino acid metabolism and glycerolphospholipid and sphingolipid metabolism. It is the first time that three important SCFAs (NaAc, NaPc, and NaB) were identified as inhibiting 5-FU-induced macrophage inflammation through inhibiting ROS/NF-κB/NLRP3 signaling pathways and regulating glycerolphospholipid and sphingolipid metabolism.
Collapse
Affiliation(s)
- Yanyan Zhang
- Testing Center, Yangzhou University, Yangzhou 225009, China
| | - Yue Xi
- Medical Laboratory Department, Huai’an Second People’s Hospital, Huai’an 223022, China
| | - Changshui Yang
- School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Weijuan Gong
- School of Medicine, Yangzhou University, Yangzhou 225009, China
- Correspondence: (W.G.); (D.W.)
| | - Chengyin Wang
- Testing Center, Yangzhou University, Yangzhou 225009, China
| | - Liang Wu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
- Correspondence: (W.G.); (D.W.)
| |
Collapse
|
12
|
Hsieh PC, Wu YK, Yang MC, Su WL, Kuo CY, Lan CC. Deciphering the role of damage-associated molecular patterns and inflammatory responses in acute lung injury. Life Sci 2022; 305:120782. [PMID: 35809663 DOI: 10.1016/j.lfs.2022.120782] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 11/29/2022]
Abstract
Acute lung injury (ALI) is characterized by diffuse pulmonary infiltrates and causes great mortality. ALI presents with overproduction of proinflammatory cytokines, cell death, destruction of alveoli-endothelial barriers, and neutrophil infiltration in lung tissues. Damage-associated molecular patterns (DAMPs) are molecules released from damaged cells due to infection, trauma, etc. DAMPs activate innate and adaptive immunity, trigger inflammatory responses, and are important in the initiation and development of ALI. We reviewed the literatures on DAMPs in ALI. Alveolar macrophages (AMs), neutrophils, and epithelial cells (AECs) are important in the pathogenesis of ALI. We comprehensively analyzed the interaction between DAMPs and AMs, alveolar neutrophils, and AECs. During the initial stage of ALI, ruptured cell membranes or destroyed mitochondria release DAMPs. DAMPs activate the inflammasome in nearby sentinel immune cells, such as AMs. AMs produce IL-1β and other cytokines. These mediators upregulate adhesion molecules of the capillary endothelium that facilitate neutrophil recruitment. The recruited neutrophils detect DAMPs using formyl peptide receptors on the membrane, guiding their migration to the injured site. The accumulation of immune cells, cytokines, chemokines, proteases, etc., results in diffuse alveolar damage and pulmonary hyperpermeability with protein-rich fluid retention. Some clinical studies have shown that patients with ALI with higher circulating DAMPs have higher mortality rates. In conclusion, DAMPs are important in the initiation and progression of ALI. The interactions between DAMPs and AMs, neutrophils, and AECs are important in ALI. This review comprehensively addresses the mechanisms of DAMPs and their interactions in ALI.
Collapse
Affiliation(s)
- Po-Chun Hsieh
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yao-Kuang Wu
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Mei-Chen Yang
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Lin Su
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Chou-Chin Lan
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
13
|
Herminghaus A, Kozlov AV, Szabó A, Hantos Z, Gylstorff S, Kuebart A, Aghapour M, Wissuwa B, Walles T, Walles H, Coldewey SM, Relja B. A Barrier to Defend - Models of Pulmonary Barrier to Study Acute Inflammatory Diseases. Front Immunol 2022; 13:895100. [PMID: 35874776 PMCID: PMC9300899 DOI: 10.3389/fimmu.2022.895100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/20/2022] [Indexed: 12/04/2022] Open
Abstract
Pulmonary diseases represent four out of ten most common causes for worldwide mortality. Thus, pulmonary infections with subsequent inflammatory responses represent a major public health concern. The pulmonary barrier is a vulnerable entry site for several stress factors, including pathogens such as viruses, and bacteria, but also environmental factors e.g. toxins, air pollutants, as well as allergens. These pathogens or pathogen-associated molecular pattern and inflammatory agents e.g. damage-associated molecular pattern cause significant disturbances in the pulmonary barrier. The physiological and biological functions, as well as the architecture and homeostatic maintenance of the pulmonary barrier are highly complex. The airway epithelium, denoting the first pulmonary barrier, encompasses cells releasing a plethora of chemokines and cytokines, and is further covered with a mucus layer containing antimicrobial peptides, which are responsible for the pathogen clearance. Submucosal antigen-presenting cells and neutrophilic granulocytes are also involved in the defense mechanisms and counterregulation of pulmonary infections, and thus may directly affect the pulmonary barrier function. The detailed understanding of the pulmonary barrier including its architecture and functions is crucial for the diagnosis, prognosis, and therapeutic treatment strategies of pulmonary diseases. Thus, considering multiple side effects and limited efficacy of current therapeutic treatment strategies in patients with inflammatory diseases make experimental in vitro and in vivo models necessary to improving clinical therapy options. This review describes existing models for studyying the pulmonary barrier function under acute inflammatory conditions, which are meant to improve the translational approaches for outcome predictions, patient monitoring, and treatment decision-making.
Collapse
Affiliation(s)
- Anna Herminghaus
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Andrey V. Kozlov
- L Boltzmann Institute for Traumatology in Cooperation with AUVA and Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Department of Human Pathology , IM Sechenov Moscow State Medical University, Moscow, Russia
| | - Andrea Szabó
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Zoltán Hantos
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Severin Gylstorff
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Kuebart
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Mahyar Aghapour
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Bianka Wissuwa
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Thorsten Walles
- Department of Thoracic Surgery, Magdeburg University Medicine, Magdeburg, Germany
| | - Heike Walles
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- Core Facility Tissue Engineering, Otto-von-Guericke-University, Magdeburg, Germany
| | - Sina M. Coldewey
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- *Correspondence: Borna Relja,
| |
Collapse
|
14
|
Zhang M, Sun Y, Ding C, Hong S, Li N, Guan Y, Zhang L, Dong X, Cao J, Yao W, Ren W, Yao S. Metformin mitigates gas explosion‑induced blast lung injuries through AMPK‑mediated energy metabolism and NOX2‑related oxidation pathway in rats. Exp Ther Med 2022; 24:529. [PMID: 35837050 PMCID: PMC9257965 DOI: 10.3892/etm.2022.11456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022] Open
Abstract
Gas explosions are a recurrent event in coal mining that cause severe pulmonary damage due to shock waves, and there is currently no effective targeted treatment. To illustrate the mechanism of gas explosion-induced lung injury and to explore strategies for blast lung injury (BLI) treatment, the present study used a BLI rat model and supplementation with metformin (MET), an AMP-activated protein kinase (AMPK) activator, at a dose of 10 mg/kg body weight by intraperitoneal injection. Protein expression levels were detected by western blotting. Significantly decreased expression of phosphorylated (p)-AMPK, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α) and metabolic activity were observed in the BLI group compared with those in the control group. However, the mitochondrial stability, metabolic activity and expression of p-AMPK and PGC1α were elevated following MET treatment. These results suggested that MET could attenuate gas explosion-induced BLI by improving mitochondrial homeostasis. Meanwhile, high expression of nicotinamide adenine dinucleotide phosphate oxidase (NOX2) and low expression of catalase (CAT) were observed in the BLI group. The expression levels of NOX2 and CAT were restored in the BLI + MET group relative to changes in the BLI group, and the accumulation of oxidative stress was successfully reversed following MET treatment. Overall, these findings revealed that MET could alleviate BLI by activating the AMPK/PGC1α pathway and inhibiting oxidative stress caused by NOX2 activation.
Collapse
Affiliation(s)
- Miao Zhang
- Research Center for Precision Prevention and Control of Occupational Hazards, School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Yunzhe Sun
- Research Center for Precision Prevention and Control of Occupational Hazards, School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Chunjie Ding
- Research Center for Precision Prevention and Control of Occupational Hazards, School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Shan Hong
- Research Center for Precision Prevention and Control of Occupational Hazards, School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Ning Li
- Department of Occupational and Environmental Health, School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Yi Guan
- Department of Occupational and Environmental Health, School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Lin Zhang
- Key Laboratory of Birth Regulation and Control Technology, National Health Commission of China, Maternal and Child Care Hospital of Shandong Province, Shandong University, Jinan, Shandong 250001, P.R. China
| | - Xinwen Dong
- Research Center for Precision Prevention and Control of Occupational Hazards, School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Jia Cao
- Institute of Toxicology, College of Preventive Medicine, Army Medical University, Chongqing 400038, P.R. China
| | - Wu Yao
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Wenjie Ren
- Research Center for Precision Prevention and Control of Occupational Hazards, School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Sanqiao Yao
- Research Center for Precision Prevention and Control of Occupational Hazards, School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
15
|
Rosser JI, Nagy N, Goel R, Kaber G, Demirdjian S, Saxena J, Bollyky JB, Frymoyer AR, Pacheco-Navarro AE, Burgener EB, Rajadas J, Wang Z, Arbach O, Dunn CE, Kalinowski A, Milla CE, Bollyky PL. Oral hymecromone decreases hyaluronan in human study participants. J Clin Invest 2022; 132:e157983. [PMID: 35499083 PMCID: PMC9057598 DOI: 10.1172/jci157983] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUNDHyaluronan (HA), an extracellular matrix glycosaminoglycan, has been implicated in the pathophysiology of COVID-19 infection, pulmonary hypertension, pulmonary fibrosis, and other diseases, but is not targeted by any approved drugs. We asked whether hymecromone (4-methylumbelliferone [4-MU]), an oral drug approved in Europe for biliary spasm treatment that also inhibits HA in vitro and in animal models, could be repurposed as an inhibitor of HA synthesis in humans.METHODSWe conducted an open-label, single-center, dose-response study of hymecromone in healthy adults. Subjects received hymecromone at 1200 (n = 8), 2400 (n = 9), or 3600 (n = 9) mg/d divided into 3 doses daily, administered orally for 4 days. We assessed safety and tolerability of hymecromone and analyzed HA, 4-MU, and 4-methylumbelliferyl glucuronide (4-MUG; the main metabolite of 4-MU) concentrations in sputum and serum.RESULTSHymecromone was well tolerated up to doses of 3600 mg/d. Both sputum and serum drug concentrations increased in a dose-dependent manner, indicating that higher doses lead to greater exposures. Across all dose arms combined, we observed a significant decrease in sputum HA from baseline after 4 days of treatment. We also observed a decrease in serum HA. Additionally, higher baseline sputum HA levels were associated with a greater decrease in sputum HA.CONCLUSIONAfter 4 days of exposure to oral hymecromone, healthy human subjects experienced a significant reduction in sputum HA levels, indicating this oral therapy may have potential in pulmonary diseases where HA is implicated in pathogenesis.TRIAL REGISTRATIONClinicalTrials.gov NCT02780752.FUNDINGStanford Medicine Catalyst, Stanford SPARK, Stanford Innovative Medicines Accelerator program, NIH training grants 5T32AI052073-14 and T32HL129970.
Collapse
Affiliation(s)
- Joelle I. Rosser
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| | - Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| | - Riya Goel
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| | - Sally Demirdjian
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| | - Jamie Saxena
- Division of Infectious Diseases, Department of Pediatrics
| | | | | | | | | | - Jayakumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute & Pulmonary and Critical Care, Department of Medicine, Stanford University, Stanford, California, USA
- Bioengineering and Therapeutic Sciences, UCSF School of Pharmacy, San Francisco, California, USA
| | - Zhe Wang
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute & Pulmonary and Critical Care, Department of Medicine, Stanford University, Stanford, California, USA
| | - Olga Arbach
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Colleen E. Dunn
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, and
| | - Anissa Kalinowski
- Department of Epidemiology, Stanford University, Stanford, California, USA
| | - Carlos E. Milla
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, and
| | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| |
Collapse
|
16
|
Bergmann-Leitner ES, Bobrov AG, Bolton JS, Rouse MD, Heyburn L, Pavlovic R, Garry BI, Alamneh Y, Long J, Swierczewski B, Tyner S, Getnet D, Sajja VS, Antonic V. Blast Waves Cause Immune System Dysfunction and Transient Bone Marrow Failure in a Mouse Model. Front Bioeng Biotechnol 2022; 10:821169. [PMID: 35392409 PMCID: PMC8980552 DOI: 10.3389/fbioe.2022.821169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/23/2022] [Indexed: 11/29/2022] Open
Abstract
Explosive devices, either conventional or improvised, are common sources of injuries during combat, civil unrest, and terror attacks, resulting in trauma from exposure to blast. A blast wave (BW), a near-instantaneous rise in pressure followed by a negative pressure, propagates through the body in milliseconds and can affect physiology for days/months after exposure. Epidemiological data show that blast-related casualties result in significantly higher susceptibility to wound infections, suggesting long-lasting immune modulatory effects from blast exposure. The mechanisms involved in BW-induced immune changes are poorly understood. We evaluated the effects of BW on the immune system using an established murine model. Animals were exposed to BWs (using an Advanced Blast Simulator), followed by longitudinally sampling for 14 days. Blood, bone marrow, and spleen were analyzed for changes in the 1) complete blood count (CBC), and 2) composition of bone marrow cells (BMC) and splenocytes, and 3) concentrations of systemic cytokines/chemokines. Our data demonstrate that BW results in transient bone marrow failure and long-term changes in the frequency and profile of progenitor cell populations. Viability progressively decreased in hematopoietic stem cells and pluripotent progenitor cells. Significant decrease of CD4+ T cells in the spleen indicates reduced functionality of adaptive immune system. Dynamic changes in the concentrations of several cytokines and chemokines such as IL-1α and IL-17 occurred potentially contributing to dysregulation of immune response after trauma. This work lays the foundation for identifying the potential mechanisms behind BW’s immunosuppressive effects to inform the recognition of this compromised status is crucial for the development of therapeutic interventions for infections to reduce recovery time of wounded patients injured by explosive devices.
Collapse
Affiliation(s)
- Elke S. Bergmann-Leitner
- Biologics Research and Development, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- *Correspondence: Elke S. Bergmann-Leitner, ; Venkatasivasai S. Sajja, ; Vlado Antonic,
| | - Alexander G. Bobrov
- Wound Infections Department, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Jessica S. Bolton
- Biologics Research and Development, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Michael D. Rouse
- Wound Infections Department, Naval Research Medical Center, Silver Spring, MD, United States
- Henry M. Jackson Foundation, Rockville, MD, United States
| | - Lanier Heyburn
- Blast Induced Neurotrauma Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Radmila Pavlovic
- Wound Infections Department, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Brittany I. Garry
- Wound Infections Department, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Yonas Alamneh
- Wound Infections Department, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Joseph Long
- Blast Induced Neurotrauma Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Brett Swierczewski
- Bacterial Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Stuart Tyner
- Military Infectious Diseases Research Program, Frederick, MD, United States
| | - Derese Getnet
- Wound Infections Department, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Venkatasivasai S. Sajja
- Blast Induced Neurotrauma Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- *Correspondence: Elke S. Bergmann-Leitner, ; Venkatasivasai S. Sajja, ; Vlado Antonic,
| | - Vlado Antonic
- Wound Infections Department, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- *Correspondence: Elke S. Bergmann-Leitner, ; Venkatasivasai S. Sajja, ; Vlado Antonic,
| |
Collapse
|
17
|
Tolg C, Messam BJA, McCarthy JB, Nelson AC, Turley EA. Hyaluronan Functions in Wound Repair That Are Captured to Fuel Breast Cancer Progression. Biomolecules 2021; 11:1551. [PMID: 34827550 PMCID: PMC8615562 DOI: 10.3390/biom11111551] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/14/2022] Open
Abstract
Signaling from an actively remodeling extracellular matrix (ECM) has emerged as a critical factor in regulating both the repair of tissue injuries and the progression of diseases such as metastatic cancer. Hyaluronan (HA) is a major component of the ECM that normally functions in tissue injury to sequentially promote then suppress inflammation and fibrosis, a duality in which is featured, and regulated in, wound repair. These essential response-to-injury functions of HA in the microenvironment are hijacked by tumor cells for invasion and avoidance of immune detection. In this review, we first discuss the numerous size-dependent functions of HA and emphasize the multifunctional nature of two of its receptors (CD44 and RHAMM) in regulating the signaling duality of HA in excisional wound healing. This is followed by a discussion of how HA metabolism is de-regulated in malignant progression and how targeting HA might be used to better manage breast cancer progression.
Collapse
Affiliation(s)
- Cornelia Tolg
- London Regional Cancer Program, Lawson Health Research Institute, London, ON N6A 5W9, Canada;
| | - Britney Jodi-Ann Messam
- Department Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada;
| | - James Benjamin McCarthy
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Andrew Cook Nelson
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Eva Ann Turley
- London Regional Cancer Program, Lawson Health Research Institute, Department Oncology, Biochemistry and Surgery, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| |
Collapse
|
18
|
Figueiredo F, Kristoffersen H, Bhat S, Zhang Z, Godfroid J, Peruzzi S, Præbel K, Dalmo RA, Xu X. Immunostimulant Bathing Influences the Expression of Immune- and Metabolic-Related Genes in Atlantic Salmon Alevins. BIOLOGY 2021; 10:980. [PMID: 34681079 PMCID: PMC8533105 DOI: 10.3390/biology10100980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 12/03/2022]
Abstract
Disease resistance of fish larvae may be improved by bath treatment in water containing immunostimulants. Pattern recognition receptors, such as TLR3, TLR7, and MDA5, work as an "early warning" to induce intracellular signaling and facilitate an antiviral response. A single bath of newly hatched larvae, with Astragalus, upregulated the expression of IFNα, IFNc, ISG15, MDA5, PKR, STAT1, TLR3, and TLR7 immune genes, on day 4 post treatment. Similar patterns were observed for Hyaluronic acid and Poly I:C. Increased expression was observed for ISG15, MDA5, MX, STAT1, TLR3, TLR7, and RSAD2, on day 9 for Imiquimod. Metabolic gene expression was stimulated on day 1 after immunostimulant bath in ULK1, MYC, SLC2A1, HIF1A, MTOR, and SIX1, in Astragalus, Hyaluronic acid, and Imiquimod. Expression of NOS2 in Poly I:C was an average fourfold above that of control at the same timepoint. Throughout the remaining sampling days (2, 4, 9, 16, 32, and 45 days post immunostimulant bath), NOS2 and IL1B were consistently overexpressed. In conclusion, the immunostimulants induced antiviral gene responses, indicating that a single bath at an early life stage could enable a more robust antiviral defense in fish. Additionally, it was demonstrated, based on gene expression data, that cell metabolism was perturbed, where several metabolic genes were co-regulated with innate antiviral genes.
Collapse
Affiliation(s)
- Filipe Figueiredo
- Norwegian College of Fishery Science, UiT—The Arctic University of Norway, N-9019 Tromsø, Norway; (H.K.); (S.B.); (K.P.); (X.X.)
| | - Harald Kristoffersen
- Norwegian College of Fishery Science, UiT—The Arctic University of Norway, N-9019 Tromsø, Norway; (H.K.); (S.B.); (K.P.); (X.X.)
| | - Shripathi Bhat
- Norwegian College of Fishery Science, UiT—The Arctic University of Norway, N-9019 Tromsø, Norway; (H.K.); (S.B.); (K.P.); (X.X.)
| | - Zuobing Zhang
- College of Life Sciences, Shanxi University, Taiyuan 030006, China;
| | - Jacques Godfroid
- Department of Arctic and Marine Biology, UiT—The Arctic University of Norway, N-9019 Tromsø, Norway; (J.G.); (S.P.)
| | - Stefano Peruzzi
- Department of Arctic and Marine Biology, UiT—The Arctic University of Norway, N-9019 Tromsø, Norway; (J.G.); (S.P.)
| | - Kim Præbel
- Norwegian College of Fishery Science, UiT—The Arctic University of Norway, N-9019 Tromsø, Norway; (H.K.); (S.B.); (K.P.); (X.X.)
| | - Roy Ambli Dalmo
- Norwegian College of Fishery Science, UiT—The Arctic University of Norway, N-9019 Tromsø, Norway; (H.K.); (S.B.); (K.P.); (X.X.)
| | - Xiaoli Xu
- Norwegian College of Fishery Science, UiT—The Arctic University of Norway, N-9019 Tromsø, Norway; (H.K.); (S.B.); (K.P.); (X.X.)
| |
Collapse
|
19
|
Land WG. Role of DAMPs in respiratory virus-induced acute respiratory distress syndrome-with a preliminary reference to SARS-CoV-2 pneumonia. Genes Immun 2021; 22:141-160. [PMID: 34140652 PMCID: PMC8210526 DOI: 10.1038/s41435-021-00140-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/28/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023]
Abstract
When surveying the current literature on COVID-19, the "cytokine storm" is considered to be pathogenetically involved in its severe outcomes such as acute respiratory distress syndrome, systemic inflammatory response syndrome, and eventually multiple organ failure. In this review, the similar role of DAMPs is addressed, that is, of those molecules, which operate upstream of the inflammatory pathway by activating those cells, which ultimately release the cytokines. Given the still limited reports on their role in COVID-19, the emerging topic is extended to respiratory viral infections with focus on influenza. At first, a brief introduction is given on the function of various classes of activating DAMPs and counterbalancing suppressing DAMPs (SAMPs) in initiating controlled inflammation-promoting and inflammation-resolving defense responses upon infectious and sterile insults. It is stressed that the excessive emission of DAMPs upon severe injury uncovers their fateful property in triggering dysregulated life-threatening hyperinflammatory responses. Such a scenario may happen when the viral load is too high, for example, in the respiratory tract, "forcing" many virus-infected host cells to decide to commit "suicidal" regulated cell death (e.g., necroptosis, pyroptosis) associated with release of large amounts of DAMPs: an important topic of this review. Ironically, although the aim of this "suicidal" cell death is to save and restore organismal homeostasis, the intrinsic release of excessive amounts of DAMPs leads to those dysregulated hyperinflammatory responses-as typically involved in the pathogenesis of acute respiratory distress syndrome and systemic inflammatory response syndrome in respiratory viral infections. Consequently, as briefly outlined in this review, these molecules can be considered valuable diagnostic and prognostic biomarkers to monitor and evaluate the course of the viral disorder, in particular, to grasp the eventual transition precociously from a controlled defense response as observed in mild/moderate cases to a dysregulated life-threatening hyperinflammatory response as seen, for example, in severe/fatal COVID-19. Moreover, the pathogenetic involvement of these molecules qualifies them as relevant future therapeutic targets to prevent severe/ fatal outcomes. Finally, a theory is presented proposing that the superimposition of coronavirus-induced DAMPs with non-virus-induced DAMPs from other origins such as air pollution or high age may contribute to severe and fatal courses of coronavirus pneumonia.
Collapse
Affiliation(s)
- Walter Gottlieb Land
- German Academy for Transplantation Medicine, Munich, Germany.
- Molecular ImmunoRheumatology, INSERM UMR_S1109, Laboratory of Excellence Transplantex, University of Strasbourg, Strasbourg, France.
| |
Collapse
|