1
|
Oterkus M, Pala M, Yilmaz SG, Tanriverdi ES, Gunduz A, Delen LA, Ozturk DA, Döger C. Evaluation of hypoxia pathway genes and serum parameters in new coronavirus pneumonia (COVID-19). Gene 2025; 955:149395. [PMID: 40086704 DOI: 10.1016/j.gene.2025.149395] [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: 10/24/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Coronavirus disease-2019 (COVID-19) causes severe hypoxemia. Unlike normal pneumonia, pneumonia due to COVID-19 causes oxygen deprivation without breathing difficulties (i.e., silent hypoxia). We evaluated the relationship between COVID-19 and hypoxemia and examined possible mechanisms of pneumonia from the perspective of gene expression (HIF1A, vascular endothelial growth factor [VEGF], NF-kB, MEKK1, and EGFR) using real-time PCR and ELISA for serum parameters. METHODS We evaluated 100 individuals (50 patients and 50 controls). The patients were individuals with respiratory symptoms and pneumonia who were COVİD-19 positive. The relative quantification of standardized samples wa s calculated according to the formula 2 -ΔΔCT. Receiver operating curve (ROC) analysis was made to define the diagnostic power of the genes. The expression changes of four genes in the hypoxia pathway were significant (excluding VEGF) and upregulated in the patients' serums. RESULTS The fold change values of the HIF1A, VEGF, NF-kB, MEKK1, and EGFR genes were 0.048, 0.688, 0.168, 0.207, and 0.171, respectively, in the cases checked against to the controls. The areas under the ROC values indicating the diagnostic power of the genes were 0.727, 0.538, 0.815, 0.734, and 0.936, respectively. Some serum parameters were significant (age, PCR, urea, LDH, WBC, ferritin, and pO2). CONCLUSIONS The upregulation of some genes in the hypoxia pathway in COVID-19 pneumonia shows that these genes and protein products are candidates for treatment targets. At the same time, the high discriminative power of two genes (NF-κB and EGFR) in patients compared to controls indicates their diagnostic potential in serum samples.
Collapse
Affiliation(s)
- Mesut Oterkus
- Malatya Turgut Ozal University, Medical Faculty, Department of Anesthesiology and Reanimation, Malatya, Turkey.
| | - Mukaddes Pala
- Malatya Turgut Ozal University, Faculty of Medicine, Department of Physiology, Malatya, Turkey.
| | - Senay Gorucu Yilmaz
- Gaziantep University, Faculty of Health Science, Department of Nutrition and Dietetics, Gaziantep, Turkey.
| | | | - Ayten Gunduz
- Malatya Turgut Ozal University, Faculty of Medicine, Department of Microbiology, Malatya, Turkey.
| | | | - Dilara Altay Ozturk
- Malatya Turgut Ozal University, Faculty of Medicine, Department of Physiology, Malatya, Turkey.
| | - Cihan Döger
- Health Sciences University, Ankara Bilkent City Hospital, Department of Anesthesiology and Reanimation, Ankara, Turkey
| |
Collapse
|
2
|
Lotsios NS, Keskinidou C, Karagiannis SP, Papavassiliou KA, Papavassiliou AG, Kotanidou A, Dimopoulou I, Orfanos SE, Vassiliou AG. Expression and Regulation of Hypoxia-Inducible Factor Signalling in Acute Lung Inflammation. Cells 2024; 14:29. [PMID: 39791730 PMCID: PMC11719729 DOI: 10.3390/cells14010029] [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: 12/12/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are central regulators of gene expression in response to oxygen deprivation, a common feature in critical illnesses. The significant burden that critical illnesses place on global healthcare systems highlights the need for a deeper understanding of underlying mechanisms and the development of innovative treatment strategies. Among critical illnesses, impaired lung function is frequently linked to hypoxic conditions. This review focuses on the expression and regulation of HIF signalling in experimental models of acute lung injury (ALI) and clinical studies in critically ill patients with acute respiratory distress syndrome (ARDS). We explore the potential dual role of HIF signalling in acute lung inflammation. Furthermore, its role in key biological processes and its potential prognostic significance in clinical scenarios are discussed. Finally, we explore recent pharmacological advancements targeting HIF signalling, which have emerged as promising alternatives to existing therapeutic approaches, potentially enabling more effective management strategies.
Collapse
Affiliation(s)
- Nikolaos S. Lotsios
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Chrysi Keskinidou
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Sotirios P. Karagiannis
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, ‘Sotiria’ Chest Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Anastasia Kotanidou
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Ioanna Dimopoulou
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Stylianos E. Orfanos
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Alice G. Vassiliou
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| |
Collapse
|
3
|
Li C, Lian Y, Lin Y, Li Z. A Network Pharmacology and Molecular Dynamics Simulation-Based Study of Qing Run Hua Jie Decoction in Interstitial Pneumonia Treatment. Infect Drug Resist 2024; 17:605-621. [PMID: 38379588 PMCID: PMC10878319 DOI: 10.2147/idr.s433755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/22/2024] [Indexed: 02/22/2024] Open
Abstract
Objective This study is dedicated to revealing the potential mechanism of Qin Run Hua Jie (QRHJ) decoction in Interstitial pneumonia (IP) treatment. Methods The TCMSP database predicted the chemical components and targets of QRHJ decoction, and the IP-related genes were from the Genecards database. Cytoscape software was used to establish the interaction network. R package clusterProfiler was utilized for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. The molecular docking analysis of target proteins and the corresponding active pharmaceutical ingredients in the core position of the interaction network was conducted. Then, molecular dynamics (MD) simulations of a potential active substance and its key targets were performed. The binding efficiency of EGFR and luteolin, HIF1A and diosgenin was detected by cellular thermal shift assay (CETSA), and protein expression was measured by Western blot. CCK-8 was used to detect cell activity. Results A total of 153 active ingredients, 127 targets and 362 IP-related genes were obtained. KEGG enrichment analysis identified IP-related signaling pathways including HIF-1 signaling pathway and TNF signaling pathway. The two key components luteolin and diosgenin stably bound to the key targets EGFR and HIF1A. Cell experiments further showed that EGFR and luteolin, HIF1A and diosgenin bound to exert anti-fibrotic effects. Conclusion As an active ingredient of QRHJ decoction, luteolin and diosgenin may exert therapeutic effect on IP through binding to the key target EGFR and HIF1A. This work initially revealed the key molecular mechanism of QRHJ decoction in IP treatment and offered theoretical evidence.
Collapse
Affiliation(s)
- Chunxiang Li
- Department of Integrative Medicine Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, 362000, People’s Republic of China
| | - Yingbin Lian
- Department of Integrative Medicine Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, 362000, People’s Republic of China
| | - Yaoshen Lin
- Department of Integrative Medicine Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, 362000, People’s Republic of China
| | - Zhihua Li
- Department of Oncology, Zhangzhou Second Hospital, Zhangzhou, Fujian, 363199, People’s Republic of China
| |
Collapse
|
4
|
Shan C, Xia Y, Wu Z, Zhao J. HIF-1α and periodontitis: Novel insights linking host-environment interplay to periodontal phenotypes. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 184:50-78. [PMID: 37769974 DOI: 10.1016/j.pbiomolbio.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/27/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
Periodontitis, the sixth most prevalent epidemic disease globally, profoundly impacts oral aesthetics and masticatory functionality. Hypoxia-inducible factor-1α (HIF-1α), an oxygen-dependent transcriptional activator, has emerged as a pivotal regulator in periodontal tissue and alveolar bone metabolism, exerts critical functions in angiogenesis, erythropoiesis, energy metabolism, and cell fate determination. Numerous essential phenotypes regulated by HIF are intricately associated with bone metabolism in periodontal tissues. Extensive investigations have highlighted the central role of HIF and its downstream target genes and pathways in the coupling of angiogenesis and osteogenesis. Within this concise perspective, we comprehensively review the cellular phenotypic alterations and microenvironmental dynamics linking HIF to periodontitis. We analyze current research on the HIF pathway, elucidating its impact on bone repair and regeneration, while unraveling the involved cellular and molecular mechanisms. Furthermore, we briefly discuss the potential application of targeted interventions aimed at HIF in the field of bone tissue regeneration engineering. This review expands our biological understanding of the intricate relationship between the HIF gene and bone angiogenesis in periodontitis and offers valuable insights for the development of innovative therapies to expedite bone repair and regeneration.
Collapse
Affiliation(s)
- Chao Shan
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - YuNing Xia
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - Zeyu Wu
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - Jin Zhao
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China; Xinjiang Uygur Autonomous Region Institute of Stomatology, Ürümqi, China.
| |
Collapse
|
5
|
Suresh MV, Aktay S, Yalamanchili G, Solanki S, Sathyarajan DT, Arnipalli MS, Pennathur S, Raghavendran K. Role of succinate in airway epithelial cell regulation following traumatic lung injury. JCI Insight 2023; 8:e166860. [PMID: 37737265 PMCID: PMC10561732 DOI: 10.1172/jci.insight.166860] [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: 11/07/2022] [Accepted: 08/17/2023] [Indexed: 09/23/2023] Open
Abstract
Lung contusion and gastric aspiration (LC and GA) are major risk factors for developing acute respiratory distress following trauma. Hypoxia from lung injury is mainly regulated by hypoxia-inducible factor 1α (HIF-1α). Published data from our group indicate that HIF-1α regulation in airway epithelial cells (AEC) drives the acute inflammatory response following LC and GA. Metabolomic profiling and metabolic flux of Type II AEC following LC revealed marked increases in glycolytic and TCA intermediates in vivo and in vitro that were HIF-1α dependent. GLUT-1/4 expression was also increased in HIF-1α+/+ mice, suggesting that increased glucose entry may contribute to increased intermediates. Importantly, lactate incubation in vitro on Type II cells did not significantly increase the inflammatory byproduct IL-1β. Contrastingly, succinate had a direct proinflammatory effect on human small AEC by IL-1β generation in vitro. This effect was reversed by dimethylmalonate, suggesting an important role for succinate dehydrogenase in mediating HIF-1α effects. We confirmed the presence of the only known receptor for succinate binding, SUCNR1, on Type II AEC. These results support the hypothesis that succinate drives HIF-1α-mediated airway inflammation following LC. This is the first report to our knowledge of direct proinflammatory activation of succinate in nonimmune cells such as Type II AEC in direct lung injury models.
Collapse
|
6
|
Sun H, Gao W, Chen R, Chen S, Gu X, Wang F, Li Q. CircRNAs in BALF exosomes and plasma as diagnostic biomarkers in patients with acute respiratory distress syndrome caused by severe pneumonia. Front Cell Infect Microbiol 2023; 13:1194495. [PMID: 37674577 PMCID: PMC10477665 DOI: 10.3389/fcimb.2023.1194495] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/29/2023] [Indexed: 09/08/2023] Open
Abstract
Background The transcriptomic studies targeting circular RNAs (circRNAs) in bronchoalveolar lavage fluid (BALF) exosomes of acute respiratory distress syndrome (ARDS) patients caused by severe pneumonia have rarely been reported. This study aimed to screen and validate abnormally expressed circRNAs in exosomes from BALF of patients with ARDS caused by severe pneumonia and then evaluate the diagnostic values of these circRNAs for ARDS. Method BALF was collected from four patients with ARDS caused by severe pneumonia and four healthy subjects. CircRNA expression profile was obtained by microarray analysis in BALF exosomes of the discovery cohort. The differentially expressed circRNAs in BALF exosomes were verified by real-time quantitative PCR (RT-qPCR) and underwent competitive endogenous RNA (ceRNA) network construction and functional enrichment analysis. Results A total of 629 circRNAs were differentially expressed in BALF exosomes between ARDS patients and healthy subjects. Nine differentially expressed circRNAs were validated by RT-qPCR, and seven were consistent with the results of microarray analysis. CeRNA network analysis was performed for hsa_circRNA_002809, hsa_circRNA_042882, and hsa_circRNA_104034. Functional enrichment analysis showed that the target genes were mainly associated with hypoxia-induced damage, inflammatory response, and the HIF-1 signaling pathway. Hsa_circRNA_042882 and hsa_circRNA_104034 can be regarded as promising diagnostic biomarkers for patients with ARDS caused by severe pneumonia, with remarkable sensitivity and specificity of the area under the curve of 0.8050 and 1 or 0.835 and 0.799, respectively. Conclusion This study obtained circRNA expression profiles of ARDS patients, and hsa_circRNA_042882 and hsa_circRNA_104034 were regarded as promising diagnostic biomarkers for patients with ARDS caused by severe pneumonia.
Collapse
Affiliation(s)
- He Sun
- *Correspondence: Qiang Li, ; Feilong Wang, ; He Sun,
| | | | | | | | | | - Feilong Wang
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Qiang Li
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
7
|
Suresh MV, Aggarwal V, Raghavendran K. The Intersection of Pulmonary Vascular Disease and Hypoxia-Inducible Factors. Interv Cardiol Clin 2023; 12:443-452. [PMID: 37290846 DOI: 10.1016/j.iccl.2023.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Hypoxia-inducible factors (HIFs) are a family of nuclear transcription factors that serve as the master regulator of the adaptive response to hypoxia. In the lung, HIFs orchestrate multiple inflammatory pathways and signaling. They have been reported to have a major role in the initiation and progression of acute lung injury, chronic obstructive pulmonary disease, pulmonary fibrosis, and pulmonary hypertension. Although there seems to be a clear mechanistic role for both HIF 1α and 2α in pulmonary vascular diseases including PH, a successful translation into a definitive therapeutic modality has not been accomplished to date.
Collapse
Affiliation(s)
| | - Vikas Aggarwal
- Division of Cardiology (Frankel Cardiovascular Center), Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Section of Cardiology, Department of Internal Medicine, Veterans Affairs Medical Center, Ann Arbor, MI, USA
| | - Krishnan Raghavendran
- Division of Acute Care Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Fesenmeier DJ, Suresh MV, Kim S, Raghavendran K, Won YY. Polymer Lung Surfactants Attenuate Direct Lung Injury in Mice. ACS Biomater Sci Eng 2023; 9:2716-2730. [PMID: 37079432 PMCID: PMC11973738 DOI: 10.1021/acsbiomaterials.3c00061] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
If not properly managed, acute lung injuries, either through direct or indirect causes, have the potential to present serious risk for many patients worldwide. One of the mechanisms for the transition from acute lung injury (ALI) to the more serious acute respiratory distress syndrome (ARDS) is the deactivation of the native lung surfactant by injury-induced infiltrates to the alveolar space. Currently, there are no surfactant replacement therapies that are used to treat ALI and subsequent ARDS. In this paper, we present an indepth efficacy study of using a novel polymer lung surfactant (PLS, composed of poly(styrene-block-ethylene glycol) (PS-PEG) block copolymer micelles), which has unique properties compared to other tested surfactant replacements, in two different mouse models of lung injury. The results demonstrate that pharyngeal administration of PLS after the instillation of either acid (HCl) or lipopolysaccharide (LPS) can decrease the severity of lung injury as measured by multiple injury markers.
Collapse
Affiliation(s)
- Daniel J. Fesenmeier
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN 47907, United States
| | | | - Seyoung Kim
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN 47907, United States
| | - Krishnan Raghavendran
- Department of Surgery, University of Michigan Medical
School, Ann Arbor, MI 48109, USA
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN 47907, United States
- Purdue University Center for Cancer Research, Purdue
University, West Lafayette, IN 47907, United States
| |
Collapse
|
9
|
Suresh MV, Balijepalli S, Solanki S, Aktay S, Choudhary K, Shah YM, Raghavendran K. Hypoxia-Inducible Factor 1α and Its Role in Lung Injury: Adaptive or Maladaptive. Inflammation 2023; 46:491-508. [PMID: 36596930 PMCID: PMC9811056 DOI: 10.1007/s10753-022-01769-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/24/2022] [Accepted: 11/18/2022] [Indexed: 01/05/2023]
Abstract
Hypoxia-inducible factors (HIFs) are transcription factors critical for the adaptive response to hypoxia. There is also an essential link between hypoxia and inflammation, and HIFs have been implicated in the dysregulated immune response to various insults. Despite the prevalence of hypoxia in tissue trauma, especially involving the lungs, there remains a dearth of studies investigating the role of HIFs in clinically relevant injury models. Here, we summarize the effects of HIF-1α on the vasculature, metabolism, inflammation, and apoptosis in the lungs and review the role of HIFs in direct lung injuries, including lung contusion, acid aspiration, pneumonia, and COVID-19. We present data that implicates HIF-1α in the context of arguments both in favor and against its role as adaptive or injurious in the propagation of the acute inflammatory response in lung injuries. Finally, we discuss the potential for pharmacological modulation of HIFs as a new class of therapeutics in the modern intensive care unit.
Collapse
Affiliation(s)
| | | | - Sumeet Solanki
- Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Sinan Aktay
- Department of Surgery, University of Michigan, Ann Arbor, USA
| | | | - Yatrik M Shah
- Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | | |
Collapse
|
10
|
Chen L, Yang J, Zhang M, Fu D, Luo H, Yang X. SPP1 exacerbates ARDS via elevating Th17/Treg and M1/M2 ratios through suppression of ubiquitination-dependent HIF-1α degradation. Cytokine 2023; 164:156107. [PMID: 36773529 DOI: 10.1016/j.cyto.2022.156107] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/24/2022] [Accepted: 12/05/2022] [Indexed: 02/11/2023]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a severe inflammatory pulmonary condition that leads to respiratory failure. The imbalance of Th17/Treg and M1/M2 is implicated in ARDS. A better understanding of the regulation of the balance of Th17/Treg and M1/M2 may provide novel therapeutic targets for ARDS. METHODS Plasma and BALF samples were collected from ARDS patients. Inflammatory cytokines were examined by ELISA. Th17, Treg, M1 and M2 were identified via immunofluorescence staining of RORγt, Foxp3, iNOS and Arg-1. H&E and Masson's trichrome staining were applied for evaluating pulmonary damage and fibrosis. A mouse model of ARDS was established through LPS administration. HIF-1α was immunoprecipitated and subjected to ubiquitination analysis via western blotting. The expression of SPP1, VHL and HIF-1α was examined by RT-qPCR and western blotting. RESULTS ARDS patients showed elevated levels of inflammatory cytokines and ratios of Th17/Treg and M1/M2. SPP1 was upregulated in ARDS mice, and silencing of SPP1 alleviated lung injury and fibrosis. SPP1 inhibited VHL expression to reduce the ubiquitination and degradation of HIF-1α in ARDS. Overexpression of SPP1 facilitated Th17, Treg and M1 polarization but inhibited M2 polarization through upregulation of HIF-1α. CONCLUSION SPP1 elevates Th17/Treg and M1/M2 ratio by suppressing VHL expression and ubiquitination-dependent HIF-1α degradation, thus exacerbating ARDS. Our study provides novel mechanistic insights into ARDS pathogenesis and promising therapeutic targets.
Collapse
Affiliation(s)
- Liang Chen
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China.
| | - Jin Yang
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China
| | - Meng Zhang
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China
| | - Donglin Fu
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China
| | - Huan Luo
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China
| | - Xiaolei Yang
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China
| |
Collapse
|
11
|
Yegen CH, Haine L, Da Costa Ferreira K, Marchant D, Bernaudin JF, Planès C, Voituron N, Boncoeur E. A New Model of Acute Exacerbation of Experimental Pulmonary Fibrosis in Mice. Cells 2022; 11:3379. [PMID: 36359778 PMCID: PMC9654438 DOI: 10.3390/cells11213379] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/07/2022] [Accepted: 10/23/2022] [Indexed: 11/27/2023] Open
Abstract
RATIONALE idiopathic pulmonary fibrosis (IPF) is the most severe form of fibrosing interstitial lung disease, characterized by progressive respiratory failure leading to death. IPF's natural history is heterogeneous, and its progression unpredictable. Most patients develop a progressive decline of respiratory function over years; some remain stable, but others present a fast-respiratory deterioration without identifiable cause, classified as acute exacerbation (AE). OBJECTIVES to develop and characterize an experimental mice model of lung fibrosis AE, mimicking IPF-AE at the functional, histopathological, cellular and molecular levels. METHODS we established in C57BL/6 male mice a chronic pulmonary fibrosis using a repetitive low-dose bleomycin (BLM) intratracheal (IT) instillation regimen (four instillations of BLM every 2 weeks), followed by two IT instillations of a simple or double-dose BLM challenge to induce AE. Clinical follow-up and histological and molecular analyses were done for fibrotic and inflammatory lung remodeling analysis. MEASUREMENTS AND MAIN RESULTS as compared with a low-dose BLM regimen, this AE model induced a late burst of animal mortality, worsened lung fibrosis and remodeling, and superadded histopathological features as observed in humans IPF-AE. This was associated with stronger inflammation, increased macrophage infiltration of lung tissue and increased levels of pro-inflammatory cytokines in lung homogenates. Finally, it induced in the remodeled lung a diffuse expression of hypoxia-inducible factor 1α, a hallmark of tissular hypoxia response and a major player in the progression of IPF. CONCLUSION this new model is a promising model of AE in chronic pulmonary fibrosis that could be relevant to mimic IPF-AE in preclinical trials.
Collapse
Affiliation(s)
- Céline-Hivda Yegen
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| | - Liasmine Haine
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| | - Kevin Da Costa Ferreira
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| | - Dominique Marchant
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| | - Jean-Francois Bernaudin
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
- Faculté de Médecine, Sorbonne Université, 75006 Paris, France
- Service de Physiologie et d’Explorations Fonctionnelles, Hôpital Avicenne, APHP, 93000 Bobigny, France
| | - Carole Planès
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
- Service de Physiologie et d’Explorations Fonctionnelles, Hôpital Avicenne, APHP, 93000 Bobigny, France
| | - Nicolas Voituron
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
- Département STAPS, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| | - Emilie Boncoeur
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| |
Collapse
|
12
|
HIF-1α inhibition attenuates severity of Achilles tendinopathy by blocking NF-κB and MAPK pathways. Int Immunopharmacol 2022; 106:108543. [DOI: 10.1016/j.intimp.2022.108543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 01/15/2023]
|
13
|
Suresh A, Rao TC, Solanki S, Suresh MV, Menon B, Raghavendran K. The holy basil administration diminishes the NF-kB expression and protects alveolar epithelial cells from pneumonia infection through interferon gamma. Phytother Res 2022; 36:1822-1835. [PMID: 35233841 PMCID: PMC9018535 DOI: 10.1002/ptr.7428] [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] [Received: 03/11/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 11/07/2022]
Abstract
Bacterial pneumonia is one of the most important causes of mortality in the United States. The bacteria Klebsiella pneumoniae (KP) accounts for a significant proportion of community and hospital-acquired infections. Here, we determine that the holy basil (Ocimum sanctum) extract improves cell viability and dampens the proinflammatory cytokine response in an in vitro model of pneumonia. For this, A549, a human alveolar basal epithelial cell line, was subjected to a lethal KP model following a 24-hr pretreatment with basil extract. Bacteremia, cell viability, apoptosis, MTT assay, phagocytic capacity, cytokines, and Khe gene expression were assessed in these cells following pneumonia. Cell morphology analysis showed that holy basil protected A549 cells from KP infection-mediated effects by inhibiting cell death due to apoptosis. Additionally, in the presence of basil, A549 cells demonstrated significantly higher bactericidal capacity and phagocytosis. Administration of holy basil led to reduced expression of hypoxia-inducible factor-1/2a, nuclear factor kappa B, and Khe in the KP-infected cells while increasing interferon (IFN)-γ expression. Our results suggest that basil significantly reduced cell death in the setting of KP infection, likely via attenuation of cytokine and IFN-γ mediated signaling pathways. Holy basil is a promising therapeutic agent for managing and treating bacterial pneumonia based on its potency.
Collapse
Affiliation(s)
| | - Tejeshwar C. Rao
- Department of Cell, Developmental, and Integrative Biology, the University of Alabama at Birmingham, Birmingham AL 35294
| | - Sumeet Solanki
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | | | - Bindu Menon
- Department of Medical Education & Physiology/Pharmacology, University of Toledo, OH
| | | |
Collapse
|
14
|
Repurposing Multiple-Molecule Drugs for COVID-19-Associated Acute Respiratory Distress Syndrome and Non-Viral Acute Respiratory Distress Syndrome via a Systems Biology Approach and a DNN-DTI Model Based on Five Drug Design Specifications. Int J Mol Sci 2022; 23:ijms23073649. [PMID: 35409008 PMCID: PMC8998971 DOI: 10.3390/ijms23073649] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) epidemic is currently raging around the world at a rapid speed. Among COVID-19 patients, SARS-CoV-2-associated acute respiratory distress syndrome (ARDS) is the main contribution to the high ratio of morbidity and mortality. However, clinical manifestations between SARS-CoV-2-associated ARDS and non-SARS-CoV-2-associated ARDS are quite common, and their therapeutic treatments are limited because the intricated pathophysiology having been not fully understood. In this study, to investigate the pathogenic mechanism of SARS-CoV-2-associated ARDS and non-SARS-CoV-2-associated ARDS, first, we constructed a candidate host-pathogen interspecies genome-wide genetic and epigenetic network (HPI-GWGEN) via database mining. With the help of host-pathogen RNA sequencing (RNA-Seq) data, real HPI-GWGEN of COVID-19-associated ARDS and non-viral ARDS were obtained by system modeling, system identification, and Akaike information criterion (AIC) model order selection method to delete the false positives in candidate HPI-GWGEN. For the convenience of mitigation, the principal network projection (PNP) approach is utilized to extract core HPI-GWGEN, and then the corresponding core signaling pathways of COVID-19-associated ARDS and non-viral ARDS are annotated via their core HPI-GWGEN by KEGG pathways. In order to design multiple-molecule drugs of COVID-19-associated ARDS and non-viral ARDS, we identified essential biomarkers as drug targets of pathogenesis by comparing the core signal pathways between COVID-19-associated ARDS and non-viral ARDS. The deep neural network of the drug–target interaction (DNN-DTI) model could be trained by drug–target interaction databases in advance to predict candidate drugs for the identified biomarkers. We further narrowed down these predicted drug candidates to repurpose potential multiple-molecule drugs by the filters of drug design specifications, including regulation ability, sensitivity, excretion, toxicity, and drug-likeness. Taken together, we not only enlighten the etiologic mechanisms under COVID-19-associated ARDS and non-viral ARDS but also provide novel therapeutic options for COVID-19-associated ARDS and non-viral ARDS.
Collapse
|
15
|
Yin L, Zhan J, Liao H, Qiu W, Hou W, Li S, Zhang J. Novel vandetanib derivative inhibited proliferation and promoted apoptosis of cancer cells under normoxia and hypoxia. Eur J Pharmacol 2022; 922:174907. [DOI: 10.1016/j.ejphar.2022.174907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/17/2022] [Accepted: 03/17/2022] [Indexed: 11/29/2022]
|
16
|
Suresh MV, Yalamanchili G, Rao TC, Aktay S, Kralovich A, Shah YM, Raghavendran K. Hypoxia‐inducible factor (HIF)‐1α‐induced regulation of lung injury in pulmonary aspiration is mediated through NF‐kB. FASEB Bioadv 2022; 4:309-328. [PMID: 35520392 PMCID: PMC9065579 DOI: 10.1096/fba.2021-00132] [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: 11/01/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/11/2022] Open
Abstract
Aspiration‐induced lung injury is a common grievance encountered in the intensive care unit (ICU). It is a significant risk factor for improving ventilator‐associated pneumonia (VAP) and acute respiratory distress syndrome (ARDS). Hypoxia‐inducible factor (HIF)‐1α is one of the primary transcription factors responsible for regulating the cellular response to changes in oxygen tension. Here, we sought to determine the role of HIF‐1α and specifically the role of type 2 alveolar epithelial cells in generating the acute inflammatory response following acid and particles (CASP) aspiration. Previous studies show HIF‐1 α is involved in regulating the hypoxia‐stimulated expression of MCP‐1 in mice and humans. The CASP was induced in C57BL/6, ODD‐Luc, HIF‐1α (+/+) control, and HIF‐1α conditional knockout (HIF‐1α (−/−) mice). Following an injury in ODD mice, explanted organs were subjected to IVIS imaging to measure the degree of hypoxia. HIF‐1α expression, BAL albumin, cytokines, and histology were measured following CASP. In C57BL/6 mice, the level of HIF‐1α was increased at 1 h after CASP. There were significantly increased levels of albumin and cytokines in C57BL/6 and ODD‐Luc mice lungs following CASP. HIF‐1α (+/+) mice given CASP demonstrated a synergistic increase in albumin leakage, increased pro‐inflammatory cytokines, and worse injury. MCP‐1 antibody neutralized HIF‐1α (+/+) mice showed reduced granuloma formation. The NF‐κB expression was increased substantially in the HIF‐1α (+/+) mice following CASP compared to HIF‐1α (−/−) mice. Our data collectively identify that HIF‐1α upregulation of the acute inflammatory response depends on NF‐κB following CASP.
Collapse
Affiliation(s)
| | | | - Tejeshwar C. Rao
- Department of Cell, Developmental, and Integrative Biology The University of Alabama at Birmingham Birmingham UK
| | - Sinan Aktay
- Department of Surgery University of Michigan Ann Arbor Michigan USA
| | - Alex Kralovich
- Department of Surgery University of Michigan Ann Arbor Michigan USA
| | - Yatrik M. Shah
- Molecular & Integrative Physiology University of Michigan Ann Arbor Michigan USA
| | | |
Collapse
|
17
|
Afshari R, Akhavan O, Hamblin MR, Varma RS. Review of Oxygenation with Nanobubbles: Possible Treatment for Hypoxic COVID-19 Patients. ACS APPLIED NANO MATERIALS 2021; 4:11386-11412. [PMID: 37556289 PMCID: PMC8565459 DOI: 10.1021/acsanm.1c01907] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/12/2021] [Indexed: 05/05/2023]
Abstract
The coronavirus disease (COVID-19) pandemic, which has spread around the world, caused the death of many affected patients, partly because of the lack of oxygen arising from impaired respiration or blood circulation. Thus, maintaining an appropriate level of oxygen in the patients' blood by devising alternatives to ventilator systems is a top priority goal for clinicians. The present review highlights the ever-increasing application of nanobubbles (NBs), miniature gaseous vesicles, for the oxygenation of hypoxic patients. Oxygen-containing NBs can exert a range of beneficial physiologic and pharmacologic effects that include tissue oxygenation, as well as tissue repair mechanisms, antiinflammatory properties, and antibacterial activity. In this review, we provide a comprehensive survey of the application of oxygen-containing NBs, with a primary focus on the development of intravenous platforms. The multimodal functions of oxygen-carrying NBs, including antimicrobial, antiinflammatory, drug carrying, and the promotion of wound healing are discussed, including the benefits and challenges of using NBs as a treatment for patients with acute hypoxemic respiratory failure, particularly due to COVID-19.
Collapse
Affiliation(s)
- Ronak Afshari
- Department of Physics, Sharif University
of Technology, P.O. Box 11155-9161, Tehran 14588-89694,
Iran
| | - Omid Akhavan
- Department of Physics, Sharif University
of Technology, P.O. Box 11155-9161, Tehran 14588-89694,
Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science,
University of Johannesburg, Doornfontein 2028, South
Africa
| | - Rajender S. Varma
- Regional Center of Advanced Technologies and Materials,
Czech Advanced Technology and Research Institute, Palacky
University, Šlechtitelů 27, Olomouc 78371, Czech
Republic
| |
Collapse
|
18
|
Lu X, Ma W, Fan B, Li P, Gao J, Liu Q, Hu C, Li Y, Yao M, Ning H, Xing L. Integrating Network Pharmacology, Transcriptome and Artificial Intelligence for Investigating Into the Effect and Mechanism of Ning Fei Ping Xue Decoction Against the Acute Respiratory Distress Syndrome. Front Pharmacol 2021; 12:731377. [PMID: 34803679 PMCID: PMC8595141 DOI: 10.3389/fphar.2021.731377] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 09/20/2021] [Indexed: 01/19/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a high-mortality disease and lacks effective pharmacotherapy. A traditional Chinese medicine (TCM) formula, Ning Fei Ping Xue (NFPX) decoction, was demonstrated to play a critical role in alleviating inflammatory responses of the lung. However, its therapeutic effectiveness in ARDS and active compounds, targets, and molecular mechanisms remain to be elucidated. The present study investigates the effects of NFPX decoction on ARDS mice induced by lipopolysaccharides (LPS). The results revealed that NFPX alleviated lung edema evaluated by lung ultrasound, decreased lung wet/Dry ratio, the total cell numbers of bronchoalveolar lavage fluid (BALF), and IL-1β, IL-6, and TNF-α levels in BALF and serum, and ameliorated lung pathology in a dose-dependent manner. Subsequently, UPLC-HRMS was performed to establish the compounds of NFPX. A total of 150 compounds in NFPX were characterized. Moreover, integrating network pharmacology approach and transcriptional profiling of lung tissues were performed to predict the underlying mechanism. 37 active components and 77 targets were screened out, and a herbs-compounds-targets network was constructed. Differentially expressed genes (DEGs) were identified from LPS-treated mice compared with LPS combined with NFPX mice. GO, KEGG, and artificial intelligence analysis indicated that NFPX might act on various drug targets. At last, potential targets, HRAS, SMAD4, and AMPK, were validated by qRT-PCR in ARDS murine model. In conclusion, we prove the efficacy of NFPX decoction in the treatment of ARDS. Furthermore, integrating network pharmacology, transcriptome, and artificial intelligence analysis contributes to illustrating the molecular mechanism of NFPX decoction on ARDS.
Collapse
Affiliation(s)
- Xiaoxiao Lu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wentao Ma
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Baofeng Fan
- Air Force General Hospital PLA, Beijing, China
| | - Peng Li
- Department of Basic Sciences, Shanxi Agricultural University, Taigu, China
| | - Jing Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiuhong Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunling Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengying Yao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hanbing Ning
- Department of Digestive Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lihua Xing
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Liu H, Hu T, Zhang C, Chen X, Zhang S, Li M, Jing H, Wang C, Hu T, Shi J. Mechanisms of COVID-19 thrombosis in an inflammatory environment and new anticoagulant targets. Am J Transl Res 2021; 13:3925-3941. [PMID: 34149990 PMCID: PMC8205677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/25/2021] [Indexed: 06/12/2023]
Abstract
COVID-19 is widely epidemic in the world and poses a great threat to our life. Coagulopathy is one of the major characteristics in the COVID-19 patients. A growing number of studies have found that the severe COVID-19 patients have thrombotic microangiopathy and thromboembolism. Coagulopathy associated with increased risk of death in the patients. Unfortunately, the mechanism of coagulopathy is not clearly addressed. Understanding the pathophysiological mechanism of COVID-19 thrombosis and improving the coagulopathy through efficient treatment may help to stop disease progression, reduce mortality and sequelae. In severe COVID-19 patients, inflammation, cytokine storm, and coagulation are closely related, which together cause blood congestion and thrombosis. Many cytokines activate blood cells, expressing activating factors or releasing activated microparticles, and then accelerating thrombosis. However, the role of blood cells is not well understood in COVID-19 patients. In addition, cytokines stimulate endothelial cells, transforming them into a procoagulant phenotype. Therefore, determine their role and propose new strategies for the prevention and treatment of thrombosis in severe COVID-19 patients. We outline the major events of coagulopathies, discuss the role of blood and endothelial cells in thrombosis, to formulate a new anticoagulation protocol.
Collapse
Affiliation(s)
- Huan Liu
- Department of Stomatology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
| | - Tianshui Hu
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
| | - Cong Zhang
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
| | - Xiaojing Chen
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
| | - Shuoqi Zhang
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
| | - Mengdi Li
- Department of Stomatology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
| | - Haijiao Jing
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
| | - Chunxu Wang
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
| | - Tenglong Hu
- Department of Stomatology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
| | - Jialan Shi
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityHarbin, China
- Department of Research and Medicine, VA Boston Healthcare System, Brigham and Women’s Hospital, Harvard Medical SchoolBoston, MA, USA
| |
Collapse
|
20
|
Al-Nimer MSM. Is COVID-19-induced liver injury different from other RNA viruses? World J Meta-Anal 2021; 9:108-127. [DOI: 10.13105/wjma.v9.i2.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/12/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 is a pandemic disease caused by a novel RNA coronavirus, SARS coronavirus 2 (SARS-CoV-2), which is implicated in the respiratory system. SARS-CoV-2 also targets extrapulmonary systems, including the gastrointestinal tract, liver, central nervous system and others. SARS-CoV-2, like other RNA viruses, targets the liver and produces liver injury. This literature review showed that SARS-CoV-2-induced liver injury is different from other RNA viruses by a transient elevation of hepatic enzymes and does not progress to liver fibrosis or other unfavorable events. Moreover, SARS-CoV-2-induced liver injury usually occurs in the presence of risk factors, such as nonalcoholic liver fatty disease. This review highlights the important differences between RNA viruses inducing liver injury taking into consideration the clinical, biochemical, histopathological, postmortem findings and the chronicity of liver injury that ultimately leads to liver fibrosis and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Marwan SM Al-Nimer
- Department of Clinical Pharmacy, Hawler Medical University, Erbil 44001, Iraq
- College of Medicine, University of Diyala, Baqubah 32001, Iraq
| |
Collapse
|
21
|
Pourfathi M, Xin Y, Rosalino M, Cereda M, Kadlecek S, Duncan I, Profka H, Hamedani H, Siddiqui S, Ruppert K, Chatterjee S, Rizi RR. Pulmonary pyruvate metabolism as an index of inflammation and injury in a rat model of acute respiratory distress syndrome. NMR IN BIOMEDICINE 2020; 33:e4380. [PMID: 32681670 DOI: 10.1002/nbm.4380] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/15/2020] [Accepted: 06/29/2020] [Indexed: 06/11/2023]
Abstract
Increased pulmonary lactate production is correlated with severity of lung injury and outcome in acute respiratory distress syndrome (ARDS) patients. This study was conducted to investigate the relative contributions of inflammation and hypoxia to the lung's metabolic shift to glycolysis in an experimental animal model of ARDS using hyperpolarized (HP) 13 C MRI. Fifty-three intubated and mechanically ventilated male rats were imaged using HP 13 C MRI before, and 1, 2.5 and 4 hours after saline (sham) or hydrochloric acid (HCl; 0.5 ml/kg) instillation in the trachea, followed by protective and nonprotective mechanical ventilation (HCl-PEEP and HCl-ZEEP) or the start of moderate or severe hypoxia (Hyp90 and Hyp75 groups). Pulmonary and cardiac HP lactate-to-pyruvate ratios were compared among groups for different time points. Postmortem histology and immunofluorescence were used to assess lung injury severity and quantify the expression of innate inflammatory markers and local tissue hypoxia. HP pulmonary lactate-to-pyruvate ratio progressively increased in rats with lung injury and moderate hypoxia (HCl-ZEEP), with no significant change in pulmonary lactate-to-pyruvate ratio in noninjured but moderately hypoxic rats (Hyp90). Pulmonary lactate-to-pyruvate ratio was elevated in otherwise healthy lung tissue only in severe systemic hypoxia (Hyp75 group). ex vivo histological and immunopathological assessment further confirmed the link between elevated glycolysis and the recruitment into and presence of activated neutrophils in injured lungs. HP lactate-to-pyruvate ratio is elevated in injured lungs predominantly as a result of increased glycolysis in activated inflammatory cells, but can also increase due to severe inflammation-induced hypoxia.
Collapse
Affiliation(s)
- Mehrdad Pourfathi
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yi Xin
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael Rosalino
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maurizio Cereda
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephen Kadlecek
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ian Duncan
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harrilla Profka
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hooman Hamedani
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sarmad Siddiqui
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kai Ruppert
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shampa Chatterjee
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rahim R Rizi
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
22
|
Abstract
The syndrome of critical illness is a complex physiological stressor that can be triggered by diverse pathologies. It is widely believed that organ dysfunction and death result from bioenergetic failure caused by inadequate cellular oxygen supply. Teleologically, life has evolved to survive in the face of stressors by undergoing a suite of adaptive changes. Adaptation not only comprises alterations in systemic physiology but also involves molecular reprogramming within cells. The concept of cellular adaptation in critically ill patients is a matter of contention in part because medical interventions mask underlying physiology, creating the artificial construct of "chronic critical illness," without which death would be imminent. Thus far, the intensive care armamentarium has not targeted cellular metabolism to preserve a temporary equilibrium but instead attempts to normalize global oxygen and substrate delivery. Here, we review adaptations to hypoxia that have been demonstrated in cellular models and in human conditions associated with hypoxia, including the hypobaric hypoxia of high altitude, the intrauterine low-oxygen environment, and adult myocardial hibernation. Common features include upregulation of glycolytic ATP production, enhancement of respiratory efficiency, downregulation of mitochondrial density, and suppression of energy-consuming processes. We argue that these innate cellular adaptations to hypoxia represent potential avenues for intervention that have thus far remained untapped by intensive care medicine.
Collapse
Affiliation(s)
- Helen T McKenna
- Division of Surgery and Interventional Science, University College London, London, United Kingdom.,Royal Free Intensive Care Unit, Royal Free Hospital, London, United Kingdom
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Daniel S Martin
- Royal Free Intensive Care Unit, Royal Free Hospital, London, United Kingdom.,Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom
| |
Collapse
|
23
|
Liu X, Ma Y, Ouyang R, Zeng Z, Zhan Z, Lu H, Cui Y, Dai Z, Luo L, He C, Li H, Zong D, Chen Y. The relationship between inflammation and neurocognitive dysfunction in obstructive sleep apnea syndrome. J Neuroinflammation 2020; 17:229. [PMID: 32738920 PMCID: PMC7395983 DOI: 10.1186/s12974-020-01905-2] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022] Open
Abstract
Obstructive sleep apnea syndrome (OSAS), a state of sleep disorder, is characterized by repetitive apnea, chronic hypoxia, oxygen desaturation, and hypercapnia. Previous studies have revealed that intermittent hypoxia (IH) conditions in OSAS patients elicited neuron injury (especially in the hippocampus and cortex), leading to cognitive dysfunction, a significant and extraordinary complication of OSAS patients. The repeated courses of airway collapse and obstruction in OSAS patients resulted in apnea and arousal during sleep, leading to IH and excessive daytime sleepiness (EDS) and subsequently contributing to the development of inflammation. IH-mediated inflammation could further trigger various types of cognitive dysfunction. Many researchers have found that, besides continuous positive airway pressure (CPAP) treatment and surgery, anti-inflammatory substances might alleviate IH-induced neurocognitive dysfunction. Clarifying the role of inflammation in IH-mediated cognitive impairment is crucial for potentially valuable therapies and future research in the related domain. The objective of this article was to critically review the relationship between inflammation and cognitive deficits in OSAS.
Collapse
Affiliation(s)
- Xiangming Liu
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Yiming Ma
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Ruoyun Ouyang
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Zihang Zeng
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Zijie Zhan
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Huanhuan Lu
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Yanan Cui
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Zhongshang Dai
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Lijuan Luo
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Chenjie He
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Herui Li
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Dandan Zong
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China. .,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China.
| | - Yan Chen
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China. .,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
24
|
Marchetti M. COVID-19-driven endothelial damage: complement, HIF-1, and ABL2 are potential pathways of damage and targets for cure. Ann Hematol 2020; 99:1701-1707. [PMID: 32583086 PMCID: PMC7312112 DOI: 10.1007/s00277-020-04138-8] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 06/09/2020] [Indexed: 02/07/2023]
Abstract
COVID-19 pandemia is a major health emergency causing hundreds of deaths worldwide. The high reported morbidity has been related to hypoxia and inflammation leading to endothelial dysfunction and aberrant coagulation in small and large vessels. This review addresses some of the pathways leading to endothelial derangement, such as complement, HIF-1α, and ABL tyrosine kinases. This review also highlights potential targets for prevention and therapy of COVID-19-related organ damage and discusses the role of marketed drugs, such as eculizumab and imatinib, as suitable candidates for clinical trials.
Collapse
Affiliation(s)
- Monia Marchetti
- Hematology Department, Az Osp SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy.
| |
Collapse
|
25
|
Low-Molecular-Weight Heparin Reduces Ventilation-Induced Lung Injury through Hypoxia Inducible Factor-1α in a Murine Endotoxemia Model. Int J Mol Sci 2020; 21:ijms21093097. [PMID: 32353952 PMCID: PMC7247708 DOI: 10.3390/ijms21093097] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/02/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Patients with sepsis frequently require mechanical ventilation (MV) to survive. However, MV has been shown to induce the production of proinflammatory cytokines, causing ventilator-induced lung injury (VILI). It has been demonstrated that hypoxia-inducible factor (HIF)-1α plays a crucial role in inducing both apoptotic and inflammatory processes. Low-molecular-weight heparin (LMWH) has been shown to have anti-inflammatory activities. However, the effects of HIF-1α and LMWH on sepsis-related acute lung injury (ALI) have not been fully delineated. We hypothesized that LMWH would reduce lung injury, production of free radicals and epithelial apoptosis through the HIF-1α pathway. Male C57BL/6 mice were exposed to 6-mL/kg or 30-mL/kg MV for 5 h. Enoxaparin, 4 mg/kg, was administered subcutaneously 30 min before MV. We observed that MV with endotoxemia induced microvascular permeability; interleukin-6, tumor necrosis factor-α, macrophage inflammatory protein-2 and vascular endothelial growth factor protein production; neutrophil infiltration; oxidative loads; HIF-1α mRNA activation; HIF-1α expression; bronchial epithelial apoptosis; and decreased respiratory function in mice (p < 0.05). Endotoxin-induced augmentation of VILI and epithelial apoptosis were reduced in the HIF-1α-deficient mice and in the wild-type mice following enoxaparin administration (p < 0.05). Our data suggest that enoxaparin reduces endotoxin-augmented MV-induced ALI, partially by inhibiting the HIF-1α pathway.
Collapse
|
26
|
Khan MS, Kim JS, Hwang J, Choi Y, Lee K, Kwon Y, Jang J, Yoon S, Yang CS, Choi J. Effective delivery of mycophenolic acid by oxygen nanobubbles for modulating immunosuppression. Theranostics 2020; 10:3892-3904. [PMID: 32226527 PMCID: PMC7086369 DOI: 10.7150/thno.41850] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/19/2020] [Indexed: 12/11/2022] Open
Abstract
Immunosuppressive drugs are crucial for preventing acute graft rejection or autoimmune diseases. They are generally small molecules that require suitable drug carriers for ensuring stability, bioavailability, and longer half-life. Mycophenolic acid (MPA) is an extensively studied immunosuppressive drug. However, it requires suitable carriers for overcoming clinical limitations. Currently, lipid-shelled micro- and nanobubbles are being thoroughly investigated for diagnostic and therapeutic applications, as they possess essential properties, such as injectability, smaller size, gaseous core, high surface area, higher drug payload, and enhanced cellular penetration. Phospholipids are biocompatible and biodegradable molecules, and can be functionalized according to specific requirements. Methods: In this study, we synthesized oxygen nanobubbles (ONBs) and loaded the hydrophobic MPA within the ONBs to generate ONB/MPA. Peripheral blood mononuclear cells (PBMCs) were treated with ONB/MPA to determine the suppression of immune response by measuring cytokine release. In vivo murine experiments were performed to evaluate the effectiveness of ONB/MPA in the presence of inflammatory stimulants. Results: Our results suggest that ONBs successfully delivered MPA and reduced the release of cytokines, thereby controlling inflammation and significantly increasing the survival rate of animals. Conclusion: This method can be potentially used for implantation and for treating autoimmune diseases, wherein immunosuppression is desired.
Collapse
|
27
|
Inflammatory response in trauma patients: are there ways to decrease the inflammatory reaction? Curr Opin Anaesthesiol 2020; 33:253-258. [PMID: 32049884 DOI: 10.1097/aco.0000000000000842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Trauma patients are considered a complex population of patients in emergency medicine and need extensive, specialized therapy. One major part is the prevention and treatment of the inflammatory response, which occurs in patients after severe injury resulting in complications like endotheliopathy. Likely as a consequence, coagulopathy occurs. Sterile inflammation is hard to address, especially because of the lack of a single activator. Moreover, it is a complex composition of factors that lead to a pathologic immune response. Our understanding of these patterns is increasing, but the complete pathophysiologic changes have yet to be investigated. Therefore, there is no specific target to treat inflammatory response in trauma patients at the moment. RECENT FINDINGS There is increasing knowledge of the pathways and mediators that are responsible for the inflammatory response in patients after severe trauma. The endothelial glycocalyx has been identified to be an integral part of these mechanisms. There have been several new therapeutic approaches to diminish the inflammatory response. SUMMARY Our increasing understanding of the immune system have led to new potential therapeutic perspectives. All of these approaches need further research to be validated. As the current therapies are based on empirical strategies and have not changed much over the years, new treatment options would be an important progress.
Collapse
|
28
|
Activation of Hypoxia-Inducible Factor-1α Via Succinate Dehydrogenase Pathway During Acute Lung Injury Induced by Trauma/Hemorrhagic Shock. Shock 2020; 53:208-216. [DOI: 10.1097/shk.0000000000001347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
29
|
Iron and Sphingolipids as Common Players of (Mal)Adaptation to Hypoxia in Pulmonary Diseases. Int J Mol Sci 2020; 21:ijms21010307. [PMID: 31906427 PMCID: PMC6981703 DOI: 10.3390/ijms21010307] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/24/2019] [Accepted: 12/31/2019] [Indexed: 12/11/2022] Open
Abstract
Hypoxia, or lack of oxygen, can occur in both physiological (high altitude) and pathological conditions (respiratory diseases). In this narrative review, we introduce high altitude pulmonary edema (HAPE), acute respiratory distress syndrome (ARDS), Chronic Obstructive Pulmonary Disease (COPD), and Cystic Fibrosis (CF) as examples of maladaptation to hypoxia, and highlight some of the potential mechanisms influencing the prognosis of the affected patients. Among the specific pathways modulated in response to hypoxia, iron metabolism has been widely explored in recent years. Recent evidence emphasizes hepcidin as highly involved in the compensatory response to hypoxia in healthy subjects. A less investigated field in the adaptation to hypoxia is the sphingolipid (SPL) metabolism, especially through Ceramide and sphingosine 1 phosphate. Both individually and in concert, iron and SPL are active players of the (mal)adaptation to physiological hypoxia, which can result in the pathological HAPE. Our aim is to identify some pathways and/or markers involved in the physiological adaptation to low atmospheric pressures (high altitudes) that could be involved in pathological adaptation to hypoxia as it occurs in pulmonary inflammatory diseases. Hepcidin, Cer, S1P, and their interplay in hypoxia are raising growing interest both as prognostic factors and therapeutical targets.
Collapse
|
30
|
What's New in Shock, December 2019? Shock 2019; 52:566-567. [PMID: 31725108 DOI: 10.1097/shk.0000000000001438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Zhang B, Swamy S, Balijepalli S, Panicker S, Mooliyil J, Sherman MA, Parkkinen J, Raghavendran K, Suresh MV. Direct pulmonary delivery of solubilized curcumin reduces severity of lethal pneumonia. FASEB J 2019; 33:13294-13309. [PMID: 31530014 DOI: 10.1096/fj.201901047rr] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute respiratory distress syndrome (ARDS), the most severe form of acute lung injury, is associated with reduced lung compliance and hypoxemia. Curcumin exhibits potent anti-inflammatory properties but has poor solubility and rapid plasma clearance. To overcome these physiochemical limitations and uncover the full therapeutic potential of curcumin in lung inflammation, in this study we utilized a novel water-soluble curcumin formulation (CDC) and delivered it directly into the lungs of C57BL/6 mice inoculated with a lethal dose of Klebsiella pneumoniae (KP). Administration of CDC led to a significant reduction in mortality, in bacterial presence within blood and lungs, as well as in lung injury, inflammation, and oxidative stress. The expression of Klebsiella hemolysin gene; TNF-α; IFN-β; nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3; hypoxia-inducible factor 1/2α; and NF-κB were also decreased following CDC treatment, suggesting modulation of the inflammasome complex and hypoxia signaling pathways as an underlying mechanism by which CDC reduces the severity of pneumonia. On a cellular level, CDC led to diminished cell death, improved viability, and protection of human lung epithelial cells in vitro. Overall, our studies demonstrate that CDC administration improves cell survival and reduces injury, inflammation, and mortality in a murine model of lethal gram-negative pneumonia. CDC, therefore, has promising anti-inflammatory potential in pneumonia and likely other inflammatory lung diseases, demonstrating the importance of optimizing the physicochemical properties of active natural products to optimize their clinical application.-Zhang, B., Swamy, S., Balijepalli, S., Panicker, S., Mooliyil, J., Sherman, M. A., Parkkinen, J., Raghavendran, K., Suresh, M. V. Direct pulmonary delivery of solubilized curcumin reduces severity of lethal pneumonia.
Collapse
Affiliation(s)
- Boya Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Samantha Swamy
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Sreehari Panicker
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Jashitha Mooliyil
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew A Sherman
- Department of Pediatrics, Children's National Medical Center, Washington, DC, USA
| | - Jaakko Parkkinen
- Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | | | | |
Collapse
|
32
|
Wu D, Birukov K. Endothelial Cell Mechano-Metabolomic Coupling to Disease States in the Lung Microvasculature. Front Bioeng Biotechnol 2019; 7:172. [PMID: 31380363 PMCID: PMC6658821 DOI: 10.3389/fbioe.2019.00172] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/03/2019] [Indexed: 12/15/2022] Open
Abstract
Lungs are the most vascular part of humans, accepting the totality of cardiac output in a volume much smaller than the body itself. Due to this cardiac output, the lung microvasculature is subject to mechanical forces including shear stress and cyclic stretch that vary with the cardiac and breathing cycle. Vessels are surrounded by extracellular matrix which dictates the stiffness which endothelial cells also sense and respond to. Shear stress, stiffness, and cyclic stretch are known to influence endothelial cell state. At high shear stress, endothelial cells exhibit cell quiescence marked by low inflammatory markers and high nitric oxide synthesis, whereas at low shear stress, endothelial cells are thought to "activate" into a pro-inflammatory state and have low nitric oxide. Shear stress' profound effect on vascular phenotype is most apparent in the arterial vasculature and in the pathophysiology of vascular inflammation. To conduct the flow of blood from the right heart, the lung microvasculature must be rigid yet compliant. It turns out that excessive substrate rigidity or stiffness is important in the development of pulmonary hypertension and chronic fibrosing lung diseases via excessive cell proliferation or the endothelial-mesenchymal transition. Recently, a new body of literature has evolved that couples mechanical sensing to endothelial phenotypic changes through metabolic signaling in clinically relevant contexts such as pulmonary hypertension, lung injury syndromes, as well as fibrosis, which is the focus of this review. Stretch, like flow, has profound effect on endothelial phenotype; metabolism studies due to stretch are in their infancy.
Collapse
Affiliation(s)
- David Wu
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Konstantin Birukov
- Department of Anesthesia, University of Maryland, Baltimore, MD, United States
| |
Collapse
|