1
|
Zhu Z, Zhang Y, Chen H, Zhang H. Cell-cell crosstalk in the pathogenesis of acute lung injury and acute respiratory distress syndrome. Tissue Barriers 2025:2452082. [PMID: 39798076 DOI: 10.1080/21688370.2025.2452082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/21/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are the result of an exaggerated inflammatory response triggered by a variety of pulmonary and systemic insults. The lung tissues are comprised of a variety of cell types, including alveolar epithelial cells, pulmonary vascular endothelial cells, macrophages, neutrophils, and others. There is mounting evidence that these diverse cell populations within the lung interact to regulate lung inflammation in response to both direct and indirect stimuli. The aim of this review is to provide a summary and discussion of recent advances in the understanding of the importance of cell-cell crosstalk in the pathogenesis of ALI/ARDS, with a specific focus on the cell-cell interactions that may offer prospective therapeutic avenues for ALI/ARDS.
Collapse
Affiliation(s)
- Zhenzhen Zhu
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Ying Zhang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Huan Chen
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Huali Zhang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| |
Collapse
|
2
|
Goswami R, Nagaraj H, Cicek YA, Nasim N, Mirza SS, Hassan MA, Mhaske R, Saravanan DM, Noonan C, Pham E, Mager J, Rotello VM. Polymer-siRNA nanovectors for treating lung inflammation. J Control Release 2025; 378:1092-1102. [PMID: 39730067 DOI: 10.1016/j.jconrel.2024.12.053] [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: 09/05/2024] [Revised: 12/07/2024] [Accepted: 12/21/2024] [Indexed: 12/29/2024]
Abstract
Uncontrolled inflammation is the driver of numerous lung diseases. Current treatments, including corticosteroids and bronchodilators, can be effective. However, they often come with notable side effects. siRNA is a promising therapeutic modality for immune regulation. However, effective delivery of siRNA is challenged by issues related to cellular uptake and localization within tissues. This study investigates a series of guanidinium-functionalized polymers (Cn-Guan) designed to explore the effects of amphiphilicity on siRNA complexation and efficiency in vitro and in vivo. Nine polymers with varying side chain lengths (C3, C5, C7) and molecular weights (17 kDa, 30 kDa, 65 kDa) were synthesized, forming polyplexes with siRNA. Characterization revealed that C7-Guan/si_scr polymers exhibited the smallest polyplex sizes and the tightest complexation with siRNA. In vitro studies showed that 65 kDa polymers had the highest gene knockdown efficiency, with C3 and C5-Guan/si_TNF-α achieving ∼70 % knockdown, while C7-Guan/si_TNF-α achieved ∼30 %. In vivo, C7-Guan/Cy5-siRNA demonstrated the highest lung accumulation, and all polymers showed ∼70 % TNF-α knockdown with a low siRNA dosage (0.14 mg/kg) in a murine lung inflammation model. C7-Guan polymers, despite lower in vitro efficiency, were quite effective in vivo, potentially due to enhanced serum stability. These findings demonstrate that Cn-Guan/siRNA polyplexes are effective and safe for attenuating pulmonary inflammation and provide important insights for the development of future siRNA delivery vectors for lung disease treatment.
Collapse
Affiliation(s)
- Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Yagiz Anil Cicek
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Nourina Nasim
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Sarah S Mirza
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, MA 01003, USA
| | - Muhammad Aamir Hassan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Rukmini Mhaske
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Deepthika M Saravanan
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Cedar Noonan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Edward Pham
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Jesse Mager
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, MA 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA.
| |
Collapse
|
3
|
Oliveira KB, de Souza FMA, de Sá LBM, Pacheco ALD, Prado MR, de Sousa Rodrigues CF, Bassi ÊJ, Santana-Melo I, Silva-Júnior A, Sabino-Silva R, Shetty AK, de Castro OW. Potential Mechanisms Underlying COVID-19-Mediated Central and Peripheral Demyelination: Roles of the RAAS and ADAM-17. Mol Neurobiol 2025; 62:1151-1164. [PMID: 38965171 DOI: 10.1007/s12035-024-04329-8] [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: 12/13/2023] [Accepted: 06/21/2024] [Indexed: 07/06/2024]
Abstract
Demyelination is among the most conspicuous neurological sequelae of SARS-CoV-2 infection (COVID-19) in both the central (CNS) and peripheral (PNS) nervous systems. Several hypotheses have been proposed to explain the mechanisms underlying demyelination in COVID-19. However, none have considered the SARS-CoV-2's effects on the renin-angiotensin-aldosterone system (RAAS). Therefore, our objective in this review is to evaluate how RAAS imbalance, caused by direct and indirect effects of SARS-CoV-2 infection, could contribute to myelin loss in the PNS and CNS. In the PNS, we propose that demyelination transpires from two significant changes induced by SARS-CoV-2 infection, which include upregulation of ADAM-17 and induction of lymphopenia. Whereas, in the CNS, demyelination could result from RAAS imbalance triggering two alterations: (1) a decrease in angiotensin type II receptor (AT2R) activity, responsible for restraining defense cells' action on myelin; (2) upregulation of ADAM-17 activity, leading to impaired maturation of oligodendrocytes and myelin formation. Thus, we hypothesize that increased ADAM-17 activity and decreased AT2R activity play roles in SARS-CoV-2 infection-mediated demyelination in the CNS.
Collapse
Affiliation(s)
- Kellysson Bruno Oliveira
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Fernanda Maria Araujo de Souza
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Letícia Barros Maurício de Sá
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Amanda Larissa Dias Pacheco
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Mariana Reis Prado
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Célio Fernando de Sousa Rodrigues
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Ênio José Bassi
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Igor Santana-Melo
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Abelardo Silva-Júnior
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Robinson Sabino-Silva
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA.
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, 77843, USA.
| | - Olagide Wagner de Castro
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil.
| |
Collapse
|
4
|
Tang J, Shi J, Han Z, Chen X. Application of Macrophage Subtype Analysis in Acute Lung Injury/Acute Respiratory Distress Syndrome. FRONT BIOSCI-LANDMRK 2024; 29:412. [PMID: 39735977 DOI: 10.31083/j.fbl2912412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/28/2024] [Accepted: 08/16/2024] [Indexed: 12/31/2024]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a common critical illness. Supportive therapy is still the main strategy for ALI/ARDS. Macrophages are the predominant immune cells in the lungs and play a pivotal role in maintaining homeostasis, regulating metabolism, and facilitating tissue repair. During ALI/ARDS, these versatile cells undergo polarization into distinct subtypes with significant variations in transcriptional profiles, developmental trajectory, phenotype, and functionality. This review discusses developments in the analysis of alveolar macrophage subtypes in the study of ALI/ARDS, and the potential value of targeting new macrophage subtypes in the diagnosis, prognostic evaluation, and treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Jiajia Tang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, 100048 Beijing, China
- School of Medicine, South China University of Technology, 510006 Guangzhou, Guangdong, China
| | - Jun Shi
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, 100048 Beijing, China
- School of Medicine, South China University of Technology, 510006 Guangzhou, Guangdong, China
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, 100048 Beijing, China
- School of Medicine, South China University of Technology, 510006 Guangzhou, Guangdong, China
| | - Xuxin Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, 100048 Beijing, China
- School of Medicine, South China University of Technology, 510006 Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Liu G, Mu KL, Ran F, Liu JM, Zhou LL, Peng LQ, Feng G, Liu YC, Wei FD, Zhu LL, Zhang XY, Zhang YP, Sun QW. The hemostatic activity and Mechanistic roles of glucosyloxybenzyl 2-isobutylmalate extract (BSCE) from Bletilla striata (Thunb.) Rchb.f. in Inhibiting pulmonary hemorrhage. Heliyon 2024; 10:e38203. [PMID: 39381249 PMCID: PMC11459001 DOI: 10.1016/j.heliyon.2024.e38203] [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: 05/03/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
Background Hemorrhagic events cause numerous deaths annually worldwide, highlighting the urgent need for effective hemostatic drugs. The glucosyloxybenzyl 2-isobutylmalates Control Extract (BSCE) from the orchid plant Bletilla striata (Thunb.) Rchb.f. has demonstrated significant hemostatic activity in both in vitro and in vivo studies. However, the effect and mechanism of BSCE on non-traumatic bleeding remain unclear. Methods Pulmonary hemorrhage was induced in 40 Sprague-Dawley rats by administering Zingiber officinale Roscoe. for 14 days. These rats were then randomly divided into five groups: model (Mod), positive control (YNBY), and BSCE low, medium, and high-dose groups. An additional 8 rats served as the control group (Con). The BSCE groups received different doses of BSCE for 10 days, while the YNBY group received Yunnan Baiyao suspension. The effects on body weight, food and water intake, red blood cell count (RBC), hemoglobin concentration (HGB), lung tissue pathology, platelet count, coagulation parameters, and fibrinolytic system markers were evaluated. Network pharmacology and molecular docking analyses were also conducted to identify potential targets and pathways involved in BSCE's effects. Results BSCE treatment significantly improved body weight, food intake, and water consumption in rats with pulmonary hemorrhage. RBC and HGB levels increased significantly in the BSCE medium and high-dose groups compared to the Mod group (P < 0.05). Pathological examination revealed that BSCE reduced lung tissue hemorrhage and inflammation, with improvements in alveolar structure. BSCE also positively affected platelet count, thrombin time (TT), activated partial thromboplastin time (APTT), fibrinogen (FIB) levels, and fibrinolytic markers (D-dimer, PAI-1, and t-PA). Network pharmacology and molecular docking identified key targets such as MMPs, CASPs, and pathways including IL-17 and TNF signaling, suggesting BSCE's involvement in hemostasis and anti-inflammatory processes. Conclusions BSCE exhibits significant hemostatic and protective effects on Z.officinale-induced pulmonary hemorrhage in rats by improving hematological parameters, reducing lung tissue damage, and modulating the coagulation and fibrinolytic systems. The study provides evidence supporting the potential of BSCE as a therapeutic agent for hemorrhagic diseases, with its efficacy linked to multi-target and multi-pathway interactions.
Collapse
Affiliation(s)
| | | | - Fei Ran
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Jin-mei Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Ling-li Zhou
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Le-qiang Peng
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Guo Feng
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yu-chen Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Fu-dao Wei
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Ling-li Zhu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Xin-yue Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yong-ping Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Qing-wen Sun
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| |
Collapse
|
6
|
Yang Z, Hao T, Ma J, Yang D, Qiu M, Wang R. Tribuloside: Mechanisms and Efficacy in Treating Acute Lung Injury Revealed by Network Pharmacology and Experimental Validation. Dose Response 2024; 22:15593258241251594. [PMID: 38725454 PMCID: PMC11080732 DOI: 10.1177/15593258241251594] [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: 12/09/2023] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Abstract
Background Acute lung injury (ALI) is a serious illness that has few treatment options available. Tribuloside, a natural flavonoid extracted from the Tribulus Terrestris plant in China, is potent in addressing many health issues such as headaches, dizziness, itching, and vitiligo. Objective This study intends to explore the mechanisms of action of Tribuloside in treating ALI through a combination of network pharmacology and experimental validation. Methods We obtained the 2D structure and SMILES number of Tribuloside from the PubChem database. We used the SwissTargetPrediction database to identify pharmacological targets. We found 1215 targets linked to ALI by examining the GeneCards database. We used the String database and Cytoscape software to create the "drug or disease-target" network as well as the protein-protein interactions (PPI). Key targets were identified by evaluating associated biological processes and pathway enrichment. A Venny Diagram showed 49 intersection points between Tribuloside and ALI. Molecular docking with AutoDockTools found that Tribuloside had a high affinity for IL6, BCL2, TNF, STAT3, IL1B, and MAPK3, the top 6 targets in the PPI network by Degree values. To test Tribuloside's therapeutic efficacy in ALI, an acute lung damage model in mice was constructed using lipopolysaccharide. Tribuloside treatment reduced inflammatory cell infiltration, decreased fibrotic area, repaired damaged alveoli, and suppressed inflammatory factors IL-6, TNF-α, and IL-1β in the lungs through many pathways and targets. Conclusion This study reveals that Tribuloside has the potential to treat ALI by targeting various pathways and targets, according to network pharmacology predictions and experimental confirmation.
Collapse
Affiliation(s)
| | | | | | - Dan Yang
- Baotou Medical College, Baotou, China
| | - Min Qiu
- Baotou Medical College, Baotou, China
- Inner Mongolia Agricultural University, Hohhot, China
| | - Rui Wang
- Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
7
|
Gan H, Zhou X, Lei Q, Wu L, Niu J, Zheng Q. RNA-dependent RNA polymerase of SARS-CoV-2 regulate host mRNA translation efficiency by hijacking eEF1A factors. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166871. [PMID: 37673357 DOI: 10.1016/j.bbadis.2023.166871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/20/2023] [Accepted: 08/31/2023] [Indexed: 09/08/2023]
Abstract
The RNA-dependent RNA polymerase (NSP12) of COVID-19 plays a significant role in the viral infection process, which promotes viral RNA replication by cooperating with NSP7 and NSP8, but little is known about its regulation on the function of host cells. We firstly found that overexpression of NSP12 had little effect on host mRNAs transcription. Using iCLIP technology, we found that NSP12 can bind a series of host RNAs with the conserved binding motif G(C/A/G)(U/G/A)UAG, especially ribosomal RNA. We found that NSP12 could directly bind to eEF1A factor via the NIRAN domain of NSP12 and N-terminal domain of eEF1A. NSP12 colocalized with eEF1A to inhibit type I interferon expression upon virus infection. In order to prove that NSP12 regulates the translation level of host cells, we found that NSP12 significantly affected the translation efficiency of many host mRNAs (such as ISG15, NF-κB2, ILK and SERPINI2) via ribosome profiling experiment, and the genes with significant upregulation in translation efficiency were mainly enriched in positive regulation of ubiquitin-dependent proteasomal process and NIK/NF-κB signaling pathway (such as NF-κB2, ILK), and negative regulation of type I interferon production, protein level of these genes were further confirmed in HEK293T and Calu3 cells upon NSP12 overexpression. These results indicate that NSP12 of SARS-CoV-2 can hijack the eEF1A factor to regulate translation efficiency of host mRNAs, which provides a new idea for us to evaluate the impact of SARS-CoV2 virus on the host and study the potential drug targets.
Collapse
Affiliation(s)
- Haili Gan
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Xiaoguang Zhou
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518000, China
| | - Qiong Lei
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518000, China
| | - Linlin Wu
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518000, China
| | - Jianmin Niu
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518000, China
| | - Qingliang Zheng
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518000, China.
| |
Collapse
|
8
|
Tanzadehpanah H, Lotfian E, Avan A, Saki S, Nobari S, Mahmoodian R, Sheykhhasan M, Froutagh MHS, Ghotbani F, Jamshidi R, Mahaki H. Role of SARS-COV-2 and ACE2 in the pathophysiology of peripheral vascular diseases. Biomed Pharmacother 2023; 166:115321. [PMID: 37597321 DOI: 10.1016/j.biopha.2023.115321] [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: 05/13/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023] Open
Abstract
The occurrence of a novel coronavirus known as severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), created a serious challenge worldwide. SARS-CoV-2 has high infectivity, the ability to be transmitted even during the asymptomatic phase, and relatively low virulence, which has resulted in rapid transmission. SARS-CoV-2 can invade epithelial cells, hence, many patients infected with SARS-CoV-2 have suffered from vascular diseases (VDs) in addition to pulmonary manifestations. Accordingly, SARS-CoV-2 may can worsen the clinical condition of the patients with pre-existing VDs. Endothelial cells express angiotensin-converting enzyme 2 (ACE2). ACE2 is a biological enzyme that converts angiotensin (Ang)- 2 to Ang-(1-7). SARS-CoV-2 uses ACE2 as a cell receptor for viral entry. Thus, the SARS-CoV-2 virus promotes downregulation of ACE2, Ang-(1-7), and anti-inflammatory cytokines, as well as, an increase in Ang-2, resulting in pro-inflammatory cytokines. SARS-CoV-2 infection can cause hypertension, and endothelial damage, which can lead to intravascular thrombosis. In this review, we have concentrated on the effect of SARS-CoV-2 in peripheral vascular diseases (PVDs) and ACE2 as an enzyme in Renin-angiotensin aldosterone system (RAAS). A comprehensive search was performed on PubMed, Google Scholar, Scopus, using related keywords. Articles focusing on ("SARS-CoV-2", OR "COVID-19"), AND ("Vascular disease", OR "Peripheral vascular disease", OR interested disease name) with regard to MeSH terms, were selected. According to the studies, it is supposed that vascular diseases may increase susceptibility to severe SARS-CoV-2 infection due to increased thrombotic burden and endothelial dysfunction. Understanding SARS-CoV-2 infection mechanism and vascular system pathogenesis is crucial for effective management and treatment in pre-existing vascular diseases.
Collapse
Affiliation(s)
- Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Lotfian
- Vascular and Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences; Medical Genetics Research Center, Mashhad University of Medical Sciences; Medical Genetics Research center, Mashhad University of Medical Sciences; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Saki
- Molecular Medicine Research Center, Hamadan University of Medical Science, Hamadan Iran
| | - Sima Nobari
- Molecular Medicine Research Center, Hamadan University of Medical Science, Hamadan Iran
| | - Roghaye Mahmoodian
- Molecular Medicine Research Center, Hamadan University of Medical Science, Hamadan Iran
| | - Mohsen Sheykhhasan
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran
| | | | - Farzaneh Ghotbani
- Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Raoufeh Jamshidi
- Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Hanie Mahaki
- Vascular and Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Russo C, Valle MS, Malaguarnera L, Romano IR, Malaguarnera L. Comparison of Vitamin D and Resveratrol Performances in COVID-19. Nutrients 2023; 15:nu15112639. [PMID: 37299603 DOI: 10.3390/nu15112639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Over the last few years, we have experienced the infection generated by severe respiratory syndrome coronavirus 2 (SARS-CoV-2) often resulting in an exaggerated immune reaction and systemic inflammation. The preferred treatments against SARS-CoV-2 were those that mitigated immunological/inflammatory dysfunction. A variety of observational epidemiological studies have reported that vitamin D deficiency is often a crucial factor in many inflammatory diseases and autoimmune diseases, as well as the susceptibility to contract infectious diseases, including acute respiratory infections. Similarly, resveratrol regulates immunity, modifying the gene expression and the release of proinflammatory cytokines in the immune cells. Therefore, it plays an immunomodulatory role that can be beneficial in the prevention and development of non-communicable diseases associated with inflammation. Since both vitamin D and resveratrol also act as immunomodulators in inflammatory pathologies, many studies have paid particular attention to an integrated treatment of either vitamin D or resveratrol in the immune reaction against SARS-CoV-2 infections. This article offers a critical evaluation of published clinical trials that have examined the use of vitamin D or resveratrol as adjuncts in COVID-19 management. Furthermore, we aimed to compare the anti-inflammatory and antioxidant properties linked to the modulation of the immune system, along with antiviral properties of both vitamin D and resveratrol.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Maria Stella Valle
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Luisa Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Ivana Roberta Romano
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
10
|
Jeon T, Luther DC, Goswami R, Bell C, Nagaraj H, Anil Cicek Y, Huang R, Mas-Rosario JA, Elia JL, Im J, Lee YW, Liu Y, Scaletti F, Farkas ME, Mager J, Rotello VM. Engineered Polymer-siRNA Polyplexes Provide Effective Treatment of Lung Inflammation. ACS NANO 2023; 17:4315-4326. [PMID: 36802503 PMCID: PMC10627429 DOI: 10.1021/acsnano.2c08690] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Uncontrolled inflammation is responsible for acute and chronic diseases in the lung. Regulating expression of pro-inflammatory genes in pulmonary tissue using small interfering RNA (siRNA) is a promising approach to combatting respiratory diseases. However, siRNA therapeutics are generally hindered at the cellular level by endosomal entrapment of delivered cargo and at the organismal level by inefficient localization in pulmonary tissue. Here we report efficient anti-inflammatory activity in vitro and in vivo using polyplexes of siRNA and an engineered cationic polymer (PONI-Guan). PONI-Guan/siRNA polyplexes efficiently deliver siRNA cargo to the cytosol for highly efficient gene knockdown. Significantly, these polyplexes exhibit inherent targeting to inflamed lung tissue following intravenous administration in vivo. This strategy achieved effective (>70%) knockdown of gene expression in vitro and efficient (>80%) silencing of TNF-α expression in lipopolysaccharide (LPS)-challenged mice using a low (0.28 mg/kg) siRNA dosage.
Collapse
Affiliation(s)
- Taewon Jeon
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, Massachusetts, 01003, USA
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - David C. Luther
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Charlotte Bell
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Yagiz Anil Cicek
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Javier A. Mas-Rosario
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, Massachusetts, 01003, USA
| | - James L. Elia
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Jungkyun Im
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
- Department of Chemical Engineering, and Department of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, 22 Soonchunhyangro, Asan, 31538, Republic of Korea
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Yuanchang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Federica Scaletti
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Michelle E. Farkas
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, Massachusetts, 01003, USA
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Jesse Mager
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Vincent M. Rotello
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, Massachusetts, 01003, USA
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
11
|
PLGA-Based Micro/Nanoparticles: An Overview of Their Applications in Respiratory Diseases. Int J Mol Sci 2023; 24:ijms24054333. [PMID: 36901762 PMCID: PMC10002081 DOI: 10.3390/ijms24054333] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Respiratory diseases, such as asthma and chronic obstructive pulmonary disease (COPD), are critical areas of medical research, as millions of people are affected worldwide. In fact, more than 9 million deaths worldwide were associated with respiratory diseases in 2016, equivalent to 15% of global deaths, and the prevalence is increasing every year as the population ages. Due to inadequate treatment options, the treatments for many respiratory diseases are limited to relieving symptoms rather than curing the disease. Therefore, new therapeutic strategies for respiratory diseases are urgently needed. Poly (lactic-co-glycolic acid) micro/nanoparticles (PLGA M/NPs) have good biocompatibility, biodegradability and unique physical and chemical properties, making them one of the most popular and effective drug delivery polymers. In this review, we summarized the synthesis and modification methods of PLGA M/NPs and their applications in the treatment of respiratory diseases (asthma, COPD, cystic fibrosis (CF), etc.) and also discussed the research progress and current research status of PLGA M/NPs in respiratory diseases. It was concluded that PLGA M/NPs are the promising drug delivery vehicles for the treatment of respiratory diseases due to their advantages of low toxicity, high bioavailability, high drug loading capacity, plasticity and modifiability. And at the end, we presented an outlook on future research directions, aiming to provide some new ideas for future research directions and hopefully to promote their widespread application in clinical treatment.
Collapse
|
12
|
Oliveira KB, de Melo IS, da Silva BRM, Oliveira KLDS, Sabino-Silva R, Anhezini L, Katayama PL, Santos VR, Shetty AK, de Castro OW. SARS-CoV-2 and Hypertension: Evidence Supporting Invasion into the Brain Via Baroreflex Circuitry and the Role of Imbalanced Renin-Angiotensin-Aldosterone-System. Neurosci Insights 2023; 18:26331055231151926. [PMID: 36756280 PMCID: PMC9900164 DOI: 10.1177/26331055231151926] [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: 11/18/2022] [Accepted: 01/04/2023] [Indexed: 02/05/2023] Open
Abstract
Hypertension is considered one of the most critical risk factors for COVID-19. Evidence suggests that SARS-CoV-2 infection produces intense effects on the cardiovascular system by weakening the wall of large vessels via vasa-vasorum. In this commentary, we propose that SARS-CoV-2 invades carotid and aortic baroreceptors, leading to infection of the nucleus tractus solitari (NTS) and paraventricular hypothalamic nucleus (PVN), and such dysregulation of NTS and PVN following infection causes blood pressure alteration at the central level. We additionally explored the hypothesis that SARS-CoV-2 favors the internalization of membrane ACE2 receptors generating an imbalance of the renin-angiotensin-aldosterone system (RAAS), increasing the activity of angiotensin II (ANG-II), disintegrin, and metalloproteinase 17 domain (ADAM17/TACE), eventually modulating the integration of afferents reaching the NTS from baroreceptors and promoting increased blood pressure. These mechanisms are related to the increased sympathetic activity, which leads to transient or permanent hypertension associated with SARS-CoV-2 invasion, contributing to the high number of deaths by cardiovascular implications.
Collapse
Affiliation(s)
- Kellysson Bruno Oliveira
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Igor Santana de Melo
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Bianca Rodrigues Melo da Silva
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Keylla Lavínia da Silva Oliveira
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Robinson Sabino-Silva
- Department of Physiology, Institute of
Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlândia, Minas
Gerais, Brazil
| | - Lucas Anhezini
- Department of Histology, Institute of
Biological Sciences and Health, Federal University of Alagoas, Maceió, Alagoas,
Brazil
| | - Pedro Lourenco Katayama
- Department of Physiology and Pathology,
Dentistry School of Araraquara, São Paulo State University, Araraquara, São Paulo,
Brazil
| | - Victor Rodrigues Santos
- Department of Morphology, Institute of
Biological Science, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas
Gerais, Brazil
| | - Ashok K Shetty
- Institute for Regenerative Medicine,
Department of Cell Biology and Genetics, Texas A&M University School of
Medicine, College Station, TX, USA
| | - Olagide Wagner de Castro
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil,Olagide Wagner de Castro, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Av.
Lourival de Melo Mota, km 14, Campus A. C. Simões, Cidade Universitária, Maceió,
Alagoas CEP 57072-970, Brazil.
| |
Collapse
|
13
|
Alsaiari AA, Allahyani M, Aljuaid A, Shafie A, Al-hazmi A, Dahlawi HA, Abdulaziz O, Alosimi E, Alzaidi A, Almehmadi M. Assessing the adverse effects of COVID-19 vaccine in different scenarios in Saudi Arabia: A cross-sectional study. Saudi Med J 2023; 44:194-201. [PMID: 36773975 PMCID: PMC9987704 DOI: 10.15537/smj.2023.44.2.20220680] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/19/2022] [Indexed: 02/13/2023] Open
Abstract
OBJECTIVES To assess the different side effects of COVID-19 vaccines at different scenarios in Saudi Arabia. METHODS This cross-sectional study sought to investigate the side effects of COVID-19 vaccines through an online survey of 2,718 participants in Saudi Arabia. RESULTS People can manage their expectations about vaccine side effects and deal with symptoms better by knowing beforehand that they are likely to experience mild side effects for a short period, symptoms that are manifested regardless of age, and infection before or after vaccination. There are certain uncommon side effects that affect more people who got infected, and not before vaccination; there are side effects that disproportionately impact women, and also the side effects that wane after the second dose. CONCLUSION These findings can assist in evaluating the concerns regarding vaccine acceptance. The public should be made aware that they are likely to experience at least one side effect, with temporary post-injection inflammation, musculoskeletal pain, fever, and headache as the most commonly reported side effects across the board. However, the common symptoms are mild to moderate, and the side effects last for a short period for most people.
Collapse
Affiliation(s)
- Ahad Amer Alsaiari
- From the Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Mamdouh Allahyani
- From the Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Abdulelah Aljuaid
- From the Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Alaa Shafie
- From the Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Ayman Al-hazmi
- From the Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Haytham A. Dahlawi
- From the Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Osama Abdulaziz
- From the Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Ebtisam Alosimi
- From the Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Albatool Alzaidi
- From the Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Mazen Almehmadi
- From the Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
- Address correspondence and reprint request to: Dr. Mazen Almehmadi, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia. E-mail: ORCID ID: https://orcid.org/0000-0002-7580-8667
| |
Collapse
|
14
|
Singh M, Pushpakumar S, Bard N, Zheng Y, Homme RP, Mokshagundam SPL, Tyagi SC. Simulation of COVID-19 symptoms in a genetically engineered mouse model: implications for the long haulers. Mol Cell Biochem 2023; 478:103-119. [PMID: 35731343 PMCID: PMC9214689 DOI: 10.1007/s11010-022-04487-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/30/2022] [Indexed: 01/24/2023]
Abstract
The ongoing pandemic (also known as coronavirus disease-19; COVID-19) by a constantly emerging viral agent commonly referred as the severe acute respiratory syndrome corona virus 2 or SARS-CoV-2 has revealed unique pathological findings from infected human beings, and the postmortem observations. The list of disease symptoms, and postmortem observations is too long to mention; however, SARS-CoV-2 has brought with it a whole new clinical syndrome in "long haulers" including dyspnea, chest pain, tachycardia, brain fog, exercise intolerance, and extreme fatigue. We opine that further improvement in delivering effective treatment, and preventive strategies would be benefited from validated animal disease models. In this context, we designed a study, and show that a genetically engineered mouse expressing the human angiotensin converting enzyme 2; ACE-2 (the receptor used by SARS-CoV-2 agent to enter host cells) represents an excellent investigative resource in simulating important clinical features of the COVID-19. The ACE-2 mouse model (which is susceptible to SARS-CoV-2) when administered with a recombinant SARS-CoV-2 spike protein (SP) intranasally exhibited a profound cytokine storm capable of altering the physiological parameters including significant changes in cardiac function along with multi-organ damage that was further confirmed via histological findings. More importantly, visceral organs from SP treated mice revealed thrombotic blood clots as seen during postmortem examination. Thus, the ACE-2 engineered mouse appears to be a suitable model for studying intimate viral pathogenesis thus paving the way for identification, and characterization of appropriate prophylactics as well as therapeutics for COVID-19 management.
Collapse
Affiliation(s)
- Mahavir Singh
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Nia Bard
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Yuting Zheng
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Rubens P Homme
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Sri Prakash L Mokshagundam
- Division of Endocrinology, Metabolism and Diabetes and Robley Rex VA Medical Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| |
Collapse
|
15
|
B.1.351 SARS-CoV-2 Variant Exhibits Higher Virulence but Less Viral Shedding than That of the Ancestral Strain in Young Nonhuman Primates. Microbiol Spectr 2022; 10:e0226322. [PMID: 36069561 PMCID: PMC9603226 DOI: 10.1128/spectrum.02263-22] [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] [Indexed: 01/04/2023] Open
Abstract
We investigated the distribution, virulence, and pathogenic characteristics of mutated SARS-CoV-2 to clarify the association between virulence and the viral spreading ability of current and future circulating strains. Chinese rhesus macaques were infected with ancestral SARS-CoV-2 strain GD108 and Beta variant B.1.351 (B.1.351) and assessed for clinical signs, viral distribution, pathological changes, and pulmonary inflammation. We found that GD108 replicated more efficiently in the upper respiratory tract, whereas B.1.351 replicated more efficiently in the lower respiratory tract and lung tissue, implying a reduced viral shedding and spreading ability of B.1.351 compared with that of GD108. Importantly, B.1.351 caused more severe lung injury and dramatically elevated the level of inflammatory cytokines compared with those observed after infection with GD108. Moreover, both B.1.351 and GD108 induced spike-specific T-cell responses at an early stage of infection, with higher levels of interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) in the B.1.351 group and higher levels of interleukin 17 (IL-17) in the GD108 group, indicating a divergent pattern in the T-cell-mediated inflammatory "cytokine storm." This study provides a basis for exploring the pathogenesis of SARS-CoV-2 variants of concern (VOCs) and establishes an applicable animal model for evaluating the efficacy and safety of vaccines and drugs. IMPORTANCE One of the priorities of the current SARS-CoV-2 vaccine and drug research strategy is to determine the changes in transmission ability, virulence, and pathogenic characteristics of SARS-CoV-2 variants. In addition, nonhuman primates (NHPs) are suitable animal models for the study of the pathogenic characteristics of SARS-CoV-2 and could contribute to the understanding of pathogenicity and transmission mechanisms. As SARS-CoV-2 variants continually emerge and the viral biological characteristics change frequently, the establishment of NHP infection models for different VOCs is urgently needed. In the study, the virulence and tissue distribution of B.1.351 and GD108 were comprehensively studied in NHPs. We concluded that the B.1.351 strain was more virulent but exhibited less viral shedding than the latter. This study provides a basis for determining the pathogenic characteristics of SARS-CoV-2 and establishes an applicable animal model for evaluating the efficacy and safety of vaccines and drugs.
Collapse
|
16
|
Jiang T, Sun L, Zhu J, Li N, Gu H, Zhang Y, Li M, Xu J. MicroRNA-23a-3p promotes macrophage M1 polarization and aggravates lipopolysaccharide-induced acute lung injury by regulating PLK1/STAT1/STAT3 signalling. Int J Exp Pathol 2022; 103:198-207. [PMID: 35739646 PMCID: PMC9482356 DOI: 10.1111/iep.12445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/07/2022] [Indexed: 12/16/2022] Open
Abstract
Macrophage polarization is an important effector process in acute lung injury (ALI) induced by sepsis. MicroRNAs (miRNAs) have emerged as important players in regulating ALI process. Here, we showed that elevated microRNA-23a-3p (miR-23a-3p) promoted LPS-induced macrophage polarization and ALI in mice, while inhibition of miR-23a-3p led to reduced macrophage response and ameliorated ALI inflammation. Mechanically, miR-23a-3p regulated macrophage M1 polarization through targeting polo-like kinase 1 (PLK1). PLK1 was downregulated in LPS-treated macrophages and ALI mouse lung tissues. Knockdown of PLK1 increased macrophage M1 polarization through promoting STAT1/STAT3 activation, while overexpression of PLK1 reduced macrophage immune response. Collectively, our results reveal a key miRNA regulon that regulates macrophage polarization for LPS-induced immune response.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of MedicineZhejiang UniversityYiwuChina
| | - Li Sun
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of MedicineZhejiang UniversityYiwuChina
| | - Jun Zhu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of MedicineZhejiang UniversityYiwuChina
| | - Ning Li
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of MedicineZhejiang UniversityYiwuChina
| | - Haibo Gu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of MedicineZhejiang UniversityYiwuChina
| | - Ying Zhang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of MedicineZhejiang UniversityYiwuChina
| | - Miaomiao Li
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of MedicineZhejiang UniversityYiwuChina
| | - Jiayao Xu
- Department of EmergencyTiantai Hospital of Hangzhou Medical CollegeTiantaiChina
| |
Collapse
|
17
|
Al-Kuraishy HM, Al-Gareeb AI, Alkazmi L, Habotta OA, Batiha GES. High-mobility group box 1 (HMGB1) in COVID-19: extrapolation of dangerous liaisons. Inflammopharmacology 2022; 30:811-820. [PMID: 35471628 PMCID: PMC9040700 DOI: 10.1007/s10787-022-00988-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/30/2022] [Indexed: 02/06/2023]
Abstract
High-mobility group box 1 (HMGB1), a multifunctional nuclear protein, exists mainly within the nucleus of all mammal eukaryotic cells. It is actively secreted by the necrotic cells as a response to the inflammatory signaling pathway. HMGB1 binds to receptor ligands as RAGE, and TLR and becomes a pro-inflammatory cytokine with a robust capacity to trigger inflammatory response. It is a critical mediator of the pathogenesis of systemic inflammation in numerous inflammatory disorders. Release of HMGB1 is associated with different viral infections and strongly participates in the regulation of viral replication cycles. In COVID-19 era, high HMGB1 serum levels were observed in COVID-19 patients and linked with the disease severity, development of cytokine storm (CS), acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). SARS-CoV-2-induced cytolytic effect may encourage release of HMGB1 due to nuclear damage. Besides, HMGB1 activates release of pro-inflammatory cytokines from immune cells and up-regulation of angiotensin I-converting enzyme 2 (ACE2). Therefore, targeting of the HMGB1 pathway by anti-HMGB1 agents, such as heparin, resveratrol and metformin, may decrease COVID-19 severity. HMGB1 signaling pathway has noteworthy role in the pathogenesis of SARS-CoV-2 infections and linked with development of ALI and ARDS in COVID-19 patients. Different endogenous and exogenous agents may affect release and activation of HMGB1 pathway. Targeting of HMGB1-mediated TLR2/TLR4, RAGE and MAPK signaling, might be a new promising drug candidate against development of ALI and/or ARDS in severely affected COVID-19 patients.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, 14132, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, 14132, Iraq
| | - Luay Alkazmi
- Biology Department, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Ola A Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
18
|
Ultrastructural examination of lung "cryobiopsies" from a series of fatal COVID-19 cases hardly revealed infected cells. Virchows Arch 2022; 480:967-977. [PMID: 35294603 PMCID: PMC8924574 DOI: 10.1007/s00428-022-03308-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/24/2022] [Accepted: 03/07/2022] [Indexed: 11/10/2022]
Abstract
Ultrastructural analysis of autopsy samples from COVID-19 patients usually suffers from significant structural impairment possibly caused by the rather long latency between death of the patient and an appropriate sample fixation. To improve structural preservation of the tissue, we obtained samples from ventilated patients using a trans-bronchial “cryobiopsy” within 30 min after their death and fixed them immediately for electron microscopy. Samples of six COVID-19 patients with a documented histopathology were systematically investigated by thin section electron microscopy. The different samples and areas inspected revealed the ultrastructural correlates of the different phases of diffuse alveolar damage, including detachment of the alveolar epithelium, hyperplasia of type 2 cells, exudates, and accumulation of extracellular material, such as the hyaline membranes and fibrin. Macrophages and neutrophilic granulocytes were regularly detected. Structural integrity of endothelium was intact in regions where the alveolar epithelium was already detached. Aggregates of erythrocytes, leukocytes with fibrin, and thrombocytes were not observed. Coronavirus particles were only found in and around very few cells in one of the six patient samples. The type and origin of these cells could not be assessed although the overall structural preservation of the samples allowed the identification of pulmonary cell types. Hence, the observed alveolar damage is not associated with virus presence or structural impairment due to ongoing replication at later stages of the disease in fatal cases, which implies that the lung damage in these patients is at least propagated by alternative mechanisms, perhaps, an inappropriate immune or stress response.
Collapse
|
19
|
Abstract
Patients in the intensive care unit (ICU) often straddle the divide between life and death. Understanding the complex underlying pathomechanisms relevant to such situations may help intensivists select broadly acting treatment options that can improve the outcome for these patients. As one of the most important defense mechanisms of the innate immune system, the complement system plays a crucial role in a diverse spectrum of diseases that can necessitate ICU admission. Among others, myocardial infarction, acute lung injury/acute respiratory distress syndrome (ARDS), organ failure, and sepsis are characterized by an inadequate complement response, which can potentially be addressed via promising intervention options. Often, ICU monitoring and existing treatment options rely on massive intervention strategies to maintain the function of vital organs, and these approaches can further contribute to an unbalanced complement response. Artificial surfaces of extracorporeal organ support devices, transfusion of blood products, and the application of anticoagulants can all trigger or amplify undesired complement activation. It is, therefore, worth pursuing the evaluation of complement inhibition strategies in the setting of ICU treatment. Recently, clinical studies in COVID-19-related ARDS have shown promising effects of central inhibition at the level of C3 and paved the way for prospective investigation of this approach. In this review, we highlight the fundamental and often neglected role of complement in the ICU, with a special focus on targeted complement inhibition. We will also consider complement substitution therapies to temporarily counteract a disease/treatment-related complement consumption.
Collapse
|
20
|
Zhou Y, Zhang F, Ding J. As a Modulator, Multitasking Roles of SIRT1 in Respiratory Diseases. Immune Netw 2022; 22:e21. [PMID: 35799705 PMCID: PMC9250864 DOI: 10.4110/in.2022.22.e21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 01/04/2023] Open
Affiliation(s)
- Yunxin Zhou
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing Institute of Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Fan Zhang
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing Institute of Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Junying Ding
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing Institute of Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| |
Collapse
|
21
|
Ma Y, Meng C, Weng L. Association between trauma exposure and respiratory disease-A Mendelian randomization study. Front Endocrinol (Lausanne) 2022; 13:1001223. [PMID: 36133309 PMCID: PMC9483852 DOI: 10.3389/fendo.2022.1001223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Trauma is a well-known risk factor for many disease, but the effect of trauma on respiratory disease is unclarified. In the present study, we aimed to evaluate the association between trauma and respiratory disease. METHOD Using both United Kingdom biobank and Finnish biobank genome-wide association study data (GWAS), we performed a two-sample Mendelian randomization (MR) analysis to evaluate the relationship between trauma and respiratory disease. We used four methods including inverse-variance weighted (IVW), weighted median, Maximum likelihood, and MR-Egger in this MR analysis. The IVW MR was selected as the main method. We also performed multivariable Mendelian randomization (MVMR) to simultaneously assess the independent impact of trauma exposure on respiratory disease. RESULTS In the main two-sample MR analysis, trauma exposure was significantly associated with increased risk of respiratory disease (OR 1.15, 95%CI: 1.05-1.25). Besides, there was no heterogeneity and horizontal pleiotropy observed in the sensitivity analysis. After adjusting for pack years of smoking and body mass index (BMI), trauma exposure retained its association with respiratory disease (OR, 1.13, 95%CI, 1.04-1.23 adjusted by pack years of smoking; and OR, 1.11, 95%CI, 1.04-1.18 adjusted by BMI). CONCLUSION Our study discovered the association between trauma exposure and the increased risk of respiratory disease, suggesting the prevention and treatment with trauma to reduce the risk of respiratory disease.
Collapse
Affiliation(s)
- Yuchao Ma
- Department of Cardiothoracic Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Changjiang Meng
- Clinical Research Center, Third Xiangya Hospital, Central South University, Changsha, China
| | - Liang Weng
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Xiangya Hospital, Central South University, Changsha, China
- Institute of Gerontological Cancer Research, National Clinical Research Center for Gerontology, Changsha, China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Liang Weng,
| |
Collapse
|
22
|
Nechipurenko YD, Semyonov DA, Lavrinenko IA, Lagutkin DA, Generalov EA, Zaitceva AY, Matveeva OV, Yegorov YE. The Role of Acidosis in the Pathogenesis of Severe Forms of COVID-19. BIOLOGY 2021; 10:852. [PMID: 34571729 PMCID: PMC8469745 DOI: 10.3390/biology10090852] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/23/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022]
Abstract
COVID-19 has specific characteristics that distinguish this disease from many other infections. We suggest that the pathogenesis of severe forms of COVID-19 can be associated with acidosis. This review article discusses several mechanisms potentially linking the damaging effects of COVID-19 with acidosis and shows the existence of a vicious cycle between the development of hypoxia and acidosis in COVID-19 patients. At the early stages of the disease, inflammation, difficulty in gas exchange in the lungs and thrombosis collectively contribute to the onset of acidosis. In accordance with the Verigo-Bohr effect, a decrease in blood pH leads to a decrease in oxygen saturation, which contributes to the exacerbation of acidosis and results in a deterioration of the patient's condition. A decrease in pH can also cause conformational changes in the S-protein of the virus and thus lead to a decrease in the affinity and avidity of protective antibodies. Hypoxia and acidosis lead to dysregulation of the immune system and multidirectional pro- and anti-inflammatory reactions, resulting in the development of a "cytokine storm". In this review, we highlight the potential importance of supporting normal blood pH as an approach to COVID-19 therapy.
Collapse
Affiliation(s)
- Yury D. Nechipurenko
- Laboratory DNA-Protein Recognition, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Denis A. Semyonov
- Institute of Molecular Medicine and Pathobiochemistry, Voyno-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia;
- Institute of Biophysics Siberian Branch of Russian Academy of Sciences, Krasnoyarsk 660036, Russia
| | - Igor A. Lavrinenko
- Department of Human and Animal Physiology, Faculty of Medicine and Biology, Voronezh State University, Voronezh 394018, Russia;
| | - Denis A. Lagutkin
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia;
| | - Evgenii A. Generalov
- Department of Biophysics, Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia;
| | - Anna Y. Zaitceva
- Laboratory of Medical Analytical Methods and Devices, Institute for Analytical Instrumentation of the Russian Academy of Sciences, St. Petersburg 198095, Russia;
| | | | - Yegor E. Yegorov
- Laboratory of Cellular Bases for the Development of Malignant Diseases, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| |
Collapse
|
23
|
Peña-Otero D, Bello Dronda S, Díaz-Pérez D, de la Rosa Carrillo D. One year on: Are we ready for COVID? Arch Bronconeumol 2021; 57:517-518. [PMID: 34059451 PMCID: PMC8163601 DOI: 10.1016/j.arbr.2021.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/15/2022]
Affiliation(s)
- David Peña-Otero
- Enfermería, Hospital Sierrallana, Subdirección de Cuidados del Servicio Cántabro de Salud, Miembro IDIVAL e IiSGM, Área de Enfermería Respiratoria-SEPAR, Torrelavega, Cantabria, Spain
| | | | - David Díaz-Pérez
- Enfermería, Hospital Universitario Nuestra Señora de Candelaria, Servicio Canario de Salud, Área de Enfermería Respiratoria-SEPAR, Santa Cruz de Tenerife, Spain
| | | |
Collapse
|
24
|
Ilias I, Goulas S, Zabuliene L. Polycystic ovary syndrome: Pathways and mechanisms for possible increased susceptibility to COVID-19. World J Clin Cases 2021; 9:2711-2720. [PMID: 33969054 PMCID: PMC8058679 DOI: 10.12998/wjcc.v9.i12.2711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/16/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023] Open
Abstract
In 75% of women with polycystic ovary syndrome (PCOS), insulin action is impaired. In obesity, visceral adipose tissue becomes dysfunctional: Chronic inflammation is favored over storage, contributing to the development of metabolic complications. PCOS, metabolic syndrome (MetSy) and non-alcoholic fatty liver disease (NAFLD) apparently share common pathogenic factors; these include abdominal adiposity, excess body weight and insulin resistance. Alterations in the gut microbiome have been noted in women with PCOS compared to controls; these may lead to deterioration of the intestinal barrier, increased gut mucosal permeability and immune system activation, hyperinsulinemia and glucose intolerance, which hamper normal ovarian function and follicular development (all being hallmarks of PCOS). It has been proposed that PCOS may entail higher susceptibility to coronavirus disease 2019 (COVID-19) via its associated comorbidities (NAFLD, obesity, MetSy and alterations in the gut microbiome). Studies have found an association between acute respiratory distress syndrome (seen in severe cases of COVID-19) and the intestinal microbiome. Furthermore, apparently, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can gain entry to the gastrointestinal tract via locally-expressed angiotensin converting enzyme type 2 receptors. Excess body weight is associated with more severe COVID-19 and increased mortality. Although robust links between SARS-CoV-2 infection and PCOS/NAFLD/gut microbiome/metabolic consequences are yet to be confirmed, it seems that strategies for adapting the intestinal microbiome could help reduce the severity of COVID-19 in women with PCOS with or without NAFLD, MetSy or obesity.
Collapse
Affiliation(s)
- Ioannis Ilias
- Department of Endocrinology, Elena Venizelou Hospital, Athens GR-11521, Greece
| | - Spyridon Goulas
- Department of Gastroenterology Unit, Elena Venizelou Hospital, Athens GR-11521, Greece
| | - Lina Zabuliene
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius LT-03101, Lithuania
| |
Collapse
|
25
|
Peña-Otero D, Bello Dronda S, Díaz-Pérez D, de la Rosa Carrillo D. One Year On: Are We Ready for COVID? Arch Bronconeumol 2021; 57:S0300-2896(21)00089-2. [PMID: 33795187 PMCID: PMC7955579 DOI: 10.1016/j.arbres.2021.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/15/2022]
Affiliation(s)
- David Peña-Otero
- Enfermería, Hospital Sierrallana, Subdirección de Cuidados del Servicio Cántabro de Salud. Miembro IDIVAL e IiSGM. Área de Enfermería Respiratoria-SEPAR, Torrelavega, Cantabria, España
| | | | - David Díaz-Pérez
- Enfermería, Hospital Universitario Nuestra Señora de Candelaria, Servicio Canario de Salud. Área de Enfermería Respiratoria-SEPAR, Santa Cruz de Tenerife, España
| | | |
Collapse
|
26
|
Zhang B, Zeng M, Li B, Kan Y, Wang S, Cao B, Huang Y, Zheng X, Feng W. Arbutin attenuates LPS-induced acute kidney injury by inhibiting inflammation and apoptosis via the PI3K/Akt/Nrf2 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 82:153466. [PMID: 33494001 DOI: 10.1016/j.phymed.2021.153466] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/14/2020] [Accepted: 01/08/2021] [Indexed: 05/23/2023]
Abstract
BACKGROUND Arbutin (Ar) has anti-oxidative and anti-inflammatory activities. However, the effects of Ar on lipopolysaccharide (LPS)-induced acute kidney injury (AKI) are not clear. PURPOSE This study aimed to investigate the effects of Ar on LPS-induced AKI in rats. METHODS The possible data regarding the effects of Ar on AKI were collected by network pharmacology research. Histological changes in the kidney and the levels of blood urea nitrogen, serum creatinine, and kidney injury molecule 1 were measured to assess the effects of Ar on renal function in LPS-induced AKI. The levels of inflammatory were detected by live small-animal imaging, cytometric bead array and enzyme linked immunosorbent assay. The levels of reactive oxygen species and apoptosis of primary kidney cells were detected by flow cytometry. The oxidative stress-related markers were detected by the cuvette assay. The TLR4/NF-κB and PI3K/Akt/Nrf2 levels and apoptosis were detected by Western blot analysis. The effects of GDC-0068 (GDC, Akt inhibitor) on Ar interposed on LPS-induced NRK-52e cell apoptosis were investigated by flow cytometry. RESULTS The data collected by network pharmacology suggested that Ar might inhibit AKI by exerting an anti-inflammatory effect and regulating the Akt signaling pathway. The experimental results showed that Ar markedly improved renal function, and attenuated inflammation and cell apoptosis via regulating PI3K/Akt/Nrf2 pathway following LPS challenge in vivo, which blocked by GDC effectively in vitro. CONCLUSION In a word, this study demonstrated that Ar attenuated LPS-induced AKI by inhibiting inflammation and apoptosis via the PI3K/Akt/Nrf2 pathway.
Collapse
Affiliation(s)
- Beibei Zhang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Mengnan Zeng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Benke Li
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Yuxuan Kan
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Shengchao Wang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Bing Cao
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Yanjie Huang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Xiaoke Zheng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China.
| | - Weisheng Feng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China.
| |
Collapse
|
27
|
Butler B, Nguyen TT, Williams A, Tucker A, Proud WG, Brown KA. Use of a Shock Tube Platform in the Replication of Blast Lung Injury. EPJ WEB OF CONFERENCES 2021. [DOI: 10.1051/epjconf/202125001024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
War and asymmetrical conflicts are becoming increasingly prevalent in the modern world. Due to improvements in conflict medicine, survivable injuries are now more severe than they once were. Therefore, it is now more important than ever that there exist scientific and engineering methods for replicating wartime injuries in the context of the laboratory. We have developed one such method: a shock tube platform for testing ex vivo samples of the porcine respiratory system. Using this platform, we can, to some extent, simulate the pathophysiological consequences of blast lung. This is a condition commonly present in victims of explosive blasts, both those due to typical armaments and Improvised Explosive Devices (IEDs). Presented here are the results of experiments conducted using porcine bronchiole tissue as ex vivo organ cultures. Data presented show epithelial damage, consistent with known trauma-induced cell injury that can lead to acute respiratory distress syndrome (ARDS).
Collapse
|
28
|
Bösmüller H, Matter M, Fend F, Tzankov A. The pulmonary pathology of COVID-19. Virchows Arch 2021; 478:137-150. [PMID: 33604758 PMCID: PMC7892326 DOI: 10.1007/s00428-021-03053-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 01/08/2023]
Abstract
The lung is the main affected organ in severe coronavirus disease 2019 (COVID-19) caused by the novel coronavirus SARS-CoV-2, and lung damage is the leading cause of death in the vast majority of patients. Mainly based on results obtained by autopsies, the seminal features of fatal COVID-19 have been described by many groups worldwide. Early changes encompass edema, epithelial damage, and capillaritis/endothelialitis, frequently combined with microthrombosis. Subsequently, patients with manifest respiratory insufficiency exhibit exudative diffuse alveolar damage (DAD) with hyaline membrane formation and pneumocyte type 2 hyperplasia, variably complicated by superinfection, which may progress to organizing/fibrotic stage DAD. These features, however, are not specific for COVID-19 and can be found in other disorders including viral infections. Clinically, the early disease stage of severe COVID-19 is characterized by high viral load, lymphopenia, massive secretion of pro-inflammatory cytokines and hypercoagulability, documented by elevated D-dimers and an increased frequency of thrombotic and thromboembolic events, whereas virus loads and cytokine levels tend to decrease in late disease stages, when tissue repair including angiogenesis prevails. The present review describes the spectrum of lung pathology based on the current literature and the authors' personal experience derived from clinical autopsies, and tries to summarize our current understanding and open questions of the pathophysiology of severe pulmonary COVID-19.
Collapse
Affiliation(s)
- Hans Bösmüller
- Institute of Pathology and Neuropathology, University Hospital Tübingen and Eberhard Karls University Tübingen, Liebermeisterstraße 8, 72076, Tübingen, Germany
| | - Matthias Matter
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital Tübingen and Eberhard Karls University Tübingen, Liebermeisterstraße 8, 72076, Tübingen, Germany.
| | - Alexandar Tzankov
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|