1
|
Lindo J, Nogueira C, Soares R, Cunha N, Almeida MR, Rodrigues L, Coelho P, Rodrigues F, Cunha RA, Gonçalves T. Genetic Polymorphisms of P2RX7 but Not of ADORA2A Are Associated with the Severity of SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:6135. [PMID: 38892324 PMCID: PMC11173306 DOI: 10.3390/ijms25116135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
SARS-CoV-2 infection ranges from mild to severe presentations, according to the intensity of the aberrant inflammatory response. Purinergic receptors dually control the inflammatory response: while adenosine A2A receptors (A2ARs) are anti-inflammatory, ATP P2X7 receptors (P2X7Rs) exert pro-inflammatory effects. The aim of this study was to assess if there were differences in allelic and genotypic frequencies of a loss-of-function SNP of ADORA2A (rs2298383) and a gain-of-function single nucleotide polymorphism (SNP) of P2RX7 (rs208294) in the severity of SARS-CoV-2-associated infection. Fifty-five individuals were enrolled and categorized according to the severity of the infection. Endpoint genotyping was performed in blood cells to screen for both SNPs. The TT genotype (vs. CT + CC) and the T allele (vs. C allele) of P2RX7 SNP were found to be associated with more severe forms of COVID-19, whereas the association between ADORA2A SNP and the severity of infection was not significantly different. The T allele of P2RX7 SNP was more frequent in people with more than one comorbidity and with cardiovascular conditions and was associated with colorectal cancer. Our findings suggest a more prominent role of P2X7R rather than of A2AR polymorphisms in SARS-CoV-2 infection, although larger population-based studies should be performed to validate our conclusions.
Collapse
Affiliation(s)
- Jorge Lindo
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Célia Nogueira
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Rui Soares
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
- Clinical Pathology Service, Portuguese Oncology Institute of Coimbra, 3004-011 Coimbra, Portugal;
| | - Nuno Cunha
- Clinical Pathology Service, Portuguese Oncology Institute of Coimbra, 3004-011 Coimbra, Portugal;
| | - Maria Rosário Almeida
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Lisa Rodrigues
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Patrícia Coelho
- IPCB/ESALD—Instituto Politécnico de Castelo Branco, Escola Superior de Saúde Dr. Lopes Dias, SPRINT-IPCB—Sport Physical Activity and Health Research & Innovation Center, 6000-767 Castelo Branco, Portugal; (P.C.); (F.R.)
| | - Francisco Rodrigues
- IPCB/ESALD—Instituto Politécnico de Castelo Branco, Escola Superior de Saúde Dr. Lopes Dias, SPRINT-IPCB—Sport Physical Activity and Health Research & Innovation Center, 6000-767 Castelo Branco, Portugal; (P.C.); (F.R.)
| | - Rodrigo A. Cunha
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Teresa Gonçalves
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
2
|
Figarella K, Kim J, Ruan W, Mills T, Eltzschig HK, Yuan X. Hypoxia-adenosine axis as therapeutic targets for acute respiratory distress syndrome. Front Immunol 2024; 15:1328565. [PMID: 38312838 PMCID: PMC10835146 DOI: 10.3389/fimmu.2024.1328565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
The human respiratory and circulatory systems collaborate intricately to ensure oxygen delivery to all cells, which is vital for ATP production and maintaining physiological functions and structures. During limited oxygen availability, hypoxia-inducible factors (HIFs) are stabilized and play a fundamental role in maintaining cellular processes for hypoxia adaptation. First discovered during investigations of erythropoietin production regulation, HIFs influence physiological and pathological processes, including development, inflammation, wound healing, and cancer. HIFs promote extracellular adenosine signaling by enhancing adenosine generation and receptor signaling, representing an endogenous feedback mechanism that curbs excessive inflammation, supports injury resolution, and enhances hypoxia tolerance. This is especially important for conditions that involve tissue hypoxia, such as acute respiratory distress syndrome (ARDS), which globally poses significant health challenges without specific treatment options. Consequently, pharmacological strategies to amplify HIF-mediated adenosine production and receptor signaling are of great importance.
Collapse
Affiliation(s)
- Katherine Figarella
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jieun Kim
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Wei Ruan
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tingting Mills
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Holger Klaus Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xiaoyi Yuan
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
3
|
Tanaka DM, Fabricio CG, Marin-Neto JA, de Barros Filho ACL, de Oliveira LFL, Mejia J, Almeida RR, de Souza Vieira R, Lopes CD, Batah SS, Moreira HT, de Lourdes Higuchi M, Neto EC, Fabro AT, Nekolla SG, Romano MMD, Simões MV. Pentoxifylline reduces inflammation and prevents myocardial perfusion derangements in experimental chronic Chagas' cardiomyopathy. J Nucl Cardiol 2023; 30:2327-2337. [PMID: 37165114 DOI: 10.1007/s12350-023-03270-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/14/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND Myocardial perfusion defect (MPD) is common in chronic Chagas cardiomyopathy (CCC) and is associated with inflammation and development of left ventricular systolic dysfunction. We tested the hypothesis that pentoxifylline (PTX) could reduce inflammation and prevent the development of MPD in a model of CCC in hamsters. METHODS AND RESULTS We investigated with echocardiogram and rest myocardial perfusion scintigraphy at baseline (6-months after T. cruzi infection/saline) and post-treatment (after additional 2-months of PTX/saline administration), female Syrian hamsters assigned to 3 groups: T. cruzi-infected animals treated with PTX (CH + PTX) or saline (CH + SLN); and uninfected control animals (CO). At the baseline, all groups showed similar left ventricular ejection fraction (LVEF) and MPD areas. At post-treatment evaluation, there was a significant increase of MPD in CH + SLN group (0.8 ± 1.6 to 9.4 ± 9.7%), but not in CH + PTX (1.9 ± 3.0% to 2.7 ± 2.7%) that exhibited MPD area similar to CO (0.0 ± 0.0% to 0.0 ± 0.0%). The LVEF decreased in both infected groups. Histological analysis showed a reduced inflammatory infiltrate in CH + PTX group (395.7 ± 88.3 cell/mm2), as compared to CH + SLN (515.1 ± 133.0 cell/mm2), but larger than CO (193.0 ± 25.7 cell/mm2). The fibrosis and TNF-α expression was higher in both infected groups. CONCLUSIONS The prolonged use of PTX is associated with positive effects, including prevention of MPD development and reduction of inflammation in the chronic hamster model of CCC.
Collapse
Affiliation(s)
| | | | - José A Marin-Neto
- Medical School of Ribeirao Preto, University of São Paulo, Sao Paulo, Brazil
| | | | | | - Jorge Mejia
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Rafael Ribeiro Almeida
- Faculty of Medicine, Heart Institute (InCor), University of Sao Paulo, Sao Paulo, Brazil
| | - Raquel de Souza Vieira
- Faculty of Medicine, Heart Institute (InCor), University of Sao Paulo, Sao Paulo, Brazil
| | - Carla Duque Lopes
- Medical School of Ribeirao Preto, University of São Paulo, Sao Paulo, Brazil
| | | | | | | | - Edecio Cunha Neto
- Faculty of Medicine, Heart Institute (InCor), University of Sao Paulo, Sao Paulo, Brazil
| | | | - Stephan G Nekolla
- Department of Nuclear Medicine at Technische Universität, Munich, Germany
| | | | - Marcus Vinícius Simões
- Medical School of Ribeirao Preto, University of São Paulo, Sao Paulo, Brazil.
- Cardiology Division, Internal Medicine Department, Hospital das Clínicas, Faculdade de Medicina de Ribeirão Preto, 3900 Bandeirantes Avenue, Ribeirão Preto, São Paulo, 14048900, Brazil.
| |
Collapse
|
4
|
Zhao CQ, Wang C, Liu MM, Cao M, Peng J, Kong DQ, Ren XT, Liu R, Hai CX, Zhang XD. Single-cell transcriptomes reveal heterogeneity of chlorine-induced mice acute lung injury and the inhibitory effect of pentoxifylline on ferroptosis. Sci Rep 2023; 13:6833. [PMID: 37100790 PMCID: PMC10131515 DOI: 10.1038/s41598-023-32093-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 03/22/2023] [Indexed: 04/28/2023] Open
Abstract
To investigate the effect of pentoxifylline (PTX) on Chlorine (Cl2)-induced acute lung injury (ALI) by single-cell RNA sequencing (scRNA-seq). Female BALB/c mice were exposed to Cl2 at 400 ppm for 15 min. H&E staining was used to observe the degree of lung injury. scRNA-seq was conducted to analysis of normal and Cl2-exposed mice lung tissues. Immunofluorescence was used to observe genes of interest. Thirty-two mice were randomly divided into four groups: Control, Cl2, Cl2+Fer-1, Cl2+PTX. TEM, WB and ELISA were used to detect ferroptosis-related indicators. The 5, 8, 10, 12, 16, 20 clusters were epithelial cells and 4, 15, 18, 19, 21 clusters were endothelial cells. Pseudo-time analysis revealed the differentiation trajectory of epithelial cells and key regulatory genes (Gclc, Bpifa1, Dnah5 and Dnah9) during the process of injury. Cell-cell communication analysis identified several important receptor-ligand complexes (Nrp1-Vegfa, Nrp2-Vegfa, Flt1-Vegfa and Flt4-Vegfa). Ferroptosis were found up-regulated in epithelial and endothelial cells by GSVA analysis. Highly expressed genes to which closely related ferroptosis were found by SCENIC analysis. PTX could significantly decrease the levels of MDA and abnormal high expression of solute carrier family 7 member 11 (SLC7A11, the key transporter of cystine) as well as increase the expression of GSH/GSSG and glutathione peroxidase 4 (GPX4) (p < 0.05). This study revealed novel molecular features of Cl2-induced ALI. PTX may be a potential specific drug by inhibiting the process of ferroptosis in epithelial and endothelial cells.
Collapse
Affiliation(s)
- Chen-Qian Zhao
- Department of Medical Experiment Center, Shaanxi University of Chinese Medicine, Xi'an, 712046, Xianyang, China
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - Chong Wang
- Department of Medical Experiment Center, Shaanxi University of Chinese Medicine, Xi'an, 712046, Xianyang, China
| | - Meng-Meng Liu
- Department of Health Service, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, China
| | - Meng Cao
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - Jie Peng
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - De-Qin Kong
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - Xiao-Ting Ren
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - Rui Liu
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - Chun-Xu Hai
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China.
| | - Xiao-di Zhang
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
5
|
Khoury W, Trus R, Chen X, Baghaie L, Clark M, Szewczuk MR, El-Diasty M. Parsimonious Effect of Pentoxifylline on Angiogenesis: A Novel Pentoxifylline-Biased Adenosine G Protein-Coupled Receptor Signaling Platform. Cells 2023; 12:cells12081199. [PMID: 37190108 DOI: 10.3390/cells12081199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Angiogenesis is the physiological process of developing new blood vessels to facilitate the delivery of oxygen and nutrients to meet the functional demands of growing tissues. It also plays a vital role in the development of neoplastic disorders. Pentoxifylline (PTX) is a vasoactive synthetic methyl xanthine derivative used for decades to manage chronic occlusive vascular disorders. Recently, it has been proposed that PTX might have an inhibitory effect on the angiogenesis process. Here, we reviewed the modulatory effects of PTX on angiogenesis and its potential benefits in the clinical setting. Twenty-two studies met the inclusion and exclusion criteria. While sixteen studies demonstrated that pentoxifylline had an antiangiogenic effect, four suggested it had a proangiogenic effect, and two other studies showed it did not affect angiogenesis. All studies were either in vivo animal studies or in vitro animal and human cell models. Our findings suggest that pentoxifylline may affect the angiogenic process in experimental models. However, there is insufficient evidence to establish its role as an anti-angiogenesis agent in the clinical setting. These gaps in our knowledge regarding how pentoxifylline is implicated in host-biased metabolically taxing angiogenic switch may be via its adenosine A2BAR G protein-coupled receptor (GPCR) mechanism. GPCR receptors reinforce the importance of research to understand the mechanistic action of these drugs on the body as promising metabolic candidates. The specific mechanisms and details of the effects of pentoxifylline on host metabolism and energy homeostasis remain to be elucidated.
Collapse
Affiliation(s)
- William Khoury
- School of Medicine, Queen's University, Kingston, ON K7L 3L4, Canada
| | - Ryan Trus
- Faculty of Arts and Science, Queen's University, Kingston, ON K7L 3N9, Canada
- School of Medicine, The Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Xingyu Chen
- School of Medicine, Queen's University, Kingston, ON K7L 3L4, Canada
| | - Leili Baghaie
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Mira Clark
- Faculty of Arts and Science, Queen's University, Kingston, ON K7L 3N9, Canada
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Myron R Szewczuk
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Mohammad El-Diasty
- Division of Cardiac Surgery, Queen's University, Kingston, ON K7L 2V7, Canada
| |
Collapse
|
6
|
Broekhuizen M, de Vries R, Smits MAW, Dik WA, Schoenmakers S, Koch BCP, Merkus D, Reiss IKM, Danser AHJ, Simons SHP, Hitzerd E. Pentoxifylline as a therapeutic option for pre-eclampsia: a study on its placental effects. Br J Pharmacol 2022; 179:5074-5088. [PMID: 35861684 PMCID: PMC9804511 DOI: 10.1111/bph.15931] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Recently pentoxifylline, a non-selective phosphodiesterase inhibitor and adenosine receptor antagonist, has attracted much interest for the treatment of the increased vascular resistance and endothelial dysfunction in pre-eclampsia. We therefore investigated the placental transfer, vascular effects and anti-inflammatory actions of pentoxifylline in healthy and pre-eclamptic human placentas. EXPERIMENTAL APPROACH The placental transfer and metabolism of pentoxifylline were studied using ex vivo placenta perfusion experiments. In wire myography experiments with chorionic plate arteries, pentoxifyllines vasodilator properties were investigated, focusing on the cGMP and cAMP pathways and adenosine receptors. Its effects on inflammatory factors were also studied in placental explants. KEY RESULTS Pentoxifylline transferred from the maternal to foetal circulation, reaching identical concentrations. The placenta metabolized pentoxifylline into its active metabolite lisofylline (M1), which was released into both circulations. In healthy placentas, pentoxifylline potentiated cAMP- and cGMP-induced vasodilation, as well as causing vasodilation by adenosine A1 antagonism and via NO synthase and PKG. Pentoxifylline also reduced inflammatory factors secretion. In pre-eclamptic placentas, we observed that its vasodilator capacity was preserved, however not via NO-PKG but likely through adenosine signalling. Pentoxifylline neither potentiated vasodilation through cAMP and cGMP, nor suppressed the release of inflammatory factors from these placentas. CONCLUSION AND IMPLICATIONS Pentoxifylline is transferred across and metabolized by the placenta. Its beneficial effects on the NO pathway and inflammation are not retained in pre-eclampsia, limiting its application in this disease, although it could be useful for other placenta-related disorders. Future studies might focus on selective A1 receptor antagonists as a new treatment for pre-eclampsia.
Collapse
Affiliation(s)
- Michelle Broekhuizen
- Division of Neonatology, Department of PaediatricsErasmus MC University Medical CenterRotterdamThe Netherlands,Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MC University Medical CenterRotterdamThe Netherlands,Division of Experimental Cardiology, Department of CardiologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Rene de Vries
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Marja A. W. Smits
- Laboratory Medical Immunology, Department of ImmunologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of ImmunologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Sam Schoenmakers
- Department of Obstetrics and GynaecologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Birgit C. P. Koch
- Department of PharmacyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of CardiologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Irwin K. M. Reiss
- Division of Neonatology, Department of PaediatricsErasmus MC University Medical CenterRotterdamThe Netherlands
| | - A. H. Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Sinno H. P. Simons
- Division of Neonatology, Department of PaediatricsErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Emilie Hitzerd
- Division of Neonatology, Department of PaediatricsErasmus MC University Medical CenterRotterdamThe Netherlands,Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MC University Medical CenterRotterdamThe Netherlands
| |
Collapse
|
7
|
Kanapeckaitė A, Mažeikienė A, Geris L, Burokienė N, Cottrell GS, Widera D. Computational pharmacology: New avenues for COVID-19 therapeutics search and better preparedness for future pandemic crises. Biophys Chem 2022; 290:106891. [PMID: 36137310 PMCID: PMC9464258 DOI: 10.1016/j.bpc.2022.106891] [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: 07/07/2022] [Revised: 09/03/2022] [Accepted: 09/04/2022] [Indexed: 01/07/2023]
Abstract
The COVID-19 pandemic created an unprecedented global healthcare emergency prompting the exploration of new therapeutic avenues, including drug repurposing. A large number of ongoing studies revealed pervasive issues in clinical research, such as the lack of accessible and organised data. Moreover, current shortcomings in clinical studies highlighted the need for a multi-faceted approach to tackle this health crisis. Thus, we set out to explore and develop new strategies for drug repositioning by employing computational pharmacology, data mining, systems biology, and computational chemistry to advance shared efforts in identifying key targets, affected networks, and potential pharmaceutical intervention options. Our study revealed that formulating pharmacological strategies should rely on both therapeutic targets and their networks. We showed how data mining can reveal regulatory patterns, capture novel targets, alert about side-effects, and help identify new therapeutic avenues. We also highlighted the importance of the miRNA regulatory layer and how this information could be used to monitor disease progression or devise treatment strategies. Importantly, our work bridged the interactome with the chemical compound space to better understand the complex landscape of COVID-19 drugs. Machine and deep learning allowed us to showcase limitations in current chemical libraries for COVID-19 suggesting that both in silico and experimental analyses should be combined to retrieve therapeutically valuable compounds. Based on the gathered data, we strongly advocate for taking this opportunity to establish robust practices for treating today's and future infectious diseases by preparing solid analytical frameworks.
Collapse
Affiliation(s)
- Austė Kanapeckaitė
- AK Consulting, Laisvės g. 7, LT 12007 Vilnius, Lithuania,Corresponding author
| | - Asta Mažeikienė
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, M. K. Čiurlionio g. 21, LT-03101 Vilnius, Lithuania
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA In Silico Medicine, University of Liège, Quartier Hôpital, Avenue de l'Hôpital 11 (B34), Liège 4000, Belgium,Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Celestijnenlaan 300C (2419), Leuven 3001, Belgium,Skeletel Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Herestraat 49 (813), Leuven 3000, Belgium
| | - Neringa Burokienė
- Clinics of Internal Diseases, Family Medicine and Oncology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, M. K. Čiurlionio str. 21/27, LT-03101 Vilnius, Lithuania
| | - Graeme S. Cottrell
- University of Reading, School of Pharmacy, Hopkins Building, Reading RG6 6UB, United Kingdom
| | - Darius Widera
- University of Reading, School of Pharmacy, Hopkins Building, Reading RG6 6UB, United Kingdom
| |
Collapse
|
8
|
Sevoflurane Dampens Acute Pulmonary Inflammation via the Adenosine Receptor A2B and Heme Oxygenase-1. Cells 2022; 11:cells11071094. [PMID: 35406657 PMCID: PMC8997763 DOI: 10.3390/cells11071094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/20/2022] [Accepted: 03/22/2022] [Indexed: 11/17/2022] Open
Abstract
Acute respiratory distress syndrome is a life-threatening disease associated with high mortality. The adenosine receptor A2B (Adora2b) provides anti-inflammatory effects, which are also associated with the intracellular enzyme heme oxygenase-1 (HO-1). Our study determined the mechanism of sevoflurane’s protective properties and investigated the link between sevoflurane and the impact of a functional Adora2b via HO-1 modulation during lipopolysaccharide (LPS)-induced lung injury. We examined the LPS-induced infiltration of polymorphonuclear neutrophils (PMNs) into the lung tissue and protein extravasation in wild-type and Adora2b−/− animals. We generated chimeric animals, to identify the impact of sevoflurane on Adora2b of hematopoietic and non-hematopoietic cells. Sevoflurane decreased the LPS-induced PMN-infiltration and diminished the edema formation in wild-type mice. Reduced PMN counts after sevoflurane treatment were detected only in chimeric mice, which expressed Adora2b exclusively on leukocytes. The Adora2b on hematopoietic and non-hematopoietic cells was required to improve the permeability after sevoflurane inhalation. Further, sevoflurane increased the protective effects of HO-1 modulation on PMN migration and microvascular permeability. These protective effects were abrogated by specific HO-1 inhibition. In conclusion, our data revealed new insights into the protective mechanisms of sevoflurane application during acute pulmonary inflammation and the link between sevoflurane and Adora2b, and HO-1 signaling, respectively.
Collapse
|
9
|
Park SJ, Choi SH, Cho YD, Kim JY, Cho HJ, Kim KH, Kim WY. Protective effects of pentoxifylline on T-cell viability under inflammatory conditions. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221120753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Introduction: Pentoxifylline (PTX) reduces the levels of pro-inflammatory cytokines; however, its effects on immune system is not well understood. The aim of this study was to investigate the effect of PTX on T cells under inflammatory conditions in co-culture with THP-1-derived macrophages. Methods: Toll-like receptor 4 (TLR4) and macrophage migration inhibitory factor (MIF) levels were measured after addition of PTX to lipopolysaccharide (LPS)-stimulated differentiated THP-1 cells. T cell viability and MIF levels were measured after PTX was added to prostaglandin E2 (PGE2)-stimulated Jurkat T-cell leukemia line. Co-culture was conducted to determine the effect of LPS-stimulated differentiated THP-1 cells that are affected by PTX on Jurkat cells. To prevent the direct effects of LPS and PTX on Jurkat cells, LPS and PTX were washed from THP-1 cells before co-culture. T cell viability and interleukin-2 (IL-2) levels were determined in Jurkat cells. Results: Increase in the MIF concentration and TLR4 expression level in differentiated THP-1 cells stimulated with LPS were reversed after PTX addition. However, PTX did not improve T cell viability in PGE2–stimulated Jurkat cells. Co-culturing Jurkat cell and LPS-stimulated differentiated THP-1 cells resulted in a decreased viability of T cells. The addition of PTX restored T cell viability to normal control levels and IL-2 expression level in Jurkat cells. Conclusion: LPS-stimulated THP-1-derived macrophages reduced the T cell viability under inflammation. However, PTX restored T cells viability and IL-2 back to normal levels. Therefore, the immunomodulatory action of PTX may be mediated by macrophage-T cell interactions.
Collapse
Affiliation(s)
- Sung-Joon Park
- Department of Emergency Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Sung-Hyuk Choi
- Department of Emergency Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Young-Duck Cho
- Department of Emergency Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Jung-Youn Kim
- Department of Emergency Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Han-Jin Cho
- Department of Emergency Medicine, Korea University Ansan Hospital, Ansan, Kyunggi-do, Korea
| | - Kyung-Hwan Kim
- Department of Emergency Medicine, Inje University Ilsanbaik Hospital, Ilsan, Kyunggi-do, Korea
| | - Won-Young Kim
- Department of Emergency Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
10
|
Pentoxifylline effects on hospitalized patients with COVID19: A randomized, double-blind clinical trial. Int Immunopharmacol 2021; 101:108227. [PMID: 34666302 PMCID: PMC8492603 DOI: 10.1016/j.intimp.2021.108227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/24/2021] [Accepted: 10/03/2021] [Indexed: 12/27/2022]
Abstract
Pentoxifylline (PTX) has broad-spectrum properties such as anti-inflammatory, anticoagulant, and antiviral effects. The aim of this study was to evaluate the efficacy and safety of PTX in hospitalized patients with COVID-19. This double-blind, placebo-controlled randomized clinical trial was conducted on hospitalized patients with COVID-19. The recruited patients were randomly (1:1) assigned to the PTX group and the placebo group. The intervention group received PTX capsules at a dose of 400 mg three times a day for 10 days along with the national regimen, including interferon plus lopinavir/ritonavir and hydroxychloroquine. The primary outcome was the improvement of clinical scores. The secondary outcomes, on the other hand, were improvement in inflammatory and oxidative stress factors and hospital complications. From a total of 102 patients who met the inclusion criteria, 72 individuals completed the study and were analyzed. No significant differences were shown in demographics and baseline clinical characteristics. Clinical scores was not significant between the two groups (P = 0.31 and 0.07 for day 5 and 11, respectively). Although the mean serum levels of interleukin-6 (IL-6) and glutathione changed significantly after 5 days in the PTX group (P = 0.03 and p = 0.04), ICU admission, intubation, and hospital stay did not differ between the two groups. The results of our study did not show any superiority of PTX over placebo in improving the clinical outcomes of patients with COVID-19. Although PTX had a beneficial effect on IL-6 and showed an acceptable safety profile, it did not offer any clinical benefit for COVID-19 complications.
Collapse
|
11
|
Zheng X, Chen D, Zhu X, Le Grange JM, Zhou L, Zhang J. Impacts of anti-inflammatory phosphodiesterase inhibitors on a murine model of chronic pulmonary inflammation. Pharmacol Res Perspect 2021; 9:e00840. [PMID: 34327862 PMCID: PMC8322673 DOI: 10.1002/prp2.840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 01/04/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) often tends to respond poorly to glucocorticoid (GC) therapy. Reduced Histone deacetylase-2 (HDAC-2) activity is an important mechanism behind this GC insensitivity. In this study, we investigated the effects of three phosphodiesterase inhibitors (PDEIs), with an anti-inflammatory propensity, on cigarette smoke (CS)-induced pulmonary inflammation and HDAC-2 activity. Male C57BL/6 mice were exposed to cigarette smoke (CS) over the course of 30 weeks. Administration of the PDEIs commenced from the 29th week and followed a schedule of once daily treatments, 5 days a week, for 2 weeks. Roflumilast (ROF) was administered intragastrically (5 mg·kg-1 ), while pentoxifylline (PTX) (10 mg·kg-1 ) and theophylline (THEO) (10 mg·kg-1 ) were administered intraperitoneally, either alone or in combination with a GC (triamcinolone acetonide or TRI, 5 mg·kg-1 , i.m., single injection). Lung morphometry, as well as the activity of HDAC-2, pro-inflammatory cytokines and reactive oxygen species (ROS) were assessed at the end of the 30-week course. CS exposure was associated with a reduction in HDAC-2 activity and the up-regulation of ROS expression. PTX, ROF, and THEO administration led to the partial restoration of HDAC-2 activity, which was favorably associated with the reduction of ROS expression. However, combining TRI to any of these PDEIs did not synergistically augment HDAC-2 activity. Inactivation of HDAC-2 due to long-term CS exposure is closely related to exaggerated oxidative stress, and this reduced HDAC-2 activity could partially be restored through the use of PDEIs. This finding provides a potential novel approach for further clinical research.
Collapse
Affiliation(s)
- Xiao‐Fang Zheng
- Department of Emergency MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Dan‐Dan Chen
- Department of Emergency MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiao‐Ling Zhu
- Department of Emergency MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jehane Michael Le Grange
- Department of Emergency MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lu‐Qian Zhou
- Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Jin‐Nong Zhang
- Department of Emergency MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
12
|
Pasquini S, Contri C, Borea PA, Vincenzi F, Varani K. Adenosine and Inflammation: Here, There and Everywhere. Int J Mol Sci 2021; 22:7685. [PMID: 34299305 PMCID: PMC8304851 DOI: 10.3390/ijms22147685] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Adenosine is a ubiquitous endogenous modulator with the main function of maintaining cellular and tissue homeostasis in pathological and stress conditions. It exerts its effect through the interaction with four G protein-coupled receptor (GPCR) subtypes referred as A1, A2A, A2B, and A3 adenosine receptors (ARs), each of which has a unique pharmacological profile and tissue distribution. Adenosine is a potent modulator of inflammation, and for this reason the adenosinergic system represents an excellent pharmacological target for the myriad of diseases in which inflammation represents a cause, a pathogenetic mechanism, a consequence, a manifestation, or a protective factor. The omnipresence of ARs in every cell of the immune system as well as in almost all cells in the body represents both an opportunity and an obstacle to the clinical use of AR ligands. This review offers an overview of the cardinal role of adenosine in the modulation of inflammation, showing how the stimulation or blocking of its receptors or agents capable of regulating its extracellular concentration can represent promising therapeutic strategies for the treatment of chronic inflammatory pathologies, neurodegenerative diseases, and cancer.
Collapse
Affiliation(s)
- Silvia Pasquini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.)
| | - Chiara Contri
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.)
| | | | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.)
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.)
| |
Collapse
|
13
|
Purinergic Regulation of Endothelial Barrier Function. Int J Mol Sci 2021; 22:ijms22031207. [PMID: 33530557 PMCID: PMC7865261 DOI: 10.3390/ijms22031207] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/10/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Increased vascular permeability is a hallmark of several cardiovascular anomalies, including ischaemia/reperfusion injury and inflammation. During both ischaemia/reperfusion and inflammation, massive amounts of various nucleotides, particularly adenosine 5'-triphosphate (ATP) and adenosine, are released that can induce a plethora of signalling pathways via activation of several purinergic receptors and may affect endothelial barrier properties. The nature of the effects on endothelial barrier function may depend on the prevalence and type of purinergic receptors activated in a particular tissue. In this review, we discuss the influence of the activation of various purinergic receptors and downstream signalling pathways on vascular permeability during pathological conditions.
Collapse
|
14
|
Cyclic AMP in human preterm infant blood is associated with increased TLR-mediated production of acute-phase and anti-inflammatory cytokines in vitro. Pediatr Res 2020; 88:717-725. [PMID: 31578034 PMCID: PMC7392158 DOI: 10.1038/s41390-019-0586-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/16/2019] [Accepted: 08/21/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Preterm infants are at high risk of infection and have distinct pathogen recognition responses. Suggested mechanisms include soluble mediators that enhance cellular levels of cAMP. The aim of this study was to assess the relationship between blood cAMP concentrations and TLR-mediated cytokine production in infants during the first month of life. METHODS Cord and serial peripheral blood samples (days of life 1-28) were obtained from a cohort of very preterm (<30 weeks' gestational age) and term human infants. Whole-blood concentrations of cAMP and FSL-1 and LPS in vitro stimulated cytokine concentrations were measured by ELISA and multiplex bead assay. RESULTS cAMP concentrations were higher in cord than in peripheral blood, higher in cord blood of female preterm infants, and lower at Days 1 and 7 in infants exposed to chorioamnionitis, even after adjusting for leukocyte counts. TLR2 and TLR4-mediated TNF-α, IL-1β, IL-6, IL-12p70, and IL-10 production in vitro increased over the first month of life in preterm infants and were positively correlated with leukocyte-adjusted cAMP levels and reduced by exposure to chorioamnionitis. CONCLUSIONS The ontogeny of blood cAMP concentrations and associations with chorioamnionitis and TLR-mediated production of cytokines suggest that this secondary messenger helps shape distinct neonatal pathogen responses in early life.
Collapse
|
15
|
Hendry BM, Stafford N, Arnold AD, Sangwaiya A, Manglam V, Rosen SD, Arnold J. Hypothesis: Pentoxifylline is a potential cytokine modulator therapeutic in COVID-19 patients. Pharmacol Res Perspect 2020; 8:e00631. [PMID: 32715661 PMCID: PMC7383088 DOI: 10.1002/prp2.631] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
We propose a new hypothesis that the established drug pentoxifylline deserves attention as a potential repurposed therapeutic for COVID-19. Pentoxifylline is an immunomodulator with anti-inflammatory properties. It is a nonselective phosphodiesterase inhibitor and through Adenosine A2A Receptor-mediated pathways reduces tumor necrosis factor alpha, interleukin 1, interleukin 6, and interferon gamma and may act to reduce tissue damage during the cytokine storm host response to SARS-CoV-2 infection. This agent has been used clinically for many years and has a favorable profile of safety and tolerability. Pre-clinical data support pentoxifylline as effective in cytokine-driven lung damage. Clinical studies of pentoxifylline in radiation and cytokine-induced lung damage in humans are positive and consistent with anti-inflammatory efficacy. Pentoxifylline is a readily available, off-patent and inexpensive drug, suitable for large-scale use including in resource-limited countries. Current trials of therapeutics are largely focused on the inhibition of viral processes. We advocate urgent randomized trials of pentoxifylline for COVID-19 as a complementary approach to target the host responses.
Collapse
Affiliation(s)
- Bruce M. Hendry
- Renal SciencesDepartment of Inflammation BiologyKing’s College LondonLondonUK
| | - Nina Stafford
- Department of GastroenterologyEaling HospitalLondon North West University Healthcare NHS TrustSouthallUK
| | - Ahran D. Arnold
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Arvind Sangwaiya
- Department of GastroenterologyEaling HospitalLondon North West University Healthcare NHS TrustSouthallUK
| | - Vijay Manglam
- Department of GastroenterologyEaling HospitalLondon North West University Healthcare NHS TrustSouthallUK
| | - Stuart D. Rosen
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Jayantha Arnold
- Department of GastroenterologyEaling HospitalLondon North West University Healthcare NHS TrustSouthallUK
| |
Collapse
|
16
|
DiNicolantonio JJ, Barroso-Aranda J. Harnessing adenosine A2A receptors as a strategy for suppressing the lung inflammation and thrombotic complications of COVID-19: Potential of pentoxifylline and dipyridamole. Med Hypotheses 2020; 143:110051. [PMID: 32650197 PMCID: PMC7330590 DOI: 10.1016/j.mehy.2020.110051] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/25/2020] [Indexed: 12/30/2022]
Abstract
Counterproductive lung inflammation and dysregulated thrombosis contribute importantly to the lethality of advanced COVID-19. Adenosine A2A receptors (A2AR), expressed by a wide range of immune cells, as well as endothelial cells and platelets, exert cAMP-mediated anti-inflammatory and anti-thrombotic effects that potentially could be highly protective in this regard. The venerable drug pentoxifylline (PTX) exerts both anti-inflammatory and antithrombotic effects that reflect its ability to boost the responsiveness of A2AR to extracellular adenosine. The platelet-stabilizing drug dipyridamole (DIP) blocks intracellular uptake of extracellularly-generated adenosine, thereby up-regulating A2AR signaling in a way that should be functionally complementary to the impact of PTX in that regard. Moreover, DIP has recently been reported to slow the cellular replication of SARS-CoV-2 in clinically feasible concentrations. Both PTX and DIP are reasonably safe, well-tolerated, widely available, and inexpensive drugs. When COVID-19 patients can be treated within several days of symptom onset, using PTX + DIP in conjunction with hydroxychloroquine (HCQ) and an antibiotic – azithromycin (AZM) or doxycycline – might be warranted. HCQ and AZM can suppress SARS-CoV-2 proliferation in vitro and may slow the cell-to-cell spread of the virus; a large case series evaluating this combination in early-stage patients reported an impressively low mortality rate. However, whereas HCQ and AZM can promote QT interval lengthening and may be contraindicated in more advanced COVID-19 entailing cardiac damage, doxycycline has no such effect and exerts a potentially beneficial anti-inflammatory action. In contrast to HCQ, we propose that the combination of PTX + DIP can be used in both early and advanced stages of COVID-19. Concurrent use of certain nutraceuticals – yeast beta-glucan, zinc, vitamin D, spirulina, phase 2 inducers, N-acetylcysteine, glucosamine, quercetin, and magnesium – might also improve therapeutic outcomes in COVID-19.
Collapse
|
17
|
Effendi WI, Nagano T, Kobayashi K, Nishimura Y. Focusing on Adenosine Receptors as a Potential Targeted Therapy in Human Diseases. Cells 2020; 9:E785. [PMID: 32213945 PMCID: PMC7140859 DOI: 10.3390/cells9030785] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Adenosine is involved in a range of physiological and pathological effects through membrane-bound receptors linked to G proteins. There are four subtypes of adenosine receptors, described as A1AR, A2AAR, A2BAR, and A3AR, which are the center of cAMP signal pathway-based drug development. Several types of agonists, partial agonists or antagonists, and allosteric substances have been synthesized from these receptors as new therapeutic drug candidates. Research efforts surrounding A1AR and A2AAR are perhaps the most enticing because of their concentration and affinity; however, as a consequence of distressing conditions, both A2BAR and A3AR levels might accumulate. This review focuses on the biological features of each adenosine receptor as the basis of ligand production and describes clinical studies of adenosine receptor-associated pharmaceuticals in human diseases.
Collapse
Affiliation(s)
- Wiwin Is Effendi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
- Department of Pulmonology and Respiratory Medicine, Medical Faculty of Airlangga University, Surabaya 60131, Indonesia
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
| | - Kazuyuki Kobayashi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
| |
Collapse
|
18
|
Systemic Administration of Calea pinnatifida Inhibits Inflammation Induced by Carrageenan in a Murine Model of Pulmonary Neutrophilia. Mediators Inflamm 2020; 2020:4620251. [PMID: 32410853 PMCID: PMC7204167 DOI: 10.1155/2020/4620251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/04/2020] [Indexed: 12/23/2022] Open
Abstract
Objective The aim of this study was to investigate the anti-inflammatory effects of the crude extract (CE), derived fraction, and isolated compounds from Calea pinnatifida leaves in a mouse model of pulmonary neutrophilia. Methods The CE and derived fractions, hexane, ethyl acetate, and methanol, were obtained from C. pinnatifida leaves. The compounds 3,5- and 4,5-di-O-E-caffeoylquinic acids were isolated from the EtOAc fraction using chromatography and were identified using infrared spectroscopic data and nuclear magnetic resonance (1H and 13C NMR). Leukocytes count, protein concentration of the exudate, myeloperoxidase (MPO) and adenosine deaminase (ADA), and nitrate/nitrite (NO x ), tumor necrosis factor-alpha (TNF-α), interleukin-1-beta (IL-1β), and interleukin-17A (IL-17A) levels were determined in the pleural fluid leakage after 4 h of pleurisy induction. We also analyzed the effects of isolated compounds on the phosphorylation of both p65 and p38 in the lung tissue. Results The CE, its fractions, and isolated compounds inhibited leukocyte activation, protein concentration of the exudate, and MPO, ADA, NO x , TNF-α, IL-1β, and IL-17A levels. 3,5- and 4,5-di-O-E-caffeoylquinic acids also inhibited phosphorylation of both p65 and p38 (P < 0.05). Conclusion This study demonstrated that C. pinnatifida presents important anti-inflammatory properties by inhibiting activated leukocytes and protein concentration of the exudate. These effects were related to the inhibition of proinflammatory mediators. The dicaffeoylquinic acids may be partially responsible for these anti-inflammatory properties through the inhibition of nuclear transcription factor kappa B and mitogen-activated protein kinase pathways.
Collapse
|
19
|
Vanderstocken G, Dvorkin-Gheva A, Shen P, Brandsma CA, Obeidat M, Bossé Y, Hassell JA, Stampfli MR. Identification of Drug Candidates to Suppress Cigarette Smoke-induced Inflammation via Connectivity Map Analyses. Am J Respir Cell Mol Biol 2019; 58:727-735. [PMID: 29256623 DOI: 10.1165/rcmb.2017-0202oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cigarette smoking is the main risk factor for chronic obstructive pulmonary disease, and to date, existing pharmacologic interventions have been ineffective at controlling inflammatory processes associated with the disease. To address this issue, we used the Connectivity Map (cMap) database to identify drug candidates with the potential to attenuate cigarette smoke-induced inflammation. We queried cMap using three independent in-house cohorts of healthy nonsmokers and smokers. Potential drug candidates were validated against four publicly available human datasets, as well as six independent datasets from cigarette smoke-exposed mice. Overall, these analyses yielded two potential drug candidates: kaempferol and bethanechol. Subsequently, the efficacy of each drug was validated in vivo in a model of cigarette smoke-induced inflammation. BALB/c mice were exposed to room air or cigarette smoke and treated with each of the two candidate drugs either prophylactically or therapeutically. We found that kaempferol, but not bethanechol, was able to reduce cigarette smoke-induced neutrophilia, both when administered prophylactically and when administered therapeutically. Mechanistically, kaempferol decreased expression of IL-1α and CXCL5 concentrations in the lung. Our data suggest that cMap analyses may serve as a useful tool to identify novel drug candidates against cigarette smoke-induced inflammation.
Collapse
Affiliation(s)
- Gilles Vanderstocken
- 1 Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre
| | - Anna Dvorkin-Gheva
- 1 Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre.,2 Department of Pathology and Molecular Medicine, Centre for Functional Genomics, and
| | - Pamela Shen
- 1 Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre
| | - Corry-Anke Brandsma
- 3 Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ma'en Obeidat
- 4 The University of British Columbia Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Yohan Bossé
- 5 Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec City, Québec, Canada; and.,6 Department of Molecular Medicine, Laval University, Québec City, Québec, Canada
| | - John A Hassell
- 2 Department of Pathology and Molecular Medicine, Centre for Functional Genomics, and
| | - Martin R Stampfli
- 1 Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre.,7 Department of Medicine, Firestone Institute for Respiratory Health at St. Joseph's Healthcare, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
20
|
Konrad FM, Zwergel C, Ngamsri KC, Reutershan J. Anti-inflammatory Effects of Heme Oxygenase-1 Depend on Adenosine A 2A- and A 2B-Receptor Signaling in Acute Pulmonary Inflammation. Front Immunol 2017; 8:1874. [PMID: 29326725 PMCID: PMC5742329 DOI: 10.3389/fimmu.2017.01874] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/08/2017] [Indexed: 12/16/2022] Open
Abstract
Acute pulmonary inflammation is still a frightening complication in intensive care units. In our previous study, we determined that heme oxygenase (HO)-1 had anti-inflammatory effects in pulmonary inflammation. Recent literature has emphasized a link between HO-1 and the nucleotide adenosine. Since adenosine A2A- and A2B-receptors play a pivotal role in pulmonary inflammation, we investigated their link to the enzyme HO-1. In a murine model of pulmonary inflammation, the activation of HO-1 by hemin significantly decreased polymorphonuclear leukocyte (PMN) migration into the lung. This anti-inflammatory reduction of PMN migration was abolished in A2A- and A2B-knockout mice. Administration of hemin significantly reduced chemokine levels in the BAL of wild-type animals but had no effects in A2A-/- and A2B-/- mice. Microvascular permeability was significantly attenuated in HO-1-stimulated wild-type mice, but not in A2A-/- and A2B-/- mice. The activity of HO-1 rose after LPS inhalation in wild-type animals and, surprisingly, also in A2A-/- and A2B-/- mice after the additional administration of hemin. Immunofluorescence images of animals revealed alveolar macrophages to be the major source of HO-1 activity in both knockout strains—in contrast to wild-type animals, where HO-1 was also significantly augmented in the lung tissue. In vitro studies on PMN migration further confirmed our in vivo findings. In conclusion, we linked the anti-inflammatory effects of HO-1 to functional A2A/A2B-receptor signaling under conditions of pulmonary inflammation. Our findings may explain why targeting HO-1 in acute pulmonary inflammation has failed to prove effective in some patients, since septic patients have altered adenosine receptor expression.
Collapse
Affiliation(s)
- Franziska M Konrad
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Constantin Zwergel
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Kristian-Christos Ngamsri
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Jörg Reutershan
- Department of Anesthesiology and Intensive Care Medicine, Hospital of Bayreuth, Bayreuth, Germany
| |
Collapse
|
21
|
Chen YM, Chiang WC, Lin SL, Tsai TJ. Therapeutic efficacy of pentoxifylline on proteinuria and renal progression: an update. J Biomed Sci 2017; 24:84. [PMID: 29132351 PMCID: PMC5683556 DOI: 10.1186/s12929-017-0390-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/30/2017] [Indexed: 01/17/2023] Open
Abstract
Blood pressure control with renin-angiotensin system (RAS) blockade has remained the gold standard for treating patients with proteinuric chronic kidney disease (CKD) up to date. Nevertheless, RAS blockade slows but does not halt the progression of kidney disease, thus highlighting the need to search for additional therapeutic approaches. The nonselective phosphodiesterase (PDE) inhibitor pentoxifylline (PTX) is an old drug that exhibits prominent anti-inflammatory, anti-proliferative and anti-fibrotic activities both in vitro and in vivo. Studies in human subjects have shown that PTX monotherapy decreases urinary protein excretion, and add-on therapy of PTX to background RAS blockade additively reduces proteinuria in patients with CKD of various etiology. More recent studies find that PTX combined with RAS blockade delays the decline of glomerular filtration rate in diabetic patients with mild to moderate CKD, and reduces the risk of end-stage renal disease in diabetic and non-diabetic patients in late stage of CKD with high proteinuria levels. In this review, we update the clinical trial results of PTX as monotherapy, or in conjunction or in comparison with RAS blockade on patients with proteinuria and CKD, and propose a mechanistic scheme explaining the renoprotective activities of this drug.
Collapse
Affiliation(s)
- Yung-Ming Chen
- Renal Division, Department of Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei, Taiwan
| | - Wen-Chih Chiang
- Renal Division, Department of Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei, Taiwan
| | - Shuei-Liong Lin
- Renal Division, Department of Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei, Taiwan
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, Section 1, Taipei, Taiwan
| | - Tun-Jun Tsai
- Renal Division, Department of Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei, Taiwan
| |
Collapse
|
22
|
Inhibition of SDF-1 receptors CXCR4 and CXCR7 attenuates acute pulmonary inflammation via the adenosine A 2B-receptor on blood cells. Cell Death Dis 2017; 8:e2832. [PMID: 28542132 PMCID: PMC5520683 DOI: 10.1038/cddis.2016.482] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 11/10/2016] [Accepted: 12/16/2016] [Indexed: 12/13/2022]
Abstract
Acute pulmonary inflammation is characterized by migration of polymorphonuclear neutrophils into the different compartments of the lung. Recent studies showed evidence that the chemokine stromal cell-derived factor (SDF)-1 and its receptors CXCR4 and CXCR7 influence migration of immune cells and their activity was linked to adenosine concentrations. We investigated the particular role of CXCR4- and CXCR7-inhibition and the potential link to the adenosine A2B-receptor, which plays an important anti-inflammatory role in the lung. After LPS-inhalation for 45 minutes, administration of the CXCR4-inhibitor (AMD3100) decreased transendothelial and transepithelial migration, whereas CXCR7-antagonism influenced epithelial migration exclusively. In A2B−/− mice, no anti-inflammatory effects were detectible through either one of the agents. Using chimeric mice, we identified A2B on hematopoietic cells to be crucial for these anti-inflammatory effects of CXCR4/7-inhibition. Both inhibitors decreased TNFα, IL6, CXCL1 and CXCL2/3 levels in the bronchoalveolar lavage of wild type mice, while not influencing the chemokine release in A2B−/− mice. Inflammation augmented the expression of both receptors and their inhibition increased A2B-levels upon inflammation. In vitro assays with human epithelium/endothelium confirmed our in vivo findings. During inflammation, inhibition of CXCR4- and CXCR7-receptors prevented microvascular permeability in wild type but not in A2B−/− mice, highlighting the pivotal role of an active A2B-receptor in this setting. The combination of both inhibitors had a synergistic effect in preventing capillary leakage. In conclusion, we determined the pivotal role of CXCR4- and CXCR7-inhibition in acute pulmonary inflammation, which depended on A2B-receptor signalling.
Collapse
|
23
|
Borea PA, Gessi S, Merighi S, Varani K. Adenosine as a Multi-Signalling Guardian Angel in Human Diseases: When, Where and How Does it Exert its Protective Effects? Trends Pharmacol Sci 2016; 37:419-434. [PMID: 26944097 DOI: 10.1016/j.tips.2016.02.006] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 12/20/2022]
Abstract
The importance of adenosine for human health cannot be overstated. Indeed, this ubiquitous nucleoside is an integral component of ATP, and regulates the function of every tissue and organ in the body. Acting via receptor-dependent and -independent mechanisms [the former mediated via four G-protein-coupled receptors (GPCRs), A1, A2A, A2B, and A3,], it has a significant role in protecting against cell damage in areas of increased tissue metabolism, and combating organ dysfunction in numerous pathological states. Accordingly, raised levels of adenosine have been demonstrated in epilepsy, ischaemia, pain, inflammation, and cancer, in which its behaviour can be likened to that of a guardian angel, even though there are instances in which overproduction of adenosine is pathological. In this review, we condense the current body of knowledge on the issue, highlighting when, where, and how adenosine exerts its protective effects in both the brain and the periphery.
Collapse
Affiliation(s)
- Pier Andrea Borea
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, Via Fossato di Mortara, 17-19, 44121 Ferrara, Italy.
| | - Stefania Gessi
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, Via Fossato di Mortara, 17-19, 44121 Ferrara, Italy.
| | - Stefania Merighi
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, Via Fossato di Mortara, 17-19, 44121 Ferrara, Italy.
| | - Katia Varani
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, Via Fossato di Mortara, 17-19, 44121 Ferrara, Italy
| |
Collapse
|
24
|
Pulmonary Protection Strategies in Cardiac Surgery: Are We Making Any Progress? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:416235. [PMID: 26576223 PMCID: PMC4630421 DOI: 10.1155/2015/416235] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/12/2015] [Indexed: 01/19/2023]
Abstract
Pulmonary dysfunction is a common complication of cardiac surgery. The mechanisms involved in the development of pulmonary dysfunction are multifactorial and can be related to the activation of inflammatory and oxidative stress pathways. Clinical manifestation varies from mild atelectasis to severe respiratory failure. Managing pulmonary dysfunction postcardiac surgery is a multistep process that starts before surgery and continues during both the operative and postoperative phases. Different pulmonary protection strategies have evolved over the years; however, the wide acceptance and clinical application of such techniques remain hindered by the poor level of evidence or the sample size of the studies. A better understanding of available modalities and/or combinations can result in the development of customised strategies for the different cohorts of patients with the potential to hence maximise patients and institutes benefits.
Collapse
|
25
|
The unrecognized effects of phosphodiesterase 4 on epithelial cells in pulmonary inflammation. PLoS One 2015; 10:e0121725. [PMID: 25909327 PMCID: PMC4409344 DOI: 10.1371/journal.pone.0121725] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/03/2015] [Indexed: 12/27/2022] Open
Abstract
Acute pulmonary inflammation is characterized by migration of polymorphonuclear neutrophils (PMNs) into the different compartments of the lung, passing an endothelial and epithelial barrier. Recent studies showed evidence that phosphodiesterase (PDE)4-inhibitors stabilized endothelial cells. PDE4B and PDE4D subtypes play a pivotal role in inflammation, whereas blocking PDE4D is suspected to cause gastrointestinal side effects. We thought to investigate the particular role of the PDE4-inhibitors roflumilast and rolipram on lung epithelium. Acute pulmonary inflammation was induced by inhalation of LPS. PDE4-inhibitors were administered i.p. or nebulized after inflammation. The impact of PDE4-inhibitors on PMN migration was evaluated in vivo and in vitro. Microvascular permeability, cytokine levels, and PDE4B and PDE4D expression were analyzed. In vivo, both PDE4-inhibitors decreased transendothelial and transepithelial migration even when administered after inflammation, whereas roflumilast showed a superior effect compared to rolipram on the epithelium. Both inhibitors decreased TNFα, IL6, and CXCL2/3. CXCL1, the strong PMN chemoattractant secreted by the epithelium, was significantly more reduced by roflumilast. In vitro assays with human epithelium also emphasized the pivotal role of roflumilast on the epithelium. Additionally, LPS-induced stress fibers, an essential requirement for a direct migration of PMNs into the alveolar space, were predominantly reduced by roflumilast. Expression of PDE4B and PDE4D were both increased in the lungs by LPS, PDE4-inhibitors decreased mainly PDE4B. The topical administration of PDE4-inhibitors was also effective in curbing down PMN migration, further highlighting the clinical potential of these compounds. In pulmonary epithelial cells, both subtypes were found coexistent around the nucleus and the cytoplasm. In these epithelial cells, LPS increased PDE4B and, to a lesser extend, PDE4D, whereas the effect of the inhibitors was prominent on the PDE4B subtype. In conclusion, we determined the pivotal role of the PDE4-inhibitor roflumilast on lung epithelium and emphasized its main effect on PDE4B in hyperinflammation.
Collapse
|
26
|
Anti-inflammatory, antioxidant, and antiparkinsonian effects of adenosine A 2A receptor antagonists. Pharmacol Biochem Behav 2015; 132:71-78. [PMID: 25735490 DOI: 10.1016/j.pbb.2015.02.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 02/18/2015] [Accepted: 02/20/2015] [Indexed: 11/21/2022]
Abstract
The purpose of the study was to examine derivatives of annelated xanthines (imidazo-, pyrimido-, and diazepino-purinediones) for potential anti-inflammatory effects in carrageenan-induced paw edema in mice. Additionally, their antioxidant activity using the FRAP (ferric-reducing ability of plasma) assay and lipid peroxidation in rat brain homogenate were analyzed. All the studied derivatives showed affinity for adenosine A2A receptor. The preliminary assays found that five (KD-114, KD-57, KD-129, KD-50, and KD-358) pyrimidopurinedione derivatives, administered intraperitoneally (i.p.) at a dose of 100mg/kg, had stronger anti-inflammatory effects. At a concentration of 10-5M, three of the derivatives KD-57, KD-114, and KD-129 most influenced the total antioxidant ability. The most efficient anti-inflammatory compound, KD-114, also showed the strongest binding to A2A receptors and when administered at a dose of 5mg/kg (i.p.), effectively reversed haloperidol-induced catalepsy and significantly increased the striatal extracellular dopamine level in the rat striatum. This effect was weaker than the one produced by CSC (1mg/kg i.p.), and only slightly weaker than that produced by ZM 241385 (3mg/kg i.p.) used as reference drugs. From the results of the present studies, it may be concluded that anti-inflammatory and antiparkinsonian effects of the examined compounds correlate with their influence on adenosine A2A receptors, the most probable antagonism to these subtype receptors.
Collapse
|
27
|
Konrad FM, Braun S, Ngamsri KC, Vollmer I, Reutershan J. Heme oxygenase-1 attenuates acute pulmonary inflammation by decreasing the release of segmented neutrophils from the bone marrow. Am J Physiol Lung Cell Mol Physiol 2014; 307:L707-17. [DOI: 10.1152/ajplung.00145.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recruiting polymorphonuclear neutrophil granulocytes (PMNs) from circulation and bone marrow to the site of inflammation is one of the pivotal mechanisms of the innate immune system. During inflammation, the enzyme heme oxygenase 1 (HO-1) has been shown to reduce PMN migration. Although these effects have been described in various models, underlying mechanisms remain elusive. Recent studies revealed an influence of HO-1 on different cells of the bone marrow. We investigated the particular role of the bone marrow in terms of HO-1-dependent pulmonary inflammation. In a murine model of LPS inhalation, stimulation of HO-1 by cobalt (III) protoporphyrin-IX-chloride (CoPP) resulted in reduced segmented PMN migration into the alveolar space. In the CoPP group, segmented PMNs were also decreased intravascularly, and concordantly, mature and immature PMN populations were higher in the bone marrow. Inhibition of the enzyme by tin protoporphyrin-IX increased segmented and banded PMN migration into the bronchoalveolar lavage fluid with enhanced PMN release from the bone marrow and aggravated parameters of tissue inflammation. Oxidative burst activity was significantly higher in immature compared with mature PMNs. The chemokine stromal-derived factor-1 (SDF-1), which mediates homing of leukocytes into the bone marrow and is decreased in inflammation, was increased by CoPP. When SDF-1 was blocked by the specific antagonist AMD3100, HO-1 activation was no longer effective in curbing PMN trafficking to the inflamed lungs. In conclusion, we show evidence that the anti-inflammatory effects of HO-1 are largely mediated by inhibiting the release of segmented PMNs from the bone marrow rather than direct effects within the lung.
Collapse
Affiliation(s)
- Franziska M. Konrad
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Stefan Braun
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Kristian-Christos Ngamsri
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Irene Vollmer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Jörg Reutershan
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, Tübingen, Germany
| |
Collapse
|
28
|
Koppe SWP. Obesity and the liver: nonalcoholic fatty liver disease. Transl Res 2014; 164:312-22. [PMID: 25028077 DOI: 10.1016/j.trsl.2014.06.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 02/08/2023]
Abstract
The increasing prevalence of nonalcoholic fatty liver disease (NAFLD) parallels the rise of obesity and its complications. NAFLD is a common cause of cirrhosis and a leading indication for liver transplant. Genetic susceptibility, dietary composition, and exercise habits influence the development of NAFLD, and insulin resistance results in widespread metabolic perturbations with a net effect of triglyceride accumulation in the liver. Some patients will develop hepatocyte cellular injury and fibrosis of the liver, which can progress to cirrhosis and require liver transplant. Treatments targeting the pathophysiological mechanisms of NAFLD exist, but carry some potential risk and are not universally effective. Weight loss and lifestyle changes remain the most effective and safest approach, but sustainable change is difficult for most patients to achieve. Future work will continue to focus on developing effective and safe interventions to prevent the development of advanced liver disease, whereas efforts in the public health domain continue to combat obesity.
Collapse
Affiliation(s)
- Sean W P Koppe
- Division of Gastroenterology and Hepatology, Northwestern University, Chicago, Ill.
| |
Collapse
|
29
|
Antonioli L, Csóka B, Fornai M, Colucci R, Kókai E, Blandizzi C, Haskó G. Adenosine and inflammation: what's new on the horizon? Drug Discov Today 2014; 19:1051-68. [DOI: 10.1016/j.drudis.2014.02.010] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 02/06/2014] [Accepted: 02/25/2014] [Indexed: 12/18/2022]
|
30
|
Gonzales JN, Gorshkov B, Varn MN, Zemskova MA, Zemskov EA, Sridhar S, Lucas R, Verin AD. Protective effect of adenosine receptors against lipopolysaccharide-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 2014; 306:L497-507. [PMID: 24414256 DOI: 10.1152/ajplung.00086.2013] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Acute lung injury and acute respiratory distress syndrome (ALI/ARDS) affect 200,000 people a year in the USA. Pulmonary vascular and specifically endothelial cell (EC) barrier compromise is a hallmark of these diseases. We have recently shown that extracellular adenosine enhances human pulmonary (EC) barrier via activation of adenosine receptors (ARs) in cell cultures. On the basis of these data, we hypothesized that activation of ARs might exert barrier-protective effects in a model of ALI/ARDS in mice. To test this hypothesis, we examined the effects of pre- and posttreatment of adenosine and 5'-N-ethylcarboxamidoadenosine (NECA), a nonselective stable AR agonist, on LPS-induced lung injury. Mice were given vehicle or LPS intratracheally followed by adenosine, NECA, or vehicle instilled via the internal jugular vein. Postexperiment cell counts, Evans Blue Dye albumin (EBDA) extravasation, levels of proteins, and inflammatory cytokines were analyzed. Harvested lungs were used for histology and myeloperoxidase studies. Mice challenged with LPS alone demonstrated an inflammatory response typical of ALI. Cell counts, EBDA extravasation, as well as levels of proteins and inflammatory cytokines were decreased in adenosine-treated mice. Histology displayed reduced infiltration of neutrophils. NECA had a similar effect on LPS-induced vascular barrier compromise. Importantly, posttreatment with adenosine or NECA recovers lung vascular barrier and reduces inflammation induced by LPS challenge. Furthermore, adenosine significantly attenuated protein degradation of A2A and A3 receptors induced by LPS. Collectively, our results demonstrate that activation of ARs protects and restores vascular barrier functions and reduces inflammation in LPS-induced ALI.
Collapse
Affiliation(s)
- Joyce N Gonzales
- Assistant Prof. of Medicine, Div. of Pulmonary and Critical Care Medicine, Georgia Regents Univ., Rm. BBR-5513, 1120 15th St., Augusta, GA 30912.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Lee CF, Lai HL, Lee YC, Chien CL, Chern Y. The A2A adenosine receptor is a dual coding gene: a novel mechanism of gene usage and signal transduction. J Biol Chem 2013; 289:1257-70. [PMID: 24293369 DOI: 10.1074/jbc.m113.509059] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The A2A adenosine receptor (A2AR) is a G protein-coupled receptor and a major target of caffeine. The A2AR gene encodes alternative transcripts that are initiated from at least two independent promoters. The different transcripts of the A2AR gene contain the same coding region and 3'-untranslated region and different 5'-untranslated regions that are highly conserved among species. We report here that in addition to the production of the A2AR protein, translation from an upstream, out-of-frame AUG of the rat A2AR gene produces a 134-amino acid protein (designated uORF5). An anti-uORF5 antibody recognized a protein of the predicted size of uORF5 in PC12 cells and rat brains. Up-regulation of A2AR transcripts by hypoxia led to increased levels of both the A2AR and uORF5 proteins. Moreover, stimulation of A2AR increased the level of the uORF5 protein via post-transcriptional regulation. Expression of the uORF5 protein suppressed the AP1-mediated transcription promoted by nerve growth factor and modulated the expression of several proteins that were implicated in the MAPK pathway. Taken together, our results show that the rat A2AR gene encodes two distinct proteins (A2AR and uORF5) in an A2AR-dependent manner. Our study reveals a new example of the complexity of the mammalian genome and provides novel insights into the function of A2AR.
Collapse
Affiliation(s)
- Chien-fei Lee
- From the Institute of Neuroscience, School of Life Sciences, National Yang Ming University, Taipei 112, Taiwan
| | | | | | | | | |
Collapse
|