1
|
Eldien HMS, Almaeen AH, El Fath AA, Taha AE, Ahmed R, Elfadil H, Hetta HF. Unlocking the Potential of RNA Sequencing in COVID-19: Toward Accurate Diagnosis and Personalized Medicine. Diagnostics (Basel) 2025; 15:229. [PMID: 39857114 PMCID: PMC11763845 DOI: 10.3390/diagnostics15020229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/08/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
COVID-19 has caused widespread morbidity and mortality, with its effects extending to multiple organ systems. Despite known risk factors for severe disease, including advanced age and underlying comorbidities, patient outcomes can vary significantly. This variability complicates efforts to predict disease progression and tailor treatment strategies. While diagnostic and therapeutic approaches are still under debate, RNA sequencing (RNAseq) has emerged as a promising tool to provide deeper insights into the pathophysiology of COVID-19 and guide personalized treatment. A comprehensive literature review was conducted using PubMed, Scopus, Web of Science, and Google Scholar. We employed Medical Subject Headings (MeSH) terms and relevant keywords to identify studies that explored the role of RNAseq in COVID-19 diagnostics, prognostics, and therapeutics. RNAseq has proven instrumental in identifying molecular biomarkers associated with disease severity in patients with COVID-19. It allows for the differentiation between asymptomatic and symptomatic individuals and sheds light on the immune response mechanisms that contribute to disease progression. In critically ill patients, RNAseq has been crucial for identifying key genes that may predict patient outcomes, guiding therapeutic decisions, and assessing the long-term effects of the virus. Additionally, RNAseq has helped in understanding the persistence of viral RNA after recovery, offering new insights into the management of post-acute sequelae, including long COVID. RNA sequencing significantly improves COVID-19 management, particularly for critically ill patients, by enhancing diagnostic accuracy, personalizing treatment, and predicting therapeutic responses. It refines patient stratification, improving outcomes, and holds promise for targeted interventions in both acute and long COVID.
Collapse
Affiliation(s)
- Heba M. Saad Eldien
- Department of Anatomy, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
| | - Abdulrahman H. Almaeen
- Department of Pathology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Ahmed Abo El Fath
- Tropical Medicine and Gastroenterology Department, Assiut University Hospital, Assiut 71515, Egypt;
| | - Ahmed E. Taha
- Microbiology and Immunology Unit, Department of Pathology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia;
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Rehab Ahmed
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| | - Hassabelrasoul Elfadil
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| | - Helal F. Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| |
Collapse
|
2
|
Yadav S, Gowda S, Agrawal-Rajput R. CSF-1R blockade to alleviate azithromycin mediated immunosuppression in a mouse model of intracellular infection. Int Immunopharmacol 2024; 143:113477. [PMID: 39476565 DOI: 10.1016/j.intimp.2024.113477] [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: 07/21/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024]
Abstract
Colony Stimulating Factor-1 Receptor (CSF-1R) signalling plays an important role in maturation, differentiation and activation of macrophages. Apposite generation and activation of macrophage phenotypes and subsequent adaptive immune response against any infection is decisive for a positive disease outcome. Antibiotic therapy is imperative for treating bacterial infections however antibiotics have off-target effects on host immune-cells. These effects could either be contextually beneficial or harmful and could potentially aid generation of infection persistence and antimicrobial resistance (AMR) via host immunosuppression. We had recently reported the immunosuppressive-mechanism of azithromycin-induced increased CSF-1R expression on murine-macrophages and bacterial-persistence in Balb/c model of intracellular infection. We further wanted to explore the molecular-mechanism behind these observations and tested GW2580-mediated CSF-1R blockade before azithromycin treatment during S. flexneri induced intracellular infection. In the presented study, we report that the azithromycin alters the protein expression or phosphorylation of transcription-factors ERK1/2, P38, AKT1, STAT3, STAT6, and EGR2 that are involved in macrophage polarisatoin and also take part in CSF-1R signalling pathways. Intrestingly, CSF-1R blockade using GW2580 abrogated or reversed the azithromycin-induced up- or down-regulated expression or phosphorylation of ERK1/2, P38, AKT1, STAT3, STAT6, and EGR2. We further validated our results in Balb/c model of S. flexneri infection. Intrestingly, the CSF-1R blocker and azithromycin treated mice showed batter recovery than the azithromycin alone treated mice and hence we report the aftermath of GW2580 with azithromycin treatment on disease and immunological outcome of an intracellular infection caused by Shigella flexneri.
Collapse
Affiliation(s)
- Shivani Yadav
- Department of Biotechnology and Bioengineering, Immunology Lab, Indian Institute of Advanced Research, Gandhinagar 382421, Gujarat, India
| | - Sharath Gowda
- Department of Biotechnology and Bioengineering, Immunology Lab, Indian Institute of Advanced Research, Gandhinagar 382421, Gujarat, India
| | - Reena Agrawal-Rajput
- Department of Biotechnology and Bioengineering, Immunology Lab, Indian Institute of Advanced Research, Gandhinagar 382421, Gujarat, India.
| |
Collapse
|
3
|
Gusovsky A, Slade E, Forrest JM, Henderson D, Abdel-Latif A, Venditto VJ, Delcher C, Feola DJ. Azithromycin Exposure in a 10-Day Window of Myocardial Infarction and Short- and Long-Term Outcomes. JACC. ADVANCES 2024; 3:101337. [PMID: 39469610 PMCID: PMC11513797 DOI: 10.1016/j.jacadv.2024.101337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/30/2024]
Abstract
Background The U.S. Food and Drug Administration warned in 2012 that azithromycin (AZM) can cause potentially fatal irregular heart rhythm, particularly in patients with known cardiac risk factors. Objectives This study aimed to examine cardiac and hospital readmission outcomes associated with AZM exposure near the time of a myocardial infarction (MI). Methods This was a retrospective cohort study using Merative MarketScan databases examining adult inpatients admitted with MI from January 1, 2010 to December 31, 2017. Patients with AZM exposure 7 days pre-MI to 3 days post-MI were compared to unexposed controls. Time to subsequent MI and incident heart failure (HF) were examined up to 5 years post-MI using Cox models. All-cause, MI-related, MI and sequelae-related readmissions and incident HF diagnosis were examined 30 days post-MI using logistic regression. Results There were 18,066 eligible patients in the full cohort (AZM, N = 3,011), and the HF-free at baseline cohort included 9,180 patients (AZM, N = 1,530). Probability of subsequent MI up to 5 years post-MI was 15.3% in the AZM group vs 9.7% in control (HR: 1.41 [95% CI: 1.10-1.81], P = 0.0076). Probability of incident HF was 39.8% in the AZM group vs 35.5% in control (HR: 1.12 [95% CI: 0.91-1.39], P = 0.2795). Odds of all 4 30-day outcomes were significantly higher in the AZM group vs control. Conclusions We found an increased risk of long-term subsequent MI, 30-day hospital readmissions, and 30-day incident HF among MI patients with AZM exposure compared to controls. Our findings are consistent with the 2012 Food and Drug Administration warning.
Collapse
Affiliation(s)
- Amanda Gusovsky
- Institute for Pharmaceutical Outcomes & Policy, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Emily Slade
- Department of Biostatistics, University of Kentucky College of Public Health, Lexington, Kentucky, USA
| | - Jasmine M. Forrest
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, Kentucky, USA
| | - Darren Henderson
- University of Kentucky, Center for Clinical and Translational Science, Lexington, Kentucky, USA
| | - Ahmed Abdel-Latif
- Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Vincent J. Venditto
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Chris Delcher
- Institute for Pharmaceutical Outcomes & Policy, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - David J. Feola
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| |
Collapse
|
4
|
Ahangari F, Soudi S, Ghaffari Khaligh S, Mirsanei Z, Soufihasanabad S, Ebadi Asl P, Mahmoud Hashemi S. Combinational therapy of mesenchymal stem cell-derived extracellular vesicles and azithromycin improves clinical and histopathological recovery in CLP sepsis model. Int Immunopharmacol 2024; 139:112732. [PMID: 39053229 DOI: 10.1016/j.intimp.2024.112732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Sepsis is a syndrome that occurs following an infection and marked by severe inflammatory responses, and if not treated in time, it can lead to multi-organ failure syndrome and death. This study examines the effects of a novel combination therapy using azithromycin and mesenchymal stem cell-derived extracellular vesicles (EVs) on a cecal ligation and puncture (CLP) model of sepsis. METHODS Human Wharton's jelly-mesenchymal stem cells were cultured, characterized, and used to extract EVs. The CLP sepsis model was induced in mice, followed by treatments: saline, AZM, EVs, and combination therapy (A+E). Clinical sepsis scores were recorded 24 h post-treatment. Serum, peritoneal fluid, and organ tissues (kidney, liver, lung) were collected and analyzed for biochemical parameters (AST ALT, and creatinine), inflammatory markers, bacterial load, and histopathological changes. RESULTS The A+E combined treatment improved the clinical scores of septic mice. The administration of A+E reduced bacterial loads in the peritoneum of septic mice, contributing to effective control of infection. Inflammatory markers of neutrophils-to-lymphocytes ratio (NLR) and TNF-α serum levels were significantly lower in the combinational therapy group, indicating significant anti-inflammatory effect of this combination. Additionally, combination of AZM and EVs alleviated organ damage mainly within liver, kidneys and lungs. Based on histopathological assessments and biochemical parameters, there was diminished tissue damage as well as reduced inflammation, which is correlated with improved functions of these vital organs. CONCLUSION The combined use of azithromycin and EVs offers a promising therapeutic approach for sepsis by effectively controlling infection and modulating the inflammatory response.
Collapse
Affiliation(s)
- Fatemeh Ahangari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Zahra Mirsanei
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Soufihasanabad
- Department of Animal Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Pedram Ebadi Asl
- Department of Medical Lab Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Melis R, Braca A, Pagnozzi D, Anedda R. The metabolic footprint of Vero E6 cells highlights the key metabolic routes associated with SARS-CoV-2 infection and response to drug combinations. Sci Rep 2024; 14:7950. [PMID: 38575586 PMCID: PMC10995198 DOI: 10.1038/s41598-024-57726-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/21/2024] [Indexed: 04/06/2024] Open
Abstract
SARS-CoV-2 burdens healthcare systems worldwide, yet specific drug-based treatments are still unavailable. Understanding the effects of SARS-CoV-2 on host molecular pathways is critical for providing full descriptions and optimizing therapeutic targets. The present study used Nuclear Magnetic Resonance-based metabolic footprinting to characterize the secreted cellular metabolite levels (exometabolomes) of Vero E6 cells in response to SARS-CoV-2 infection and to two candidate drugs (Remdesivir, RDV, and Azithromycin, AZI), either alone or in combination. SARS-CoV-2 infection appears to force VE6 cells to have increased glucose concentrations from extra-cellular medium and altered energetic metabolism. RDV and AZI, either alone or in combination, can modify the glycolic-gluconeogenesis pathway in the host cell, thus impairing the mitochondrial oxidative damage caused by the SARS-CoV-2 in the primary phase. RDV treatment appears to be associated with a metabolic shift toward the TCA cycle. Our findings reveal a metabolic reprogramming produced by studied pharmacological treatments that protects host cells against virus-induced metabolic damage, with an emphasis on the glycolytic-gluconeogenetic pathway. These findings may help researchers better understand the relevant biological mechanisms involved in viral infection, as well as the creation of mechanistic hypotheses for such candidate drugs, thereby opening up new possibilities for SARS-CoV-2 pharmacological therapy.
Collapse
Affiliation(s)
- Riccardo Melis
- Porto Conte Ricerche s.r.l., S.P. 55 Porto Conte-Capo Caccia, Km 8.400 Loc. Tramariglio, Alghero, SS, Italy
| | - Angela Braca
- Porto Conte Ricerche s.r.l., S.P. 55 Porto Conte-Capo Caccia, Km 8.400 Loc. Tramariglio, Alghero, SS, Italy
| | - Daniela Pagnozzi
- Porto Conte Ricerche s.r.l., S.P. 55 Porto Conte-Capo Caccia, Km 8.400 Loc. Tramariglio, Alghero, SS, Italy
| | - Roberto Anedda
- Porto Conte Ricerche s.r.l., S.P. 55 Porto Conte-Capo Caccia, Km 8.400 Loc. Tramariglio, Alghero, SS, Italy.
| |
Collapse
|
6
|
Rodríguez-Moreno CB, Cañeque-Rufo H, Flor-García M, Terreros-Roncal J, Moreno-Jiménez EP, Pallas-Bazarra N, Bressa C, Larrosa M, Cafini F, Llorens-Martín M. Azithromycin preserves adult hippocampal neurogenesis and behavior in a mouse model of sepsis. Brain Behav Immun 2024; 117:135-148. [PMID: 38211636 PMCID: PMC7615685 DOI: 10.1016/j.bbi.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/11/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024] Open
Abstract
The mammalian hippocampus can generate new neurons throughout life. Known as adult hippocampal neurogenesis (AHN), this process participates in learning, memory, mood regulation, and forgetting. The continuous incorporation of new neurons enhances the plasticity of the hippocampus and contributes to the cognitive reserve in aged individuals. However, the integrity of AHN is targeted by numerous pathological conditions, including neurodegenerative diseases and sustained inflammation. In this regard, the latter causes cognitive decline, mood alterations, and multiple AHN impairments. In fact, the systemic administration of Lipopolysaccharide (LPS) from E. coli to mice (a model of sepsis) triggers depression-like behavior, impairs pattern separation, and decreases the survival, maturation, and synaptic integration of adult-born hippocampal dentate granule cells. Here we tested the capacity of the macrolide antibiotic azithromycin to neutralize the deleterious consequences of LPS administration in female C57BL6J mice. This antibiotic exerted potent neuroprotective effects. It reversed the increased immobility time during the Porsolt test, hippocampal secretion of pro-inflammatory cytokines, and AHN impairments. Moreover, azithromycin promoted the synaptic integration of adult-born neurons and functionally remodeled the gut microbiome. Therefore, our data point to azithromycin as a clinically relevant drug with the putative capacity to ameliorate the negative consequences of chronic inflammation by modulating AHN and hippocampal-related behaviors.
Collapse
Affiliation(s)
- Carla B Rodríguez-Moreno
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Héctor Cañeque-Rufo
- Department of Chemistry and Biochemistry, School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - Miguel Flor-García
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - Julia Terreros-Roncal
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - Elena P Moreno-Jiménez
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - Noemí Pallas-Bazarra
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Carlo Bressa
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Ctra. Pozuelo-Majadahonda Km 1,800, 28223, Pozuelo de Alarcón, Madrid
| | - Mar Larrosa
- Department of Food Science and Nutrition, Faculty of Pharmacy, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Fabio Cafini
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain.
| | - María Llorens-Martín
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
7
|
Ao D, He X, Liu J, Xu L. Strategies for the development and approval of COVID-19 vaccines and therapeutics in the post-pandemic period. Signal Transduct Target Ther 2023; 8:466. [PMID: 38129394 PMCID: PMC10739883 DOI: 10.1038/s41392-023-01724-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/24/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
The spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in significant casualties and put immense strain on public health systems worldwide, leading to economic recession and social unrest. In response, various prevention and control strategies have been implemented globally, including vaccine and drug development and the promotion of preventive measures. Implementing these strategies has effectively curbed the transmission of the virus, reduced infection rates, and gradually restored normal social and economic activities. However, the mutations of SARS-CoV-2 have led to inevitable infections and reinfections, and the number of deaths continues to rise. Therefore, there is still a need to improve existing prevention and control strategies, mainly focusing on developing novel vaccines and drugs, expediting medical authorization processes, and keeping epidemic surveillance. These measures are crucial to combat the Coronavirus disease (COVID-19) pandemic and achieve sustained, long-term prevention, management, and disease control. Here, we summarized the characteristics of existing COVID-19 vaccines and drugs and suggested potential future directions for their development. Furthermore, we discussed the COVID-19-related policies implemented over the past years and presented some strategies for the future.
Collapse
Affiliation(s)
- Danyi Ao
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China
| | - Jian Liu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China
| | - Li Xu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
8
|
Mustafa T, Niazi MRK, Lakdawala Z, Mirza S. Regional and National Trends in Consumption of Antimicrobials in Pakistan; Pre and Post-COVID (2019-2021). Clin Infect Dis 2023; 77:S569-S577. [PMID: 38118009 PMCID: PMC10732562 DOI: 10.1093/cid/ciad647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023] Open
Abstract
BACKGROUND Efforts to combat antimicrobial resistance, a growing public health problem in Pakistan, have been hampered by the lack of high-quality national and provincial-level antimicrobial consumption data. The singular objective of this retrospective study was to measure antimicrobial consumption over 3 years between 2019 and 2021. METHODS The study was designed to estimate antimicrobial consumption at National and Regional levels. Antimicrobial consumption data was collected by IQVIA covering 110 districts of Pakistan in which 88% of sales are census (accurate sales collected directly from distributors), whereas 12% of sales (sales of 300 pharmacies) are projected on the national level. To determine the usage for 3 consecutive years, the consumption of antibiotics was calculated as defined daily doses (DDD) of antibiotics per 1000 inhabitants per day (DID). RESULTS The results of our study demonstrated a steep increase in the consumption of antimicrobials from 2019 to 2021. An increase in consumption of most classes of antibiotics was observed both nationally and Regionally. Quinolones, penicillins (co-amoxiclav), macrolides, and third-generation cephalosporins remained the most frequently used antibiotics nationally. A 40% increase in intravenous use of antimicrobials was observed between 2019 and 2021 at the national level. Moxifloxacin, Levofloxacin, Ciprofloxacin, and linezolid were the most commonly used intravenous antibiotics. Region 7 (Peshawar) demonstrated the highest consumption, followed by Region 1 (Karachi) and Region 6 (Faisalabad). Among the most commonly used antibiotics, the use of third-generation cephalosporin (cefixime), quinolones, penicillins (amoxicillin + clavulanic acid), and macrolides (azithromycin) was most noticeable in all regions, particularly in those with the higher consumption of antibiotics. CONCLUSIONS Although the increase in consumption of all antibiotics is concerning, the steep increase in the use of watch and reserve category antibiotics during the study period calls for immediate actions to limit and regulate their usage.
Collapse
Affiliation(s)
| | | | - Zahra Lakdawala
- Numerical Yield and Site Assessment Group, Fraunhofer Institute for Wind Energy Systems, Oldenburg, Germany
| | - Shaper Mirza
- Department of Life Sciences, SBASSE-LUMS, Lahore, Pakistan
| |
Collapse
|
9
|
Petcharat K, Munkong N, Thongboontho R, Chartarrayawadee W, Thim-Uam A. Synergistic Effects of Azithromycin and STING Agonist Promote IFN-I Production by Enhancing the Activation of STING-TBK1 Signaling. J Exp Pharmacol 2023; 15:407-421. [PMID: 37933302 PMCID: PMC10625772 DOI: 10.2147/jep.s433181] [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: 08/25/2023] [Accepted: 10/28/2023] [Indexed: 11/08/2023] Open
Abstract
Background Azithromycin (AZM) is a macrolide antibiotic that exhibits anti-inflammatory and anti-viral infection properties by enhancing type-I interferon (IFN-I) responses. The stimulator of interferon genes (STING) can directly induce IFN-I production. However, elevated IFN-I induces auto-immune phenotypes such as systemic lupus erythematosus (SLE). The effects of AZM and STING on the production of IFN-I are unclear. Objective Therefore, this study aims to evaluate the role of AZM and STING on IFN-I responses in macrophages. Methods RAW 264.7 macrophages were treated with AZM with and without a STING-agonist (DMXAA), and the maturation of macrophages was determined using flow cytometry. Gene expression and pro-inflammatory cytokines were analyzed using qPCR and ELISA, respectively. Moreover, protein expression was investigated using Western blot assays and immunofluorescence. Results Our results show that AZM significantly induced M1 phenotypes, promoting surface molecule expansion of CD80 and MHC-II and production of IL-6 and TNF-α cytokines on DMXAA-stimulated macrophages. Furthermore, we found that AZM-increased mRNA levels of interferon-stimulated genes (ISGs) could be due to the high expression of STNG-TBK1 signaling in the presence of DMXAA. Conclusion Our data suggest that AZM enhancement of IFN-I responses was STING dependent in DMXAA-stimulated macrophages. These data underline a novel approach to AZM action-mediated STING-TBK1 signaling for regulating IFN-I responses and may further augment the scientific basis and potential use of AZM in clinical applications.
Collapse
Affiliation(s)
- Kanoktip Petcharat
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, 56000, Thailand
| | - Narongsuk Munkong
- Department of Pathology, School of Medicine, University of Phayao, Phayao, 56000, Thailand
| | - Rungthip Thongboontho
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, 56000, Thailand
| | | | - Arthid Thim-Uam
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, 56000, Thailand
| |
Collapse
|
10
|
Bigelow RA, Richeson JT, McClurg M, Valeris-Chacin R, Morley PS, Funk JL, Scott MA. Characterizing the influence of various antimicrobials used for metaphylaxis against bovine respiratory disease on host transcriptome responses. Front Vet Sci 2023; 10:1272940. [PMID: 37869487 PMCID: PMC10585045 DOI: 10.3389/fvets.2023.1272940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023] Open
Abstract
Currently, control against bovine respiratory disease (BRD) primarily consists of mass administration of an antimicrobial upon arrival to facility, termed "metaphylaxis." The objective of this study was to determine the influence of six different antimicrobials used as metaphylaxis on the whole blood host transcriptome in healthy steers upon and following arrival to the feedlot. One hundred and five steers were stratified by arrival body weight (BW = 247 ± 28 kg) and randomly and equally allocated to one of seven treatments: negative control (NC), ceftiofur (CEFT), enrofloxacin (ENRO), florfenicol (FLOR), oxytetracycline (OXYT), tildipirosin (TILD), or tulathromycin (TULA). On day 0, whole blood samples and BW were collected prior to a one-time administration of the assigned antimicrobial. Blood samples were collected again on days 3, 7, 14, 21, and 56. A subset of cattle (n = 6) per treatment group were selected randomly for RNA sequencing across all time points. Isolated RNA was sequenced (NovaSeq 6,000; ~35 M paired-end reads/sample), where sequenced reads were processed with ARS-UCD1.3 reference-guided assembly (HISAT2/StringTie2). Differential expression analysis comparing treatment groups to NC was performed with glmmSeq (FDR ≤ 0.05) and edgeR (FDR ≤ 0.1). Functional enrichment was performed with KOBAS-i (FDR ≤ 0.05). When compared only to NC, unique differentially expressed genes (DEGs) found within both edgeR and glmmSeq were identified for CEFT (n = 526), ENRO (n = 340), FLOR (n = 56), OXYT (n = 111), TILD (n = 3,001), and TULA (n = 87). At day 3, CEFT, TILD, and OXYT shared multiple functional enrichment pathways related to T-cell receptor signaling and FcεRI-mediated NF-kappa beta (kB) activation. On day 7, Class I major histocompatibility complex (MHC)-mediated antigen presentation pathways were enriched in ENRO and CEFT groups, and CEFT and FLOR had DEGs that affected IL-17 signaling pathways. There were no shared pathways or Gene Ontology (GO) terms among treatments at day 14, but TULA had 19 pathways and eight GO terms enriched related to NF- κβ activation, and interleukin/interferon signaling. Pathways related to cytokine signaling were enriched by TILD on day 21. Our research demonstrates immunomodulation and potential secondary therapeutic mechanisms induced by antimicrobials commonly used for metaphylaxis, providing insight into the beneficial anti-inflammatory properties antimicrobials possess.
Collapse
Affiliation(s)
- Rebecca A. Bigelow
- Department of Agricultural Sciences, West Texas A&M University, Canyon, TX, United States
| | - John T. Richeson
- Department of Agricultural Sciences, West Texas A&M University, Canyon, TX, United States
| | - Molly McClurg
- Veterinary, Education, Research, and Outreach Program, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, Canyon, TX, United States
| | - Robert Valeris-Chacin
- Veterinary, Education, Research, and Outreach Program, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, Canyon, TX, United States
| | - Paul S. Morley
- Veterinary, Education, Research, and Outreach Program, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, Canyon, TX, United States
| | - Jenna L. Funk
- Veterinary, Education, Research, and Outreach Program, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, Canyon, TX, United States
| | - Matthew A. Scott
- Veterinary, Education, Research, and Outreach Program, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, Canyon, TX, United States
| |
Collapse
|
11
|
Uematsu T, Takano T, Matsui H, Kobayashi N, Ōmura S, Hanaki H. Prophylactic administration of ivermectin attenuates SARS-CoV-2 induced disease in a Syrian Hamster Model. J Antibiot (Tokyo) 2023; 76:481-488. [PMID: 37185581 PMCID: PMC10127164 DOI: 10.1038/s41429-023-00623-0] [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: 01/05/2023] [Revised: 03/22/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023]
Abstract
COVID-19, caused by SARS-CoV-2 infection, is currently among the most important public health concerns worldwide. Although several effective vaccines have been developed, there is an urgent clinical need for effective pharmaceutical treatments for treatment of COVID-19. Ivermectin, a chemical derivative of avermectin produced by Streptomyces avermitilis, is a macrocyclic lactone with antiparasitic activity. Recent studies have shown that ivermectin inhibits SARS-CoV-2 replication in vitro. In the present study, we investigated the in vivo effects of ivermectin in a hamster model of SARS-CoV-2 infection. The results of the present study demonstrate oral administration of ivermectin prior to SARS-CoV-2 infection in hamsters was associated with decreased weight loss and pulmonary inflammation. In addition, the administration of ivermectin reduced pulmonary viral titers and mRNA expression level of pro-inflammatory cytokines associated with severe COVID-19 disease. The administration of ivermectin rapidly induced the production of virus-specific neutralizing antibodies in the late stage of viral infection. Zinc concentrations leading to immune quiescence were also significantly higher in the lungs of ivermectin-treated hamsters compared to controls. These results indicate that ivermectin may have efficacy in reducing the development and severity of COVID-19 by affecting host immunity in a hamster model of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Takayuki Uematsu
- Biomedical Laboratory, Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Saitama, Japan.
| | - Tomomi Takano
- Laboratory of Veterinary Infectious Disease, Department of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Hidehito Matsui
- Infection Control Research Center, Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo, Japan
| | - Noritada Kobayashi
- Biomedical Laboratory, Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Saitama, Japan
| | - Satoshi Ōmura
- Drug Discovery Project from Natural Products, Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo, Japan
| | - Hideaki Hanaki
- Infection Control Research Center, Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo, Japan
| |
Collapse
|
12
|
Parveen S, Ali MS, Al-Lohedan HA, Hoti N, Tabassum S. Molecular interaction of lysozyme with therapeutic drug azithromycin: Effect of sodium dodecyl sulfate on binding profile. Int J Biol Macromol 2023; 242:124844. [PMID: 37210056 DOI: 10.1016/j.ijbiomac.2023.124844] [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: 01/11/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/22/2023]
Abstract
This paper describes an inclusive biophysical study elucidating the interaction of therapeutic drug azithromycin (Azith) with hen egg white lysozyme (HEWL). Spectroscopic and computational tools have been employed to study the interaction of Azith with HEWL at pH 7.4. The fluorescence quenching constant values (Ksv) exhibited a decrease with the increase in temperature which revealed the occurrence of static quenching mechanism between Azith and HEWL. The thermodynamic data demonstrated that hydrophobic interactions were predominantly involved in the Azith-HEWL interaction. The negative value of standard Gibbs free energy (ΔG°) stated that the Azith-HEWL complex formed via spontaneous molecular interactions. The effect of sodium dodecyl sulfate (SDS) surfactant monomers on the binding propensity of Azith with HEWL was insignificant at lower concentrations however the binding significantly decreased at increased concentrations of the former. Far-UV CD data revealed alteration in the secondary structure of HEWL in the presence of Azith and the overall HEWL conformation changed. Molecular docking results revealed that the binding of Azith with HEWL takes place through hydrophobic interactions and hydrogen bonds.
Collapse
Affiliation(s)
- Sabiha Parveen
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Mohd Sajid Ali
- Department of Chemistry, College of Sciences, King Saud University, Riyadh 11451, the Kingdom of Saudi Arabia
| | - Hamad A Al-Lohedan
- Department of Chemistry, College of Sciences, King Saud University, Riyadh 11451, the Kingdom of Saudi Arabia
| | | | - Sartaj Tabassum
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
13
|
Toussi SS, Hammond JL, Gerstenberger BS, Anderson AS. Therapeutics for COVID-19. Nat Microbiol 2023; 8:771-786. [PMID: 37142688 DOI: 10.1038/s41564-023-01356-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 03/09/2023] [Indexed: 05/06/2023]
Abstract
Vaccines and monoclonal antibody treatments to prevent severe coronavirus disease 2019 (COVID-19) illness were available within a year of the pandemic being declared but there remained an urgent need for therapeutics to treat patients who were not vaccinated, were immunocompromised or whose vaccine immunity had waned. Initial results for investigational therapies were mixed. AT-527, a repurposed nucleoside inhibitor for hepatitis C virus, enabled viral load reduction in a hospitalized cohort but did not reduce viral load in outpatients. The nucleoside inhibitor molnupiravir prevented death but failed to prevent hospitalization. Nirmatrelvir, an inhibitor of the main protease (Mpro), co-dosed with the pharmacokinetic booster ritonavir, reduced hospitalization and death. Nirmatrelvir-ritonavir and molnupiravir received an Emergency Use Authorization in the United States at the end of 2021. Immunomodulatory drugs such as baricitinib, tocilizumab and corticosteroid, which target host-driven COVID-19 symptoms, are also in use. We highlight the development of COVID-19 therapies and the challenges that remain for anticoronavirals.
Collapse
|
14
|
Čačić A, Amidžić Klarić D, Keser S, Radiković M, Rukavina Z, Jøraholmen MW, Uzelac L, Kralj M, Škalko-Basnet N, Šegvić Klarić M, Vanić Ž. A Novel Approach for the Treatment of Aerobic Vaginitis: Azithromycin Liposomes-in-Chitosan Hydrogel. Pharmaceutics 2023; 15:pharmaceutics15051356. [PMID: 37242598 DOI: 10.3390/pharmaceutics15051356] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/22/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Biocompatible mucoadhesive formulations that enable a sustained drug delivery at the site of action, while exhibiting inherent antimicrobial activity, are of great importance for improved local therapy of vaginal infections. The aim of this research was to prepare and evaluate the potential of the several types of azithromycin (AZM)-liposomes (180-250 nm) incorporated into chitosan hydrogel (AZM-liposomal hydrogels) for the treatment of aerobic vaginitis. AZM-liposomal hydrogels were characterized for in vitro release, and rheological, texture, and mucoadhesive properties under conditions simulating the vaginal site of application. The role of chitosan as a hydrogel-forming polymer with intrinsic antimicrobial properties was explored against several bacterial strains typical for aerobic vaginitis as well as its potential effect on the anti-staphylococcal activity of AZM-liposomes. Chitosan hydrogel prolonged the release of the liposomal drug and exhibited inherent antimicrobial activity. Additionally, it boosted the antibacterial effect of all tested AZM-liposomes. All AZM-liposomal hydrogels were biocompatible with the HeLa cells and demonstrated mechanical properties suitable for vaginal application, thus confirming their potential for enhanced local therapy of aerobic vaginitis.
Collapse
Affiliation(s)
- Ana Čačić
- Microbiology and Biology Laboratory, PLIVA Croatia Ltd., Prilaz Baruna Filipovića 25, 10000 Zagreb, Croatia
| | - Daniela Amidžić Klarić
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10000 Zagreb, Croatia
| | - Sabina Keser
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10000 Zagreb, Croatia
| | - Maja Radiković
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10000 Zagreb, Croatia
| | - Zora Rukavina
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10000 Zagreb, Croatia
| | - May Wenche Jøraholmen
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø The Arctic University of Norway, Universitetsveien 57, 5037 Tromsø, Norway
| | - Lidija Uzelac
- Department of Molecular Medicine, Ruđer Bošković Institute, Bijenička Cesta 54, 10000 Zagreb, Croatia
| | - Marijeta Kralj
- Department of Molecular Medicine, Ruđer Bošković Institute, Bijenička Cesta 54, 10000 Zagreb, Croatia
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø The Arctic University of Norway, Universitetsveien 57, 5037 Tromsø, Norway
| | - Maja Šegvić Klarić
- Department of Microbiology, Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10000 Zagreb, Croatia
| | - Željka Vanić
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10000 Zagreb, Croatia
| |
Collapse
|
15
|
Tsalik EL, Rouphael NG, Sadikot RT, Rodriguez-Barradas MC, McClain MT, Wilkins DM, Woods CW, Swamy GK, Walter EB, El Sahly HM, Keitel WA, Mulligan MJ, Tuyishimire B, Serti E, Hamasaki T, Evans SR, Ghazaryan V, Lee MS, Lautenbach E. Efficacy and safety of azithromycin versus placebo to treat lower respiratory tract infections associated with low procalcitonin: a randomised, placebo-controlled, double-blind, non-inferiority trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:484-495. [PMID: 36525985 PMCID: PMC10040424 DOI: 10.1016/s1473-3099(22)00735-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Lower respiratory tract infections are frequently treated with antibiotics, despite a viral cause in many cases. It remains unknown whether low procalcitonin concentrations can identify patients with lower respiratory tract infection who are unlikely to benefit from antibiotics. We aimed to compare the efficacy and safety of azithromycin versus placebo to treat lower respiratory tract infections in patients with low procalcitonin. METHODS We conducted a randomised, placebo-controlled, double-blind, non-inferiority trial at five health centres in the USA. Adults aged 18 years or older with clinically suspected non-pneumonia lower respiratory tract infection and symptom duration from 24 h to 28 days were eligible for enrolment. Participants with a procalcitonin concentration of 0·25 ng/mL or less were randomly assigned (1:1), in blocks of four with stratification by site, to receive over-encapsulated oral azithromycin 250 mg or matching placebo (two capsules on day 1 followed by one capsule daily for 4 days). Participants, non-study clinical providers, investigators, and study coordinators were masked to treatment allocation. The primary outcome was efficacy of azithromycin versus placebo in terms of clinical improvement at day 5 in the intention-to-treat population. The non-inferiority margin was -12·5%. Solicited adverse events (abdominal pain, vomiting, diarrhoea, allergic reaction, or yeast infections) were recorded as a secondary outcome. This trial is registered with ClinicalTrials.gov, NCT03341273. FINDINGS Between Dec 8, 2017, and March 9, 2020, 691 patients were assessed for eligibility and 499 were enrolled and randomly assigned to receive azithromycin (n=249) or placebo (n=250). Clinical improvement at day 5 was observed in 148 (63%, 95% CI 54 to 71) of 238 participants with full data in the placebo group and 155 (69%, 61 to 77) of 227 participants with full data in the azithromycin group in the intention-to-treat analysis (between-group difference -6%, 95% CI -15 to 2). The 95% CI for the difference did not meet the non-inferiority margin. Solicited adverse events and the severity of solicited adverse events were not significantly different between groups at day 5, except for increased abdominal pain associated with azithromycin (47 [23%, 95% CI 18 to 29] of 204 participants) compared with placebo (35 [16%, 12 to 21] of 221; between-group difference -7% [95% CI -15 to 0]; p=0·066). INTERPRETATION Placebo was not non-inferior to azithromycin in terms of clinical improvement at day 5 in adults with lower respiratory tract infection and a low procalcitonin concentration. After accounting for both the rates of clinical improvement and solicited adverse events at day 5, it is unclear whether antibiotics are indicated for patients with lower respiratory tract infection and a low procalcitonin concentration. FUNDING National Institute of Allergy and Infectious Diseases, bioMérieux.
Collapse
Affiliation(s)
- Ephraim L Tsalik
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Center for Applied Genomics and Precision Medicine, Duke University School of Medicine, Durham, NC, USA; Emergency Medicine Service, Durham VA Health Care System, Durham, NC, USA.
| | - Nadine G Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Ruxana T Sadikot
- Atlanta VA Health Care System, Atlanta, GA, USA; Medical Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maria C Rodriguez-Barradas
- Infectious Diseases Section, Michael E DeBakey VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Micah T McClain
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Center for Applied Genomics and Precision Medicine, Duke University School of Medicine, Durham, NC, USA; Medical Service, Durham VA Health Care System, Durham, NC, USA
| | | | - Christopher W Woods
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Center for Applied Genomics and Precision Medicine, Duke University School of Medicine, Durham, NC, USA; Medical Service, Durham VA Health Care System, Durham, NC, USA
| | - Geeta K Swamy
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA; Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Emmanuel B Walter
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA; Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Hana M El Sahly
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Wendy A Keitel
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mark J Mulligan
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA; Division of Infectious Diseases and Immunology, NYU Langone Health, New York, NY, USA
| | | | | | - Toshimitsu Hamasaki
- Biostatistics Center, Milken Institute School of Public Health, George Washington University, Rockville, MD, USA; Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville, MD, USA
| | - Scott R Evans
- Biostatistics Center, Milken Institute School of Public Health, George Washington University, Rockville, MD, USA; Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville, MD, USA
| | - Varduhi Ghazaryan
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Marina S Lee
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Ebbing Lautenbach
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
16
|
Rofael SAD, Brown J, Lipman MCI, Lowe DM, Spratt D, Quaderi S, Hurst JR, McHugh TD. Impact of prophylactic and 'rescue pack' antibiotics on the airway microbiome in chronic lung disease. BMJ Open Respir Res 2023; 10:10/1/e001335. [PMID: 37085283 PMCID: PMC10124267 DOI: 10.1136/bmjresp-2022-001335] [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: 06/14/2022] [Accepted: 03/24/2023] [Indexed: 04/23/2023] Open
Abstract
The management of many chronic lung diseases involves multiple antibiotic prescriptions either to treat acute exacerbations or as prophylactic therapy to reduce the frequency of exacerbations and improve patients' quality of life. AIM To investigate the effects of antibiotics on the homeostasis of bacterial communities in the airways, and how this may contribute to antimicrobial resistance (AMR) among respiratory pathogens and microbiota. METHODS Within an observational cohort study, sputum was collected from 84 patients with chronic obstructive pulmonary disease and/or bronchiectasis at stable state: 47 were receiving antibiotic prophylaxis therapy. V3-V4 16S-rRNA sequencing on Illumina MiSeq, quantitative PCR for typical respiratory pathogens, bacteriology cultures and antimicrobial susceptibility testing of sputum isolates, resistome analysis on a subset of 17 sputum samples using MinION metagenomics sequencing were performed. FINDING The phylogenetic α-diversity and the total bacterial density in sputum were significantly lower in patients receiving prophylactic antibiotics (p=0.014 and 0.029, respectively). Antibiotic prophylaxis was associated with significantly lower relative abundance of respiratory pathogens such as Pseudomonas aeruginosa, Moraxella catarrhalis and members of family Enterobacteriaceae in the airway microbiome, but not Haemophilus influenzae and Streptococcus pneumoniae. No major definite directional shifts in the microbiota composition were identified with prophylactic antibiotic use at the cohort level. Surveillance of AMR and resistome analysis revealed a high frequency of resistance to macrolide and tetracycline in the cohort. AMR expressed by pathogenic bacterial isolates was associated with antibiotics prescribed as 'rescue packs' for prompt initiation of self-treatment of exacerbations (Spearman's rho=0.408, p=0.02). CONCLUSIONS Antibiotic prophylactic therapy suppresses recognised pathogenic bacteria in the sputum of patients with chronic lung disease. The use of antibiotic rescue packs may be driving AMR in this cohort rather than prophylactic antibiotics.
Collapse
Affiliation(s)
- Sylvia A D Rofael
- UCL Centre for Clinical Microbiology, Division of Infection & Immunity, University College London, London, UK
- Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - James Brown
- UCL Respiratory, Divison of Medicine, University College London, London, UK
- Respiratory Medicine, Royal Free NHS Foundation Trust, London, UK
| | - Marc C I Lipman
- UCL Respiratory, Divison of Medicine, University College London, London, UK
- Respiratory Medicine, Royal Free NHS Foundation Trust, London, UK
| | - David M Lowe
- Institute for Immunity and Transplantation, Divison of Infection and Immunity, University College London, London, UK
| | - David Spratt
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, UK
| | - Shumonta Quaderi
- UCL Respiratory, Divison of Medicine, University College London, London, UK
- Respiratory Medicine, Royal Free NHS Foundation Trust, London, UK
| | - John R Hurst
- UCL Respiratory, Divison of Medicine, University College London, London, UK
- Respiratory Medicine, Royal Free NHS Foundation Trust, London, UK
| | - Timothy D McHugh
- UCL Centre for Clinical Microbiology, Division of Infection & Immunity, University College London, London, UK
| |
Collapse
|
17
|
Fernandez-Guzman D, Caira-Chuquineyra B, Baca-Rondan F, Yucra-Sosa MC, Ccami-Bernal F, Soriano-Moreno DR, Nieto-Gutierrez W, Benites-Zapata VA. Association between self-reported evidence-based medicine competencies and prescribing of drugs without scientific evidence against mild COVID-19 among recently graduated physicians in Peru. Heliyon 2023; 9:e15366. [PMID: 37064449 PMCID: PMC10082469 DOI: 10.1016/j.heliyon.2023.e15366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/20/2023] [Accepted: 04/04/2023] [Indexed: 04/18/2023] Open
Abstract
Objective To evaluate the association between self-rated evidence-based medicine (EBM) competencies and the prescription of drugs without scientific evidence against mild COVID-19 (present with any of the signs and symptoms of COVID-19 but who do not have shortness of breath, dyspnea, or abnormal chest imaging) among recently graduated physicians in Peru. Methods We conducted an analytical cross-sectional study where we evaluated a non-probability sample of recently graduated physicians during June and July 2021 (end of second wave of COVID-19 in Peru). Self-rated EBM competencies were assessed by four domains (formulation of a clinical question, search, analysis, and application) using a Likert scale with scores from zero to four ("Very inadequate" = 0, to "Very Adequate" = 4), it was considered as "Adequate" if the score was three or four. In addition, the variable "General competence on EBM" was rated as "Adequate" if in all domains evaluated it presented an adequate self-rating. For the outcome, drug prescription, we considered the use of ivermectin, azithromycin, other antibiotics, hydroxychloroquine, dexamethasone, and anticoagulants (drugs with no efficacy demonstrated for patients with mild COVID-19). To assess the association, we used Poisson regression models with robust variances and obtaining crude (cPR) and adjusted (aPR) prevalence ratios with their 95% confidence intervals (95%CI). Results Of a total of 239 physicians included 70.7% prescribed at least one drug without scientific evidence. A total of 51.1% reported adequate ratings in all evaluated domains of EBM. Self-rating the "Clinical Question Formulation" competency as adequate was associated with a lower frequency of prescribing medications for mild COVID-19 (aPR: 0.93; 95% CI: 0.91-0.95). While self-rating as adequate the competency of "Identify possible implications of investigations" was associated with an increase in the prescription of such drugs (aPR: 1.14; 95% CI: 1.09-1.20). Additionally, self-rating all domains as adequate were associated with less prescription (aPR: 0.93; 95% CI: 0.90-0.96). Conclusion Seven out of ten recently graduated physicians prescribed some type of medication without scientific evidence to treat patients with mild COVID-19. Having adequate self-perceived EBM competencies was associated with a lower frequency of prescribing medications without scientific evidence to manage patients with mild COVID-19.
Collapse
Affiliation(s)
| | | | - Fiorella Baca-Rondan
- Escuela Profesional de Medicina Humana, Universidad Nacional de San Antonio Abad del Cusco, Cusco, Peru
| | - Maria Cristina Yucra-Sosa
- Escuela Profesional de Medicina Humana, Universidad Nacional de San Antonio Abad del Cusco, Cusco, Peru
| | - Fabricio Ccami-Bernal
- Facultad de Medicina, Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru
| | - David R Soriano-Moreno
- Unidad de Investigación Clínica y Epidemiológica, Escuela de Medicina, Universidad Peruana Unión, Lima, Peru
| | | | - Vicente A Benites-Zapata
- Unidad para la Generación y Síntesis de Evidencias en Salud, Universidad San Ignacio de Loyola, Lima, Peru
| |
Collapse
|
18
|
The Association between Previous Antibiotic Consumption and SARS-CoV-2 Infection: A Population-Based Case-Control Study. Antibiotics (Basel) 2023; 12:antibiotics12030587. [PMID: 36978453 PMCID: PMC10044412 DOI: 10.3390/antibiotics12030587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Background: The susceptibility to SARS-CoV-2 infection is complex and not yet fully elucidated, being related to many variables; these include human microbiome and immune status, which are both affected for a long period by antibiotic use. We therefore aimed to examine the association of previous antibiotic consumption and SARS-CoV-2 infection in a large-scale population-based study with control of known confounders. Methods: A matched case–control study was performed utilizing the electronic medical records of a large Health Maintenance Organization. Cases were subjects with confirmed SARS-CoV-2 infection (n = 31,260), matched individually (1:4 ratio) to controls without a positive SARS-CoV-2 test (n = 125,039). The possible association between previous antibiotic use and SARS-CoV-2 infection was determined by comparing antibiotic consumption in the previous 6 and 12 months between the cases and controls. For each antibiotic consumed we calculated the odds ratio (OR) for documented SARS-CoV-2 infection, 95% confidence interval (CI), and p-value using univariate and multivariate analyses. Results: The association between previous antibiotic consumption and SARS-CoV-2 infection was complex and bi-directional. In the multivariate analysis, phenoxymethylpenicillin was associated with increased rate of SARS-CoV-2 infection (OR 1.110, 95% CI: 1.036–1.191) while decreased rates were associated with previous consumption of trimethoprim-sulfonamides (OR 0.783, 95% CI: 0.632–0.971) and azithromycin (OR 0.882, 95% CI: 0.829–0.938). Fluroquinolones were associated with decreased rates (OR 0.923, 95% CI: 0.861–0.989) only in the univariate analysis. Previous consumption of other antibiotics had no significant association with SARS-CoV-2 infection. Conclusions: Previous consumption of certain antibiotic agents has an independent significant association with increased or decreased rates of SARS-CoV-2 infection. Plausible mechanisms, that should be further elucidated, are mainly antibiotic effects on the human microbiome and immune modulation.
Collapse
|
19
|
Hameiri-Bowen D, Yindom LM, Sovershaeva E, Bandason T, Mayini J, M Rehman A, Simms V, Gift Ngwira L, Flagestad T, Jarl Gutteberg T, McHugh G, Abbas Ferrand R, Rowland-Jones SL. "The effect of 48-weeks azithromycin therapy on levels of soluble biomarkers associated with HIV-associated chronic lung disease". Int Immunopharmacol 2023; 116:109756. [PMID: 36682262 PMCID: PMC10914635 DOI: 10.1016/j.intimp.2023.109756] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/04/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
OBJECTIVES HIV-associated immune activation contributes to chronic lung disease (CLD) in children and adolescents living with HIV. Azithromycin has immunomodulatory and anti-microbial properties that may be useful for treating HIV-associated CLD (HCLD). This study describes the effect of azithromycin on expression of plasma soluble biomarkers in children and adolescents with HCLD. METHODS This study was nested within a multi-site double-blind, placebo controlled, randomised controlled trial (RCT) of azithromycin in individuals aged 6-19 years with HCLD (defined as FEV1 z-score < -1) in Malawi and Zimbabwe (BREATHE (NCT02426112)). Participants were randomized 1:1 to once-weekly oral azithromycin with weight-based dosing, for 48 weeks, or placebo. Twenty-six plasma soluble biomarkers were measured on a MagPix Luminex instrument at enrolment, after 48-weeks of treatment and 24-weeks after treatment cessation. Mixed effects models were constructed to compare biomarker expression across treatment and placebo groups. RESULTS Weekly azithromycin was associated with reduced levels of C-Reactive Protein (CRP), E-Selectin, Matrix metalloproteinase 10 (MMP-10). Treatment effects for all soluble biomarkers were not sustained 24-weeks after treatment cessation with biomarker expression returning to pre-treatment levels. CONCLUSIONS We observed real-world effects of azithromycin on acute inflammation, neutrophil accumulation, and extracellular matrix degradation, that were not sustained after treatment cessation. These results are pertinent when using azithromycin for its immunomodulatory properties, or targeting pathways represented by the soluble biomarkers in this study.
Collapse
Affiliation(s)
- Dan Hameiri-Bowen
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| | - Louis-Marie Yindom
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Evgeniya Sovershaeva
- UiT The Arctic University of Norway, University Hospital of North Norway, Tromsø, Norway
| | - Tsitsi Bandason
- Biomedical Research and Training Institute, Harare, Zimbabwe
| | - Justin Mayini
- Biomedical Research and Training Institute, Harare, Zimbabwe
| | - Andrea M Rehman
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Victoria Simms
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Lucky Gift Ngwira
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom; Malawi-Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi
| | - Trond Flagestad
- UiT The Arctic University of Norway, University Hospital of North Norway, Tromsø, Norway
| | - Tore Jarl Gutteberg
- UiT The Arctic University of Norway, University Hospital of North Norway, Tromsø, Norway
| | - Grace McHugh
- Biomedical Research and Training Institute, Harare, Zimbabwe
| | - Rashida Abbas Ferrand
- Biomedical Research and Training Institute, Harare, Zimbabwe; Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | |
Collapse
|
20
|
Zhang R, Sun C, Han Y, Huang L, Sheng H, Wang J, Zhang Y, Lai J, Yuan J, Chen X, Jiang C, Wu F, Wang J, Fan X, Wang J. Neutrophil autophagy and NETosis in COVID-19: perspectives. Autophagy 2023; 19:758-767. [PMID: 35951555 PMCID: PMC9980466 DOI: 10.1080/15548627.2022.2099206] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic has caused substantial losses worldwide in people's lives, health, and property. Currently, COVID-19 is still prominent worldwide without any specific drug treatment. The SARS-CoV-2 pathogen is the cause of various systemic diseases, mainly acute pneumonia. Within the pathological process, neutrophils are recruited to infected sites, especially in the lungs, for the first stage of removing invading SARS-CoV-2 through a range of mechanisms. Macroautophagy/autophagy, a conserved autodegradation process in neutrophils, plays a crucial role in the neutrophil phagocytosis of pathogens. NETosis refers to neutrophil cell death, while auto-inflammatory factors and antigens release NETs. This review summarizes the latest research progress and provides an in-depth explanation of the underlying mechanisms of autophagy and NETosis in COVID-19. Furthermore, after exploring the relationship between autophagy and NETosis, we discuss potential targets and treatment options. This review keeps up with the latest research on COVID-19 from neutrophil autophagy and NETosis with a new perspective, which can guide the urgent development of antiviral drugs and provide guidance for the clinical treatment of COVID-19.Abbreviations: AKT1: AKT serine/threonine kinase 1; AMPK: AMP-activated protein kinase; AP: autophagosome; ARDS: acute respiratory distress syndrome; ATG: autophagy related; BECN1: beclin 1; cfDNA: cell-free DNA; COVID-19: coronavirus disease 2019; CQ: chloroquine; DMVs: double-membrane vesicles; ELANE/NE: elastase, neutrophil expressed; F3: coagulation factor III, tissue factor; HCQ: hydroxychloroquine; MAP1LC3/LC3: microtubule associated protein 1 light chain of 3; MPO: myeloperoxidase; MTORC1: mechanistic target of rapamycin kinase complex 1; NETs: neutrophil traps; NSP: nonstructural protein; PI3K: class I phosphoinositide 3-kinase; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; ROS: reactive oxygen species; SARS-CoV-2: severe acute respiratory syndrome coronavirus 2; SKP2: S-phase kinase associated protein 2; TCC: terminal complement complex; ULK1: unc-51 like.
Collapse
Affiliation(s)
- Ruoyu Zhang
- Department of Pain Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chen Sun
- Department of Pain Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yunze Han
- Department of Pain Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Honghui Sheng
- Department of Pain Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing Wang
- Department of Pain Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yuqing Zhang
- Department of Pain Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jonathan Lai
- Premed track majoring in Biology, Baylor University, Waco, Texas, USA
| | - Jiahao Yuan
- Department of Pain Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chao Jiang
- Department of Neurology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Fuyuan Wu
- Department of Pain Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xiaochong Fan
- Department of Pain Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jian Wang
- Department of Pain Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
21
|
Yan Y, Wu L, Li X, Zhao L, Xu Y. Immunomodulatory role of azithromycin: Potential applications to radiation-induced lung injury. Front Oncol 2023; 13:966060. [PMID: 36969016 PMCID: PMC10030824 DOI: 10.3389/fonc.2023.966060] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 02/13/2023] [Indexed: 03/29/2023] Open
Abstract
Radiation-induced lung injury (RILI) including radiation-induced pneumonitis and radiation-induced pulmonary fibrosis is a side effect of radiotherapy for thoracic tumors. Azithromycin is a macrolide with immunomodulatory properties and anti-inflammatory effects. The immunopathology of RILI that results from irradiation is robust pro-inflammatory responses with high levels of chemokine and cytokine expression. In some patients, pulmonary interstitial fibrosis results usually due to an overactive immune response. Growing clinical studies recently proposed that the anti-inflammatory and immunomodulatory effects of azithromycin may benefit patients with acute lung injury. It has been shown potential benefits for patients with RILI in preclinical studies. Azithromycin has a variety of immunomodulatory effect to improve the process of disease, including inhibition of pro-inflammatory cytokines production participating in the regulatory function of macrophages, changes in autophagy, and inhibition of neutrophil influx. We review the published evidence of mechanisms of azithromycin, and focus on the potential effect of azithromycin on the immune response to RILI.
Collapse
Affiliation(s)
- Yujie Yan
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Leilei Wu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Yaping Xu, ; Xuefei Li, ; Lan Zhao,
| | - Lan Zhao
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Yaping Xu, ; Xuefei Li, ; Lan Zhao,
| | - Yaping Xu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Yaping Xu, ; Xuefei Li, ; Lan Zhao,
| |
Collapse
|
22
|
Azithromycin Mitigates Cisplatin-Induced Lung Oxidative Stress, Inflammation and Necroptosis by Upregulating SIRT1, PPARγ, and Nrf2/HO-1 Signaling. Pharmaceuticals (Basel) 2022; 16:ph16010052. [PMID: 36678549 PMCID: PMC9861532 DOI: 10.3390/ph16010052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/26/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Acute lung injury (ALI) is one of the adverse effects of the antineoplastic agent cisplatin (CIS). Oxidative stress, inflammation, and necroptosis are linked to the emergence of lung injury in various disorders. This study evaluated the effect of the macrolide antibiotic azithromycin (AZM) on oxidative stress, inflammatory response, and necroptosis in the lungs of CIS-administered rats, pinpointing the involvement of PPARγ, SIRT1, and Nrf2/HO-1 signaling. The rats received AZM for 10 days and a single dose of CIS on the 7th day. CIS provoked bronchial and alveolar injury along with increased levels of ROS, MDA, NO, MPO, NF-κB p65, TNF-α, and IL-1β, and decreased levels of GSH, SOD, GST, and IL-10, denoting oxidative and inflammatory responses. The necroptosis-related proteins RIP1, RIP3, MLKL, and caspase-8 were upregulated in CIS-treated rats. AZM effectively prevented lung tissue injury, ameliorated oxidative stress and NF-κB p65 and pro-inflammatory markers levels, boosted antioxidants and IL-10, and downregulated necroptosis-related proteins in CIS-administered rats. AZM decreased the concentration of Ang II and increased those of Ang (1-7), cytoglobin, PPARγ, SIRT1, Nrf2, and HO-1 in the lungs of CIS-treated rats. In conclusion, AZM attenuated the lung injury provoked by CIS in rats through the suppression of inflammation, oxidative stress, and necroptosis. The protective effect of AZM was associated with the upregulation of Nrf2/HO-1 signaling, cytoglobin, PPARγ, and SIRT1.
Collapse
|
23
|
Frontera JA, Sabadia S, Yang D, de Havenon A, Yaghi S, Lewis A, Lord AS, Melmed K, Thawani S, Balcer LJ, Wisniewski T, Galetta SL. Life stressors significantly impact long-term outcomes and post-acute symptoms 12-months after COVID-19 hospitalization. J Neurol Sci 2022; 443:120487. [PMID: 36379135 PMCID: PMC9637014 DOI: 10.1016/j.jns.2022.120487] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/12/2022] [Accepted: 11/01/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Limited data exists evaluating predictors of long-term outcomes after hospitalization for COVID-19. METHODS We conducted a prospective, longitudinal cohort study of patients hospitalized for COVID-19. The following outcomes were collected at 6 and 12-months post-diagnosis: disability using the modified Rankin Scale (mRS), activities of daily living assessed with the Barthel Index, cognition assessed with the telephone Montreal Cognitive Assessment (t-MoCA), Neuro-QoL batteries for anxiety, depression, fatigue and sleep, and post-acute symptoms of COVID-19. Predictors of these outcomes, including demographics, pre-COVID-19 comorbidities, index COVID-19 hospitalization metrics, and life stressors, were evaluated using multivariable logistic regression. RESULTS Of 790 COVID-19 patients who survived hospitalization, 451(57%) completed 6-month (N = 383) and/or 12-month (N = 242) follow-up, and 77/451 (17%) died between discharge and 12-month follow-up. Significant life stressors were reported in 121/239 (51%) at 12-months. In multivariable analyses, life stressors including financial insecurity, food insecurity, death of a close contact and new disability were the strongest independent predictors of worse mRS, Barthel Index, depression, fatigue, and sleep scores, and prolonged symptoms, with adjusted odds ratios ranging from 2.5 to 20.8. Other predictors of poor outcome included older age (associated with worse mRS, Barthel, t-MoCA, depression scores), baseline disability (associated with worse mRS, fatigue, Barthel scores), female sex (associated with worse Barthel, anxiety scores) and index COVID-19 severity (associated with worse Barthel index, prolonged symptoms). CONCLUSIONS Life stressors contribute substantially to worse functional, cognitive and neuropsychiatric outcomes 12-months after COVID-19 hospitalization. Other predictors of poor outcome include older age, female sex, baseline disability and severity of index COVID-19.
Collapse
Affiliation(s)
- Jennifer A Frontera
- Departments of Neurology, New York University Grossman School of Medicine, New York, NY, USA.
| | - Sakinah Sabadia
- Departments of Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Dixon Yang
- Department of Neurology, New York Presbyterian, Columbia Medical Center, New York, NY, USA
| | - Adam de Havenon
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Shadi Yaghi
- Department of Neurology, Brown University School of Medicine, Providence, RI, USA
| | - Ariane Lewis
- Departments of Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Aaron S Lord
- Departments of Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Kara Melmed
- Departments of Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sujata Thawani
- Departments of Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Laura J Balcer
- Departments of Neurology, New York University Grossman School of Medicine, New York, NY, USA; Population Health, New York University Grossman School of Medicine, New York, NY, USA; Ophthalmology, New York University Grossman School of Medicine, New York, NY, USA
| | - Thomas Wisniewski
- Departments of Neurology, New York University Grossman School of Medicine, New York, NY, USA; Pathology, New York University Grossman School of Medicine, New York, NY, USA; Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Steven L Galetta
- Departments of Neurology, New York University Grossman School of Medicine, New York, NY, USA; Ophthalmology, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
24
|
Ceramella J, Iacopetta D, Sinicropi MS, Andreu I, Mariconda A, Saturnino C, Giuzio F, Longo P, Aquaro S, Catalano A. Drugs for COVID-19: An Update. Molecules 2022; 27:8562. [PMID: 36500655 PMCID: PMC9740261 DOI: 10.3390/molecules27238562] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was the seventh known human coronavirus, and it was identified in Wuhan, Hubei province, China, in 2020. It caused the highly contagious disease called coronavirus disease 2019 (COVID-19), declared a global pandemic by the World Health Organization (WHO) on 11 March 2020. A great number of studies in the search of new therapies and vaccines have been carried out in these three long years, producing a series of successes; however, the need for more effective vaccines, therapies and other solutions is still being pursued. This review represents a tracking shot of the current pharmacological therapies used for the treatment of COVID-19.
Collapse
Affiliation(s)
- Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, 87036 Rende, Italy
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, 87036 Rende, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, 87036 Rende, Italy
| | - Inmaculada Andreu
- Departamento de Química, Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
- Unidad Mixta de Investigación UPV-IIS La Fe, Hospital Universitari i Politècnic La Fe, Avenida de Fernando Abril Martorell 106, 46026 Valencia, Spain
| | | | - Carmela Saturnino
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Federica Giuzio
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Pasquale Longo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Stefano Aquaro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, 87036 Rende, Italy
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy
| |
Collapse
|
25
|
Molaaghaee‐Rouzbahani S, Asri N, Jahani‐Sherafat S, Amani D, Masotti A, Baghaei K, Yadegar A, Mirjalali H, Rostami‐Nejad M. The modulation of macrophage subsets in celiac disease pathogenesis. Immun Inflamm Dis 2022; 10:e741. [PMID: 36444633 PMCID: PMC9667199 DOI: 10.1002/iid3.741] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/12/2022] [Accepted: 11/06/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND So far, limited studies have focused on the role of Macrophages (MQs) in the development or progression of celiac disease (CD). Researchers believe that increasing knowledge about the function of MQs in inflammatory disorders plays a critical role in finding a new treatment for these kinds of diseases. MAIN BODY CD is a permanent autoimmune intestinal disorder triggered by gluten exposure in predisposed individuals. This disorder happens due to the loss of intestinal epithelial barrier integrity characterized by dysregulated innate and adaptive immune responses. MQs are known as key players of the innate immune system that link innate and adaptive immunity. MQs of human intestinal lamina propria participate in maintaining tissue homeostasis, and also intestinal inflammation development. Previous studies suggested that gliadin triggers a proinflammatory phenotype (M1 MQ) in human primary MQs. Moreover, M2-related immunosuppressive mediators are also present in CD. In fact, CD patients present an impaired transition from pro-inflammatory to anti-inflammatory responses due to inappropriate responses to gliadin peptides. CONCLUSION The M1/M2 MQs polarization balancing regulators can be considered novel therapeutic targets for celiac disease.
Collapse
Affiliation(s)
| | - Nastaran Asri
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Somayeh Jahani‐Sherafat
- Laser Application in Medical Sciences Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Davar Amani
- Department of Immunology, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Andrea Masotti
- Bambino Gesù Children's Hospital‐IRCCSResearch LaboratoriesRomeItaly
| | - Kaveh Baghaei
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Hamed Mirjalali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Mohammad Rostami‐Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
26
|
|
27
|
Alrashedi MG, Ali AS, Ahmed OA, Ibrahim IM. Local Delivery of Azithromycin Nanoformulation Attenuated Acute Lung Injury in Mice. Molecules 2022; 27:8293. [PMID: 36500388 PMCID: PMC9739299 DOI: 10.3390/molecules27238293] [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: 11/01/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Humanity has suffered from the coronavirus disease 2019 (COVID-19) pandemic over the past two years, which has left behind millions of deaths. Azithromycin (AZ), an antibiotic used for the treatment of several bacterial infections, has shown antiviral activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as well as against the dengue, Zika, Ebola, and influenza viruses. Additionally, AZ has shown beneficial effects in non-infective diseases such as cystic fibrosis and bronchiectasis. However, the systemic use of AZ in several diseases showed low efficacy and potential cardiac toxicity. The application of nanotechnology to formulate a lung delivery system of AZ could prove to be one of the solutions to overcome these drawbacks. Therefore, we aimed to evaluate the attenuation of acute lung injury in mice via the local delivery of an AZ nanoformulation. The hot emulsification-ultrasonication method was used to prepare nanostructured lipid carrier of AZ (AZ-NLC) pulmonary delivery systems. The developed formulation was evaluated and characterized in vitro and in vivo. The efficacy of the prepared formulation was tested in the bleomycin (BLM) -mice model for acute lung injury. AZ-NLC was given by the intratracheal (IT) route for 6 days at a dose of about one-eighth oral dose of AZ suspension. Samples of lung tissues were taken at the end of the experiment for immunological and histological assessments. AZ-NLC showed an average particle size of 453 nm, polydispersity index of 0.228 ± 0.07, zeta potential of -30 ± 0.21 mV, and a sustained release pattern after the initial 50% drug release within the first 2 h. BLM successfully induced a marked increase in pro-inflammatory markers and also induced histological changes in pulmonary tissues. All these alterations were significantly reversed by the concomitant administration of AZ-NLC (IT). Pulmonary delivery of AZ-NLC offered delivery of the drug locally to lung tissues. Its attenuation of lung tissue inflammation and histological injury induced by bleomycin was likely through the downregulation of the p53 gene and the modulation of Bcl-2 expression. This novel strategy could eventually improve the effectiveness and diminish the adverse drug reactions of AZ. Lung delivery could be a promising treatment for acute lung injury regardless of its cause. However, further work is needed to explore the stability of the formulation, its pharmacokinetics, and its safety.
Collapse
Affiliation(s)
- Mohsen G. Alrashedi
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Ministry of Health, Riyadh 12628, Saudi Arabia
| | - Ahmed Shaker Ali
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Osama Abdelhakim Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ibrahim M. Ibrahim
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
28
|
Gao J, Hu X, Xu C, Guo M, Li S, Yang F, Pan X, Zhou F, Jin Y, Bai F, Cheng Z, Wu Z, Chen S, Huang X, Wu W. Neutrophil-mediated delivery of the combination of colistin and azithromycin for the treatment of bacterial infection. iScience 2022; 25:105035. [PMID: 36117992 PMCID: PMC9474925 DOI: 10.1016/j.isci.2022.105035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/29/2022] [Accepted: 08/24/2022] [Indexed: 11/21/2022] Open
Abstract
Novel treatment strategies are in urgent need to deal with the rapid development of antibiotic-resistant superbugs. Combination therapies and targeted drug delivery have been exploited to promote treatment efficacies. In this study, we loaded neutrophils with azithromycin and colistin to combine the advantages of antibiotic combinations, targeted delivery, and immunomodulatory effect of azithromycin to treat infections caused by Gram-negative pathogens. Delivery of colistin into neutrophils was mediated by fusogenic liposome, while azithromycin was directly taken up by neutrophils. Neutrophils loaded with the drugs maintained the abilitity to generate reactive oxygen species and migrate. In vitro assays demonstrated enhanced bactericidal activity against multidrug-resistant pathogens and reduced inflammatory cytokine production by the drug-loaded neutrophils. A single intravenous administration of the drug-loaded neutrophils effectively protected mice from Pseudomonas aeruginosa infection in an acute pneumonia model. This study provides a potential effective therapeutic approach for the treatment of bacterial infections. Neutrophils are loaded with colistin and azithromycin in vitro The loaded drugs enhance the bactericidal effect and reduce the inflammatory response Drug-loaded neutrophils conferred effective protection against bacterial infection
Collapse
Affiliation(s)
- Jiacong Gao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xueyan Hu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Congjuan Xu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mingming Guo
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shouyi Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fan Yang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaolei Pan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fangyu Zhou
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fang Bai
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhihui Cheng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhenzhou Wu
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shuiping Chen
- Department of Laboratory Medicine, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
29
|
Masud AA, Alsharif FM, Creameans JW, Perdeh J, Feola DJ, Venditto VJ. Optimization and Characterization of a Liposomal Azithromycin Formulation for Alternative Macrophage Activation. FRONTIERS IN DRUG DELIVERY 2022; 2:908709. [PMID: 36407498 PMCID: PMC9670256 DOI: 10.3389/fddev.2022.908709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Liposomal azithromycin (L-AZM) promotes macrophage polarization toward an M2-like phenotype in the context of myocardial infarction that results in improved cardiovascular outcomes in mice. To improve upon this formulation, we sought to identify optimized formulation, stability, and biological activity parameters necessary to enhance the immunomodulatory activity and efficacy of L-AZM. While our parent formulation contains a mixture of long-chain saturated phosphatidylcholine and phosphatidylglycerol lipids, we evaluated a series of formulations with different amounts of unsaturated lipids and cholesterol with the goal of improving the loading capacity and stability of the formulations. We also introduce fusogenic lipids to improve the cytosolic delivery to enhance the immune modulatory properties of the drug. To achieve these goals, we initially prepared a library of 24 formulations using thin film hydration and assessed the resultant liposomes for size and polydispersity. Five lead formulations were identified based on low polydispersity (<0.3) and stability over time. The lead formulations were then evaluated for stability in serum using dialysis and macrophage polarization activity in vitro as measured by decreased IL-12 expression. Collectively, our data indicate that the formulation components drive the balance between encapsulation efficiency and stability and that all the lead liposomal formulations improve in vitro alternative macrophage activation as compared to free AZM.
Collapse
Affiliation(s)
- Abdullah A. Masud
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Fahd M. Alsharif
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Jarrod W. Creameans
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Jasmine Perdeh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - David J. Feola
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Vincent J. Venditto
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
30
|
Venditto VJ, Feola DJ. Delivering macrolide antibiotics to heal a broken heart - And other inflammatory conditions. Adv Drug Deliv Rev 2022; 184:114252. [PMID: 35367307 PMCID: PMC9063468 DOI: 10.1016/j.addr.2022.114252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/07/2022] [Accepted: 03/28/2022] [Indexed: 12/17/2022]
Abstract
Drug carriers to deliver macrolide antibiotics, such as azithromycin, show promise as antibacterial agents. Macrolide drug carriers have largely focused on improving the drug stability and pharmacokinetics, while reducing adverse reactions and improving antibacterial activity. Recently, macrolides have shown promise in treating inflammatory conditions by promoting a reparative effect and limiting detrimental pro-inflammatory responses, which shifts the immunologic setpoint from suppression to balance. While macrolide drug carriers have only recently been investigated for their ability to modulate immune responses, the previous strategies that deliver macrolides for antibacterial therapy provide a roadmap for repurposing the macrolide drug carriers for therapeutic interventions targeting inflammatory conditions. This review describes the antibacterial and immunomodulatory activity of macrolides, while assessing the past in vivo evaluation of drug carriers used to deliver macrolides with the intention of presenting a case for increased effort to translate macrolide drug carriers into the clinic.
Collapse
|
31
|
Heidary M, Ebrahimi Samangani A, Kargari A, Kiani Nejad A, Yashmi I, Motahar M, Taki E, Khoshnood S. Mechanism of action, resistance, synergism, and clinical implications of azithromycin. J Clin Lab Anal 2022; 36:e24427. [PMID: 35447019 PMCID: PMC9169196 DOI: 10.1002/jcla.24427] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/20/2022] [Accepted: 03/24/2022] [Indexed: 12/02/2022] Open
Abstract
Background Azithromycin (AZM), sold under the name Zithromax, is classified as a macrolide. It has many benefits due to its immunomodulatory, anti‐inflammatory, and antibacterial effects. This review aims to study different clinical and biochemisterial aspects and properties of this drug which has a priority based on literature published worldwide. Methods Several databases including Web of Science, Google Scholar, PubMed, and Scopus were searched to obtain the relevant studies. Results AZM mechanism of action including the inhibition of bacterial protein synthesis, inhibition of proinflammatory cytokine production, inhibition of neutrophil infestation, and macrophage polarization alteration, gives it the ability to act against a wide range of microorganisms. Resistant organisms are spreading and being developed because of the irrational use of the drug in the case of dose and duration. AZM shows synergistic effects with other drugs against a variety of organisms. This macrolide is considered a valuable antimicrobial agent because of its use as a treatment for a vast range of diseases such as asthma, bronchiolitis, COPD, cystic fibrosis, enteric infections, STIs, and periodontal infections. Conclusions Our study shows an increasing global prevalence of AZM resistance. Thus, synergistic combinations are recommended to treat different pathogens. Moreover, continuous monitoring of AZM resistance by registry centers and the development of more rapid diagnostic assays are urgently needed.
Collapse
Affiliation(s)
- Mohsen Heidary
- Department of Laboratory Sciences, School of Paramedical Sciences, Sabzevar University of Medical Sciences, Sabzevar, Iran.,Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | | | - Abolfazl Kargari
- Student Research Committee, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Aliakbar Kiani Nejad
- Student Research Committee, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Ilya Yashmi
- Student Research Committee, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Moloudsadat Motahar
- Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elahe Taki
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Khoshnood
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
32
|
Antiviral effects of azithromycin: A narrative review. Biomed Pharmacother 2022; 147:112682. [PMID: 35131658 PMCID: PMC8813546 DOI: 10.1016/j.biopha.2022.112682] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 12/20/2022] Open
Abstract
Viral infections have a great impact on human health. The urgent need to find a cure against different viruses led us to investigations in a vast range of drugs. Azithromycin (AZT), classified as a macrolide, showed various effects on different known viruses such as severe acute respiratory syndrome coronavirus (SARS-CoV), Zika, Ebola, Enterovirus (EVs) and Rhinoviruses (RVs), and Influenza A previously; namely, these viruses, which caused global concerns, are considered as targets for AZT different actions. Due to AZT background in the treatment of known viral infections mentioned above (which is described in this study), in the early stages of COVID-19 (a new zoonotic disease caused by a novel coronavirus called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)) development, AZT drew attention to itself due to its antiviral and immunomodulatory effects as a valuable candidate for COVID-19 treatment. AZT usage instructions for treating different viral infections have always been under observation, and COVID-19 is no exception. There are still debates about the use of AZT in COVID-19 treatment. However, eventually, novel researches convinced WHO to announce the discontinuation of AZT use (alone or in combination with hydroxychloroquine) in treating SARS-CoV-2 infection. This research aims to study the structure of all of the viruses mentioned above and the molecular and clinical effects of AZT against the virus.
Collapse
|
33
|
Liu X, Wu Y, Mao C, Shen J, Zhu K. Host-acting antibacterial compounds combat cytosolic bacteria. Trends Microbiol 2022; 30:761-777. [DOI: 10.1016/j.tim.2022.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/22/2021] [Accepted: 01/12/2022] [Indexed: 01/25/2023]
|
34
|
Gyselinck I, Liesenborghs L, Belmans A, Engelen MM, Betrains A, Van Thillo Q, Nguyen PAH, Goeminne P, Soenen AC, De Maeyer N, Pilette C, Papleux E, Vanderhelst E, Derweduwen A, Alexander P, Bouckaert B, Martinot JB, Decoster L, Vandeurzen K, Schildermans R, Verhamme P, Janssens W, Vos R. Azithromycin for Treatment of Hospitalised COVID-19 Patients: a randomised, multicentre, open-label clinical trial (DAWn-AZITHRO). ERJ Open Res 2022; 8:00610-2021. [PMID: 35233389 PMCID: PMC8801156 DOI: 10.1183/23120541.00610-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/28/2021] [Indexed: 12/23/2022] Open
Abstract
Background and objectives Azithromycin was rapidly adopted as a repurposed drug to treat coronavirus disease 2019 (COVID-19) early in the pandemic. We aimed to evaluate its efficacy in patients hospitalised for COVID-19. Methods In a series of randomised, open-label, phase 2 proof-of-concept, multicentre clinical trials (Direct Antivirals Working against the novel coronavirus (DAWn)), several treatments were compared with standard of care. In 15 Belgian hospitals, patients hospitalised with moderate to severe COVID-19 were allocated 2:1 to receive standard of care plus azithromycin or standard of care alone. The primary outcome was time to live discharge or sustained clinical improvement, defined as a two-point improvement on the World Health Organization (WHO) ordinal scale sustained for at least 3 days. Results Patients were included between April 22 and December 17, 2020. When 15-day follow-up data were available for 160 patients (56% of preset cohort), an interim analysis was performed at request of the independent Data Safety and Monitoring Board. Subsequently, DAWn-AZITHRO was stopped for futility. In total, 121 patients were allocated to the treatment arm and 64 patients to the standard-of-care arm. We found no effect of azithromycin on the primary outcome with a hazard ratio of 1.044 (95% CI 0.772–1.413; p=0.7798). None of the predefined subgroups showed significant interaction as covariates in the Fine–Gray regression analysis. No benefit of azithromycin was found on any of the short- and longer-term secondary outcomes. Conclusion Time to clinical improvement is not influenced by azithromycin in patients hospitalised with moderate to severe COVID-19. Previous randomised controlled studies with azithromycin in hospitalised COVID-19 patients assessed end-points at fixed timepoints. Complementary to this, DAWn-AZITHRO assessed time to sustained improvement. No benefit of azithromycin was shown.https://bit.ly/3FapyC7
Collapse
|
35
|
Muflihah H, Bhekti Rahimah S, Widiyanto T, Mahwati Y, Parumasivam T, Sastramihardja HS. Clinical use of antiviral, antibiotic and immunomodulatory drugs in hospitalized COVID-19 patients: a retrospective study in Bandung, Indonesia. F1000Res 2021. [DOI: 10.12688/f1000research.73606.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Evidence of highly effective repurposed drugs for coronavirus disease 2019 (COVID-19) is insufficient. However, empirical therapy using antiviral, antibiotic and immunomodulatory drugs is massive. Studies evaluating the clinical use of these drugs in Indonesia are sparse. Methods: We performed a retrospective study using medical records of hospitalized COVID-19 patients from July 2020 to March 2021 in Bandung, Indonesia. Data were collected at relevant timelines: age, sex, comorbid condition, peripheral oxygen saturation (SpO2), and hematology at admission; antiviral, antibiotic, and immunomodulator treatment during hospitalization; length of stay hospitalization (LOS) and death at discharge. Clinical use of the drug regimens included dose, frequency, and duration of therapy. The main outcome of hospitalization care was LOS and death. Results: Out of 249 patients, 43.3% had a comorbid condition, 74.7% had non-severe COVID-19 (SpO2 ≥ 90%), and almost all received antiviral or antibiotic agents. Remdesivir was the most frequent drug composing various antiviral regimens. Patients receiving a combination of remdesivir and favipiravir had lower SpO2 compared to those receiving oseltamivir (p=0.01). The short LOS was associated with remdesivir alone (p=0.03), the combination of favipiravir and oseltamivir (p=0.01), and the combination of intravenous levofloxacin and ceftriaxone (p<0.0001). Immunomodulatory drugs (methylprednisolone, dexamethasone, tocilizumab) were used in 47.1% of patients with low SpO2 (p=0.001). Its use was associated with prolonged LOS (p=0.0043). The increased risk of death in patients treated with the combination of remdesivir and favipiravir (OR 4.1;95%CI 1.4-12.2), and immunomodulatory drugs (OR 6.2; 95%CI 1.7-23.3) was confounded by the baseline characteristics of older age, comorbid condition, SpO2 level, and low lymphocyte number. Conclusions: Some treatment regimens were associated with short LOS, but there were drug regimens which might increase the risk of death. Further study should control the clinical conditions of COVID-19 patients at admission to confirm the outcome of death following drug therapy.
Collapse
|
36
|
Ezeokoli OT, Gcilitshana O, Pohl CH. Risk Factors for Fungal Co-Infections in Critically Ill COVID-19 Patients, with a Focus on Immunosuppressants. J Fungi (Basel) 2021; 7:545. [PMID: 34356924 PMCID: PMC8304654 DOI: 10.3390/jof7070545] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Severe cases of coronavirus disease 2019 (COVID-19) managed in the intensive care unit are prone to complications, including secondary infections with opportunistic fungal pathogens. Systemic fungal co-infections in hospitalized COVID-19 patients may exacerbate COVID-19 disease severity, hamper treatment effectiveness and increase mortality. Here, we reiterate the role of fungal co-infections in exacerbating COVID-19 disease severity as well as highlight emerging trends related to fungal disease burden in COVID-19 patients. Furthermore, we provide perspectives on the risk factors for fungal co-infections in hospitalized COVID-19 patients and highlight the potential role of prolonged immunomodulatory treatments in driving fungal co-infections, including COVID-19-associated pulmonary aspergillosis (CAPA), COVID-19-associated candidiasis (CAC) and mucormycosis. We reiterate the need for early diagnosis of suspected COVID-19-associated systemic mycoses in the hospital setting.
Collapse
Affiliation(s)
| | | | - Carolina H. Pohl
- Yeast Research Group, Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein 9300, South Africa or (O.T.E.); (O.G.)
| |
Collapse
|
37
|
Siddiqui M, Gültekingil A, Bakırcı O, Uslu N, Baskın E. Comparison of clinical features and laboratory findings of coronavirus disease 2019 and influenza A and B infections in children: a single-center study. Clin Exp Pediatr 2021; 64:364-369. [PMID: 34015895 PMCID: PMC8255514 DOI: 10.3345/cep.2021.00066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/07/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND As the coronavirus disease 2019 (COVID-19) outbreak continues to evolve, it is crucially important for pediatricians to be aware of the differences in demographic and clinical features between COVID-19 and influenza A and B infections. PURPOSE This study analyzed and compared the clinical features and laboratory findings of COVID-19 and influenza A and B infections in children. METHODS This retrospective study evaluated the medical data of 206 pediatric COVID-19 and 411 pediatric seasonal influenza A or B patients. RESULTS COVID-19 patients were older than seasonal influenza patients (median [interquartile range], 7.75 [2-14] years vs. 4 [2-6] years). The frequency of fever and cough in COVID-19 patients was lower than that of seasonal influenza patients (80.6% vs. 94.4%, P<0.001 and 22.8 % vs. 71.5%, P<0.001, respectively). Ageusia (4.9%) and anosmia (3.4%) were present in only COVID-19 patients. Leukopenia, lymphopenia, and thrombocytopenia were encountered more frequently in influenza patients than in COVID-19 patients (22.1% vs. 8.5%, P=0.029; 17.6% vs. 5.6%, P=0.013; and 13.2% vs. 5.6%, P= 0.048, respectively). Both groups showed significantly elevated monocyte levels in the complete blood count (70.4% vs. 69.9%, P=0.511). Major chest x-ray findings in COVID-19 patients included mild diffuse ground-glass opacity and right lower lobe infiltrates. There were no statistically significant intergroup differences in hospitalization or mortality rates; however, the intensive care unit admission rate was higher among COVID-19 patients (2.4% vs. 0.5%, P=0.045). CONCLUSION In this study, pediatric COVID-19 patients showed a wide range of clinical presentations ranging from asymptomatic/mild to severe illness. We found no intergroup differences in hospitalization rates, oxygen requirements, or hospital length of stay; however, the intensive care unit admission rate was higher among COVID-19 patients.
Collapse
Affiliation(s)
- Meraj Siddiqui
- Department of Pediatrics, Baskent University, Ankara, Turkey
| | - Ayşe Gültekingil
- Department of Pediatric Emergency, Baskent University, Ankara, Turkey
| | - Oğuz Bakırcı
- Department of Pediatrics, Baskent University, Ankara, Turkey
| | - Nihal Uslu
- Department of Radiodiagnostics, Baskent University, Ankara, Turkey
| | - Esra Baskın
- Department of Pediatric Nephrology and Rheumatology, Baskent University, Ankara, Turkey
| |
Collapse
|
38
|
Systems Biology and Bile Acid Signalling in Microbiome-Host Interactions in the Cystic Fibrosis Lung. Antibiotics (Basel) 2021; 10:antibiotics10070766. [PMID: 34202495 PMCID: PMC8300688 DOI: 10.3390/antibiotics10070766] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022] Open
Abstract
The study of the respiratory microbiota has revealed that the lungs of healthy and diseased individuals harbour distinct microbial communities. Imbalances in these communities can contribute to the pathogenesis of lung disease. How these imbalances occur and establish is largely unknown. This review is focused on the genetically inherited condition of Cystic Fibrosis (CF). Understanding the microbial and host-related factors that govern the establishment of chronic CF lung inflammation and pathogen colonisation is essential. Specifically, dissecting the interplay in the inflammation–pathogen–host axis. Bile acids are important host derived and microbially modified signal molecules that have been detected in CF lungs. These bile acids are associated with inflammation and restructuring of the lung microbiota linked to chronicity. This community remodelling involves a switch in the lung microbiota from a high biodiversity/low pathogen state to a low biodiversity/pathogen-dominated state. Bile acids are particularly associated with the dominance of Proteobacterial pathogens. The ability of bile acids to impact directly on both the lung microbiota and the host response offers a unifying principle underpinning the pathogenesis of CF. The modulating role of bile acids in lung microbiota dysbiosis and inflammation could offer new potential targets for designing innovative therapeutic approaches for respiratory disease.
Collapse
|
39
|
Bai G, Furushima D, Niki T, Matsuba T, Maeda Y, Takahashi A, Hattori T, Ashino Y. High Levels of the Cleaved Form of Galectin-9 and Osteopontin in the Plasma Are Associated with Inflammatory Markers That Reflect the Severity of COVID-19 Pneumonia. Int J Mol Sci 2021; 22:ijms22094978. [PMID: 34067072 PMCID: PMC8125627 DOI: 10.3390/ijms22094978] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/01/2021] [Accepted: 05/03/2021] [Indexed: 01/08/2023] Open
Abstract
Numbers of patients with coronavirus disease 2019 (COVID-19) have increased rapidly worldwide. Plasma levels of full-length galectin-9 (FL-Gal9) and osteopontin (FL-OPN) as well as their truncated forms (Tr-Gal9, Ud-OPN, respectively), are representative inflammatory biomarkers. Here, we measured FL-Gal9, FL-OPN, Tr-Gal9, and Ud-OPN in 94 plasma samples obtained from 23 COVID-19-infected patients with mild clinical symptoms (CV), 25 COVID-19 patients associated with pneumonia (CP), and 14 patients with bacterial infection (ID). The four proteins were significantly elevated in the CP group when compared with healthy individuals. ROC analysis between the CV and CP groups showed that C-reactive protein had the highest ability to differentiate, followed by Tr-Gal9 and ferritin. Spearman's correlation analysis showed that Tr-Gal9 and Ud-OPN but not FL-Gal9 and FL-OPN, had a significant association with laboratory markers for lung function, inflammation, coagulopathy, and kidney function in CP patients. CP patients treated with tocilizumab had reduced levels of FL-Gal9, Tr-Gal9, and Ud-OPN. It was suggested that OPN is cleaved by interleukin-6-dependent proteases. These findings suggest that the cleaved forms of OPN and galectin-9 can be used to monitor the severity of pathological inflammation and the therapeutic effects of tocilizumab in CP patients.
Collapse
Affiliation(s)
- Gaowa Bai
- Research Institute of Health and Welfare, Kibi International University, Takahashi 716-8508, Japan; (G.B.); (A.T.)
| | - Daisuke Furushima
- Department of Drug Evaluation and Informatics, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan;
| | - Toshiro Niki
- Department of Immunology, Kagawa University, Kagawa 761-0793, Japan;
| | - Takashi Matsuba
- Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Tottori 683-8503, Japan;
- Department of Animal Pharmaceutical Science, School of Pharmaceutical Science, Kyusyu University of Health and Welfare, Nobeoka, Miyazaki 882-8508, Japan
| | - Yosuke Maeda
- Viral Section, Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan;
| | - Atsushi Takahashi
- Research Institute of Health and Welfare, Kibi International University, Takahashi 716-8508, Japan; (G.B.); (A.T.)
| | - Toshio Hattori
- Research Institute of Health and Welfare, Kibi International University, Takahashi 716-8508, Japan; (G.B.); (A.T.)
- Correspondence: (T.H.); (Y.A.); Tel.: +81-866-22-9469 (T.H.); +81-22-308-7111 (Y.A.); Fax: +81-866-22-9469 (T.H.); +81-22-308-9921 (Y.A.)
| | - Yugo Ashino
- Department of Respiratory Medicine, Sendai City Hospital, Miyagi 982-8502, Japan
- Correspondence: (T.H.); (Y.A.); Tel.: +81-866-22-9469 (T.H.); +81-22-308-7111 (Y.A.); Fax: +81-866-22-9469 (T.H.); +81-22-308-9921 (Y.A.)
| |
Collapse
|