51
|
Mehri Z, Mehr AJ, Molavynejad S, Navarbafzadeh N, Adineh M, Nazari M, Nematollahzadeh Z. Investigating Some Effective Factors on the Prediction of Continuous Positive Airway Pressure Failure Rate in COVID-19-Related Hypoxemia. IRANIAN JOURNAL OF NURSING AND MIDWIFERY RESEARCH 2024; 29:697-702. [PMID: 39759919 PMCID: PMC11694580 DOI: 10.4103/ijnmr.ijnmr_392_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/14/2023] [Accepted: 09/07/2024] [Indexed: 01/07/2025]
Abstract
Background Considering the importance of using Non-Invasive Ventilation (NIV) in COVID-19-related hypoxemia, the present study was conducted to determine the effective factors on Continuous Positive Airway Pressure (CPAP) failure rate in COVID-19-related hypoxemia. Materials and Methods This research was a retrospective cross-sectional study (2021) investigating the records of 200 adult patients with the medical diagnosis of acute respiratory failure (ARF) of COVID-19, admitted to the Intensive Care Unit (ICU) in Shoushtar (southwestern Iran) who underwent CPAP therapy. The Heart rate, Acidosis, Consciousness, Oxygenation, and Respiratory rate (HACOR) scores were measured before the treatment and 1 h after undergoing CPAP treatment. Moreover, patients' demographic and clinical data were recorded. Data were analyzed using the Mann-Whitney, Chi-square, Wilcoxon, and logistic regression tests. The significance level was set at p ≤ 0.05. Results The mean standard deviation [SD]) age of patients was 63.96 (16.23) years. Among all 200 patients, 78.50% (n = 157) experienced CPAP failure and the remaining 21.50% (n = 43) underwent successful CPAP therapy. Failure chance was 7.10% higher in patients with higher HACOR scores undergoing 1 h CPAP treatment than others. It was also 14.92% higher among patients with diabetes mellitus (DM) than non-DM patients. Additionally, old age (z = 2591.50, p value = 0.02), obesity (z = 2433.00, p value = 0.024), and elevated Blood Urea Nitrogen (BUN) (z = 2620.00, p value = 0.0) impacted CPAP failure rates among patients. Conclusions The HACOR score 1 h after CPAP, DM, old age, obesity, and elevated BUN favor increased CPAP failure rates among patients.
Collapse
Affiliation(s)
- Zahra Mehri
- Scientometrics Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | | | - Shahram Molavynejad
- Nursing Care Research Center in Chronic Diseases, School of Nursing and Midwifery, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmeh Navarbafzadeh
- Knowledge and Information Science, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Mohammad Adineh
- Nursing Care Research Center in Chronic Diseases, School of Nursing and Midwifery, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Nazari
- Student Research Committee, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | | |
Collapse
|
52
|
Rahman MS, Hossain MS. Eicosanoids Signals in SARS-CoV-2 Infection: A Foe or Friend. Mol Biotechnol 2024; 66:3025-3041. [PMID: 37878227 DOI: 10.1007/s12033-023-00919-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023]
Abstract
SARS-CoV-2 mediated infection instigated a scary pandemic state since 2019. They created havoc comprising death, imbalanced social structures, and a wrecked global economy. During infection, the inflammation and associated cytokine storm generate a critical pathological situation in the human body, especially in the lungs. By the passage of time of infection, inflammatory disorders, and multiple organ damage happen which might lead to death, if not treated properly. Until now, many pathological parameters have been used to understand the progress of the severity of COVID-19 but with limited success. Bioactive lipid mediators have the potential of initiating and resolving inflammation in any disease. The connection between lipid storm and inflammatory states of SARS-CoV-2 infection has surfaced and got importance to understand and mitigate the pathological states of COVID-19. As the role of eicosanoids in COVID-19 infection is not well defined, available information regarding this issue has been accumulated to address the possible network of eicosanoids related to the initiation of inflammation, promotion of cytokine storm, and resolution of inflammation, and highlight possible strategies for treatment and drug discovery related to SARS-CoV-2 infection in this study. Understanding the involvement of eicosanoids in exploration of cellular events provoked by SARS-CoV-2 infection has been summarized as an important factor to deescalate any upcoming catastrophe imposed by the lethal variants of this micro-monster. Additionally, this study also recognized the eicosanoid based drug discovery, treatment, and strategies for managing the severity of SARS-COV-2 infection.
Collapse
Affiliation(s)
- Mohammad Sharifur Rahman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Mohammad Salim Hossain
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh.
| |
Collapse
|
53
|
Majolo JH, Gonçalves JIB, Souza RP, González LC, Sperotto N, Silveira MD, Oliveira SD, Bizarro CV, Machado P, Basso LA, Souza APD, Oliveira JR, Ferreira CAS. Losartan and enalapril maleate differently influence SARS-CoV-2-infected vero cells. Sci Rep 2024; 14:24801. [PMID: 39433817 PMCID: PMC11493994 DOI: 10.1038/s41598-024-76657-7] [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: 07/01/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic has posed significant challenges to global healthcare systems, particularly impacting individuals with pre-existing conditions like hypertension. This study sought to assess the impact of the antihypertensive medications, losartan and enalapril maleate on SARS-CoV-2 infected cells. Vero E6 cells were infected and treated in vitro, evaluating cell viability via the MTT colorimetric assay. Additionally, the study measured relative levels of viral RNA and selected gene messenger RNAs using reverse transcriptase followed by quantitative real-time polymerase chain reaction. RESULTS The findings revealed that losartan substantially reduced nucleocapsid RNA levels of SARS-CoV-2 to nearly undetectable levels, while enalapril maleate did not demonstrate a significant effect. In response to viral infection, the expression of il-18, p53, p21, and p62 increased compared to uninfected-untreated cells. Notably, il-6 expression was upregulated by both infection and treatments. A comparison between infected cells treated with losartan or enalapril maleate highlighted the presence of distinct profiles in the expression of il-6, p53, p21, and p62. CONCLUSIONS The data from our study suggest that these medications could interfere with certain effects triggered by SARS-CoV-2 infection in Vero E6 cells. However, their influence appears to vary both quantitatively and qualitatively in the modulation of metabolic and signal transduction pathways.
Collapse
Affiliation(s)
- Julia H Majolo
- Laboratory of Immunology and Microbiology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - João I B Gonçalves
- Laboratory of Clinical and Experimental Immunology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Renata P Souza
- Laboratory of Immunology and Microbiology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Laura C González
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Nathalia Sperotto
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Maiele D Silveira
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Sílvia D Oliveira
- Laboratory of Immunology and Microbiology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Cristiano V Bizarro
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Pablo Machado
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Luiz A Basso
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Ana P D Souza
- Laboratory of Clinical and Experimental Immunology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Jarbas R Oliveira
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil.
| | - Carlos A S Ferreira
- Laboratory of Immunology and Microbiology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil.
| |
Collapse
|
54
|
Ochar K, Iwar K, Nair VD, Chung YJ, Ha BK, Kim SH. The Potential of Glucosinolates and Their Hydrolysis Products as Inhibitors of Cytokine Storms. Molecules 2024; 29:4826. [PMID: 39459194 PMCID: PMC11510469 DOI: 10.3390/molecules29204826] [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: 08/23/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
A cytokine storm is an intense inflammatory response characterized by the overproduction of proinflammatory cytokines, resulting in tissue damage, and organ dysfunction. Cytokines play a crucial role in various conditions, such as coronavirus disease, in which the immune system becomes overactive and releases excessive levels of cytokines, including interleukins, tumor necrosis factor-alpha (TNF-α), and interferon-gamma (IFN-γ). This anomalous response often leads to acute respiratory distress syndrome (ARDS), disseminated intravascular coagulation (DIC), and multiple organ injury (MOI). Glucosinolates are plant secondary metabolites predominantly found in Brassica vegetables, but are also present in other species, such as Moringa Adens and Carica papaya L. When catalyzed by the enzyme myrosinase, glucosinolates produce valuable products, including sulforaphane, phenethyl isothiocyanate, 6-(methylsulfinyl) hexyl isothiocyanate, erucin, goitrin, and moringin. These hydrolyzed products regulate proinflammatory cytokine production by inhibiting the nuclear factor kappa-light-chain-enhancer of activated B-cell (NF-κB) signaling pathway and stimulating the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. This action can alleviate hyperinflammation in infected cells and modulate cytokine storms. In this review, we aimed to examine the potential role of glucosinolates in modulating cytokine storms and reducing inflammation in various conditions, such as coronavirus disease. Overall, we found that glucosinolates and their hydrolysis products can potentially attenuate cytokine production and the onset of cytokine storms in diseased cells. In summary, glucosinolates could be beneficial in regulating cytokine production and preventing complications related to cytokine storms.
Collapse
Affiliation(s)
- Kingsley Ochar
- Council for Scientific and Industrial Research, Plant Genetic Resources Research Institute, Bunso P.O. Box 7, Ghana;
- National Agrobiodiversity Center, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea;
| | - Kanivalan Iwar
- National Agrobiodiversity Center, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea;
| | - Vadakkemuriyil Divya Nair
- Department of Plant Sciences, Central University of Himachal Pradesh, Shahpur Campus, Kangra District, Shahpur 176206, HP, India;
| | - Yun-Jo Chung
- National Creative Research Laboratory for Ca Signaling Network, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea;
| | - Bo-Keun Ha
- Department of Applied Plant Science, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seong-Hoon Kim
- National Agrobiodiversity Center, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea;
| |
Collapse
|
55
|
Vieira J, de Oliveira TVV, Queiroz LRR, Camargo CTS, Nardy A, Monteiro FR, do Amaral JB, Paixão V, Vaisberg M, Amirato GR, Dos Santos CAF, Durigon EL, Oliveira DBL, Aguiar AS, Alvares-Saraiva AM, Heller D, Mantoanelli PGV, Siqueira MF, da Silva Nali LH, Bachi ALL. Salivary assessment of the immune/inflammatory responses and oxidative stress in older adults vaccinated with CoronaVac or ChadOx-1. BMC Geriatr 2024; 24:807. [PMID: 39363197 PMCID: PMC11448442 DOI: 10.1186/s12877-024-05357-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 09/04/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Although important information concerning COVID-19 vaccination is available, the effects of the CoronaVac and ChadOx-1 vaccines on immunity and the redox balance in the upper airway mucosa of the aged population are not fully understood. Therefore, the aim of this study was to investigate the impacts of two doses of the CoronaVac or ChadOx-1 vaccine on immune/inflammatory responses and oxidative stress in the airway mucosa of older adults. METHODS Seventy-six older adults of both sexes, with a mean age of 75.1 ± 6.4 years, were separated according to vaccination status into the CoronaVac (n = 52) and ChadOx-1 (n = 24) groups. Saliva samples were collected before (pre) and 30 days after (post) the administration of the second dose of the CoronaVac or ChadOx-1 vaccine to assess the levels of antibodies (sIgA and IgG), antimicrobial peptides, cytokines, and oxidant/antioxidant agents. RESULTS The immunogenicity in the ChadOx-1 group was 37.5% for sIgA and 25% for IgG, while that in the CoronaVac group was 18.9% for sIgA and 13.2% for IgG. Intergroup analysis revealed that (1) lower levels of IFN-α, IFN-γ, and IL-10 and a greater IFN-γ/IL-10 ratio, in addition to a greater IL-6/IL-10 ratio, were found in both the pre- and postvaccination periods, and (2) lower levels of total sIgA, IL-12p70, IL-17A, TNF-α, and the IL-12p70/IL-10 ratio, in addition to higher levels of specific sIgA for SARS-CoV-2 antigens and lysozyme, were observed only in the postvaccination period in the ChadOx-1 group than in the CoronaVac group. Intragroup analysis revealed (1) a significant increase in the salivary levels of total peroxides in the postvaccination period compared to those in the prevaccination period in both volunteer groups; (2) a decrease in the levels of lysozyme and the ratio between total antioxidant capacity (TAC) and total peroxides in the postvaccination period in the CoronaVac group compared with those in the prevaccination period; and (3) decreases in the TNF-α, IL-6, and IL-12p70 levels, and the IL-12p70/IL-10 ratio in the ChadoX-1 group, as well as a higher lactoferrin concentration in the postvaccination period than in the prevaccination period. Several positive and negative correlations between the parameters assessed here were found. CONCLUSIONS In general, the ChadOx-1 group exhibited improvements in both immune/inflammatory responses and redox balance and greater immunogenicity than did the CoronaVac group.
Collapse
Affiliation(s)
- Jeniffer Vieira
- Faculty of Dentistry, Campus 1, Santo Amaro University (UNISA), São Paulo, Brazil
| | | | | | | | - Ariane Nardy
- Postgraduate Program in Health Sciences, Santo Amaro University (UNISA), São Paulo, Brazil
| | | | - Jônatas Bussador do Amaral
- ENT Research Lab, Department of Otorhinolaryngology-Head and Neck Surgery, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Vitória Paixão
- ENT Research Lab, Department of Otorhinolaryngology-Head and Neck Surgery, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Mauro Vaisberg
- ENT Research Lab, Department of Otorhinolaryngology-Head and Neck Surgery, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Gislene Rocha Amirato
- Mane Garrincha Sport Education Center, Sports Department of the Municipality of São Paulo (SEME), São Paulo, Brazil
| | - Carlos André Freitas Dos Santos
- Discipline of Geriatrics and Gerontology, Department of Medicine, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Postgraduate Program in Translational Medicine, Department of Medicine, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Edison Luiz Durigon
- Institute of Biomedical Science of University of São Paulo (USP), São Paulo, Brazil
- Scientific Platform Pasteur USP, São Paulo, Brazil
| | - Danielle Bruna Leal Oliveira
- Institute of Biomedical Science of University of São Paulo (USP), São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Andressa Simões Aguiar
- Institute of Biomedical Science of University of São Paulo (USP), São Paulo, Brazil
- Infection Control Service, São Luiz Gonzaga Hospital of Santa Casa de Misericordia of São Paulo, São Paulo, Brazil
| | | | - Débora Heller
- Postgraduate Program in Dentistry, Cruzeiro do Sul University, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
56
|
Meto A, Ndreu A, Tragaj E, D'Amico C, Meto A, Fiorillo L. Assessment of oral tissue alterations in patients diagnosed with SARS-CoV-2. Minerva Dent Oral Sci 2024; 73:272-278. [PMID: 37878242 DOI: 10.23736/s2724-6329.23.04870-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
BACKGROUND The aim of this study was to investigate oral mucosal changes in patients with confirmed moderate-scale severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. METHODS We analyzed 85 COVID-19 patients (50 males, 35 females) with an average age of 53.6 years, treated at the prehospital infectious disease center and Aldent University Clinic in Tirana, Albania, from May 2021 to June 2022. RESULTS Elevated C-reactive protein levels were observed in 82 patients (±44.20), with 20 patients showing significant fibrinogen increase (mean ± 5.85 g/L), and 22 patients having elevated D-Dimer (mean ± 336.6 mg/mL). Despite the absence of anticoagulant history, 13 patients exhibited bleeding. Xerodermia, xerostomia, and angular cheilitis were noted, with 41 patients displaying angular cheilitis. In 82 patients, oral mucosal and tongue examinations revealed color changes from white to bright yellow, with brown edema. Pigmentation in the fixed gingiva of upper and lower front teeth was observed in 35 patients. CONCLUSIONS Oral mucosal changes during COVID-19 appear more influenced by drug treatment and disease progression than the infection itself, suggesting that secondary factors play a significant role. Despite the oral cavity's potential for viral entry, these changes seem connected to other underlying causes.
Collapse
Affiliation(s)
- Aida Meto
- Department of Dentistry, Faculty of Dental Sciences, University of Aldent, Tirana, Albania
- School of Dentistry, Department of Clinical Microbiology, University of Modena and Reggio Emilia, Modena, Italy
- Department of Conservative Dentistry and Endodontics, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - Arben Ndreu
- Intensive Care Unit, Service of Infectious Diseases, Mother Theresa University Hospital Center, Tirana, Albania
- Department of Prosthetics and Dental Technology, Faculty of Dental Sciences, University of Aldent, Tirana, Albania
| | - Emiljano Tragaj
- Department of Dentistry, Faculty of Dental Sciences, University of Aldent, Tirana, Albania
| | - Cesare D'Amico
- Department of Dentistry, Faculty of Dental Sciences, University of Aldent, Tirana, Albania
- Department of Biomedical and Dental Sciences, and Morphological and Functional Imaging, University of Messina, Messina, Italy
| | - Agron Meto
- Department of Dentistry, Faculty of Dental Sciences, University of Aldent, Tirana, Albania
| | - Luca Fiorillo
- Department of Dentistry, Faculty of Dental Sciences, University of Aldent, Tirana, Albania -
- Department of Biomedical and Dental Sciences, and Morphological and Functional Imaging, University of Messina, Messina, Italy
- Department of Prosthodontics, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, India
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, Luigi Vanvitelli University of Campania, Naples, Italy
| |
Collapse
|
57
|
Yacoub MR, Ferlito A, Nettis E. Chronic urticaria: not only after COVID-19 vaccination. Curr Opin Allergy Clin Immunol 2024; 24:404-408. [PMID: 39079165 DOI: 10.1097/aci.0000000000001019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
PURPOSE OF REVIEW To resume the current literature about vaccination and the onset of chronic urticaria. RECENT FINDINGS The recent large-scale vaccination against SARS-CoV-2 targeting elderly, adult and children, has highlighted how vaccines can trigger onset or exacerbation of chronic urticaria. SUMMARY COVID-19 vaccines may act as triggers in the development of chronic spontaneous urticaria. More data regarding the other vaccines are necessary to evaluate a similar potential role. Proper education of patients with vaccine-induced chronic urticaria is essential to avoid vaccination hesitancy.
Collapse
Affiliation(s)
- Mona-Rita Yacoub
- Vita-Salute San Raffaele University
- IRCCS San Raffaele hospital, Internal Medicine Department, Milan
| | | | - Eustachio Nettis
- Policlinico di Bari, Department of Emergency and Organ Transplantation, School of Allergology and Clinical Immunology, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
58
|
Antoniou T, McCormack D, Tadrous M, Gomes T. Alpha-1 adrenergic antagonists and the risk of hospitalization or death in non-hospitalized patients with COVID-19: A population-based study. Fundam Clin Pharmacol 2024; 38:998-1007. [PMID: 38575851 DOI: 10.1111/fcp.13004] [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: 07/05/2023] [Revised: 01/29/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Alpha-1 receptor antagonists may interfere with IL-6 signaling and could therefore be a potential treatment for COVID-19. However, the effectiveness of these drugs in mitigating the risk of clinical deterioration among non-hospitalized patients with COVID-19 is unknown. OBJECTIVES The aim of this study is to examine the association between alpha-1 antagonist exposure and the 30-day risk of a hospital encounter or death in nonhospitalized patients with COVID-19. METHODS We conducted a population-based cohort study of Ontario residents aged 35 years and older who were eligible for public drug coverage and who had a positive test for SARS-CoV-2 between January 1, 2020, and March 1, 2021. We matched each individual receiving an alpha-1 antagonist at the time of their positive test with two non-exposed individuals using propensity scores. Our outcome was a composite of a hospital admission, emergency department visit, or death, 1 to 30 days following the positive test. RESULTS We matched 3289 alpha-1 antagonist exposed patients to 6189 unexposed patients. Overall, there was no difference in the 30-day risk of the primary outcome among patients exposed to alpha-1 antagonists at the time of their diagnosis relative to unexposed individuals (28.8% vs. 28.0%; OR 1.00, 95% CI 0.91 to 1.11). In a secondary analysis, individuals exposed to alpha-1 antagonists had a lower risk of death in the 30 days following a COVID diagnosis (OR 0.79; 95% CI 0.66 to 0.93). CONCLUSION Alpha-1 antagonists did not mitigate the 30-day risk of clinical deterioration in non-hospitalized patients with COVID-19. Our findings do not support the general repurposing of alpha-1 antagonists as a treatment for such patients, although there may be subgroups of patients in whom further research is warranted.
Collapse
Affiliation(s)
- Tony Antoniou
- Department of Family and Community Medicine, Unity Health Toronto, Toronto, Ontario, Canada
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Daniel McCormack
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Mina Tadrous
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Women's College Research Institute, Toronto, Ontario, Canada
| | - Tara Gomes
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Institute for Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
59
|
de Carvalho Braga G, Francisco GR, Bagatini MD. Current treatment of Psoriasis triggered by Cytokine Storm and future immunomodulation strategies. J Mol Med (Berl) 2024; 102:1187-1198. [PMID: 39212718 DOI: 10.1007/s00109-024-02481-1] [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: 03/16/2024] [Revised: 07/14/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Psoriasis is a chronic condition caused by an inflammation mediated mainly by cytokines and T cells. In COVID-19, the same type of imbalance is common, generating the Cytokine Storm and promoting a worsening in the skin conditions of patients with autoimmune disorders, such as Psoriasis. In this context, one of the main mediators of immune responses presented by SARS-CoV-2 infected patients is the Purinergic System. This immunological resource is capable of stimulating the hyperinflammatory state presented by infected individuals, mainly by the activity of the P2X7 receptor, culminating in the Cytokine Storm and consequently in the Psoriasis crisis. Currently, different drugs are used for patients with Psoriasis, such as immunosuppressants and small molecules; however, the safety of these drugs in infected patients has not been analyzed yet. In this context, studies are being developed to evaluate the possible administration of these traditional drugs to COVID-19 patients with Psoriasis crisis. Along with that, researchers must evaluate the potential of administrating P2X7 antagonists to these patients as well, improving both the systemic and the dermatological prognostics of patients, by reducing the Cytokine Storm and its general effects, but also avoiding the provocation of Psoriasis crisis.
Collapse
|
60
|
Furman S, Green K, Lane TE. COVID-19 and the impact on Alzheimer's disease pathology. J Neurochem 2024; 168:3415-3429. [PMID: 37850241 PMCID: PMC11024062 DOI: 10.1111/jnc.15985] [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: 08/18/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has rapidly escalated into a global pandemic that primarily affects older and immunocompromised individuals due to underlying clinical conditions and suppressed immune responses. Furthermore, COVID-19 patients exhibit a spectrum of neurological symptoms, indicating that COVID-19 can affect the brain in a variety of manners. Many studies, past and recent, suggest a connection between viral infections and an increased risk of neurodegeneration, raising concerns about the neurological effects of COVID-19 and the possibility that it may contribute to Alzheimer's disease (AD) onset or worsen already existing AD pathology through inflammatory processes given that both COVID-19 and AD share pathological features and risk factors. This leads us to question whether COVID-19 is a risk factor for AD and how these two conditions might influence each other. Considering the extensive reach of the COVID-19 pandemic and the devastating impact of the ongoing AD pandemic, their combined effects could have significant public health consequences worldwide.
Collapse
Affiliation(s)
- Susana Furman
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
| | - Kim Green
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
| | - Thomas E. Lane
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
- Department of Molecular Biology & Biochemistry, School of Biological Sciences, University of California, Irvine 92697, USA
- Center for Virus Research, University of California, Irvine 92697, USA
| |
Collapse
|
61
|
Shen Z, Yin Z, Wei S, Cong Z, Zhao F, Zhang H, Zhu X. LIPS and PaO 2/FiO 2 Combined Plasma Biomarkers Predict Onset of Acute Respiratory Distress Syndrome in Patients of High Risks in SICU: A Prospective Exploratory Study. Mediators Inflamm 2024; 2024:4936265. [PMID: 39319361 PMCID: PMC11421942 DOI: 10.1155/2024/4936265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/10/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024] Open
Abstract
Objective To explore and validate the value of clinical parameters combined with plasma biomarkers for predicting acute respiratory distress syndrome (ARDS) in patients of high risks in the surgical intensive care unit (SICU). Materials and Methods We conducted a prospective, observational study from January 2020 to December 2023, which enrolled 263 patients of high risks in the SICU of Peking University Third Hospital consecutively; they were classified as ARDS and non-ARDS according to whether ARDS occurred after enrollment. Collected clinical characteristics and blood samples within 24 hr of admission to SICU. Blood samples from the first day to the seventh day of SICU were collected from patients without ARDS, and patients with ARDS were collected until 1 day after ARDS onset, forming data based on time series. ELISA and CBA were used to measure plasma biomarkers. Endpoint of the study was the onset of ARDS. Cox proportional hazard regression analysis was used to find independent risk factors of the onset of ARDS, then constructed a nomogram and tested its goodness-of-fit. Results About 84 of 263 patients ended with ARDS. Univariate analysis found 15 risk factors showed differences between ARDS and non-ARDS, namely, interleukin 6, interleukin 8 (IL-8), angiopoietin Ⅱ, LIPS, APACHEⅡ, SOFA, PaO2/FiO2, age, sex, shock, sepsis, acute abdomen, pulmonary contusion, pneumonia, hepatic dysfunction. We included factors with p < 0.2 in multivariate analysis and showed LIPS, PaO2/FiO2, IL-8, and receptor for advanced glycation end-products (RAGE) of the first day were independent risk factors for ARDS in SICU, a model combining them was good in predicting ARDS (C-index was 0.864 in total patients of high risks). The median of the C-index was 0.865, showed by fivefold cross-validation in the train cohort or validation cohort. The calibration curve shows an agreement between the probability of predicting ARDS and the actual probability of occurrence. Decision curve analysis indicated that the model had clinical use value. We constructed a nomogram that had the ability to predict ARDS in patients of high risks in SICU. Conclusions LIPS, PaO2/FiO2, plasma IL-8, and RAGE of the first day were independent risk factors of the onset of ARDS. The predictive ability for ARDS can be greatly improved when combining clinical parameters and plasma biomarkers.
Collapse
Affiliation(s)
- Ziyuan Shen
- Department of Critical Care MedicinePeking University Third Hospital, Beijing 100191, China
| | - Zhongnan Yin
- BiobankPeking University Third Hospital, Beijing 100191, China
| | - Senhao Wei
- Department of Critical Care MedicinePeking University Third Hospital, Beijing 100191, China
| | - Zhukai Cong
- Department of Critical Care MedicinePeking University Third Hospital, Beijing 100191, China
| | - Feng Zhao
- Department of Critical Care MedicinePeking University Third Hospital, Beijing 100191, China
| | - Hua Zhang
- Center of EpidemiologyPeking University Third Hospital, Beijing 100191, China
| | - Xi Zhu
- Department of Critical Care MedicinePeking University Third Hospital, Beijing 100191, China
| |
Collapse
|
62
|
Li B, Bai J, Xiong Y, Guo D, Fu B, Deng G, Wu H. Understanding the mechanisms and treatments of long COVID to address future public health risks. Life Sci 2024; 353:122938. [PMID: 39084516 DOI: 10.1016/j.lfs.2024.122938] [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: 02/20/2024] [Revised: 07/20/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
The 2019 coronavirus disease (COVID-19), triggered by the severe acute respiratory syndrome coronavirus (SARS-CoV-2), has seen numerous individuals undergo and recover from it, drawing extensive attention to their health conditions. Extensive studies indicate that even after surpassing the acute phase of infection, patients continue to experience persistent symptoms such as fatigue, pain, depression, weakening, and anosmia. COVID-19 appears not to have concluded but rather to persist long-term in certain individuals, termed as "long COVID." This represents a heterogeneous ailment involving multiple organ systems, with a perceived complex and still elusive pathogenesis. Among patients with long COVID, observations reveal immune dysregulation, coagulation impairments, and microbial dysbiosis, considered potential mechanisms explaining sustained adverse outcomes post COVID-19. Based on the multifactorial nature, varied symptoms, and heterogeneity of long COVID, we have summarized several categories of current therapeutic approaches. Furthermore, the symptoms of long COVID resemble those of other viral illnesses, suggesting that existing knowledge may offer novel insights into long-term COVID implications. Here, we provide an overview of existing literature associated with long COVID and summarize potential mechanisms, treatment modalities, and other analogous conditions. Lastly, we underscore the inadequacies in long COVID treatment approaches and emphasize the significance of conducting further research and clinical trials.
Collapse
Affiliation(s)
- Bohao Li
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Junlu Bai
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Beibei Fu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Guohong Deng
- Department of Infectious Diseases, First Affiliated Hospital, Army Medical University, Chongqing, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
63
|
Timofeeva AM, Nikitin AO, Nevinsky GA. Circulating miRNAs in the Plasma of Post-COVID-19 Patients with Typical Recovery and Those with Long-COVID Symptoms: Regulation of Immune Response-Associated Pathways. Noncoding RNA 2024; 10:48. [PMID: 39311385 PMCID: PMC11417918 DOI: 10.3390/ncrna10050048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024] Open
Abstract
Following the acute phase of SARS-CoV-2 infection, certain individuals experience persistent symptoms referred to as long COVID. This study analyzed the patients categorized into three distinct groups: (1) individuals presenting rheumatological symptoms associated with long COVID, (2) patients who have successfully recovered from COVID-19, and (3) donors who have never contracted COVID-19. A notable decline in the expression of miR-200c-3p, miR-766-3p, and miR-142-3p was identified among patients exhibiting rheumatological symptoms of long COVID. The highest concentration of miR-142-3p was found in healthy donors. One potential way to reduce miRNA concentrations is through antibody-mediated hydrolysis. Not only can antibodies possessing RNA-hydrolyzing activity recognize the miRNA substrate specifically, but they also catalyze its hydrolysis. The analysis of the catalytic activity of plasma antibodies revealed that antibodies from patients with long COVID demonstrated lower hydrolysis activity against five fluorescently labeled oligonucleotide sequences corresponding to the Flu-miR-146b-5p, Flu-miR-766-3p, Flu-miR-4742-3p, and Flu-miR-142-3p miRNAs and increased activity against the Flu-miR-378a-3p miRNA compared to other patient groups. The changes in miRNA concentrations and antibody-mediated hydrolysis of miRNAs are assumed to have a complex regulatory mechanism that is linked to gene pathways associated with the immune system. We demonstrate that all six miRNAs under analysis are associated with a large number of signaling pathways associated with immune response-associated pathways.
Collapse
Affiliation(s)
- Anna M. Timofeeva
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Artem O. Nikitin
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
| | - Georgy A. Nevinsky
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
64
|
Mattos Pereira V, Thakar A, Nair S. Targeting iRhom2/ADAM17 attenuates COVID-19-induced cytokine release from cultured lung epithelial cells. Biochem Biophys Rep 2024; 39:101811. [PMID: 39253056 PMCID: PMC11382212 DOI: 10.1016/j.bbrep.2024.101811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/28/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, continues to pose a significant global health challenge, with acute respiratory distress syndrome (ARDS) being a major cause of mortality. Excessive cytokine release (cytokine storm) has been causally related to COVID-19-associated ARDS. While TNF-α inhibitors have shown potential in reducing inflammation, their broad effects on TNF-α signaling, including both pro- and anti-inflammatory pathways, present significant challenges and side effects in clinical use. Therefore, more precise therapeutic targets are urgently needed. ADAM17 is a key enzyme driving cytokine release, but its broad presence complicates direct inhibition. Targeting iRhom2, a regulator specific to immune cells that controls ADAM17's activity, offers a more focused and effective approach to reducing cytokine release. In this study, we hypothesized that targeted inhibition of ADAM-17/iRhom2 attenuates COVID-19-induced cytokine release in cultured lung epithelial cells. Human primary bronchial/tracheal epithelial cells challenged with COVID-19 pseudo-viral particles resulted in elevated cytokine release, which was attenuated following siRNA-mediated silencing of ADAM17 and iRhom2. Targeting ADAM-17/iRhom2 pathway may thus represent a strategy to overcome the COVID-19-associated ARDS.
Collapse
Affiliation(s)
- Vitoria Mattos Pereira
- University of Wyoming, School of Pharmacy and the Biomedical Sciences Interdisciplinary Graduate Program, Laramie, WY, 82071, USA
| | - Amit Thakar
- University of Wyoming, School of Pharmacy and the Biomedical Sciences Interdisciplinary Graduate Program, Laramie, WY, 82071, USA
| | - Sreejayan Nair
- University of Wyoming, School of Pharmacy and the Biomedical Sciences Interdisciplinary Graduate Program, Laramie, WY, 82071, USA
| |
Collapse
|
65
|
Kal M, Brzdęk M, Karska-Basta I, Rzymski P, Pinna A, Zarębska-Michaluk D. Characteristics of the radial peripapillary capillary network in patients with COVID-19 based on optical coherence tomography angiography: A literature review. Adv Med Sci 2024; 69:312-319. [PMID: 38972386 DOI: 10.1016/j.advms.2024.07.001] [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: 01/03/2024] [Revised: 03/20/2024] [Accepted: 07/05/2024] [Indexed: 07/09/2024]
Abstract
PURPOSE This review aimed to evaluate the significance of assessing radial peripheral capillary (RPC) network parameters by optical coherence tomography angiography (OCTA) in patients with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection confirmed by polymerase chain reaction. METHODS A literature search was conducted in the PubMed database to select high-quality reviews and original articles on the use of OCTA for visualizing the RPC network and calculating RPC parameters. RESULTS The study revealed that systemic hypoxia, hypercoagulable state, and inflammation affect the RPC network in patients with coronavirus disease 2019 (COVID-19). Reduced RPC parameters were observed early in the course of SARS-CoV-2 infection and after several months of follow-up. Additionally, there was a correlation between reduced RPC parameters and subsequent thinning of the retinal nerve fiber layer. CONCLUSIONS The OCTA examination of the retina and optic disc should be considered in patients with a history of COVID-19 to assess the impact of systemic hypoxia and inflammation on ocular function. Follow-up assessment of these patients is also necessary to understand the potential consequences of ischemia affecting the optic nerve, retina, and choroid.
Collapse
Affiliation(s)
- Magdalena Kal
- Collegium Medicum of Jan Kochanowski University in Kielce, Kielce, Poland; Ophthalmic Clinic of the Voivodeship Hospital in Kielce, Kielce, Poland
| | - Michał Brzdęk
- Collegium Medicum of Jan Kochanowski University in Kielce, Kielce, Poland.
| | - Izabella Karska-Basta
- Jagiellonian University Medical College, Faculty of Medicine, Department of Ophthalmology, Krakow, Poland; University Hospital, Clinic of Ophthalmology and Ocular Oncology, Krakow, Poland
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznan' University of Medical Sciences, Poznan, Poland
| | - Antonio Pinna
- Department of Medicine, Surgery, and Pharmacy, Ophthalmology Unit, University of Sassari, Sassari, Italy
| | - Dorota Zarębska-Michaluk
- Collegium Medicum of Jan Kochanowski University in Kielce, Kielce, Poland; Department of Infectious Disease, Provincial Hospital in Kielce, Kielce, Poland
| |
Collapse
|
66
|
Song X, Yu Y, Zhou H, Zhang Y, Mao Y, Wang H, Cao X, Zhu X, Li Z, Li L, Liu J, Peng X, Li Q. Acute Macular Neuroretinopathy Associated with COVID-19 Pandemic: A Real-world Observation Study. Asia Pac J Ophthalmol (Phila) 2024; 13:100103. [PMID: 39326527 DOI: 10.1016/j.apjo.2024.100103] [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: 03/21/2024] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 09/28/2024] Open
Abstract
PURPOSE To evaluate the clinical and retinal imaging features of Chinese patients with acute macular neuroretinopathy (AMN) associated with COVID-19. DESIGN A prospective observational study. METHODS Retinal imaging, including color fundus photography, near-infrared imaging (NIR), swept-source optical coherence tomography (SS-OCT), optical coherence tomography angiography (OCTA), and Humphrey perimetry, were conducted for each case. RESULTS All cases were included within the first three months following the pandemic outbreak. A total of 12 male patients (36.36 %) and 21 female patients (63.64 %) were prospectively recruited, and 29 cases (87.88 %) were bilaterally affected. The median interval between the onset of fever and the appearance of ocular symptoms was two days (range, 0.5-5.0 days). Apart from the outer retinal changes typical of AMN, changes in the inner retinal layers were observed, including intraretinal hemorrhage (8.06 %), cotton wool spots (9.68 %), and paracentral acute middle maculopathy (PAMM) (8.06 %). Smaller retinal inner nuclear layer hyperreflective speckles (RIHS) (41.94 %) were identified as a distinguishing feature from typical PAMM. Voids of vessel signals were found in the superficial (11.54 %), intermediate (82.69 %), and deep capillary plexus (98.08 %), and in the choriocapillaris (19.23 %) on OCTA. Humphrey perimetry illustrated central, paracentral, and peripheral scotomas. The occult lesions associated with AMN, PAMM, and some of the RIHS illustrated by OCT were visualized topographically and further confirmed by OCTA as perfusion defects. CONCLUSION An increase in AMN cases correlated with the SARS-CoV-2 outbreak. Additional features, including widespread inner retinal perfusion deficits, were observed and may serve as potential biomarkers for systemic microcirculation dysregulation in COVID-19.
Collapse
Affiliation(s)
- Xiaojia Song
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China; Department of Ophthalmology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, China
| | - Yajie Yu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Haiying Zhou
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Yongpeng Zhang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Yu Mao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Hong Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Xusheng Cao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Xiaoqing Zhu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Zhihua Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Lin Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Jinghua Liu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Xiaoyan Peng
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China.
| | - Qian Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China.
| |
Collapse
|
67
|
Polat HU, Yalcin HA, Köm D, Aksoy Ö, Abaci I, Ekiz AT, Serhatli M, Onarici S. Antiviral effect of cannabidiol on K18-hACE2 transgenic mice infected with SARS-CoV-2. J Cell Mol Med 2024; 28:e70030. [PMID: 39267200 PMCID: PMC11392655 DOI: 10.1111/jcmm.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 06/03/2024] [Accepted: 07/09/2024] [Indexed: 09/14/2024] Open
Abstract
The aim of this study was to determine the antiviral activity of cannabidiol (CBD) against SARS-CoV-2 infection. CBD is the second most studied cannabinoid obtained from Cannabis plants. We investigated the potential use of CBD, which has so far proven to have a positive effect on different diseases, in the SARS-CoV-2 infection. To test this, in vivo studies were carried out using K18-hACE2 transgenic mice. To reveal the potential therapeutic effect of the CBD at the histopathological and molecular level challenge experiments were performed. The study was designed with two groups (n = 10) and in the treatment group animals were infected with SARS-CoV-2 virus strain B.1.1.7 alpha before the administration of CBD. While the disease progressed and resulted in death in the control group that was infected by the virus alone, it was observed that the infection slowed down and the survival rate increased in the mice treated with CBD along with the virus. In this study, K18-hACE2 transgenic mice infected with the wild SARS-CoV-2 virus were used to investigate and prove the antiviral activity of CBD.
Collapse
Affiliation(s)
| | | | - Deniz Köm
- TUBITAK Marmara Research Center, Life Sciences, TUBITAKKocaeliTurkey
| | - Özge Aksoy
- TUBITAK Marmara Research Center, Life Sciences, TUBITAKKocaeliTurkey
- Molecular Biology and Genetics, Institute of SciencesYildiz Technical UniversityIstanbulTurkey
| | - Irem Abaci
- TUBITAK Marmara Research Center, Life Sciences, TUBITAKKocaeliTurkey
- Department of Biotechnology, Institute of BiotechnologyGebze Technical UniversityKocaeliTurkey
| | - Arzu Tas Ekiz
- TUBITAK Marmara Research Center, Life Sciences, TUBITAKKocaeliTurkey
| | - Müge Serhatli
- TUBITAK Marmara Research Center, Life Sciences, TUBITAKKocaeliTurkey
| | - Selma Onarici
- TUBITAK Marmara Research Center, Life Sciences, TUBITAKKocaeliTurkey
| |
Collapse
|
68
|
Liang Y, Quan X, Gu R, Meng Z, Gan H, Wu Z, Sun Y, Pan H, Han P, Liu S, Dou G. Repurposing existing drugs for the treatment ofCOVID-19/SARS-CoV-2: A review of pharmacological effects and mechanism of action. Heliyon 2024; 10:e35988. [PMID: 39247343 PMCID: PMC11379597 DOI: 10.1016/j.heliyon.2024.e35988] [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: 02/26/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Following the coronavirus disease-2019 outbreak caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), there is an ongoing need to seek drugs that target COVID-19. First off, novel drugs have a long development cycle, high investment cost, and are high risk. Second, novel drugs must be evaluated for activity, efficacy, safety, and metabolic performance, contributing to the development cycle, investment cost, and risk. We searched the Cochrane COVID-19 Study Register (including PubMed, Embase, CENTRAL, ClinicalTrials.gov, WHO ICTRP, and medRxiv), Web of Science (Science Citation Index, Emerging Citation Index), and WHO COVID-19 Coronaviral Disease Global Literature to identify completed and ongoing studies as of February 20, 2024. We evaluated the pharmacological effects, in vivo and in vitro data of the 16 candidates in the paper. The difficulty of studying these candidates in clinical trials involving COVID-19 patients, dosage of repurposed drugs, etc. is discussed in detail. Ultimately, Metformin is more suitable for prophylactic administration or mildly ill patients; the combination of Oseltamivir, Tamoxifen, and Dexamethasone is suitable for moderately and severely ill patients; and more clinical trials are needed for Azvudine, Ribavirin, Colchicine, and Cepharanthine to demonstrate efficacy.
Collapse
Affiliation(s)
- Yutong Liang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiaoxiao Quan
- Beijing Institute of Radiation Medicine, Beijing, China
- Scientific Experimental Center of Guangxi University of Chinese Medicine, Nanning, China
| | - Ruolan Gu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhiyun Meng
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui Gan
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhuona Wu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yunbo Sun
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Huajie Pan
- General Internal Medicine Department, Jingnan Medical District, PLA General Hospital, Beijing, China
| | - Peng Han
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Shuchen Liu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Guifang Dou
- Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
69
|
Fernandez SA, Pelaez-Prestel HF, Ras-Carmona A, Mozas-Gutierrez J, Reyes-Manzanas R, Reche PA. Eucalyptus Essential Oil Inhibits Cell Infection by SARS-CoV-2 Spike Pseudotyped Lentivirus. Biomedicines 2024; 12:1885. [PMID: 39200349 PMCID: PMC11351113 DOI: 10.3390/biomedicines12081885] [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: 07/19/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 09/02/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a public health concern due to infections with new SARS-CoV-2 variants. Therefore, finding effective preventive and therapeutic treatments against all SARS-CoV-2 variants is of great interest. In this study, we examined the capacity of eucalyptus essential oil (EEO) and eucalyptol (EOL) to prevent SARS-CoV-2 infection, using as a model SARS-CoV-2 Spike pseudotyped lentivirus (SARS-CoV-2 pseudovirus) and 293T cells transfected with human angiotensin-converting enzyme 2 (hACE2-293T cells). First, we determined the cytotoxicity of EEO and EOL using the MTT colorimetric assay, selecting non-cytotoxic concentrations ≤ 0.1% (v/v) for further analysis. Subsequently, we evaluated the capacity of EEO and EOL in cell cultures to preclude infection of hACE2-293T cells by SARS-CoV-2 pseudovirus, using a luciferase-based assay. We found that EEO and EOL significantly reduced SARS-CoV-2 pseudovirus infection, obtaining IC50 values of 0.00895% and 0.0042% (v/v), respectively. Likewise, EEO and EOL also reduced infection by vesicular stomatitis virus (VSV) pseudovirus, although higher concentrations were required. Hence, EEO and EOL may be able to inhibit SARS-CoV-2 infection, at least partially, through a Spike-independent pathway, supporting the implementation of aromatherapy with these agents as a cost-effective antiviral measure.
Collapse
Affiliation(s)
| | | | | | | | - Raquel Reyes-Manzanas
- Department of Immunology & O2, Faculty of Medicine, Complutense University of Madrid, Pza Ramon y Cajal, S/N, 28040 Madrid, Spain; (S.A.F.); (H.F.P.-P.); (A.R.-C.); (J.M.-G.)
| | - Pedro A. Reche
- Department of Immunology & O2, Faculty of Medicine, Complutense University of Madrid, Pza Ramon y Cajal, S/N, 28040 Madrid, Spain; (S.A.F.); (H.F.P.-P.); (A.R.-C.); (J.M.-G.)
| |
Collapse
|
70
|
Elnagi EA, Al-Maqati TN, Maawadh RM, AlBahrani S, Al Khalaf FS, Alzahrani FM, Nazzal W, Alanazi M, Abdali AS, Al Atawi AS, Al-Jamea LH, Alshehri AM, ALshammari AA, Suliman RS, Al Bassam I. A Retrospective Study: Evaluating the Impact of the COVID-19 Pandemic on Inflammatory Markers in Hospitalized Patients. Infect Dis Rep 2024; 16:735-749. [PMID: 39195007 DOI: 10.3390/idr16040056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/27/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic has had a significant impact globally, and understanding the relationship between inflammatory markers and disease progression is crucial for effective management. This retrospective study aimed to examine the association between various inflammatory markers, such as C-reactive protein (CRP), the erythrocyte sedimentation rate (ESR), lactate dehydrogenase (LDH), D-dimer, ferritin, and procalcitonin (PCT), and the characteristics of disease progression and outcomes in individuals affected by COVID-19. METHODS This study collected raw data from 470 patients who tested positive for SARS-CoV-2 using RT-PCR. RESULTS The logistic regression analysis revealed that elevated LDH levels were associated with male gender, ICU admission, low oxygen saturation (O2 < 93%), the need for mechanical ventilation, death, and the presence of lung infiltrates. Higher D-dimer levels were associated with older age, diabetes mellitus, cardiac disease, and low oxygen saturation. Ferritin levels were significantly associated with older age, ICU admission, low oxygen saturation, mechanical ventilation, and lung infiltrates. In contrast, CRP was only significant regarding lung infiltrates and procalcitonin levels were not significantly associated with any of the examined factors. CONCLUSION This study highlights the importance of monitoring key inflammatory markers, such as LDH, D-dimer, and ferritin, as they are significantly associated with the severity of COVID-19 illness. These findings can inform clinical decision-making and guide the development of targeted interventions to improve patient outcomes.
Collapse
Affiliation(s)
- Elmoeiz A Elnagi
- Department of Clinical Laboratory Sciences, Prince Sultan Military College of Health Sciences, Dhahran 34313, Saudi Arabia
| | - Thekra N Al-Maqati
- Department of Clinical Laboratory Sciences, Prince Sultan Military College of Health Sciences, Dhahran 34313, Saudi Arabia
| | - Rawan M Maawadh
- Department of Clinical Laboratory Sciences, Prince Sultan Military College of Health Sciences, Dhahran 34313, Saudi Arabia
| | - Salma AlBahrani
- Internal Medicine Department, King Fahad Military Medical Complex, Dhahran 34313, Saudi Arabia
| | - Faisal Salem Al Khalaf
- Department of Clinical Laboratory Sciences, Prince Sultan Military College of Health Sciences, Dhahran 34313, Saudi Arabia
| | - Faisal M Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Wael Nazzal
- Department of Obstetrics and Gynecology, King Fahad Military Medical Complex, Dhahran 34313, Saudi Arabia
| | - Maha Alanazi
- Department of Pathology and Laboratory Medicine, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia
| | - Abdullah S Abdali
- Department of Clinical Laboratory Sciences, Prince Sultan Military College of Health Sciences, Dhahran 34313, Saudi Arabia
| | - Amjad Saleh Al Atawi
- Department of Clinical Laboratory Sciences, Prince Sultan Military College of Health Sciences, Dhahran 34313, Saudi Arabia
| | - Lamiaa H Al-Jamea
- Department of Medical Laboratory, King Fahad Military Medical Complex, Dhahran 34313, Saudi Arabia
| | - Ahmad Mohammad Alshehri
- Department of Medical Laboratory, King Fahad Military Medical Complex, Dhahran 34313, Saudi Arabia
| | - Adnan Awad ALshammari
- Department of Medical Laboratory, King Fahad Military Medical Complex, Dhahran 34313, Saudi Arabia
| | - Rania Saad Suliman
- Department of Clinical Laboratory Sciences, Prince Sultan Military College of Health Sciences, Dhahran 34313, Saudi Arabia
| | - Ibrahim Al Bassam
- Plastic and Reconstructive Surgery Unit, King Fahd Military Medical Complex, Dhahran 34313, Saudi Arabia
| |
Collapse
|
71
|
Huang C, Yang W, Wang H, Huang S, Gao S, Li D, Liu J, Hou S, Feng W, Wang Z, Li F, Hao Z, Zhao X, Hu P, Pan Y. Flexible/Regenerative Nanosensor with Automatic Sweat Collection for Cytokine Storm Biomarker Detection. ACS NANO 2024; 18:21198-21210. [PMID: 39099110 DOI: 10.1021/acsnano.4c04456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
The real-time monitoring of low-concentration cytokines such as TNF-α in sweat can aid clinical physicians in assessing the severity of inflammation. The challenges associated with the collection and the presence of impurities can significantly impede the detection of proteins in sweat. This issue is addressed by incorporating a nanosphere array designed for automatic sweat transportation, coupled with a reusable sensor that employs a Nafion/aptamer-modified MoS2 field-effect transistor. The nanosphere array with stepwise wettability enables automatic collection of sweat and blocks impurities from contaminating the detection zone. This device enables direct detection of TNF-α proteins in undiluted sweat, within a detection range of 10 fM to 1 nM. The use of an ultrathin, ultraflexible substrate ensures stable electrical performance, even after up to 30 extreme deformations. The findings indicate that in clinical scenarios, this device could potentially provide real-time evaluation and management of patients' immune status via sweat testing.
Collapse
Affiliation(s)
- Cong Huang
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, China
- Key Laboratory of Micro-systems and Micro-structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, China
| | - Weisong Yang
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Hao Wang
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
- Key Laboratory of Micro-systems and Micro-structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, China
| | - Suichu Huang
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Shanshan Gao
- School of Electrical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Dongliang Li
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Jialin Liu
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Siyu Hou
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Weihao Feng
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Ziran Wang
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
- Key Laboratory of High-effciency and Clean Mechanical Manufacture of MOE, School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Feiran Li
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
- Key Laboratory of Micro-systems and Micro-structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, China
| | - Zhuang Hao
- School of Mechanical Engineering & Automation, Beihang University, Beijing 100191, China
| | - Xuezeng Zhao
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
- Key Laboratory of Micro-systems and Micro-structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, China
| | - PingAn Hu
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, China
- Key Laboratory of Micro-systems and Micro-structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, China
| | - Yunlu Pan
- State Key Laboratory of Robotics and Systems, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
- Key Laboratory of Micro-systems and Micro-structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
72
|
Nascimento RR, Aquino CC, Sousa JK, Gadelha KL, Cajado AG, Schiebel CS, Dooley SA, Sousa PA, Rocha JA, Medeiros JR, Magalhães PC, Maria-Ferreira D, Gois MB, C P Lima-Junior R, V T Wong D, Lima AM, Engevik AC, Nicolau LD, Vale ML. SARS-CoV-2 Spike protein triggers gut impairment since mucosal barrier to innermost layers: From basic science to clinical relevance. Mucosal Immunol 2024; 17:565-583. [PMID: 38555027 DOI: 10.1016/j.mucimm.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
Studies have reported the occurrence of gastrointestinal (GI) symptoms, primarily diarrhea, in COVID-19. However, the pathobiology regarding COVID-19 in the GI tract remains limited. This work aimed to evaluate SARS-CoV-2 Spike protein interaction with gut lumen in different experimental approaches. Here, we present a novel experimental model with the inoculation of viral protein in the murine jejunal lumen, in vitro approach with human enterocytes, and molecular docking analysis. Spike protein led to increased intestinal fluid accompanied by Cl- secretion, followed by intestinal edema, leukocyte infiltration, reduced glutathione levels, and increased cytokine levels [interleukin (IL)-6, tumor necrosis factor-α, IL-1β, IL-10], indicating inflammation. Additionally, the viral epitope caused disruption in the mucosal histoarchitecture with impairment in Paneth and goblet cells, including decreased lysozyme and mucin, respectively. Upregulation of toll-like receptor 2 and toll-like receptor 4 gene expression suggested potential activation of local innate immunity. Moreover, this experimental model exhibited reduced contractile responses in jejunal smooth muscle. In barrier function, there was a decrease in transepithelial electrical resistance and alterations in the expression of tight junction proteins in the murine jejunal epithelium. Additionally, paracellular intestinal permeability increased in human enterocytes. Finally, in silico data revealed that the Spike protein interacts with cystic fibrosis transmembrane conductance regulator (CFTR) and calcium-activated chloride conductance (CaCC), inferring its role in the secretory effect. Taken together, all the events observed point to gut impairment, affecting the mucosal barrier to the innermost layers, establishing a successful experimental model for studying COVID-19 in the GI context.
Collapse
Affiliation(s)
- Renata R Nascimento
- Post Graduation Program in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Cristhyane C Aquino
- Institute of Biomedicine for Brazilian Semi-Arid and Clinical Research Unit, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - José K Sousa
- Institute of Biomedicine for Brazilian Semi-Arid and Clinical Research Unit, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil; Division of Infectious Diseases & International Health, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kalinne L Gadelha
- Post Graduation Program in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Aurilene G Cajado
- Post Graduation Program in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Carolina S Schiebel
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba, Brazil
| | - Sarah A Dooley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Paulo A Sousa
- Biotechnology and Biodiversity Center Research, Lab of Inflammation and Translational Gastroenterology (LIGAT), Parnaíba Delta Federal University, Parnaíba, Brazil
| | - Jefferson A Rocha
- Biotechnology and Biodiversity Center Research, Lab of Inflammation and Translational Gastroenterology (LIGAT), Parnaíba Delta Federal University, Parnaíba, Brazil
| | - Jand R Medeiros
- Biotechnology and Biodiversity Center Research, Lab of Inflammation and Translational Gastroenterology (LIGAT), Parnaíba Delta Federal University, Parnaíba, Brazil
| | - Pedro C Magalhães
- Post Graduation Program in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Daniele Maria-Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba, Brazil
| | - Marcelo B Gois
- Faculty of Health Sciences, Federal University of Rondonópolis, Rondonópolis, Brazil
| | - Roberto C P Lima-Junior
- Post Graduation Program in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Deysi V T Wong
- Post Graduation Program in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Aldo M Lima
- Institute of Biomedicine for Brazilian Semi-Arid and Clinical Research Unit, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil; Division of Infectious Diseases & International Health, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Amy C Engevik
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lucas D Nicolau
- Institute of Biomedicine for Brazilian Semi-Arid and Clinical Research Unit, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil; Biotechnology and Biodiversity Center Research, Lab of Inflammation and Translational Gastroenterology (LIGAT), Parnaíba Delta Federal University, Parnaíba, Brazil; Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Brazil.
| | - Mariana L Vale
- Post Graduation Program in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
73
|
Ralchev N, Bradyanova SL, Doneva YV, Mihaylova N, Elefterova-Florova EV, Tchorbanov AI, Muñoz-Valle JF, Petralia MC, Nicoletti F, Fagone P. Cytokine Signatures and Immune Dysregulation in COVID-19 Patients: Transcriptomic and Serum Analysis. J Interferon Cytokine Res 2024; 44:379-385. [PMID: 38949969 DOI: 10.1089/jir.2024.0083] [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] [Indexed: 07/03/2024] Open
Abstract
COVID-19, caused by the SARS-CoV-2 virus, has caused a global health crisis, necessitating a deeper understanding of its pathophysiology. In this study, we explored the immune and hematological dynamics in COVID-19 patients to gain insights into disease severity and prognosis. Our findings revealed distinct cytokine profiles in moderate and severe cases. IL12A was significantly upregulated in peripheral blood mononuclear cells from moderate cases, suggesting a potential role in initiating an effective immune response. Conversely, severe cases exhibited downregulation of key pro-inflammatory cytokines (IL23A, TNFalpha, IL1B, and IFNG) alongside an upregulation of the immunosuppressive IL10, indicative of a dysregulated immune environment. Serum analysis showed elevated IL6 and IL10 levels in both moderate and severe cases, emphasizing their potential as markers for disease severity. Notably, no significant differences in serum cytokines were found between recovery and lethal cases. In lethal cases of COVID-19, elevated D-dimer, urea, and creatinine correlated with IL6 and IL10. This study contributes valuable information to the ongoing efforts to understand and manage the dysregulated immune responses underlying COVID-19 pathology.
Collapse
Affiliation(s)
- Nikola Ralchev
- Department of Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | - Yana Valerieva Doneva
- Department of Clinical Laboratory and Immunology, Military Medical Academy, Sofia, Bulgaria
| | - Nikolina Mihaylova
- Department of Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | - Andrey Ivanov Tchorbanov
- Department of Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - José Francisco Muñoz-Valle
- Institute of Research in Biomedical Sciences, University Center for Health Science, University of Guadalaja, Jalisco, Mexico
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
74
|
Anand P, D’Andrea E, Feldman W, Wang SV, Liu J, Brill G, DiCesare E, Lin KJ. A Dynamic Prognostic Model for Identifying Vulnerable COVID-19 Patients at High Risk of Rapid Deterioration. Pharmacoepidemiol Drug Saf 2024; 33:e5872. [PMID: 39135513 PMCID: PMC11418916 DOI: 10.1002/pds.5872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 09/25/2024]
Abstract
PURPOSE We aimed to validate and, if performance was unsatisfactory, update the previously published prognostic model to predict clinical deterioration in patients hospitalized for COVID-19, using data following vaccine availability. METHODS Using electronic health records of patients ≥18 years, with laboratory-confirmed COVID-19, from a large care-delivery network in Massachusetts, USA, from March 2020 to November 2021, we tested the performance of the previously developed prediction model and updated the prediction model by incorporating data after availability of COVID-19 vaccines. We randomly divided data into development (70%) and validation (30%) cohorts. We built a model predicting worsening in a published severity scale in 24 h by LASSO regression and evaluated performance by c-statistic and Brier score. RESULTS Our study cohort consisted of 8185 patients (Development: 5730 patients [mean age: 62; 44% female] and Validation: 2455 patients [mean age: 62; 45% female]). The previously published model had suboptimal performance using data after November 2020 (N = 4973, c-statistic = 0.60. Brier score = 0.11). After retraining with the new data, the updated model included 38 predictors including 18 changing biomarkers. Patients hospitalized after Jun 1st, 2021 (when COVID-19 vaccines became widely available in Massachusetts) were younger and had fewer comorbidities than those hospitalized before. The c-statistic and Brier score were 0.77 and 0.13 in the development cohort, and 0.73 and 0.14 in the validation cohort. CONCLUSION The characteristics of patients hospitalized for COVID-19 differed substantially over time. We developed a new dynamic model for rapid progression with satisfactory performance in the validation set.
Collapse
Affiliation(s)
- Priyanka Anand
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Elvira D’Andrea
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - William Feldman
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Shirley V. Wang
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Jun Liu
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Gregory Brill
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Elyse DiCesare
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Kueiyu Joshua Lin
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School
| |
Collapse
|
75
|
Hajiesmaeil Memar E, Tahghighi F, Yousefzadegan S, Sadeghirad P, Mousavi A, Zare Mahmoudabadi R, Saeidi H, Ayati M, Naderi S, Memarian S, Zeinalabedin S, Ashjaei B, Raji H, Tahernia L, Alimadadi H, Ziaee V. Acute Abdomen as a Clinical Presentation of COVID-19-Associated Multisystem Inflammatory Syndrome in Children. Case Rep Surg 2024; 2024:9941131. [PMID: 39119587 PMCID: PMC11306682 DOI: 10.1155/2024/9941131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Background On December 2019, a novel coronavirus disease (COVID-19) spread worldwide and became a pandemic. Multisystem inflammatory syndrome in children (MIS-C) due to cytokine release syndrome following COVID-19 presents with various manifestations. We hypothesize that one of the rare manifestations is acute abdomen. Case Presentation. In this case series, eight cases (five girls and three boys) of gastrointestinal (GI) involvement and acute abdomen were reported to be associated with the cytokine storm due to COVID-19 infection. All patients were of Iranian nationality (Caucasian ethnicity), with a mean age of 8.9 years (range 3.5-14). They all presented with fever and acute abdominal pain. Additionally, maculopapular rash and edema of the extremities were common presentations. Free fluid on abdominal ultrasound or computerized tomography (CT) scan was observed in all patients. All cases tested positive for COVID-19. In six cases, laparotomy or abdominal surgery was performed for a diagnosis of acute abdomen, but appendicitis was confirmed in only one case. None of the cases presented with phlegmon. Elevated serum lipase and amylase levels were noted in two cases. Seven patients received corticosteroid pulse therapy. Clinical symptoms improved after one or two doses, and all patients were discharged after 4 weeks of follow-up with no mortality or morbidity. Conclusion Patients experiencing unexplained acute abdominal pain along with fever, skin rash, and peripheral edema, who had a history of COVID-19 infection prior to or during the episode of acute abdomen symptoms, should be considered to have MIS-C. Furthermore, methylprednisolone pulse therapy could be a safe treatment option, reducing hospitalization duration in this patient population.
Collapse
Affiliation(s)
- Elmira Hajiesmaeil Memar
- Department of PediatricsTehran University of Medical Sciences, Tehran, Iran
- Children's Medical CenterPediatric Center of Excellence, Tehran, Iran
| | - Fatemeh Tahghighi
- Department of PediatricsTehran University of Medical Sciences, Tehran, Iran
- Children's Medical CenterPediatric Center of Excellence, Tehran, Iran
| | - Sedigheh Yousefzadegan
- Department of PediatricIran University of Medical Sciences, Tehran, Iran
- Firoozabadi HospitalIran University of Medical Sciences, Tehran, Iran
| | - Parisa Sadeghirad
- Department of PediatricsTehran University of Medical Sciences, Tehran, Iran
- Children's Medical CenterPediatric Center of Excellence, Tehran, Iran
| | - Ashraf Mousavi
- Department of PediatricIran University of Medical Sciences, Tehran, Iran
- Firoozabadi HospitalIran University of Medical Sciences, Tehran, Iran
| | - Ramin Zare Mahmoudabadi
- Department of PediatricIran University of Medical Sciences, Tehran, Iran
- Firoozabadi Clinical Research and Development Unit (FACRDU)Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Saeidi
- Department of PediatricIran University of Medical Sciences, Tehran, Iran
| | - Mehri Ayati
- Department of PediatricsSemnan University of Medical Sciences, Semnan, Iran
| | - Sahar Naderi
- Children's Medical CenterPediatric Center of Excellence, Tehran, Iran
| | - Sara Memarian
- Department of PediatricsTehran University of Medical Sciences, Tehran, Iran
- Children's Medical CenterPediatric Center of Excellence, Tehran, Iran
| | | | - Bahar Ashjaei
- Children's Medical CenterPediatric Center of Excellence, Tehran, Iran
- Department of SurgeryTehran University of Medical Sciences, Tehran, Iran
| | - Hojatollah Raji
- Children's Medical CenterPediatric Center of Excellence, Tehran, Iran
- Department of SurgeryTehran University of Medical Sciences, Tehran, Iran
| | - Leila Tahernia
- Children's Medical CenterPediatric Center of Excellence, Tehran, Iran
| | - Hosein Alimadadi
- Department of PediatricsTehran University of Medical Sciences, Tehran, Iran
- Children's Medical CenterPediatric Center of Excellence, Tehran, Iran
| | - Vahid Ziaee
- Department of PediatricsTehran University of Medical Sciences, Tehran, Iran
- Children's Medical CenterPediatric Center of Excellence, Tehran, Iran
- Pediatric Rheumatology research groupRheumatology Research CenterTehran University of Medical Science, Tehran, Iran
- Pediatric Rheumatology Society of Iran, Tehran, Iran
| |
Collapse
|
76
|
Hawryłkowicz V, Stasiewicz B, Maciejewska D, Sołek-Pastuszka J, Komorniak N, Skonieczna-Żydecka K, Martynova-Van Kley A, Stachowska E. The Link between Inflammation, Lipid Derivatives, and Microbiota Metabolites in COVID-19 Patients: Implications on Eating Behaviors and Nutritional Status. Int J Mol Sci 2024; 25:7899. [PMID: 39063142 PMCID: PMC11276903 DOI: 10.3390/ijms25147899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Extreme inflammation that continues even after infections can lead to a cytokine storm. In recent times, one of the most common causes of cytokine storm activation has been SARS-CoV-2 infection. A cytokine storm leads to dysregulation and excessive stimulation of the immune system, producing symptoms typical of post-COVID syndrome, including chronic fatigue, shortness of breath, joint pain, trouble concentrating (known as "brain fog"), and even direct organ damage in the heart, lungs, kidneys, and brain. This work summarizes the current knowledge regarding inflammation and the cytokine storm related to SARS-CoV-2 infection. Additionally, changes in lipid metabolism and microbiota composition under the influence of inflammation in COVID-19, along with the possible underlying mechanisms, are described. Finally, this text explores potential health implications related to changes in eating behaviors and nutritional status in COVID-19 patients. Although research on the cytokine storm is still ongoing, there is convincing evidence suggesting that severe immune and inflammatory responses during the acute phase of COVID-19 may lead to long-term health consequences. Understanding these links is key to developing treatment strategies and supporting patients after infection.
Collapse
Affiliation(s)
- Viktoria Hawryłkowicz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| | - Beata Stasiewicz
- Department of Human Nutrition, The Faculty of Food Science, University of Warmia and Mazury in Olsztyn, Sloneczna 45f, 10-718 Olsztyn, Poland
| | - Dominika Maciejewska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| | - Joanna Sołek-Pastuszka
- Department of Anesthesiology and Intensive Care, Pomeranian Medical University, 71-242 Szczecin, Poland;
| | - Natalia Komorniak
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| | | | | | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| |
Collapse
|
77
|
Ilczak T, Skoczynski S, Oclon E, Kucharski M, Strejczyk T, Jagosz M, Jedynak A, Wita M, Ćwiertnia M, Jędrzejek M, Dutka M, Waksmańska W, Bobiński R, Pakuła R, Kawecki M, Kukla P, Białka S. Assessment of the Severity of COVID-19 on the Basis of Examination and Laboratory Diagnostics in Relation to Computed Tomography Imagery of Patients Hospitalised Due to COVID-19-Single-Centre Study. Healthcare (Basel) 2024; 12:1436. [PMID: 39057579 PMCID: PMC11276777 DOI: 10.3390/healthcare12141436] [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: 06/20/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
From the moment the SARS-CoV-2 virus was identified in December 2019, the COVID-19 disease spread around the world, causing an increase in hospitalisations and deaths. From the beginning of the pandemic, scientists tried to determine the major cause that led to patient deaths. In this paper, the background to creating a research model was diagnostic problems related to early assessment of the degree of damage to the lungs in patients with COVID-19. The study group comprised patients hospitalised in one of the temporary COVID hospitals. Patients admitted to the hospital had confirmed infection with SARS-CoV-2. At the moment of admittance, arterial blood was taken and the relevant parameters noted. The results of physical examinations, the use of oxygen therapy and later test results were compared with the condition of the patients in later computed tomography images and descriptions. The point of reference for determining the severity of the patient's condition in the computer imagery was set for a mild condition as consisting of a percentage of total lung parenchyma surface area affected no greater than 30%, an average condition of between 30% and 70%, and a severe condition as greater than 70% of the lung parenchyma surface area affected. Patients in a mild clinical condition most frequently had mild lung damage on the CT image, similarly to patients in an average clinical condition. Patients in a serious clinical condition most often had average levels of damage on the CT image. On the basis of the collected data, it can be said that at the moment of admittance, BNP, PE and HCO3- levels, selected due to the form of lung damage, on computed tomography differed from one another in a statistically significant manner (p < 0.05). Patients can qualify for an appropriate group according to the severity of COVID-19 on the basis of a physical examination and applied oxygen therapy. Patients can qualify for an appropriate group according to the severity of COVID-19 on the basis of BNP, HCO3 and BE parameters obtained from arterial blood.
Collapse
Affiliation(s)
- Tomasz Ilczak
- Department of Emergency Medicine, Faculty of Health Sciences, University of Bielsko-Biala, 43-309 Bielsko-Biała, Poland; (M.Ć.); (M.K.)
| | - Szymon Skoczynski
- Department of Lung Diseases and Tuberculosis, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Ewa Oclon
- Centre for Experimental and Innovative Medicine, Laboratory of Recombinant Proteins Production, University of Agriculture in Krakow, 30-059 Kraków, Poland
| | - Mirosław Kucharski
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Al Mickiewicza 24/28, 30-059 Krakow, Poland;
| | - Tomasz Strejczyk
- Leszek Giec Upper-Silesian Medical Centre, Medical University of Silesia in Katowice, 40-287 Katowice, Poland;
| | - Marta Jagosz
- Students’ Scientific Association, Department of Anaesthesiology and Intensive Care, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-287 Katowice, Poland;
| | - Antonina Jedynak
- Students’ Scientific Association, Department of Pneumonology, School of Medicine in Katowice, Medical University of Silesia, 40-287 Katowice, Poland;
| | - Michał Wita
- Department of Cardiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-287 Katowice, Poland;
| | - Michał Ćwiertnia
- Department of Emergency Medicine, Faculty of Health Sciences, University of Bielsko-Biala, 43-309 Bielsko-Biała, Poland; (M.Ć.); (M.K.)
| | - Marek Jędrzejek
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, 40-287 Katowice, Poland;
| | - Mieczysław Dutka
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, 43-309 Bielsko-Biała, Poland; (M.D.); (R.B.)
| | - Wioletta Waksmańska
- Department of Public Health, Faculty of Health Sciences, University of Bielsko-Biala, 43-309 Bielsko-Biała, Poland;
| | - Rafał Bobiński
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, 43-309 Bielsko-Biała, Poland; (M.D.); (R.B.)
| | - Roch Pakuła
- Department of Cardiac Surgery, Cardiac and Lung Transplantation, Mechanical Circulatory Support, Silesian Centre for Heart Diseases, 41-800 Zabrze, Poland
| | - Marek Kawecki
- Department of Emergency Medicine, Faculty of Health Sciences, University of Bielsko-Biala, 43-309 Bielsko-Biała, Poland; (M.Ć.); (M.K.)
| | - Paweł Kukla
- Medical College, Jagiellonian University, 31-001 Kraków, Poland;
| | - Szymon Białka
- Department of Anesthesia and Intensive Therapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-287 Katowice, Poland;
| |
Collapse
|
78
|
Franco-Moreno A, Acedo-Gutiérrez MS, Casado-Suela MÁ, Labrador-San Martín N, de Carranza-López M, Ibáñez-Estéllez F, Hernández-Blanco C, Jiménez-Torres J, Vallejo-Maroto I, Romero-Pareja R, Peña-Lillo G, Escobar-Rodríguez I, Torres-Macho J. Effect of early administration of dexamethasone in patients with COVID-19 pneumonia without acute hypoxemic respiratory failure and risk of development of acute respiratory distress syndrome: EARLY-DEX COVID-19 trial. Front Med (Lausanne) 2024; 11:1385833. [PMID: 39086948 PMCID: PMC11290468 DOI: 10.3389/fmed.2024.1385833] [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: 02/13/2024] [Accepted: 06/03/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction and objectives Corticosteroids are among the drugs demonstrating a mortality benefit for coronavirus disease 2019 (COVID-19). The RECOVERY trial highlighted that dexamethasone reduced 28-day mortality for hospitalized COVID-19 patients requiring either supplemental oxygen or mechanical ventilation. It is noted that approximately 30% of COVID-19 patients, initially presenting with mild symptoms, will advance to acute respiratory distress syndrome (ARDS), especially those with detectable laboratory markers of inflammation indicative of disease progression. Our research aimed to explore the efficacy of dexamethasone in preventing the progression to ARDS in patients hospitalized with COVID-19 pneumonia who do not yet require additional oxygen but are at high risk of developing ARDS, potentially leading to a reduction in morbimortality. Methods In this multicenter, randomized, controlled trial, we evaluated the impact of dexamethasone on adult patients diagnosed with COVID-19 pneumonia who did not need supplementary oxygen at admission but were identified as having risk factors for ARDS. The risk of ARDS was determined based on specific criteria: elevated lactate dehydrogenase levels over 245 U/L, C-reactive protein levels exceeding 100 mg/L, and a lymphocyte count below 0.80 × 109/L. Participants were randomly allocated to either receive dexamethasone or the standard care. The primary endpoints included the incidence of moderate or severe ARDS and all-cause mortality within 30 days post-enrollment. Results One hundred twenty-six patients were randomized. Among them, 41 were female (30.8%), with a mean age of 48.8 ± 14.4 years. Ten patients in the dexamethasone group (17.2%) and ten patients in the control group (14.7%) developed moderate ARDS with no significant differences. Mechanical ventilation was required in six patients (4.7%), with four in the treatment group and two in the control group. There were no deaths during hospitalization or during follow-up. An intermediate analysis for futility showed some differences between the control and treatment groups (Z = 0.0284). However, these findings were within the margins close to the region where the null hypothesis would not be rejected. Conclusion In patients with COVID-19 pneumonia without oxygen needs but at risk of progressing to severe disease, early dexamethasone administration did not lead to a decrease in ARDS development. Clinical trial registration ClinicalTrials.gov, identifier NCT04836780.
Collapse
Affiliation(s)
- Anabel Franco-Moreno
- Internal Medicine Department, Hospital Universitario Infanta Leonor–Virgen de la Torre, Madrid, Spain
| | | | - Miguel Ángel Casado-Suela
- Internal Medicine Department, Hospital Universitario Infanta Leonor–Virgen de la Torre, Madrid, Spain
| | | | - María de Carranza-López
- Internal Medicine Department, Hospital Universitario Infanta Leonor–Virgen de la Torre, Madrid, Spain
| | | | | | | | | | - Rodolfo Romero-Pareja
- Hospital de Emergencias Enfermera Isabel Zendal, Madrid, Spain
- European University, Madrid, Spain
| | - Gabriela Peña-Lillo
- Emergency Department, Hospital Universitario Infanta Leonor–Virgen de la Torre, Madrid, Spain
| | - Ismael Escobar-Rodríguez
- Pharmacology Department, Hospital Universitario Infanta Leonor–Virgen de la Torre, Madrid, Spain
| | - Juan Torres-Macho
- Internal Medicine Department, Hospital Universitario Infanta Leonor–Virgen de la Torre, Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
79
|
Tekin A, Devarajan A, Sakata KK, Qamar S, Sharma M, Valencia Morales DJ, Malinchoc M, Talaei F, Welle S, Taji J, Khosa S, Sharma N, Brown M, Lal A, Bansal V, Khan SA, La Nou AT, Sanghavi D, Cartin-Ceba R, Kashyap R, Gajic O, Domecq JP, Azadeh N. Pneumomediastinum and pneumothorax in coronavirus disease-2019: Description of a case series and a matched cohort study. Heliyon 2024; 10:e33679. [PMID: 39055836 PMCID: PMC11269848 DOI: 10.1016/j.heliyon.2024.e33679] [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: 10/05/2023] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
OBJECTIVE To describe the characteristics of COVID-19 patients with pneumothorax and pneumomediastinum (PTX/PM) and their association with patient outcomes. PATIENTS AND METHODS Adults admitted to five Mayo Clinic hospitals with COVID-19 between 03/2020-01/2022 were evaluated. PTX/PM was defined by imaging. Descriptive analyses and a matched (age, sex, admission month, COVID-19 severity) cohort comparison was performed. Hospital mortality, length of stay (LOS), and predisposing factors were assessed. RESULTS Among 6663 patients, 197 had PTX/PM (3 %) (75 PM, 40 PTX, 82 both). The median age was 59, with 71 % males. Exposure to invasive and non-invasive mechanical ventilation and high-flow nasal cannula before PTX/PM were 42 %, 17 %, and 20 %, respectively. Among isolated PTX and PM/PTX patients 70 % and 53.7 % underwent an intervention, respectively, while 96 % of the PM-only group was followed conservatively.A total of 171 patients with PTX/PM were compared to 171 matched controls. PTX/PM patients had more underlying lung disease (40.9 vs. 23.4 %, p < 0.001) and lower median body mass index (BMI) (29.5 vs. 31.3 kg/m2, p = .007) than controls. Among patients with available data, PTX/PM patients had higher median positive end-expiratory and plateau pressures than controls; however, differences were not significant (10 vs. 8 cmH2O; p = 0.38 and 28 vs. 22 cmH2O; p = 0.11, respectively). PTX/PM patients had a higher odds of mortality (adjusted odds ratio [95%CI]: 3.37 [1.61-7.07]) and longer mean LOS (percent change [95%CI]: 39 [9-77]) than controls. CONCLUSION In COVID-19 patients with similar severity, PTX/PM patients had more underlying lung disease and lower BMI. They had significantly increased mortality and LOS.
Collapse
Affiliation(s)
- Aysun Tekin
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Anusha Devarajan
- Division of Pulmonary, Department of Medicine and Department of Critical Care Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Kenneth K. Sakata
- Division of Pulmonary, Department of Medicine and Department of Critical Care Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Shahraz Qamar
- Post-Baccalaureate Research Education Program, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mayank Sharma
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Fahimeh Talaei
- Division of Pulmonary, Department of Medicine and Department of Critical Care Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Stephanie Welle
- Division of Critical Care Medicine, Mayo Clinic Health System, Mankato, MN, USA
| | - Jamil Taji
- Division of Pulmonary Medicine, Division of Critical Care Medicine, Mayo Clinic Health Systems, Mankato, MN, USA
| | - Sandeep Khosa
- Division of Pulmonary Medicine, Division of Critical Care Medicine, Mayo Clinic Health Systems, Mankato, MN, USA
| | - Nikhil Sharma
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Meghan Brown
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Amos Lal
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Vikas Bansal
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Syed Anjum Khan
- Division of Critical Care Medicine, Mayo Clinic Health System, Mankato, MN, USA
| | - Abigail T. La Nou
- Division of Critical Care Medicine, Mayo Clinic Health System, Eau Claire, WI, USA
| | - Devang Sanghavi
- Department of Critical Care Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Rodrigo Cartin-Ceba
- Division of Pulmonary, Department of Medicine and Department of Critical Care Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Rahul Kashyap
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Research, WellSpan Health, York, PA, USA
| | - Ognjen Gajic
- Division of Critical Care Medicine, Mayo Clinic Health System, Eau Claire, WI, USA
| | - Juan P. Domecq
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Natalya Azadeh
- Division of Pulmonary, Department of Medicine and Department of Critical Care Medicine, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
80
|
Silva-Santos Y, Pagni RL, Gamon THM, de Azevedo MSP, Bielavsky M, Darido MLG, de Oliveira DBL, de Souza EE, Wrenger C, Durigon EL, Luvizotto MCR, Ackerman HC, Marinho CRF, Epiphanio S, Carvalho LJM. Lisinopril increases lung ACE2 levels and SARS-CoV-2 viral load and decreases inflammation but not disease severity in experimental COVID-19. Front Pharmacol 2024; 15:1414406. [PMID: 39070798 PMCID: PMC11282493 DOI: 10.3389/fphar.2024.1414406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/04/2024] [Indexed: 07/30/2024] Open
Abstract
COVID-19 causes more severe and frequently fatal disease in patients with pre-existing comorbidities such as hypertension and heart disease. SARS-CoV-2 virus enters host cells through the angiotensin-converting enzyme 2 (ACE2), which is fundamental in maintaining arterial pressure through the renin-angiotensin system (RAS). Hypertensive patients commonly use medications such as angiotensin-converting enzyme inhibitors (ACEi), which can modulate the expression of ACE2 and, therefore, potentially impact the susceptibility and severity of SARS-CoV-2 infection. Here we assessed whether treatment of ACE2-humanized (K18-hACE2) mice with the ACEi Lisinopril affects lung ACE2 levels and the outcome of experimental COVID-19. K18-hACE2 mice were treated for 21 days with Lisinopril 10 mg/kg and were then infected with 105 PFU of SARS-CoV-2 (Wuhan strain). Body weight, clinical score, respiratory function, survival, lung ACE2 levels, viral load, lung histology, and cytokine (IL-6, IL-33, and TNF-α) levels were assessed. Mice treated with Lisinopril for 21 days showed increased levels of ACE2 in the lungs. Infection with SARS-CoV-2 led to massive decrease in lung ACE2 levels at 3 days post-infection (dpi) in treated and untreated animals, but Lisinopril-treated mice showed a fast recovery (5dpi) of ACE2 levels. Higher ACE2 levels in Lisinopril-treated mice led to remarkably higher lung viral loads at 3 and 6/7dpi. Lisinopril-treated mice showed decreased levels of the pro-inflammatory cytokines IL-6 and TNF-α in the serum and lungs at 6/7dpi. Marginal improvements in body weight, clinical score and survival were observed in Lisinopril-treated mice. No differences between treated and untreated infected mice were observed in respiratory function and lung histology. Lisinopril treatment showed both deleterious (higher viral loads) and beneficial (anti-inflammatory and probably anti-constrictory and anti-coagulant) effects in experimental COVID-19. These effects seem to compensate each other, resulting in marginal beneficial effects in terms of outcome for Lisinopril-treated animals.
Collapse
Affiliation(s)
- Yasmin Silva-Santos
- Laboratory of Malaria Cellular and Molecular Immunopathology, Faculty of Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, University of São Paulo, São Paulo, Brazil
- Laboratory of Malaria Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Roberta Liberato Pagni
- Immunology Laboratory, Heart Institute, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Thais Helena Martins Gamon
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
| | - Marcela Santiago Pacheco de Azevedo
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
- Laboratory of Experimental Immunoparasitology, Institute of Biomedical Sciences, Department of Parasitology, University of São Paulo, São Paulo, Brazil
| | - Mônica Bielavsky
- Laboratory of Malaria Cellular and Molecular Immunopathology, Faculty of Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, University of São Paulo, São Paulo, Brazil
| | - Maria Laura Goussain Darido
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
| | - Danielle Bruna Leal de Oliveira
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Edmarcia Elisa de Souza
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edson Luiz Durigon
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
| | | | - Hans Christian Ackerman
- Physiology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, United States
| | - Claudio Romero Farias Marinho
- Laboratory of Experimental Immunoparasitology, Institute of Biomedical Sciences, Department of Parasitology, University of São Paulo, São Paulo, Brazil
| | - Sabrina Epiphanio
- Laboratory of Malaria Cellular and Molecular Immunopathology, Faculty of Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
81
|
Lipski D, Radziemski A, Wasiliew S, Wyrwa M, Szczepaniak-Chicheł L, Stryczyński Ł, Olasińska-Wiśniewska A, Urbanowicz T, Perek B, Tykarski A, Komosa A. Assessment of COVID-19 risk factors of early and long-term mortality with prediction models of clinical and laboratory variables. BMC Infect Dis 2024; 24:685. [PMID: 38982355 PMCID: PMC11234702 DOI: 10.1186/s12879-024-09592-7] [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: 04/07/2024] [Accepted: 07/04/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Coronavirus disease (COVID-19) may lead to serious complications and increased mortality. The outcomes of patients who survive the early disease period are burdened with persistent long-term symptoms and increased long-term morbidity and mortality. The aim of our study was to determine which baseline parameters may provide the best prediction of early and long-term outcomes. METHODS The study group comprised 141 patients hospitalized for COVID-19. Demographic data, clinical data and laboratory parameters were collected. The main study endpoints were defined as in-hospital mortality and 1-year mortality. The associations between the baseline data and the study endpoints were evaluated. Prediction models were created. RESULTS The in-hospital mortality rate was 20.5% (n = 29). Compared with survivors, nonsurvivors were significantly older (p = 0.001) and presented comorbidities, including diabetes (0.027) and atrial fibrillation (p = 0.006). Assessment of baseline laboratory markers and time to early death revealed negative correlations between time to early death and higher IL-6 levels (p = 0.032; Spearman rho - 0.398) and lower lymphocyte counts (p = 0.018; Pearson r -0.438). The one-year mortality rate was 35.5% (n = 50). The 1-year nonsurvivor subgroup was older (p < 0.001) and had more patients with arterial hypertension (p = 0.009), diabetes (p = 0.023), atrial fibrillation (p = 0.046) and active malignancy (p = 0.024) than did the survivor subgroup. The model composed of diabetes and atrial fibrillation and IL-6 with lymphocyte count revealed the highest value for 1-year mortality risk prediction. CONCLUSIONS Diabetes and atrial fibrillation, as clinical factors, and LDH, IL-6 and lymphocyte count, as laboratory determinants, are the best predictors of COVID-19 mortality risk.
Collapse
Affiliation(s)
- Dawid Lipski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland.
| | - Artur Radziemski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Stanisław Wasiliew
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał Wyrwa
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Ludwina Szczepaniak-Chicheł
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Łukasz Stryczyński
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Anna Olasińska-Wiśniewska
- Department of Cardiac Surgery and Transplantology, Chair of Cardio-Thoracic Surgery, Poznan University of Medical Sciences, ul. Długa 1/2, Poznan, 61-848, Poland
| | - Tomasz Urbanowicz
- Department of Cardiac Surgery and Transplantology, Chair of Cardio-Thoracic Surgery, Poznan University of Medical Sciences, ul. Długa 1/2, Poznan, 61-848, Poland
| | - Bartłomiej Perek
- Department of Cardiac Surgery and Transplantology, Chair of Cardio-Thoracic Surgery, Poznan University of Medical Sciences, ul. Długa 1/2, Poznan, 61-848, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Anna Komosa
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
82
|
Su L, Yu T, Zhang C, Huo P, Zhao Z. A prediction model for secondary invasive fungal infection among severe SARS-CoV-2 positive patients in ICU. Front Cell Infect Microbiol 2024; 14:1382720. [PMID: 39040601 PMCID: PMC11260608 DOI: 10.3389/fcimb.2024.1382720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Background The global COVID-19 pandemic has resulted in over seven million deaths, and IFI can further complicate the clinical course of COVID-19. Coinfection of COVID-19 and IFI (secondary IFI) pose significant threats not only to healthcare systems but also to patient lives. After the control measures for COVID-19 were lifted in China, we observed a substantial number of ICU patients developing COVID-19-associated IFI. This creates an urgent need for predictive assessment of COVID-19 patients in the ICU environment for early detection of suspected fungal infection cases. Methods This study is a single-center, retrospective research endeavor. We conducted a case-control study on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) positive patients. The cases consisted of patients who developed any secondary IFI during their ICU stay at Jilin University China-Japan Union Hospital in Changchun, Jilin Province, China, from December 1st, 2022, to August 31st, 2023. The control group consisted of SARS-CoV-2 positive patients without secondary IFI. Descriptive and comparative analyses were performed, and a logistic regression prediction model for secondary IFI in COVID-19 patients was established. Additionally, we observed an increased incidence of COVID-19-associated pulmonary aspergillosis (CAPA) during this pandemic. Therefore, we conducted a univariate subgroup analysis on top of IFI, using non-CAPA patients as the control subgroup. Results From multivariate analysis, the prediction model identified 6 factors that are significantly associated with IFI, including the use of broad-spectrum antibiotics for more than 2 weeks (aOR=4.14, 95% CI 2.03-8.67), fever (aOR=2.3, 95%CI 1.16-4.55), elevated log IL-6 levels (aOR=1.22, 95% CI 1.04-1.43) and prone position ventilation (aOR=2.38, 95%CI 1.15-4.97) as independent risk factors for COVID-19 secondary IFI. High BMI (BMI ≥ 28 kg/m2) (aOR=0.85, 95% CI 0.75-0.94) and the use of COVID-19 immunoglobulin (aOR=0.45, 95% CI 0.2-0.97) were identified as independent protective factors against COVID-19 secondary IFI. The Receiver Operating Curve (ROC) area under the curve (AUC) of this model was 0.81, indicating good classification. Conclusion We recommend paying special attention for the occurrence of secondary IFI in COVID-19 patients with low BMI (BMI < 28 kg/m2), elevated log IL-6 levels and fever. Additionally, during the treatment of COVID-19 patients, we emphasize the importance of minimizing the duration of broad-spectrum antibiotic use and highlight the potential of immunoglobulin application in reducing the incidence of IFI.
Collapse
Affiliation(s)
- Leilei Su
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Tong Yu
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, United States
| | - Chunmei Zhang
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Pengfei Huo
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhongyan Zhao
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
83
|
Sadowski J, Klaudel T, Rombel-Bryzek A, Bułdak RJ. Cognitive dysfunctions in the course of SARS‑CoV‑2 virus infection, including NeuroCOVID, frontal syndrome and cytokine storm (Review). Biomed Rep 2024; 21:103. [PMID: 38800038 PMCID: PMC11117100 DOI: 10.3892/br.2024.1791] [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: 12/11/2023] [Accepted: 04/05/2024] [Indexed: 05/29/2024] Open
Abstract
During the coronavirus disease 2019 (COVID-19) pandemic, cognitive impairment of varying degrees of severity began to be observed in a significant percentage of patients. The present study discussed the impact of immunological processes on structural and functional changes in the central nervous system and the related cognitive disorders. The purpose of the present review was to analyse and discuss available information from the scientific literature considering the possible relationship between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral infection and cognitive impairment, including NeuroCOVID, frontal syndrome and cytokine storm. A systematic literature review was conducted using: Google Scholar, Elsevier and the PubMed database. When searching for materials, the following keywords were used: 'cognitive dysfunctions', 'SARS-CoV-2', 'COVID-19', 'Neuro-SARS2', 'NeuroCOVID', 'frontal syndrome', 'cytokine storm', 'Long COVID-19'. A total of 96 articles were included in the study. The analysis focused on the characteristics of each study's materials, methods, results and conclusions. SARS-CoV-2 infection may induce or influence existing cognitive disorders of various nature and severity. The influence of immunological factors related to the response against SARS-CoV-2 on the disturbance of cerebral perfusion, the functioning of nerve cells and the neuroprotective effect has been demonstrated. Particular importance is attached to the cytokine storm and the related difference between pro- and anti-inflammatory effects, oxidative stress, disturbances in the regulation of the hypothalamic-pituitary-adrenal axis and the stress response of the body.
Collapse
Affiliation(s)
- Jakub Sadowski
- Student Scientific Society of Clinical Biochemistry and Regenerative Medicine, Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, 45-050 Opole, Poland
| | - Tomasz Klaudel
- Student Scientific Society of Clinical Biochemistry and Regenerative Medicine, Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, 45-050 Opole, Poland
| | - Agnieszka Rombel-Bryzek
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, 45-050 Opole, Poland
| | - Rafał Jakub Bułdak
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, 45-050 Opole, Poland
| |
Collapse
|
84
|
Negaresh M, Ghobadi H, Hoseininia S, Samadi Takaldani AH, Javanshir N, Iranijam E, Aslani MR. Evaluation of the Efficacy of Therapeutic and Prophylactic Anticoagulation in COVID-19 Patients With Venous Catheter and Its Correlation With Clinical Outcomes. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2024; 32. [DOI: 10.1097/ipc.0000000000001382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
IntroductionThe COVID-19 disease was first detected in December 2019, and since then, various treatments have been used to manage it. One such treatment method is therapeutic plasma exchange. This method involves implanting a venous catheter, which increases the risk of venous thromboembolism (VTE). Other risk factors for VTE include infections like COVID-19, inflammation, or malignancy.Materials and MethodsIn this retrospective study, patients with acute respiratory syndrome caused by COVID-19 who were hospitalized and had venous catheters inserted for therapeutic plasma exchange were enrolled. The prophylactic anticoagulant dose was started for all patients, and after the diagnosis of VTE, it was changed to the therapeutic dose. Patients' information, including demographic data, clinical information, and laboratory findings, was extracted from patients' records and recorded in a checklist designed for each patient.ResultsFrom a total of 168 patients, 26 were diagnosed with VTE (pulmonary embolism in 5 patients and deep vein thrombosis in 21 patients). The prevalence of VTE in COVID-19 patients with the venous catheter was 15.4%. The right femoral vein was the most used route for catheterization and had the highest occurrence of venous thromboses. The patients diagnosed with thrombosis showed a lower mortality rate, higher D-dimer and lactate dehydrogenase levels, and lower platelet counts.ConclusionsThis study showed a higher risk of VTE and subclinical thrombosis in COVID-19 patients with venous catheters. Continuous screening, higher doses of anticoagulants, and early removal of venous catheters are critical in preventing VTE and mortality.
Collapse
Affiliation(s)
- Mohammad Negaresh
- Department of Internal Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hassan Ghobadi
- Department of Internal Medicine (Pulmonology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Saeed Hoseininia
- Department of Internal Medicine (Pulmonology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Hossein Samadi Takaldani
- Department of Internal Medicine (Pulmonology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nima Javanshir
- Faculty of Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Effat Iranijam
- Department of Internal Medicine (Hematology Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Reza Aslani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
85
|
Wang X, Geng Z, Bao Y, Zhong J, Ma J, Cui X, Shi Y. Shufeng Jiedu capsule alleviates influenza A (H1N1) virus induced acute lung injury by regulating the lung inflammatory microenvironment. Heliyon 2024; 10:e33237. [PMID: 39021925 PMCID: PMC11252743 DOI: 10.1016/j.heliyon.2024.e33237] [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: 01/02/2024] [Revised: 05/13/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Background Death caused by respiratory tract infection is one of the leading causes of death in the world today. Shufeng Jiedu Capsule (SFJDC) is a traditional Chinese medicine that has been widely used clinically for coronavirus disease 2019 (COVID-19), H1N1 influenza virus pneumonia and other diseases. Its pharmacological effect is to inhibit inflammation and improve the body's ability to clear viruses. However, the mechanism of SFJDC in the treatment of viral pneumonia, especially its effect on the inflammatory-immune microenvironment of lung tissue remains unclear. Methods Mice with H1N1 influenza virus pneumonia were used as a model to verify the efficacy of SFJDC through death protection, lung index, viral load, and HE staining of lung tissue. The levels of inflammatory cytokines and chemokines in lung tissue were investigated by multi-analyte immunoassay. The number and proportion of cells in peripheral blood were detected by blood routine. The percentage of infiltrating immune cells in lung tissue was detected by flow cytometry and immunofluorescence. Results SFJDC (2.2 g/kg·d-1 and 1.1 g/kg·d-1) increased survival rate (P<0.01, P<0.05), prolonged the survival period of mice, and alleviated the histopathological damage in lung (P<0.01). SFJDC (2.2 g/kg·d-1, 1.1 g/kg·d-1 and 0.055 g/kg·d-1) increased body weight(P<0.01, P<0.05), improved activity status, reduced the lung index (P<0.01, P<0.05) and viral load (P<0.01). SFJDC (2.2 g/kg·d-1 and 1.1 g/kg·d-1) reduced interleukin-1β (IL-1β), interleukin-18(IL-18), tumour necrosis factor α (TNF-α), monocyte chemoattractant protein (MCP), chemokine (C-X-C motif) ligand 1 (CXCL1) (P<0.01, P<0.05), and SFJDC (2.2 g/kg·d-1) increased IL-10 levels (P<0.05) to regulate inflammation. SFJDC (2.2 g/kg·d-1) increased the percentages of CD4+ T cells (P<0.01), CD8+ T cells (P<0.05), and B cells(P<0.05), and decreased F4/80+ macrophages (P<0.05). Conclusion Our findings indicated that SFJDC could inhibit inflammation and lung injury while maintaining the function of the adaptive immune response mediated by T and B cells, and promote the clearance of the virus, thereby treating influenza A (H1N1) virus-induced pneumonia.
Collapse
Affiliation(s)
- Xiaorui Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Zihan Geng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Yanyan Bao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Juying Zhong
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Jing Ma
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Xiaolan Cui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Yujing Shi
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| |
Collapse
|
86
|
Liang P, Wei Z, Li R, Zhou E, Chen Z. Predictive value of hematocrit, serum albumin level difference, and fibrinogen-to-albumin ratio for COVID-19-associated acute respiratory failure. Heliyon 2024; 10:e33326. [PMID: 39021974 PMCID: PMC11253537 DOI: 10.1016/j.heliyon.2024.e33326] [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: 08/08/2023] [Revised: 06/16/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Background Acute respiratory failure is the main clinical manifestation and a major cause of death in patients with COVID-19. However, few reports on its prevention and control have been published because of the need for laboratory predictive indicators. This study aimed to evaluate the predictive value of hematocrit level, serum albumin level difference, and fibrinogen-to-albumin ratio for COVID-19-associated acute respiratory failure. Material and methods A total of 120 patients with COVID-19 from the First Affiliated Hospital of Anhui Medical University were selected between December 2022 and March 2023. Patients were divided into acute respiratory failure and non-acute respiratory failure groups and compared patient-related indicators between them using univariate and multivariate logistic regression analyses. Receiver operating characteristic analysis was performed to determine the discrimination accuracy. Results In total, 48 and 72 patients were enrolled in the acute respiratory failure and non-acute respiratory failure groups, respectively. The Quick COVID-19 Severity Index scores, fibrinogen-to-albumin ratio, hematocrit and serum albumin level difference, fibrinogen, and hematocrit levels were significantly higher in the acute respiratory failure group than in the non-acute respiratory failure group. A Quick COVID-19 Severity Index >7, fibrinogen-to-albumin ratio >0.265, and hematocrit and serum albumin level difference >12.792 had a 96.14 % positive predictive rate and a 94.06 % negative predictive rate. Conclusion Both fibrinogen-to-albumin ratio and hematocrit and serum albumin level difference are risk factors for COVID-19-associated acute respiratory failure. The Quick COVID-19 Severity Index score combined with fibrinogen-to-albumin ratio, and hematocrit and serum albumin level difference predict high and low risks with better efficacy and sensitivity than those of the Quick COVID-19 Severity Index score alone; therefore, these parameters can be used collectively as a risk stratification method for assessing patients with COVID-19.
Collapse
Affiliation(s)
| | | | | | - Enze Zhou
- Department of Emergency Intensive Care Unit, The First Affiliated Hospital of AnHui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| | - Zheng Chen
- Department of Emergency Intensive Care Unit, The First Affiliated Hospital of AnHui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| |
Collapse
|
87
|
Han H, Kim JE, Lee HJ. Effect of apigetrin in pseudo-SARS-CoV-2-induced inflammatory and pulmonary fibrosis in vitro model. Sci Rep 2024; 14:14545. [PMID: 38914619 PMCID: PMC11196261 DOI: 10.1038/s41598-024-65447-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024] Open
Abstract
SARS-CoV-2 has become a global public health problem. Acute respiratory distress syndrome (ARDS) is the leading cause of death due to the SARS-CoV-2 infection. Pulmonary fibrosis (PF) is a severe and frequently reported COVID-19 sequela. In this study, an in vitro model of ARDS and PF caused by SARS-CoV-2 was established in MH-S, THP-1, and MRC-5 cells using pseudo-SARS-CoV-2 (PSCV). Expression of proinflammatory cytokines (IL-6, IL-1β, and TNF-α) and HIF-1α was increased in PSCV-infected MH-S and THP-1 cells, ARDS model, consistent with other profiling data in SARS-CoV-2-infected patients have been reported. Hypoxia-inducible factor-1 alpha (HIF-1α) siRNA and cobalt chloride were tested using this in vitro model. HIF-1α knockdown reduces inflammation caused by PSCV infection in MH-S and THP-1 cells and lowers elevated levels of CTGF, COLA1, and α-SMA in MRC-5 cells exposed to CPMSCV. Furthermore, apigetrin, a glycoside bioactive dietary flavonoid derived from several plants, including Crataegus pinnatifida, which is reported to be a HIF-1α inhibitor, was tested in this in vitro model. Apigetrin significantly reduced the increased inflammatory cytokine (IL-6, IL-1β, and TNF-α) expression and secretion by PSCV in MH-S and THP-1 cells. Apigetrin inhibited the binding of the SARS-CoV-2 spike protein RBD to the ACE2 protein. An in vitro model of PF induced by SARS-CoV-2 was produced using a conditioned medium of THP-1 and MH-S cells that were PSCV-infected (CMPSCV) into MRC-5 cells. In a PF model, CMPSCV treatment of THP-1 and MH-S cells increased cell growth, migration, and collagen synthesis in MRC-5 cells. In contrast, apigetrin suppressed the increase in cell growth, migration, and collagen synthesis induced by CMPSCV in THP-1 and MH-S MRC-5 cells. Also, compared to control, fibrosis-related proteins (CTGF, COLA1, α-SMA, and HIF-1α) levels were over two-fold higher in CMPSV-treated MRC-5 cells. Apigetrin decreased protein levels in CMPSCV-treated MRC-5 cells. Thus, our data suggest that hypoxia-inducible factor-1 alpha (HIF-1α) might be a novel target for SARS-CoV-2 sequela therapies and apigetrin, representative of HIF-1alpha inhibitor, exerts anti-inflammatory and PF effects in PSCV-treated MH-S, THP-1, and CMPVSC-treated MRC-5 cells. These findings indicate that HIF-1α inhibition and apigetrin would have a potential value in controlling SARS-CoV-2-related diseases.
Collapse
Affiliation(s)
- Hengmin Han
- Department of Cancer Preventive Material Development, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Jung-Eun Kim
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Hyo-Jeong Lee
- Department of Cancer Preventive Material Development, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
| |
Collapse
|
88
|
Geanes ES, McLennan R, Pierce SH, Menden HL, Paul O, Sampath V, Bradley T. SARS-CoV-2 envelope protein regulates innate immune tolerance. iScience 2024; 27:109975. [PMID: 38827398 PMCID: PMC11140213 DOI: 10.1016/j.isci.2024.109975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/01/2024] [Accepted: 05/10/2024] [Indexed: 06/04/2024] Open
Abstract
Severe COVID-19 often leads to secondary infections and sepsis that contribute to long hospital stays and mortality. However, our understanding of the precise immune mechanisms driving severe complications after SARS-CoV-2 infection remains incompletely understood. Here, we provide evidence that the SARS-CoV-2 envelope (E) protein initiates innate immune inflammation, via toll-like receptor 2 signaling, and establishes a sustained state of innate immune tolerance following initial activation. Monocytes in this tolerant state exhibit reduced responsiveness to secondary stimuli, releasing lower levels of cytokines and chemokines. Mice exposed to E protein before secondary lipopolysaccharide challenge show diminished pro-inflammatory cytokine expression in the lung, indicating that E protein drives this tolerant state in vivo. These findings highlight the potential of the SARS-CoV-2 E protein to induce innate immune tolerance, contributing to long-term immune dysfunction that could lead to susceptibility to subsequent infections, and uncovers therapeutic targets aimed at restoring immune function following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Eric S. Geanes
- Genomic Medicine Center, Children’s Mercy Research Institute, Kansas City, MO, USA
| | - Rebecca McLennan
- Genomic Medicine Center, Children’s Mercy Research Institute, Kansas City, MO, USA
| | - Stephen H. Pierce
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Heather L. Menden
- Division of Neonatology, Children’s Mercy Research Institute, Kansas City, MO, USA
| | - Oishi Paul
- Genomic Medicine Center, Children’s Mercy Research Institute, Kansas City, MO, USA
| | - Venkatesh Sampath
- Division of Neonatology, Children’s Mercy Research Institute, Kansas City, MO, USA
- Department of Pediatrics, University of Missouri- Kansas City, Kansas City, MO, USA
| | - Todd Bradley
- Genomic Medicine Center, Children’s Mercy Research Institute, Kansas City, MO, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Pediatrics, University of Missouri- Kansas City, Kansas City, MO, USA
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, MO, USA
| |
Collapse
|
89
|
Chittasupho C, Umsumarng S, Srisawad K, Arjsri P, Phongpradist R, Samee W, Tingya W, Ampasavate C, Dejkriengkraikul P. Inhibition of SARS-CoV-2-Induced NLRP3 Inflammasome-Mediated Lung Cell Inflammation by Triphala-Loaded Nanoparticle Targeting Spike Glycoprotein S1. Pharmaceutics 2024; 16:751. [PMID: 38931873 PMCID: PMC11206841 DOI: 10.3390/pharmaceutics16060751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/24/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, poses a significant global health threat. The spike glycoprotein S1 of the SARS-CoV-2 virus is known to induce the production of pro-inflammatory mediators, contributing to hyperinflammation in COVID-19 patients. Triphala, an ancient Ayurvedic remedy composed of dried fruits from three plant species-Emblica officinalis (Family Euphorbiaceae), Terminalia bellerica (Family Combretaceae), and Terminalia chebula (Family Combretaceae)-shows promise in addressing inflammation. However, the limited water solubility of its ethanolic extract impedes its bioavailability. In this study, we aimed to develop nanoparticles loaded with Triphala extract, termed "nanotriphala", as a drug delivery system. Additionally, we investigated the in vitro anti-inflammatory properties of nanotriphala and its major compounds, namely gallic acid, chebulagic acid, and chebulinic acid, in lung epithelial cells (A549) induced by CoV2-SP. The nanotriphala formulation was prepared using the solvent displacement method. The encapsulation efficiency of Triphala in nanotriphala was determined to be 87.96 ± 2.60% based on total phenolic content. In terms of in vitro release, nanotriphala exhibited a biphasic release profile with zero-order kinetics over 0-8 h. A549 cells were treated with nanotriphala or its active compounds and then induced with 100 ng/mL of spike S1 subunit (CoV2-SP). The results demonstrate that chebulagic acid and chebulinic acid are the active compounds in nanotriphala, which significantly reduced cytokine release (IL-6, IL-1β, and IL-18) and suppressed the expression of inflammatory genes (IL-6, IL-1β, IL-18, and NLRP3) (p < 0.05). Mechanistically, nanotriphala and its active compounds notably attenuated the expression of inflammasome machinery proteins (NLRP3, ASC, and Caspase-1) (p < 0.05). In conclusion, the nanoparticle formulation of Triphala enhances its stability and exhibits anti-inflammatory properties against CoV2-SP-induction. This was achieved by suppressing inflammatory mediators and the NLRP3 inflammasome machinery. Thus, nanotriphala holds promise as a supportive preventive anti-inflammatory therapy for COVID-19-related chronic inflammation.
Collapse
Affiliation(s)
- Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Sonthaya Umsumarng
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand;
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kamonwan Srisawad
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (P.A.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Punnida Arjsri
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (P.A.)
| | - Rungsinee Phongpradist
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Weerasak Samee
- Department of Pharmaceutical Chemistry, Srinakharinwirot University, Ongkharak, Nakhon Nayok 26120, Thailand;
| | - Wipawan Tingya
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Chadarat Ampasavate
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Pornngarm Dejkriengkraikul
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (P.A.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
90
|
Marquez-Curtis LA, Elliott JAW. Mesenchymal stromal cells derived from various tissues: Biological, clinical and cryopreservation aspects: Update from 2015 review. Cryobiology 2024; 115:104856. [PMID: 38340887 DOI: 10.1016/j.cryobiol.2024.104856] [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: 11/28/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Mesenchymal stromal cells (MSCs) have become one of the most investigated and applied cells for cellular therapy and regenerative medicine. In this update of our review published in 2015, we show that studies continue to abound regarding the characterization of MSCs to distinguish them from other similar cell types, the discovery of new tissue sources of MSCs, and the confirmation of their properties and functions that render them suitable as a therapeutic. Because cryopreservation is widely recognized as the only technology that would enable the on-demand availability of MSCs, here we show that although the traditional method of cryopreserving cells by slow cooling in the presence of 10% dimethyl sulfoxide (Me2SO) continues to be used by many, several novel MSC cryopreservation approaches have emerged. As in our previous review, we conclude from these recent reports that viable and functional MSCs from diverse tissues can be recovered after cryopreservation using a variety of cryoprotectants, freezing protocols, storage temperatures, and periods of storage. We also show that for logistical reasons there are now more studies devoted to the cryopreservation of tissues from which MSCs are derived. A new topic included in this review covers the application in COVID-19 of MSCs arising from their immunomodulatory and antiviral properties. Due to the inherent heterogeneity in MSC populations from different sources there is still no standardized procedure for their isolation, identification, functional characterization, cryopreservation, and route of administration, and not likely to be a "one-size-fits-all" approach in their applications in cell-based therapy and regenerative medicine.
Collapse
Affiliation(s)
- Leah A Marquez-Curtis
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada, T6G 1H9; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada, T6G 1C9
| | - Janet A W Elliott
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada, T6G 1H9; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada, T6G 1C9.
| |
Collapse
|
91
|
De Lorenzo R, Mazza MG, Sciorati C, Leone R, Scavello F, Palladini M, Merolla A, Ciceri F, Bottazzi B, Garlanda C, Benedetti F, Rovere-Querini P, Manfredi AA. Post-COVID Trajectory of Pentraxin 3 Plasma Levels Over 6 Months and Their Association with the Risk of Developing Post-Acute Depression and Anxiety. CNS Drugs 2024; 38:459-472. [PMID: 38658499 DOI: 10.1007/s40263-024-01081-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND AND OBJECTIVES Clinical manifestations of coronavirus disease 2019 (COVID-19) often persist after acute disease resolution. Underlying molecular mechanisms are unclear. The objective of this original article was to longitudinally measure plasma levels of markers of the innate immune response to investigate whether they associate with and predict post-COVID symptomatology. METHODS Adult patients with previous severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection during the first pandemic wave who underwent the 6-month multidisciplinary follow-up were included. Plasma levels of pentraxin 3 (PTX3), the complement components C3a and C5a, and chitinase-3 like-protein-1 (CHI3L1) were measured at hospital admission during acute disease (baseline) and at 1 and 6 months after hospital discharge. Associations with post-COVID-19 sequelae at 6 months were investigated using descriptive statistic and multiple regression models. RESULTS Ninety-four COVID-19 patients were included. Baseline PTX3, C5a, C3a, and CHI3L1 did not predict post-COVID-19 sequelae. The extent of the reduction of PTX3 over time (delta PTX3) was associated with lower depressive and anxiety symptoms at 6 months (both p < 0.05). When entering sex, age, need for intensive care unit or non-invasive ventilation during hospital stay, psychiatric history, and baseline PTX3 as nuisance covariates into a generalized linear model (GLM), the difference between baseline and 6-month PTX3 levels (delta PTX3) significantly predicted depression (χ2 = 4.66, p = 0.031) and anxiety (χ2 = 4.68, p = 0.031) at 6 months. No differences in PTX3 levels or PTX3 delta were found in patients with or without persistent or new-onset other COVID-19 symptoms or signs at 6 months. Plasma levels of C3a, C5a, and CHI3L1 did not correlate with PTX3 levels at either time point and failed to associate with residual or de novo respiratory or systemic clinical manifestations of the disease at 6 months. CONCLUSIONS A lower reduction of plasma PTX3 after acute COVID-19 associates with the presence of depression and anxiety, suggesting an involvement of inflammation in post-COVID-19 psychopathology and a potential role of PTX3 as a biomarker.
Collapse
Affiliation(s)
- Rebecca De Lorenzo
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Innate Immunity and Tissue Remodeling, Department of Internal Medicine, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Mario G Mazza
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, San Raffaele Turro, Via Stamira d'Ancona 20, 20127, Milan, Italy.
| | - Clara Sciorati
- Unit of Innate Immunity and Tissue Remodeling, Department of Internal Medicine, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | | | - Mariagrazia Palladini
- Vita-Salute San Raffaele University, Milan, Italy
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, San Raffaele Turro, Via Stamira d'Ancona 20, 20127, Milan, Italy
| | - Aurora Merolla
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Innate Immunity and Tissue Remodeling, Department of Internal Medicine, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy
- Hematology and Bone Marrow Transplant Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Cecilia Garlanda
- IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Francesco Benedetti
- Vita-Salute San Raffaele University, Milan, Italy
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, San Raffaele Turro, Via Stamira d'Ancona 20, 20127, Milan, Italy
| | - Patrizia Rovere-Querini
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Innate Immunity and Tissue Remodeling, Department of Internal Medicine, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Angelo A Manfredi
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Autoimmunity and Vascular Inflammation, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
92
|
Chen M, Shu W, Zhang J, Huang H, Liu J. Mechanisms and clinical application of Xuebijing injection, a traditional Chinese herbal medicine–a systematic review. ADVANCES IN TRADITIONAL MEDICINE 2024; 24:403-412. [DOI: 10.1007/s13596-023-00702-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/10/2023] [Indexed: 01/04/2025]
|
93
|
Özcan D, Özçelik F, Mammadov R, Aktaş M, Altındağ F, Alkan AA, Karapapak M, Altuner D, Süleyman H. Biochemical and histopathological evaluation of systemic and ocular toxicity of favipiravir in rats. Cutan Ocul Toxicol 2024; 43:105-112. [PMID: 38174703 DOI: 10.1080/15569527.2023.2300788] [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: 07/20/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
Purpose: Favipiravir (FAV) used against COVID-19 is an antiviral drug that causes adverse reactions, such as hyperuricaemia, liver damage, and hematopoetic toxicity. The aim of the study was to investigate the systemic and ocular side-effects of FAV in rats, for the first time.Materials and methods: A total of 18 albino male Wistar rats were used in the study. The rats were divided into 3 groups as the healthy group (HG), the group given 50 mg/kg/day favipiravir (FAV50), and the group given 200 mg/kg/d favipiravir (FAV200). These doses were given to the experimental groups for one week. At the end of the experiment histopathological examinations were performed on the conjunctiva and sclera of the eye. In addition, malondialdehyde (MDA), total glutathione (tGSH), superoxide dismutase (SOD), interleukin-1β (IL-1β), and tumor necrosis factor alpha (TNF-α) levels were measured in blood samples taken from rats. Results: Compared to HG, the MDA (1.37 ± 0.61 vs. 4.82 ± 1.40 µmol/mL), IL-1β (2.52 ± 1.14 vs. 6.67 ± 1.99 pg/mL), and TNF-α levels (3.28 ± 1.42 vs. 8.53 ± 3.06 pg/mL) of the FAV200 group were higher. The levels of tGSH (7.58 ± 1.98 vs. 2.50 ± 0.98 nmol/mL) and SOD (13.63 ± 3.43 vs. 3.81 ± 1.43 U/mL) the FAV200 group were lower than the HG (p < 0.05, for all). The degree of damage to the cornea and sclera of the FAV200 group was quite high according to HG (p < 0.001). Conclusions: FAV can cause damage to rat conjunctiva and sclera by increasing oxidant stress and inflammation at high dose.
Collapse
Affiliation(s)
- Delil Özcan
- Department of Ophthalmology, University of Health Sciences, Seyrantepe Hamidiye Etfal Training and Research Hospital, İstanbul, Turkey
| | - Fatih Özçelik
- Department of Medical Biochemistry, University of Health Sciences, Seyrantepe Hamidiye Etfal Training and Research Hospital, İstanbul, Turkey
| | - Renad Mammadov
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Mehmet Aktaş
- Department of Biochemistry, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Fikret Altındağ
- Department of Histology and Embryology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | | | - Murat Karapapak
- Department of Ophthalmology, Başakşehir Çam and Sakura City Hospital, İstanbul, Turkey
| | - Durdu Altuner
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Halis Süleyman
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| |
Collapse
|
94
|
Lukhna K, do Carmo HRP, Castillo AR, Davidson SM, Geffen H, Giesz S, Golforoush P, Bovi TG, Gorag D, Salama A, Imamdin A, Kalkhoran S, Lecour S, Perroud MW, Ntsekhe M, Sposito AC, Yellon DM. Effect of Remote Ischaemic Conditioning on the Inflammatory Cytokine Cascade of COVID-19 (RIC in COVID-19): a Randomized Controlled Trial. Cardiovasc Drugs Ther 2024; 38:433-445. [PMID: 36445625 PMCID: PMC9707178 DOI: 10.1007/s10557-022-07411-2] [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] [Accepted: 11/20/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Patients hospitalized with COVID-19 may develop a hyperinflammatory, dysregulated cytokine "storm" that rapidly progresses to acute respiratory distress syndrome, multiple organ dysfunction, and even death. Remote ischaemic conditioning (RIC) has elicited anti-inflammatory and cytoprotective benefits by reducing cytokines following sepsis in animal studies. Therefore, we investigated whether RIC would mitigate the inflammatory cytokine cascade induced by COVID-19. METHODS We conducted a prospective, multicentre, randomized, sham-controlled, single-blind trial in Brazil and South Africa. Non-critically ill adult patients with COVID-19 pneumonia were randomly allocated (1:1) to receive either RIC (intermittent ischaemia/reperfusion applied through four 5-min cycles of inflation (20 mmHg above systolic blood pressure) and deflation of an automated blood-pressure cuff) or sham for approximately 15 days. Serum was collected following RIC/sham administration and analyzed for inflammatory cytokines using flow cytometry. The endpoint was the change in serum cytokine concentrations. Participants were followed for 30 days. RESULTS Eighty randomized participants (40 RIC and 40 sham) completed the trial. Baseline characteristics according to trial intervention were overall balanced. Despite downward trajectories of all cytokines across hospitalization, we observed no substantial changes in cytokine concentrations after successive days of RIC. Time to clinical improvement was similar in both groups (HR 1.66; 95% CI, 0.938-2.948, p 0.08). Overall RIC did not demonstrate a significant impact on the composite outcome of all-cause death or clinical deterioration (HR 1.19; 95% CI, 0.616-2.295, p = 0.61). CONCLUSION RIC did not reduce the hypercytokinaemia induced by COVID-19 or prevent clinical deterioration to critical care. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04699227.
Collapse
Affiliation(s)
- Kishal Lukhna
- Division of Cardiology, Faculty of Health Sciences, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Helison R P do Carmo
- Atherosclerosis and Vascular Biology Laboratory, State University of Campinas, Campinas, Brazil
| | | | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Hayli Geffen
- Division of Cardiology, Faculty of Health Sciences, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Sara Giesz
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Pelin Golforoush
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Ticiane Gonçalez Bovi
- Atherosclerosis and Vascular Biology Laboratory, State University of Campinas, Campinas, Brazil
| | - Diana Gorag
- Atherosclerosis and Vascular Biology Laboratory, State University of Campinas, Campinas, Brazil
| | - Alan Salama
- Cape Heart Institute, University of Cape Town, Cape Town, South Africa
- The Royal Free Hospital, University College London, Pond St, London, NW3 2QG, UK
| | - Aqeela Imamdin
- Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Siavash Kalkhoran
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sandrine Lecour
- Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Mauricio W Perroud
- Atherosclerosis and Vascular Biology Laboratory, State University of Campinas, Campinas, Brazil
| | - Mpiko Ntsekhe
- Division of Cardiology, Faculty of Health Sciences, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Andrei C Sposito
- Atherosclerosis and Vascular Biology Laboratory, State University of Campinas, Campinas, Brazil
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
95
|
Pierre CN, Adams LE, Higgins JS, Anasti K, Goodman D, Mielke D, Stanfield-Oakley S, Powers JM, Li D, Rountree W, Wang Y, Edwards RJ, Alam SM, Ferrari G, Tomaras GD, Haynes BF, Baric RS, Saunders KO. Non-neutralizing SARS-CoV-2 N-terminal domain antibodies protect mice against severe disease using Fc-mediated effector functions. PLoS Pathog 2024; 20:e1011569. [PMID: 38900807 PMCID: PMC11218955 DOI: 10.1371/journal.ppat.1011569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 07/02/2024] [Accepted: 04/26/2024] [Indexed: 06/22/2024] Open
Abstract
Antibodies perform both neutralizing and non-neutralizing effector functions that protect against certain pathogen-induced diseases. A human antibody directed at the SARS-CoV-2 Spike N-terminal domain (NTD), DH1052, was recently shown to be non-neutralizing, yet it protected mice and cynomolgus macaques from severe disease. The mechanisms of NTD non-neutralizing antibody-mediated protection are unknown. Here we show that Fc effector functions mediate NTD non-neutralizing antibody (non-nAb) protection against SARS-CoV-2 MA10 viral challenge in mice. Though non-nAb prophylactic infusion did not suppress infectious viral titers in the lung as potently as neutralizing antibody (nAb) infusion, disease markers including gross lung discoloration were similar in nAb and non-nAb groups. Fc functional knockout substitutions abolished non-nAb protection and increased viral titers in the nAb group. Fc enhancement increased non-nAb protection relative to WT, supporting a positive association between Fc functionality and degree of protection from SARS-CoV-2 infection. For therapeutic administration of antibodies, non-nAb effector functions contributed to virus suppression and lessening of lung discoloration, but the presence of neutralization was required for optimal protection from disease. This study demonstrates that non-nAbs can utilize Fc-mediated mechanisms to lower viral load and prevent lung damage due to coronavirus infection.
Collapse
Affiliation(s)
- Camille N. Pierre
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Lily E. Adams
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jaclyn S. Higgins
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Kara Anasti
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Derrick Goodman
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Dieter Mielke
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sherry Stanfield-Oakley
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - John M. Powers
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Dapeng Li
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Wes Rountree
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Yunfei Wang
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Robert J. Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - S. Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, United States of America
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, United States of America
- Department of Immunology, Duke University, Durham, North Carolina, United States of America
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University, Durham, North Carolina, United States of America
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Kevin O. Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, United States of America
- Department of Immunology, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
96
|
Ameri A, Farashahinejad M, Davoodian P, Safa O, Hassaniazad M, Parsaii M, Heidari B, Hassanipour S, Akhlaghi B, Fathalipour M. The efficacy and safety of ginger (Zingiber officinale) rhizome extract in outpatients with COVID-19: A randomized double-blind placebo-control clinical trial. Medicine (Baltimore) 2024; 103:e38289. [PMID: 39259072 PMCID: PMC11142819 DOI: 10.1097/md.0000000000038289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/13/2024] [Accepted: 04/26/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Ginger, a potent antiviral, anti-inflammatory, and antioxidant remedy, is a potential therapeutic option for COVID-19. However, there was not enough clinical evidence about ginger and COVID-19. We evaluated the efficacy and safety of ginger on clinical and paraclinical features in outpatients with COVID-19. METHODS In this randomized controlled trial, the outpatients with confirmed COVID-19 were randomly assigned in a 1:1 ratio to receive ginger (1000 mg 3 times a day for 7 days) or placebo. The primary outcome was viral clearance after the end of the intervention. Oxygen saturation (SPO2), body temperature, respiratory rate (RR), hospital admission, and the incidence of adverse events were also assessed. RESULTS A total of 84 patients (42 in the ginger and 42 in the control groups) were randomized. The viral clearance was not statistically improved in the ginger group (41.6%) compared to the placebo group (42.8%). The findings indicated that SPO2, body temperature, and RR had no significant difference between the groups at the end of the intervention. The imaging finding indicated pulmonary infiltrate significantly reduced on the 7th day of the intervention in the ginger group. The percentage of patients with SPO2 <96% in the ginger group decreased over the study compared to the placebo group. Moreover, the need for hospital admission and the incidence of adverse drug events were not different between the groups over the follow-up period. CONCLUSIONS Ginger had no significant impact on the clinical and paraclinical parameters of patients. However, this intervention demonstrated a safe profile of adverse events and reduced pulmonary infiltrate. TRIAL REGISTRATION The trial was registered as IRCT20200506047323N1.
Collapse
Affiliation(s)
- Ali Ameri
- Student Research Committee, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mehdi Farashahinejad
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Parivash Davoodian
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Omid Safa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mehdi Hassaniazad
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohsen Parsaii
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Behnoosh Heidari
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Soheil Hassanipour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Boshra Akhlaghi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad Fathalipour
- Endocrinology and Metabolic Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
97
|
Ogiwara M, Ihara H, Muto Y, Haba M, Nakazawa H, Hotta S, Jo H, Hayama N, Honma Y, Hoshi S, Fujii M, Takahashi K. The inciting factor for bradycardia in COVID-19 patients: a potential harm of steroid treatment. J Thorac Dis 2024; 16:2835-2844. [PMID: 38883622 PMCID: PMC11170389 DOI: 10.21037/jtd-23-1382] [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: 09/02/2023] [Accepted: 03/15/2024] [Indexed: 06/18/2024]
Abstract
Background The coronavirus disease 2019 (COVID-19) is a condition caused by the novel severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2). Although several papers have reported the presence bradycardia in patients with COVID-19, the pathophysiology behind this remains unclear. Therefore, we investigated the presence of bradycardia in patients with COVID-19. Methods We conducted a retrospective cohort study in a total of 153 patients with COVID-19 and 90 patients with influenza who were hospitalized in our hospital from January 1, 2020 to December 31, 2021 and from January 1, 2014 to December 31, 2021, respectively. Data were collected from patient medical records, which included sex, age, duration of hospitalization, pneumonia complications, supplemental oxygen therapy, antiviral treatment, past history, and vital signs. Results After adjustment, the incidence of bradycardia and steroid use in patients with COVID-19 were significantly higher than those in patients with influenza (P=0.007 and P<0.001, respectively). We then compared the detailed characteristics of patients with COVID-19 to evaluate risk factors for bradycardia. Multivariate logistic regression analysis revealed that steroid use was significantly related to bradycardia [P=0.031; odds ratio (OR): 3.67; 95% confidence interval (CI): 1.12-11.96]. Overall, results showed a higher incidence of bradycardia in patients with COVID-19 who received steroid treatment. Conclusions Our results showed that steroid treatment in patients with COVID-19 may be associated with the incidence of bradycardia.
Collapse
Affiliation(s)
- Misa Ogiwara
- Department of Respiratory Medicine, Juntendo University, Faculty of Medicine & Graduate School of Medicine, Tokyo, Japan
| | - Hiroaki Ihara
- Department of Respiratory Medicine, Juntendo University, Faculty of Medicine & Graduate School of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Koto Hospital, Tokyo, Japan
| | - Yuki Muto
- Department of Respiratory Medicine, Juntendo University, Faculty of Medicine & Graduate School of Medicine, Tokyo, Japan
| | - Manami Haba
- Department of Respiratory Medicine, Juntendo University, Faculty of Medicine & Graduate School of Medicine, Tokyo, Japan
| | - Hiroki Nakazawa
- Department of Respiratory Medicine, Juntendo University, Faculty of Medicine & Graduate School of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Koto Hospital, Tokyo, Japan
| | - Saori Hotta
- Department of Respiratory Medicine, Juntendo University, Faculty of Medicine & Graduate School of Medicine, Tokyo, Japan
| | - Hitomi Jo
- Department of Respiratory Medicine, Juntendo University, Faculty of Medicine & Graduate School of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Koto Hospital, Tokyo, Japan
| | - Noriko Hayama
- Department of Respiratory Medicine, Koto Hospital, Tokyo, Japan
| | - Yuichiro Honma
- Department of Respiratory Medicine, Juntendo University, Faculty of Medicine & Graduate School of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Koto Hospital, Tokyo, Japan
| | - Sakuo Hoshi
- Department of Respiratory Medicine, Koto Hospital, Tokyo, Japan
| | - Mitsuhiro Fujii
- Department of Respiratory Medicine, Juntendo University, Faculty of Medicine & Graduate School of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Koto Hospital, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University, Faculty of Medicine & Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
98
|
Shen J, Li J, Lei Y, Chen Z, Wu L, Lin C. Frontiers and hotspots evolution in cytokine storm: A bibliometric analysis from 2004 to 2022. Heliyon 2024; 10:e30955. [PMID: 38774317 PMCID: PMC11107250 DOI: 10.1016/j.heliyon.2024.e30955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/24/2024] Open
Abstract
Background As a fatal disease, cytokine storm has garnered research attention in recent years. Nonetheless, as the body of related studies expands, a thorough and impartial evaluation of the current status of research on cytokine storms remains absent. Consequently, this study aimed to thoroughly explore the research landscape and evolution of cytokine storm utilizing bibliometric and knowledge graph approaches. Methods Research articles and reviews centered on cytokine storms were retrieved from the Web of Science Core Collection database. For bibliometric analysis, tools such as Excel 365, CiteSpace, VOSviewer, and the Bibliometrix R package were utilized. Results This bibliometric analysis encompassed 6647 articles published between 2004 and 2022. The quantity of pertinent articles and citation frequency exhibited a yearly upward trend, with a sharp increase starting in 2020. Network analysis of collaborations reveals that the United States holds a dominant position in this area, boasting the largest publication count and leading institutions. Frontiers in Immunology ranks as the leading journal for the largest publication count in this area. Stephan A. Grupp, a prominent researcher in this area, has authored the largest publication count and has the second-highest citation frequency. Research trends and keyword evaluations show that the connection between cytokine storm and COVID-19, as well as cytokine storm treatment, are hot topics in research. Furthermore, research on cytokine storm and COVID-19 sits at the forefront in this area. Conclusion This study employed bibliometric analysis to create a visual representation of cytokine storm research, revealing current trends and burgeoning topics in this area for the first time. It will provide valuable insights, helping scholars pinpoint critical research areas and potential collaborators.
Collapse
Affiliation(s)
- Junyi Shen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiaming Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yuqi Lei
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhengrui Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Lingling Wu
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Chunyan Lin
- Department of Teaching and Research Section of Internal Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
99
|
Xie Y, Xu Z, Zhang Y, Li Y, Du P, Wang C. First-episode psychiatric disorder risk from SARS-CoV-2 infection: A clinical analysis with Chinese psychiatric inpatients. J Biomed Res 2024; 39:50-60. [PMID: 38807424 PMCID: PMC11873594 DOI: 10.7555/jbr.38.20240005] [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: 01/07/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 05/30/2024] Open
Abstract
The extensive spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) throughout China in late 2022 has underscored the correlation between this virus and severe psychiatric disorders. However, there remains a lack of reported clinical and pathological features. Accordingly, we retrospectively reviewed the electronic medical records of psychiatric inpatients for seven days from early January 2023. Twenty-one inpatients who developed first-episode psychiatric disorders within two weeks after SARS-CoV-2 infection were recruited, while 24 uninfected first-episode psychiatric inpatients were selected as controls. Comparative analyses of clinical manifestations, routine laboratory tests, and imaging examinations were performed. Our investigation demonstrated a 330% increase in the incidence of first-episode psychiatric inpatients after SARS-CoV-2 infection in 2023, compared with the preceding year without SARS-CoV-2 infections. Most cases exhibited psychiatric symptoms within one week of SARS-CoV-2 infection, which resolved after approximately two weeks, with no residual symptoms after three months. One-way ANOVA demonstrated a significant difference in the highest fever temperature between inpatients with and without psychotic symptoms. Infected inpatients displayed elevated levels of interleukin-4, interleukin-8, and interferon-α, but decreased levels of eosinophils and basophils. These findings suggest that SARS-CoV-2 may contribute to the development of psychiatric disorders, likely mediated by the virus-induced inflammatory response and neuronal dysfunction in the context of psychological distress.
Collapse
Affiliation(s)
- Ya Xie
- Clinical Mental Health Center, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Zifeng Xu
- Clinical Mental Health Center, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yumin Zhang
- Clinical Mental Health Center, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yisheng Li
- Clinical Mental Health Center, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Pengyu Du
- Medical School of Nanjing University, Nanjing, Jiangsu 210008, China
| | - Chun Wang
- Clinical Mental Health Center, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
100
|
Doğan HO, Budak M, Doğan K, Zararsız GE, Yerlitaş Sİ, Bolat S, Şenol O, Büyüktuna SA, Pınarbaşı E, Sarıismailoğlu R, Yavuz H. Dysregulated Leukotriene Metabolism in Patients with COVID-19. Jpn J Infect Dis 2024; 77:129-136. [PMID: 38171849 DOI: 10.7883/yoken.jjid.2023.211] [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] [Indexed: 01/05/2024]
Abstract
This study aimed to examine the leukotriene metabolism during COVID-19. In total, 180 participants were included in this study, of which 60 were healthy controls, 60 required intensive care units (ICU), and 60 did not require intensive care (non-ICU). The serum levels of 5-lipoxygenase (5-LO), 5-LO activating protein (ALOX5AP), and cysteinyl leukotriene (CYSLT) were measured, and the mRNA expression levels of 5-LO, ALOX5AP, and cysteinyl leukotriene receptor 1 (CYSLTR1) were investigated. Compared with the control group, both the non-ICU and ICU groups had lower levels of 5-LO and mRNA expression. ICU patients had lower levels of 5-LO and mRNA expression than non-ICU patients. CYSLTR1 mRNA expression was highest in the ICU group, followed by the non-ICU group, and healthy controls had the lowest mRNA expression levels. CYSLT levels were higher in the control group than in the non-ICU and ICU groups. CYSLTR1 expression was higher in patients than in controls; therefore, selective leukotriene receptor blockers can be used as treatment options. CYSLTR1 expression was higher in the ICU group than in the non-ICU group. Furthermore, CYSLTR1 mRNA expression may be a promising biomarker of COVID-19 severity.
Collapse
Affiliation(s)
- Halef Okan Doğan
- Department of Biochemistry, School of Medicine, Sivas Cumhuriyet University, Turkey
| | - Mahir Budak
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Turkey
| | - Kübra Doğan
- Department of Biochemistry, Sivas Numune Hospital, Turkey
| | - Gözde Ertürk Zararsız
- Department of Biostatistics, School of Medicine, Erciyes University, Turkey
- Drug Application and Research Center (ERFARMA), Erciyes University, Turkey
| | - Serra İlayda Yerlitaş
- Department of Biostatistics, School of Medicine, Erciyes University, Turkey
- Drug Application and Research Center (ERFARMA), Erciyes University, Turkey
| | - Serkan Bolat
- Department of Biochemistry, School of Medicine, Sivas Cumhuriyet University, Turkey
| | - Onur Şenol
- Department of Analytical Chemistry, Faculty of Pharmacy, Atatürk University, Turkey
| | - Seyit Ali Büyüktuna
- Department of Infectious Diseases and Clinic Microbiology, School of Medicine, Sivas Cumhuriyet University, Turkey
| | - Ergun Pınarbaşı
- Department of Medical Biology, School of Medicine, Sivas Cumhuriyet University, Turkey
| | | | - Hayrettin Yavuz
- Division of Pediatric Nephrology, Department of Pediatrics, University of Virginia, VA, USA
| |
Collapse
|