1
|
Mebrahtom G, Hailay A, Aberhe W, Zereabruk K, Haile TG, Tadesse DB. Admission and outcomes of COVID-19 among chronic obstructive pulmonary diseases patients in Africa: protocol for a systematic review and meta-analysis. Int Health 2025; 17:245-250. [PMID: 39360405 PMCID: PMC12045080 DOI: 10.1093/inthealth/ihae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 05/03/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
When the coronavirus case was originally reported in Wuhan, China, in December 2019, it quickly spread throughout the world and became a global public health problem. Evidence of the admission and outcomes of coronavirus disease among patients with chronic obstructive pulmonary disease (COPD) has not been reported in Africa. Consequently, this research protocol uses a systematic review and meta-analysis of the admission and outcomes of COVID-19 in patients with COPD in Africa. All observational studies published in the English language and reporting on the prevalence, admission and outcomes of COVID-19 among patients with COPD in Africa will be included. A search strategy will be implemented using electronic databases and the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Protocol recommendations. The findings of this review will be reported to health program designers, decision-makers and healthcare providers.
Collapse
Affiliation(s)
- Guesh Mebrahtom
- Department of Adult Health Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| | - Abrha Hailay
- Department of Adult Health Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| | - Woldu Aberhe
- Department of Adult Health Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| | - Kidane Zereabruk
- Department of Adult Health Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| | - Teklehaimanot Gereziher Haile
- Department of Maternity and Neonatal Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| | - Degena Bahrey Tadesse
- Department of Adult Health Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| |
Collapse
|
2
|
Ramalho-Pinto CH, Ventura LHA, Camatta GC, Silveira-Nunes G, Gomes MS, Sato HI, Costa MS, Guimarães HC, Barbuto RC, Martins-Filho OA, Amaral LR, Bertarini PLL, Teixeira SMR, Tupinambás U, Teixeira-Carvalho A, Faria AMC. Machine learning algorithm approach to complete blood count can be used as early predictor of COVID-19 outcome. J Leukoc Biol 2025; 117:qiae223. [PMID: 39432758 DOI: 10.1093/jleuko/qiae223] [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: 07/22/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024] Open
Abstract
Although the SARS-CoV-2 infection has established risk groups, identifying biomarkers for disease outcomes is still crucial to stratify patient risk and enhance clinical management. Optimal efficacy of COVID-19 antiviral medications relies on early administration within the initial 5 d of symptoms, assisting high-risk patients in avoiding hospitalization and improving survival chances. The complete blood count (CBC) can be an efficient and affordable option to find biomarkers that predict the COVID-19 prognosis due to infection-induced alterations in various blood parameters. This study aimed to associate hematological parameters with different COVID-19 clinical forms and utilizes them as disease outcome predictors. We performed a CBC in blood samples from 297 individuals with COVID-19 from Belo Horizonte, Brazil. Statistical analysis, as well as ROC Curves and machine learning Decision Tree algorithms were used to identify correlations, and their accuracy, between blood parameters and disease severity. In the initial 4 d of infection, traditional hematological COVID-19 alterations, such as lymphopenia, were not yet apparent. However, the monocyte percentage and granulocyte-to-lymphocyte ratio (GLR) proved to be reliable predictors for hospitalization, even in cases where patients exhibited mild symptoms that later progressed to hospitalization. Thus, our findings demonstrate that COVID-19 patients with monocyte percentages lower than 7.7% and a GLR higher than 8.75 are assigned to the hospitalized group with a precision of 86%. This suggests that these variables can serve as important biomarkers in predicting disease outcomes and could be used to differentiate patients at hospital admission for managing therapeutic interventions, including early antiviral administration. Moreover, they are simple parameters that can be useful in minimally equipped health care units.
Collapse
Affiliation(s)
- Cecília Horta Ramalho-Pinto
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| | - Lucas Haniel Araújo Ventura
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| | - Giovanna Caliman Camatta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| | - Gabriela Silveira-Nunes
- Departamento de Medicina, Universidade Federal de Juiz de Fora, Av. Doutor Raimundo Monteiro Resende, 330, 35010-177, Governador Valadares, Brazil
| | - Matheus Souza Gomes
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, R. Padre Pavoni, 290, 38701-002, Patos de Minas, Brazil
| | - Hugo Itaru Sato
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| | - Murilo Soares Costa
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190, 30130-100, Belo Horizonte, Brazil
| | | | - Rafael Calvão Barbuto
- Hospital Universitário Risoleta Tolentino Neves, R. das Gabirobas, 1, 31744-012, Belo Horizonte, Brazil
| | - Olindo Assis Martins-Filho
- Instituto de Pesquisa René Rachou, Fundação Oswaldo Cruz - MG, Av. Augusto de Lima, 1715, 30190-002, Belo Horizonte, Brazil
| | - Laurence Rodrigues Amaral
- Faculdade de Computação, Universidade Federal de Uberlândia, R. Padre Pavoni, 290, 38701-002, Patos de Minas, Brazil
| | - Pedro Luiz Lima Bertarini
- Faculdade de Engenharia Elétrica, Universidade Federal de Uberlândia, R. Padre Pavoni, 290, 38701-002, Patos de Minas, Brazil
| | - Santuza Maria Ribeiro Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| | - Unaí Tupinambás
- Departamento de Medicina, Universidade Federal de Juiz de Fora, Av. Doutor Raimundo Monteiro Resende, 330, 35010-177, Governador Valadares, Brazil
| | - Andrea Teixeira-Carvalho
- Instituto de Pesquisa René Rachou, Fundação Oswaldo Cruz - MG, Av. Augusto de Lima, 1715, 30190-002, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| |
Collapse
|
3
|
Wang P, Yao J, Li Y, Zhang Z, Zhang R, Lu S, Sun M, Huang X. Immune responses in children with secondary infection of mycoplasma pneumoniae after COVID-19: focus on eosinophils and IgE. BMC Infect Dis 2025; 25:134. [PMID: 39875814 PMCID: PMC11773693 DOI: 10.1186/s12879-025-10534-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The COVID-19 (SARS-CoV-2) epidemic has posed a major challenge to global public health, especially in children. Some children may experience secondary infection with Mycoplasma pneumoniae after SARS-CoV-2 infection, which has attracted widespread attention. Studies have shown that eosinophils play an important role in respiratory tract infections and are involved in regulating immune responses and inflammatory processes. However, there is a lack of systematic research on the specific manifestations and mechanisms of eosinophils in secondary infection with Mycoplasma pneumoniae after SARS-CoV-2 infection. OBJECTIVE This study aims to explore the characteristics of immune response in children with SARS-CoV-2 infection and Mycoplasma pneumoniae infection, focusing on the changes in immune indicators such as eosinophils (EOS), immunoglobulin E (IgE), interleukin-6 (IL-6), C-reactive protein (CRP), and procalcitonin (PCT). METHODS This study is a retrospective observational study, and a total of pediatric patients who were treated in our hospital from January 2023 to December 2023 were included. The study group included children who were diagnosed with SARS-CoV-2 infection and further infected with Mycoplasma pneumoniae, and the control group included children who were only infected with SARS-CoV-2 and had no other pathogens. The clinical data of the two groups of patients, including absolute eosinophil value, IgE quantification, IL-6, CRP and PCT levels, were collected and analyzed, and statistical comparisons were performed. RESULTS A total of 134 children were included, including 79 in the study group and 55 in the control group. The absolute eosinophil value [0.17 (0.09, 0.31) vs. 0.09 (0.06, 0.23), P < 0.01] and IgE level [59.28 (37.54, 256.88) vs. 22.00 (11.00, 113.10) P < 0.01] of the children in the study group were significantly higher than those in the control group, while IL-6 [16.81(4.72,31.86) vs. 9.5(3,57.3), P = 0.602], CRP [2.82(1.10,6.13) vs. 1.94(0.50,8.94), P = 0.528] and PCT[0.12(0.08,0.20) vs. 0.12(0.10,0.24), P = 0.329] were no significant difference between the two groups. Binary logistic regression analysis showed that the absolute value of eosinophils and IgE were independent risk factors for secondary infection of Mycoplasma pneumoniae after SARS-CoV-2 infection. CONCLUSION This study shows that after SARS-CoV-2 infection, the increase in eosinophils and the increase in related immune indicators IgE may be closely related to secondary infection with Mycoplasma pneumoniae. This study provides an important basis for understanding the immune response of children after SARS-CoV-2 infection and its related clinical management, suggesting that clinicians should closely monitor the eosinophil count and IgE level of children after SARS-CoV-2 infection, especially for children at risk of secondary infection, so as to take timely intervention measures to prevent secondary infection with Mycoplasma pneumoniae and improve the prognosis of children.
Collapse
Affiliation(s)
- Pingping Wang
- Department of Medical Laboratory, Luoyang Maternal and Child Health Hospital, 206 Tongqu Road, Luolong District, Luoyang City, Henan Province, China.
| | - Jin Yao
- Department of Infection and Public Health Management, The Second Affiliated Hospital of Henan University of Science and Technology, No. 80 Jinguyuan Road, Xigong District, Luoyang City, Henan Province, China
| | - Yaqiong Li
- Medical Imaging Department, Luoyang Maternal and Child Health Hospital, 206 Tongqu Road, Luolong District, Luoyang City, Henan Province, China
| | - Zhanjun Zhang
- Department of Medical Laboratory, Luoyang Maternal and Child Health Hospital, 206 Tongqu Road, Luolong District, Luoyang City, Henan Province, China
| | - Ruiling Zhang
- School of Humanities and Social Sciences, Luoyang Institute of Technology, No. 90 Wangcheng Avenue, Luolong District, Luoyang City, Henan Province, China
| | - Shouting Lu
- Luoyang Community Construction and Social Development Research Center, Luoyang Institute of Science and Technology School of Marxism (LIT), No. 90 Wangcheng Avenue, Luolong District, Luoyang City, Henan Province, China
| | - Meixia Sun
- Research Center of Theoretical Innovation and Think Tank Construction, Luoyang Institute of Science and Technology School of Marxism (LIT), No. 90 Wangcheng Avenue, Luolong District, Luoyang City, Henan Province, China
| | - Xiaorong Huang
- Luoyang Research Center for Inheritance and Innovation of Chinese Historical Civilization, Luoyang Institute of Science and Technology School of Marxism (LIT), No. 90 Wangcheng Avenue, Luolong District, Luoyang City, Henan Province, China
| |
Collapse
|
4
|
Wang J, Li X, Ren J, Rao Y, Qiao Y, Sun L, Liang Y, Chang C, Zhou Q, Sun Y. The Association of Blood Eosinophils and Neutrophils Expressing Eosinophilic Surface Markers with the Severity and Outcome of COVID-19. Microorganisms 2024; 12:2503. [PMID: 39770705 PMCID: PMC11727756 DOI: 10.3390/microorganisms12122503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 01/16/2025] Open
Abstract
(1) Background: The implication of type 2 (T2) inflammatory response in COVID-19 remains controversial. This study aimed to evaluate the association of eosinophils, neutrophils expressing eosinophilic surface markers and T2 cytokines with the severity and outcome of COVID-19. (2) Methods: Patients who were admitted to hospital due to COVID-19 from 18 December 2022 to 31 January 2023 were enrolled. Peripheral blood WBC and differentials, T2 cellular markers (subsets of eosinophils and neutrophils expressing eosinophilic surface markers) and cytokines at admission were measured and compared between subjects with different disease severities and outcomes. (3) Results: Ten mild-to-moderate and 22 severe-to-very severe cases were enrolled for analysis. Of these patients, seven died of severe-to-very severe disease. The severe-to-very severe patients showed a higher number of neutrophils, but lower numbers of eosinophils, lymphocytes cells and neutrophils expressing eosinophilic surface markers. Similarly, deceased cases were also characterized by increased neutrophils, but decreased eosinophils and neutrophils expressing eosinophilic surface markers. The levels of T2 cytokines failed to demonstrate a significant correlation with the severity or outcome of COVID-19. (4) Conclusions: Eosinophils and neutrophils expressing eosinophilic surface markers were associated with milder disease and better outcomes of COVID-19, suggesting that a T2 inflammatory response may confer a potential protective effect against the disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing 100191, China; (J.W.)
| |
Collapse
|
5
|
Shu W, Yang Q, Le J, Cai Q, Dai H, Luo L, Tong J, Song Y, Chen B, Tang Y, Jin D. Analysis of coinfections in patients with hematologic malignancies and COVID-19 by next-generation sequencing of bronchoalveolar lavage fluid. Eur J Med Res 2024; 29:576. [PMID: 39623478 PMCID: PMC11613933 DOI: 10.1186/s40001-024-02180-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Coinfections in patients with coronavirus disease 2019 (COVID-19) affect patient prognosis. Patients with hematologic malignancies (HMs) are usually immunosuppressed and may be at high risk of coinfection, but few related data have been reported. Here, we conducted a retrospective study to explore coinfections in patients with HMs and COVID-19 by next-generation sequencing (NGS) of bronchoalveolar lavage fluid (BALF). METHODS The data of hospitalized patients with pneumonia who underwent NGS analysis of BALF were reviewed. COVID-19 patients with HMs were enrolled in the HM group, and those without HMs were enrolled in the non-HM group. The coinfections of the two groups identified by NGS were analyzed. RESULTS Fifteen patients were enrolled in the HM group, and 14 patients were enrolled in the non-HM group. The coinfection rates in the HM group and non-HM group were 80.0% and 85.7%, respectively. The percentage of coinfected bacteria in the HM group was significantly lower than that in the non-HM group (20.0% vs 71.4%, p = 0.005). The coinfection rates of fungi and viruses were 60.0% and 35.7%, respectively, in the HM group and 35.7% and 78.6%, respectively, in the non-HM group, with no significant differences. The most common coexisting pathogen in patients with HMs was Pneumocystis jirovecii (33.3%), and the most common coexisting pathogen in patients without HMs was human gammaherpesvirus 4 (50%). Coinfection with herpesviruses occurred frequently in both groups. CONCLUSIONS Our study showed that the majority of hospitalized patients with COVID-19 are likely to be co-infected with other pathogens. Pneumocystis jiroveci and herpesvirus are commonly coinfected pathogens in patients with HMs. Bacterial coinfection is rare in patients with HMs but is more common in patients without HMs.
Collapse
Affiliation(s)
- Wenxiu Shu
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Qianqian Yang
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Jing Le
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Qianqian Cai
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Hui Dai
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Liufei Luo
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Jiaqi Tong
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Yanping Song
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Bingrong Chen
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Yaodong Tang
- Department of Respiratory Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Dian Jin
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China.
| |
Collapse
|
6
|
Aqillouch S, Zerrad C, Laazaazia O, Ouladlahsen A, El Bissati K, Akarid K, Pineau P, Benjelloun S, Ezzikouri S. Genetic variations in Interferon-Induced with Helicase C Domain 1: Impact on COVID-19 risk and severity in the Moroccan population. Hum Immunol 2024; 85:111149. [PMID: 39342923 DOI: 10.1016/j.humimm.2024.111149] [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: 06/19/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND The clinical spectrum of COVID-19 varies considerably, ranging from asymptomatic cases to severe disease and even death. This variability can partly be attributed to genetic differences in genes associated with inflammation and immune responses. Among these genes, Interferon Induced with Helicase C Domain 1 (IFIH1), which codes for a cytoplasmic sensor, plays a crucial role in detecting SARS-CoV-2 viral RNA and initiating the antiviral interferon (IFN) response, thereby constituting a key element of innate immune defense. AIM This study aims to examine the association between genetic variants in the IFIH1 gene and susceptibility to, as well as the severity of, COVID-19 in the Moroccan population. MATERIALS AND METHODS We conducted a case-control study involving 299 COVID-19 positive patients (149 severe, 150 benign) and 145 uninfected-SARS-CoV-2 controls. We determined the genotypes of two functional variants, rs1990760 (Ala946Thr) and rs3747517 (His843Arg), in the IFIH1 gene using predesigned TaqMan real-time allelic discrimination assay. RESULTS Our results indicated that the TT genotype of rs1990760 was associated with increased susceptibility to SARS-CoV-2 under a recessive model (odds ratio [OR] = 2.22, 95 % confidence interval [CI] 1.28-3.84, P=0.003). Conversely, the CT genotype appeared to confer protection against SARS-CoV-2 infection (OR=0.58, 95 % CI 0.38-0.91, P=0.016) and COVID-19 severity (OR=0.56, 95 % CI 0.34-0.91, P=0.019). No significant association was found between rs3747517 and the risk of hospitalization or infection susceptibility. CONCLUSION These findings underscore the significance of genetic variability in the IFIH1 gene in shaping individual responses to SARS-CoV-2 in the Moroccan population.
Collapse
Affiliation(s)
- Safaa Aqillouch
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca 20360, Morocco; Biochemistry, Biotechnology and Immunophysiopathology Research Team, Health and Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca 20000, Morocco
| | - Chaimaa Zerrad
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca 20360, Morocco
| | - Oumaima Laazaazia
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca 20360, Morocco
| | - Ahd Ouladlahsen
- Service des Maladies Infectieuses, CHU Ibn Rochd, Casablanca 20360, Morocco; Faculté de médecine et de pharmacie, Université Hassan II, Casablanca 20360, Morocco
| | - Kamal El Bissati
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
| | - Khadija Akarid
- Biochemistry, Biotechnology and Immunophysiopathology Research Team, Health and Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca 20000, Morocco
| | - Pascal Pineau
- Institut Pasteur, Université Paris Cité, Unité "Organisation Nucléaire et Oncogenèse", INSERM U993, Paris, France
| | - Soumaya Benjelloun
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca 20360, Morocco
| | - Sayeh Ezzikouri
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca 20360, Morocco.
| |
Collapse
|
7
|
Matsuyama E, Miyata J, Terai H, Miyazaki N, Iwasaki T, Nagashima K, Watase M, Sunata K, Namkoong H, Asakura T, Masaki K, Chubachi S, Ohgino K, Kawada I, Minami K, Hagiwara R, Ueda S, Yoshiyama T, Kokuto H, Kusumoto T, Oashi A, Miyawaki M, Saito F, Tani T, Ishioka K, Takahashi S, Nakamura M, Ishii M, Sato Y, Fukunaga K. Chronic obstructive pulmonary disease, asthma, and mechanical ventilation are risk factors for dyspnea in patients with long COVID: A Japanese nationwide cohort study. Respir Investig 2024; 62:1094-1101. [PMID: 39342666 DOI: 10.1016/j.resinv.2024.09.009] [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: 07/24/2024] [Revised: 09/15/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Patients often experience multiple prolonged symptoms following acute coronavirus disease 2019 (COVID-19) recovery, defined as long coronavirus disease (COVID). Patients with long COVID may experience dyspnea during acute and post-acute phases. Therefore, this study aimed to identify specific risk factors for dyspnea in patients with long COVID. METHODS Hospitalized patients with COVID-19, aged ≥18 years, were enrolled in this multicenter cohort study conducted at 26 medical institutions across Japan. Clinical data during hospitalization and patient-reported outcomes after discharge at the 3, 6, and 12-month follow-ups were retrieved from medical records and paper-based or smartphone application-based questionnaires, respectively. RESULTS Generalized linear mixed model (GLMM) analysis of prolonged dyspnea at each time point during follow-up showed that this symptom was associated with chronic obstructive pulmonary disease (COPD) (odds ratio [OR], 2.74; 95% confidence interval [CI], 1.31-5.74), asthma (OR, 2.21; 95%CI, 1.17-4.16), and ventilator management (OR, 3.10; 95%CI, 1.65-5.83). In addition, patients with COPD (44.4%) and ventilator management (25.0%) were more frequently associated with delayed dyspnea onset. The generalized estimating equations analysis results with multiple imputed datasets, conducted as a sensitivity analysis, confirmed the adjusted GLMM analysis results. CONCLUSIONS Prolonged dyspnea was associated with COPD, asthma, and severe infection that required mechanical ventilation in the Japanese population with long COVID. Further investigation is needed to clarify its mechanism and develop prophylactic and therapeutic strategies for dyspnea in patients with long COVID.
Collapse
Affiliation(s)
- Emiko Matsuyama
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Jun Miyata
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan.
| | - Hideki Terai
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan; Keio Cancer Center, Keio University School of Medicine, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Naoki Miyazaki
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Toshiki Iwasaki
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Kengo Nagashima
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Mayuko Watase
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan; Department of Respiratory Medicine, National Hospital Organization Tokyo Medical Center, 5-1, Higashigaoka 2-chome, Meguro-ku, Tokyo, 152-8902, Japan
| | - Keeya Sunata
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Ho Namkoong
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan; Department of Infectious Diseases, Keio University School of Medicine, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Takanori Asakura
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan; Department of Respiratory Medicine, Kitasato University Kitasato Institute Hospital, 9-1, Shirokane 5-chome, Minato-ku, Tokyo, 108-8642, Japan
| | - Katsunori Masaki
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Shotaro Chubachi
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Keiko Ohgino
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Ichiro Kawada
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan; Research Centers and Institutes, Health Center, Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Kazuhiro Minami
- Department of Internal Medicine, Saitama Medical Center, 4-9-3 Kita-Urawa, Urawa-ku, Saitama-shi, Saitama, 330-0074, Japan
| | - Rie Hagiwara
- Department of Internal Medicine, Saitama Medical Center, 4-9-3 Kita-Urawa, Urawa-ku, Saitama-shi, Saitama, 330-0074, Japan
| | - Soichiro Ueda
- Department of Internal Medicine, Saitama Medical Center, 4-9-3 Kita-Urawa, Urawa-ku, Saitama-shi, Saitama, 330-0074, Japan
| | - Takashi Yoshiyama
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, 1-24, Matsuyama 3-chome, Kiyose-shi, Tokyo, 204-8522, Japan
| | - Hiroyuki Kokuto
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, 1-24, Matsuyama 3-chome, Kiyose-shi, Tokyo, 204-8522, Japan
| | - Tatsuya Kusumoto
- Department of Pulmonary Medicine, Eiju General Hospital, 3-16, Higashi-Ueno 2-chome, Taito-ku, Tokyo, 110-8645, Japan
| | - Ayano Oashi
- Department of Pulmonary Medicine, Eiju General Hospital, 3-16, Higashi-Ueno 2-chome, Taito-ku, Tokyo, 110-8645, Japan
| | - Masayoshi Miyawaki
- Department of Pulmonary Medicine, Eiju General Hospital, 3-16, Higashi-Ueno 2-chome, Taito-ku, Tokyo, 110-8645, Japan
| | - Fumitake Saito
- Department of Pulmonary Medicine, Eiju General Hospital, 3-16, Higashi-Ueno 2-chome, Taito-ku, Tokyo, 110-8645, Japan
| | - Tetsuo Tani
- Department of Pulmonary Medicine, Tokyo Saiseikai Central Hospital, 4-17, Mita 1-chome, Minato-ku, Tokyo, 108-0073, Japan
| | - Kota Ishioka
- Department of Pulmonary Medicine, Tokyo Saiseikai Central Hospital, 4-17, Mita 1-chome, Minato-ku, Tokyo, 108-0073, Japan
| | - Saeko Takahashi
- Department of Pulmonary Medicine, Tokyo Saiseikai Central Hospital, 4-17, Mita 1-chome, Minato-ku, Tokyo, 108-0073, Japan
| | - Morio Nakamura
- Department of Pulmonary Medicine, Tokyo Saiseikai Central Hospital, 4-17, Mita 1-chome, Minato-ku, Tokyo, 108-0073, Japan; Department of Pulmonary Medicine, National Hospital Organization Kanagawa Hospital, 666-1 Ochiai, Hadano, Kanagawa, 257-8585, Japan
| | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Yasunori Sato
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicin, 35 Shinano-cho, Shinjuku-ku, Tokyo, 160-0016, Japan
| |
Collapse
|
8
|
Bernardinello N, Castelli G, Pasin D, Grisostomi G, Cola M, Giraudo C, Cocconcelli E, Cattelan A, Spagnolo P, Balestro E. Blood Eosinophils Matter in Post-COVID-19 Pneumonia. Diagnostics (Basel) 2024; 14:2320. [PMID: 39451643 PMCID: PMC11506628 DOI: 10.3390/diagnostics14202320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/06/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Even after the development of vaccines, SARS-CoV-2 continues to cause severe pneumonia all over the world. Consequently, in order to improve the management of patients and optimize the use of resources, predictors of disease severity and lung complications after COVID-19 pneumonia are urgently needed. Blood cell count is an easily available and reproducible biomarker. With this study, we aimed to explore the role of eosinophils in predicting disease behavior and pulmonary sequelae at first follow-up with computed tomography (CT). METHODS we evaluated blood cell count and other inflammatory markers, both at baseline and during hospitalization, in a large population of hospitalized COVID-19 patients. RESULTS 327 patients were finally enrolled, 214 were classified as low-intensity medical care (LIMC) and 113 as high-intensity medical care. Eosinophils were higher at discharge in the HIMC group [0.1 (0-0.72) vs. 0.05 (0-0.34) × 109/L; p < 0.0001]. Moreover, in the multivariable analysis, age ≥ 62 years (OR 1.76 (1.05-2.8) p = 0.03) and Δ eosinophils ≥ 0.05 (OR 1.75 (1.05-2.9) p = 0.03) were two independent predictors of residual lung abnormalities in the whole patient population at first follow-up. CONCLUSIONS an eosinophil increase during hospitalization could be a potential predictor of pulmonary sequelae in surviving patients after COVID-19 pneumonia.
Collapse
Affiliation(s)
- Nicol Bernardinello
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, 35128 Padova, Italy; (N.B.); (G.C.); (D.P.); (G.G.); (E.C.); (P.S.)
| | - Gioele Castelli
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, 35128 Padova, Italy; (N.B.); (G.C.); (D.P.); (G.G.); (E.C.); (P.S.)
| | - Dylan Pasin
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, 35128 Padova, Italy; (N.B.); (G.C.); (D.P.); (G.G.); (E.C.); (P.S.)
| | - Giulia Grisostomi
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, 35128 Padova, Italy; (N.B.); (G.C.); (D.P.); (G.G.); (E.C.); (P.S.)
| | - Marco Cola
- Infectious Diseases Unit, Department of Internal Medicine, Azienda Ospedaliera Universitaria di Padova, 35128 Padova, Italy; (M.C.); (A.C.)
| | - Chiara Giraudo
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy;
| | - Elisabetta Cocconcelli
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, 35128 Padova, Italy; (N.B.); (G.C.); (D.P.); (G.G.); (E.C.); (P.S.)
| | - Annamaria Cattelan
- Infectious Diseases Unit, Department of Internal Medicine, Azienda Ospedaliera Universitaria di Padova, 35128 Padova, Italy; (M.C.); (A.C.)
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, 35128 Padova, Italy; (N.B.); (G.C.); (D.P.); (G.G.); (E.C.); (P.S.)
| | - Elisabetta Balestro
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, 35128 Padova, Italy; (N.B.); (G.C.); (D.P.); (G.G.); (E.C.); (P.S.)
| |
Collapse
|
9
|
Wu K, Yin L, Han J, Cai Q, Guo Y, Jin X, Wu J, Cheng Y. Case-control study on risk factors for in-hospital mortality in patients with severe COVID-19. Front Public Health 2024; 12:1424720. [PMID: 39440172 PMCID: PMC11493594 DOI: 10.3389/fpubh.2024.1424720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024] Open
Abstract
Objective The purpose of this study was to identify independent risk factors affecting patient survival and explore predictors of severe cases of coronavirus disease 2019 (COVID-19). Methods We conducted a retrospective, observational, case-control study on adult patients with severe COVID-19 who were admitted to affiliated hospitals in Tianjin between December 18, 2022, and January 31, 2023. We used univariate and multifactorial logistic regression analyses to analyze demographic indicators, comorbidity profiles, and laboratory parameters in two groups of patients (deceased and surviving) to identify independent risk factors for death in patients with severe COVID-19. Results Patients in the deceased group were older than those in the survival group (p = 0.018), and there were more cases of coexisting respiratory insufficiency in the deceased group (p = 0.002). Additionally, laboratory test results for white blood cell count (WBC) and creatine kinase (CK) showed significantly higher values in the deceased group (p = 0.047 and p = 0.029, respectively), while arterial oxygen partial pressure (PAO2) showed significantly lower values compared to the survival group (p = 0.021). Age, respiratory insufficiency, WBCH (highest WBC value), CKH (highest CK value), and PAO2F (first PAO2 value) had area under curve (AUC) values of 0.698, 0.838, 0.721, 0.744, and 0.633, respectively. Conclusion The main risk factors for mortality in patients with severe COVID-19 that we identified in this study were the advanced age of patients, coexisting respiratory insufficiency, elevated levels of WBC and CK, and decreased levels of PAO2. Elevated WBC and CK laboratory parameters, in particular, demonstrated good predictive value for in-hospital mortality risk.
Collapse
Affiliation(s)
- Kemei Wu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lili Yin
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiangqin Han
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiuhan Cai
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yang Guo
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin Jin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinling Wu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yupei Cheng
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
10
|
Opalska A, Gardarsdottir H, Kwa M, Wójkowska-Mach J, Sabate M, Ballarin ME, de Groot M, Leufkens H. Use of antibiotics in the early COVID-19 pandemic in Poland, the Netherlands and Spain, from erraticism to (more) logic. Eur J Clin Pharmacol 2024; 80:1581-1589. [PMID: 39017693 PMCID: PMC11393094 DOI: 10.1007/s00228-024-03726-1] [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: 02/02/2024] [Accepted: 06/30/2024] [Indexed: 07/18/2024]
Abstract
INTRODUCTION In the Spring of 2020, the world was hit with unparalleled impact by the coronavirus pandemic. Antibiotics were widely used, even without good rationale. The aim of our study was to compare the use of antibiotics in patients with confirmed COVID-19 from three hospitals across Europe (Poland, the Netherlands and Spain) between two subsequent periods in the early days of the pandemic. METHOD We analysed data (antibiotics used and variation in the use of antibiotics, patients, admission and disease-related characteristics) from 300 patients admitted in three hospitals (University Hospital in Cracow, University Medical Center in Utrecht and Vall d'Hebron University Hospital in Barcelona) with confirmed infection of SARS-CoV-2 during Q1 2020 and Q4 2020. RESULTS There was ample variation in terms of patient mix and outcomes across the 3 hospitals. The majority of patients (225 out of 300) in all 3 hospitals received at least 1 antibiotic during the hospitalisation period. A minority of patients (68 out of 300) had their bacterial test results positive during their hospitalisation period. Throughout the 2 study periods, third-generation cephalosporins (ceftriaxone in 170 out of 300 patients) emerged as the most commonly used class of antibiotics. There was an apparent shift towards more rational utilisation of antibiotics, in all three hospitals. CONCLUSIONS Our study shows that during the early stage of COVID-19 pandemic in 2020, antibiotics were frequently used in three European teaching hospitals despite the relatively low incidence of microbiologically confirmed bacterial infections. While in the early days of the COVID-19 pandemic antibiotic prescribing was full of trial and error, we could also confirm a learning curve over time.
Collapse
Affiliation(s)
- Aleksandra Opalska
- Division Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Directorate-General for Health and Food Safety, European Commission, Brussels, Belgium
| | - Helga Gardarsdottir
- Division Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marcel Kwa
- Department of Pharmacovigilance, Medicines Evaluation Board, Utrecht, Netherlands
| | - Jadwiga Wójkowska-Mach
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Crakow, Poland
| | - Monica Sabate
- Department of Clinical Pharmacology, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Clinical Pharmacology Research Group, Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Maria Elena Ballarin
- Department of Clinical Pharmacology, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Clinical Pharmacology Research Group, Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Mark de Groot
- UPOD, Central Diagnostic Laboratory, Division Laboratories, Pharmacy and Biomedical Genetics, Utrecht, Netherlands
| | - Hubert Leufkens
- Division Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.
| |
Collapse
|
11
|
Engin MMN, Özdemir Ö. Role of vitamin D in COVID-19 and other viral infections. World J Virol 2024; 13:95349. [PMID: 39323448 PMCID: PMC11401007 DOI: 10.5501/wjv.v13.i3.95349] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/14/2024] [Accepted: 07/19/2024] [Indexed: 08/29/2024] Open
Abstract
Vitamin D is a steroid hormone that is naturally produced in the body or obtained through dietary sources, primarily under the influence of UVB radiation. This essential nutrient has a vital role in numerous physiological processes, encompassing immune function, cell growth, differentiation, insulin regulation, and cardiovascular well-being, along with its pivotal role in sustaining the delicate equilibrium of calcium and phosphate concentrations in the body. Moreover, vitamin D reinforces mucosal defense and bolsters the immune system through immunomodulation, making it a critical component of overall health. Numerous studies have unveiled the profound connection between vitamin D and the predisposition to respiratory tract infections, including well-known viruses such as influenza and the novel severe acute respiratory syndrome coronavirus 2. Vitamin D deficiency has been consistently linked to increased severity of coronavirus disease 2019 (COVID-19) and a heightened risk of mortality among afflicted individuals. Retrospective observational studies have further substantiated these findings, indicating that levels of vitamin D are linked with both the occurrence and severity of COVID-19 cases. Vitamin D has its influence on viral infections through a multitude of mechanisms, such as promoting the release of antimicrobial peptides and fine-tuning the responses of the immune system. Additionally, vitamin D is intertwined with the intricate network of the renin-angiotensin system, suggesting a potential impact on the development of complications related to COVID-19. While further clinical trials and extensive research are warranted, the existing body of evidence strongly hints at the possible use of vitamin D as a valuable tool in the prophylaxis and management of COVID-19 and other viral infectious diseases.
Collapse
Affiliation(s)
| | - Öner Özdemir
- Division of Allergy and Immunology, Department of Pediatrics, Sakarya Research and Training Hospital, Sakarya University, Faculty of Medicine, Sakarya 54100, Türkiye
| |
Collapse
|
12
|
Aboras SI, Maher HM, Alzoman NZ, Elbordiny HS. Sustainable and technically smart spectrophotometric determination of PAXLOVID: a comprehensive ecological and analytical performance rating. BMC Chem 2024; 18:184. [PMID: 39304939 DOI: 10.1186/s13065-024-01275-3] [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: 04/17/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
The Food and Drug Administration (FDA) authorized the administration of ritonavir (RIT)-boosted nirmatrelvir (NMV) on May 25, 2023, for the treatment of mild to moderate COVID-19 in patients who are at high risk of developing severe COVID-19. In accordance with sustainability and environmental friendliness, simple, eco-friendly, and sustainable spectrophotometric methods were established for concurrently estimating RIT and NMV in newly launched copackaged pills. The suggested solutions for resolving the spectral overlap between RIT and NMV involve the following mathematical methods: the first derivative method (1D), second derivative method (2D), and dual-wavelength zero-order method (DWZ). When ethanol was used as a green dilution solvent, the linearity range was adjusted (10-250 µg/mL) for both drugs. The procedures resulted in a high correlation coefficient (not less than 0.9996) and satisfactory levels of detection and quantification. Additionally, method validation was performed in accordance with International Council for Harmonization norms. Moreover, a detailed ecological and sustainability evaluation protocol was established to confirm the greenness and whiteness of the methods. Finally, the proposed method, along with previously reported methods for analysing NMV and RIT, were reviewed analytically and ecologically.
Collapse
Affiliation(s)
- Sara I Aboras
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Al-Mesallah, Alexandria University, Alexandria, 21521, Egypt.
| | - Hadir M Maher
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Al-Mesallah, Alexandria University, Alexandria, 21521, Egypt
| | - Nourah Z Alzoman
- College of Pharmacy, Department of Pharmaceutical Chemistry, King Saud University, P.O. Box 22452, 11495, Riyadh, Saudi Arabia
| | - Haydi S Elbordiny
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| |
Collapse
|
13
|
Spiesshoefer J, Regmi B, Senol M, Jörn B, Gorol O, Elfeturi M, Walterspacher S, Giannoni A, Kahles F, Gloeckl R, Dreher M. Potential Diaphragm Muscle Weakness-related Dyspnea Persists 2 Years after COVID-19 and Could Be Improved by Inspiratory Muscle Training: Results of an Observational and an Interventional Clinical Trial. Am J Respir Crit Care Med 2024; 210:618-628. [PMID: 38763165 PMCID: PMC11389583 DOI: 10.1164/rccm.202309-1572oc] [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: 09/07/2023] [Accepted: 05/19/2024] [Indexed: 05/21/2024] Open
Abstract
Rationale: Diaphragm muscle weakness might underlie persistent exertional dyspnea, despite normal lung and cardiac function in individuals who were previously hospitalized for acute coronavirus disease (COVID-19) illness. Objectives: The authors sought, first, to determine the persistence and pathophysiological nature of diaphragm muscle weakness and its association with exertional dyspnea 2 years after hospitalization for COVID-19 and, second, to investigate the impact of inspiratory muscle training (IMT) on diaphragm and inspiratory muscle weakness and exertional dyspnea in individuals with long COVID. Methods: Approximately 2 years after hospitalization for COVID-19, 30 individuals (11 women, 19 men; median age, 58 years; interquartile range [IQR] = 51-63) underwent comprehensive (invasive) respiratory muscle assessment and evaluation of dyspnea. Eighteen with persistent diaphragm muscle weakness and exertional dyspnea were randomized to 6 weeks of IMT or sham training; assessments were repeated immediately after and 6 weeks after IMT completion. The primary endpoint was change in inspiratory muscle fatiguability immediately after IMT. Measurements and Main Results: At a median of 31 months (IQR = 23-32) after hospitalization, 21 of 30 individuals reported relevant persistent exertional dyspnea. Diaphragm muscle weakness on exertion and reduced diaphragm cortical activation were potentially related to exertional dyspnea. Compared with sham control, IMT improved diaphragm and inspiratory muscle function (sniff transdiaphragmatic pressure, 83 cm H2O [IQR = 75-91] vs. 100 cm H2O [IQR = 81-113], P = 0.02), inspiratory muscle fatiguability (time to task failure, 365 s [IQR = 284-701] vs. 983 s [IQR = 551-1,494], P = 0.05), diaphragm voluntary activation index (79% [IQR = 63-92] vs. 89% [IQR = 75-94], P = 0.03), and dyspnea (Borg score, 7 [IQR = 5.5-8] vs. 6 [IQR = 4-7], P = 0.03). Improvements persisted for 6 weeks after IMT completion. Conclusions: To the best of the authors' knowledge, this study is the first to identify a potential treatment for persisting exertional dyspnea in long COVID and provide a possible pathophysiological explanation for the treatment benefit. Clinical trial registered with www.clinicaltrials.gov (NCT04854863, NCT05582642).
Collapse
Affiliation(s)
| | - Binaya Regmi
- Department of Pneumology and Intensive Care Medicine and
| | - Mehdi Senol
- Department of Pneumology and Intensive Care Medicine and
| | - Benedikt Jörn
- Department of Pneumology and Intensive Care Medicine and
| | - Oscar Gorol
- Department of Pneumology and Intensive Care Medicine and
| | | | - Stephan Walterspacher
- Faculty of Health/School of Medicine, Witten/Herdecke University, Witten, Germany
- Medical Clinic II, Department of Pneumology, Cardiology and Intensive Care Medicine, Klinikum Konstanz, Konstanz, Germany
| | - Alberto Giannoni
- Interdisciplinary Health Science Center, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Florian Kahles
- Department of Cardiology, Vascular Medicine and Intensive Care Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Rainer Gloeckl
- Philipps-University of Marburg, German Center for Lung Research, Marburg, Germany; and
- Institute for Pulmonary Rehabilitation Research, Schoen Klinik Berchtesgadener Land, Schoenau am Koenigssee, Germany
| | - Michael Dreher
- Department of Pneumology and Intensive Care Medicine and
| |
Collapse
|
14
|
Essex M, Millet Pascual-Leone B, Löber U, Kuhring M, Zhang B, Brüning U, Fritsche-Guenther R, Krzanowski M, Fiocca Vernengo F, Brumhard S, Röwekamp I, Anna Bielecka A, Lesker TR, Wyler E, Landthaler M, Mantei A, Meisel C, Caesar S, Thibeault C, Corman VM, Marko L, Suttorp N, Strowig T, Kurth F, Sander LE, Li Y, Kirwan JA, Forslund SK, Opitz B. Gut microbiota dysbiosis is associated with altered tryptophan metabolism and dysregulated inflammatory response in COVID-19. NPJ Biofilms Microbiomes 2024; 10:66. [PMID: 39085233 PMCID: PMC11291933 DOI: 10.1038/s41522-024-00538-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
The clinical course of COVID-19 is variable and often unpredictable. To test the hypothesis that disease progression and inflammatory responses associate with alterations in the microbiome and metabolome, we analyzed metagenome, metabolome, cytokine, and transcriptome profiles of repeated samples from hospitalized COVID-19 patients and uninfected controls, and leveraged clinical information and post-hoc confounder analysis. Severe COVID-19 was associated with a depletion of beneficial intestinal microbes, whereas oropharyngeal microbiota disturbance was mainly linked to antibiotic use. COVID-19 severity was also associated with enhanced plasma concentrations of kynurenine and reduced levels of several other tryptophan metabolites, lysophosphatidylcholines, and secondary bile acids. Moreover, reduced concentrations of various tryptophan metabolites were associated with depletion of Faecalibacterium, and tryptophan decrease and kynurenine increase were linked to enhanced production of inflammatory cytokines. Collectively, our study identifies correlated microbiome and metabolome alterations as a potential contributor to inflammatory dysregulation in severe COVID-19.
Collapse
Affiliation(s)
- Morgan Essex
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Belén Millet Pascual-Leone
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulrike Löber
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mathias Kuhring
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, BIH Metabolomics Platform, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, Core Unit Bioinformatics, Berlin, Germany
| | - Bowen Zhang
- Department of Computational Biology for Individualized Infection Medicine, Center for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz-Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- TWINCORE, joint ventures between the Helmholtz Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- College of Life Sciences, Beijing Normal University, Beijing, China
| | - Ulrike Brüning
- Berlin Institute of Health (BIH) at Charité, BIH Metabolomics Platform, Berlin, Germany
| | | | - Marta Krzanowski
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Facundo Fiocca Vernengo
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sophia Brumhard
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ivo Röwekamp
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Agata Anna Bielecka
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany
| | - Till Robin Lesker
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Christian Meisel
- Labor Berlin-Charité Vivantes GmbH, Berlin, Germany
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sandra Caesar
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Charlotte Thibeault
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Victor M Corman
- Labor Berlin-Charité Vivantes GmbH, Berlin, Germany
- Institute of Virology, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Berlin, Germany
| | - Lajos Marko
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Till Strowig
- Department of Computational Biology for Individualized Infection Medicine, Center for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz-Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany
| | - Florian Kurth
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leif E Sander
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Yang Li
- Department of Computational Biology for Individualized Infection Medicine, Center for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz-Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- TWINCORE, joint ventures between the Helmholtz Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Jennifer A Kirwan
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, BIH Metabolomics Platform, Berlin, Germany
- University of Nottingham School of Veterinary Medicine and Science, Loughborough, UK
| | - Sofia K Forslund
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Bastian Opitz
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Labor Berlin-Charité Vivantes GmbH, Berlin, Germany.
- German Center for Lung Research (DZL), Berlin, Germany.
| |
Collapse
|
15
|
Dulkadir R, Oztelcan Gunduz B. Differentiating COVID-19 and influenza in children: hemogram parameters as diagnostic tools. Front Public Health 2024; 12:1377785. [PMID: 39056079 PMCID: PMC11269124 DOI: 10.3389/fpubh.2024.1377785] [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: 02/01/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Introduction It is not always possible to differentiate between influenza and COVID-19 based on symptoms alone. This is a topic of significant importance as it aims to determine whether there are specific hematological parameters that can be used to distinguish between influenza and COVID-19 in children. Methodology Two hundred thirty-one children between the ages of 1 month and 18 years who presented to the children's outpatient clinic between June 2021 and June 2022 with similar symptoms and were tested with an influenza test and a COVID-19 PCR test were included in the study. Of the patients included in the study, 130 tested positive for COVID-19 and 101 positive for influenza. The patients were evaluated for hematological parameters. Results Age, eosinophils and monocyte factors were shown to be statistically significantly effective in COVID-19. The risk of COVID-19 increased 1,484-fold with age, 10,708-fold with increasing eosinophil count, and 1,591-fold with increasing monocyte count. The performance of the monocyte count and eosinophil count was assessed by receiver operating characteristic curve (ROC) analysis. According to the performed ROC analysis, the area under the curve (AUC) value was observed to be 0.990 for monocytes. According to the cutoff point >1.50, the sensitivity value was determined as 98.4% and the specificity value as 97.0%. AUC significance for eosinophils was found to be 0.989. According to the cutoff point >0.02, the sensitivity value was determined as 99.2% and the specificity value as 93.1%. Conclusion In the diagnosis of COVID-19, the eosinophil count and monocyte count are easily accessible, inexpensive, and important parameters in terms of differential diagnosis and can help in the differentiation of COVID-19 from influenza during seasonal outbreaks of the latter. Developing parameters for clinicians to use in diagnosing COVID-19 and influenza can facilitate their work in practice.
Collapse
|
16
|
Ismail NH, Siddig A, Hasenan M'A, Ramli M, Mohd Noor NH, Hassan MN, Johan MF, Ramli M, Bahar R, Mohamed Yusoff S. Hematological Markers as Predictors of ICU Admission in COVID-19 Patients: A Case-Control Study From a Tertiary Hospital. Cureus 2024; 16:e64213. [PMID: 39130863 PMCID: PMC11310825 DOI: 10.7759/cureus.64213] [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] [Accepted: 07/04/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND COVID-19 illness severity ranges from mild- to life-threatening cases necessitating critical care. Rapid prediction of disease severity and the need for critical care support in COVID-19 patients remain essential, not only for current management but also for preparedness in future pandemics. This study aimed to assess hematological parameters as predictors of intensive care unit (ICU) admission and survival in COVID-19 patients, providing insights applicable to a broad range of infectious diseases. METHODS A case-control study was conducted at Hospital Raja Perempuan Zainab II, a tertiary referral hospital in Kelantan, Malaysia, from March 2020 to August 2021. Demographics, clinical, and laboratory data were retrieved from patients' medical records. Statistical analyses, including the Chi-square (χ2) test, independent t-tests, and simple and multiple logistic regressions, were used to analyze the data. A receiver operating characteristic (ROC) curve analysis was conducted to assess the accuracy of the predictors. RESULTS The median age was 51 years, with females comprising 56.7% (n=148) and males 43.3% (n=113). A total of 88.5% of patients were admitted to non-ICU wards, with a mortality rate of 5.7%. Significant differences were observed in the distribution of hematological parameters between ICU-admitted and non-admitted patients. Neutrophil (OR: 23.96, 95% CI: 7.296-78.675) and white blood cell (WBC) count (OR: 36.677, 95% CI: 2.086-644.889) were the most significant predictors for ICU admission and survival, respectively. CONCLUSIONS WBC and neutrophil counts exhibited high predictive value for ICU admission, while WBC, neutrophil, lymphocyte, and immature granulocyte (IG) counts were significant predictors of survival status among COVID-19 patients. These findings underscore the continued relevance of hematological markers in managing severe respiratory infections and improving critical care triage, with implications for current and future healthcare challenges.
Collapse
Affiliation(s)
- Nor Hayati Ismail
- Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, MYS
| | - Alaa Siddig
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, MYS
| | - Muhammad 'Akif Hasenan
- Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, MYS
- Department of Pathology, Hospital Raja Perempuan Zainab II, Kota Bharu, MYS
| | - Majdan Ramli
- Department of Pathology, Hospital Raja Perempuan Zainab II, Kota Bharu, MYS
| | - Noor Haslina Mohd Noor
- Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, MYS
| | - Mohd Nazri Hassan
- Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, MYS
| | - Muhammad Farid Johan
- Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, MYS
| | - Marini Ramli
- Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, MYS
| | - Rosnah Bahar
- Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, MYS
| | - Shafini Mohamed Yusoff
- Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, MYS
| |
Collapse
|
17
|
Dong J, Su D, Zhao B, Han J, Tu M, Zhang K, Wang F, An Y. Potential Protective Factors for Allergic Rhinitis Patients Infected with COVID-19. Curr Issues Mol Biol 2024; 46:6633-6645. [PMID: 39057037 PMCID: PMC11275266 DOI: 10.3390/cimb46070395] [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: 05/09/2024] [Revised: 06/10/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
At the beginning of the 2019 coronavirus disease (COVID-19) pandemic, airway allergic diseases such as asthma and allergic rhinitis (AR) were considered as risk factors for COVID-19, as they would aggravate symptoms. With further research, more and more literature has shown that airway allergic disease may not be a high-risk factor, but may be a protective factor for COVID-19 infection, which is closely related to its low-level expression of the ACE2 receptor and the complex cytokines network as underlying molecular regulatory mechanisms. In addition, steroid hormones and age factors could not be ignored. In this review, we have summarized some current evidence on the relationship between COVID-19 and allergic rhinitis to highlight the underlying mechanisms of COVID-19 infection and provide novel insights for its prevention and treatment. The key findings show that allergic rhinitis and its related molecular mechanisms may have a protective effect against COVID-19 infection.
Collapse
Affiliation(s)
- Jiaoyue Dong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Dingyuan Su
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Kaifeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Fengling Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| |
Collapse
|
18
|
Jin Q, Ma W, Zhang W, Wang H, Geng Y, Geng Y, Zhang Y, Gao D, Zhou J, Li L, Gou Y, Zhong B, Li J, Hou W, Lu S. Clinical and hematological characteristics of children infected with the omicron variant of SARS-CoV-2: role of the combination of the neutrophil: lymphocyte ratio and eosinophil count in distinguishing severe COVID-19. Front Pediatr 2024; 12:1305639. [PMID: 38978839 PMCID: PMC11228319 DOI: 10.3389/fped.2024.1305639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 05/30/2024] [Indexed: 07/10/2024] Open
Abstract
Purpose Investigate the clinical/hematological characteristics of children infected with the Omicron variant of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) and identify an effective indicator to distinguish coronavirus disease 2019 (COVID-19) severity in children. Methods A retrospective study was conducted through electronic medical records from pediatric patients. The demographic, clinical, and routine blood test (RBT) features of children diagnosed by real-time PCR for SARS-CoV-2 were collected. Results Data of 261 patients were analyzed. The most common abnormality shown by RBTs was increased monocyte count (68%). Children had "mild-moderate" or "severe" forms of COVID-19. Prevalence of abnormal neutrophil count (p = 0.048), eosinophil count (p = 0.006), mean corpuscular volume (p = 0.033), mean platelet volume (p = 0.006), platelet-large cell ratio (p = 0.043), and red blood cell distribution width-standard deviation (p = 0.031) were significantly different in the two types. A combination of the neutrophil: lymphocyte ratio (NLR) and eosinophil count for diagnosing severe COVID-19 presented the largest AUC (0.688, 95% CI = 0.599-0.777; p < 0.001), and the AUC increased with a decrease in age. Conclusions Combination of the NLR and eosinophil count might be a promising indicator for identifying severe COVID-19 in children at infection onset.
Collapse
Affiliation(s)
- Qiaoyan Jin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Biochemistry and Molecular Biology, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Wenxian Ma
- Department of Biochemistry and Molecular Biology, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Wei Zhang
- Xijing 986 Hospital Department, Air Force Medical University, Xi’an, China
| | - Huiyuan Wang
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yiongxiang Geng
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yan Geng
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yang Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Dan Gao
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jing Zhou
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lin Li
- Xijing 986 Hospital Department, Air Force Medical University, Xi’an, China
| | - Yaping Gou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xi’an, China
| | - Bo Zhong
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jing Li
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Wei Hou
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, Xi’an Jiaotong University Health Science Center, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, China
| |
Collapse
|
19
|
Ranjbar M, Cusack RP, Whetstone CE, Brister DL, Wattie J, Wiltshire L, Alsaji N, Le Roux J, Cheng E, Srinathan T, Ho T, Sehmi R, O’Byrne PM, Snow-Smith M, Makiya M, Klion AD, Duong M, Gauvreau GM. Immune Response Dynamics and Biomarkers in COVID-19 Patients. Int J Mol Sci 2024; 25:6427. [PMID: 38928133 PMCID: PMC11204302 DOI: 10.3390/ijms25126427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The immune response dynamics in COVID-19 patients remain a subject of intense investigation due to their implications for disease severity and treatment outcomes. We examined changes in leukocyte levels, eosinophil activity, and cytokine profiles in patients hospitalized with COVID-19. METHODS Serum samples were collected within the first 10 days of hospitalization/confirmed infection and analyzed for eosinophil granule proteins (EGP) and cytokines. Information from medical records including comorbidities, clinical symptoms, medications, and complete blood counts were collected at the time of admission, during hospitalization and at follow up approximately 3 months later. RESULTS Serum levels of eotaxin, type 1 and type 2 cytokines, and alarmin cytokines were elevated in COVID-19 patients, highlighting the heightened immune response (p < 0.05). However, COVID-19 patients exhibited lower levels of eosinophils and eosinophil degranulation products compared to hospitalized controls (p < 0.05). Leukocyte counts increased consistently from admission to follow-up, indicative of recovery. CONCLUSION Attenuated eosinophil activity alongside elevated chemokine and cytokine levels during active infection, highlights the complex interplay of immune mediators in the pathogenesis COVID-19 and underscores the need for further investigation into immune biomarkers and treatment strategies.
Collapse
Affiliation(s)
- Maral Ranjbar
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Ruth P. Cusack
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Christiane E. Whetstone
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Danica L. Brister
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Jennifer Wattie
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Lesley Wiltshire
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Nadia Alsaji
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | | | - Eric Cheng
- St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada; (E.C.); (T.S.)
| | - Thivya Srinathan
- St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada; (E.C.); (T.S.)
| | - Terence Ho
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
- The Research Institute of St. Joe’s Hamilton, Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada
| | - Roma Sehmi
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
- The Research Institute of St. Joe’s Hamilton, Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada
| | - Paul M. O’Byrne
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
- Hamilton Health Sciences, Hamilton, ON L8N 3Z5, Canada;
- St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada; (E.C.); (T.S.)
| | - Maryonne Snow-Smith
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (M.S.-S.); (M.M.); (A.D.K.)
| | - Michelle Makiya
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (M.S.-S.); (M.M.); (A.D.K.)
| | - Amy D. Klion
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (M.S.-S.); (M.M.); (A.D.K.)
| | - MyLinh Duong
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
- The Research Institute of St. Joe’s Hamilton, Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada
- Population Health Research Institute, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Gail M. Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| |
Collapse
|
20
|
Fekri MS, Najminejad Z, Karami Robati F, Dalfardi B, Lashkarizadeh M, Najafzadeh MJ. Eosinophils and chronic obstructive pulmonary diseases (COPD) in hospitalized COVID-19 patients. BMC Infect Dis 2024; 24:553. [PMID: 38831292 PMCID: PMC11149342 DOI: 10.1186/s12879-024-09373-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/03/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND The emergence of coronavirus disease 2019 (COVID-19) as a global health emergency necessitates continued investigation of the disease progression. This study investigated the relationship between eosinophilia and the severity of COVID-19 in chronic obstructive pulmonary disease (COPD) patients. METHODS This cross-sectional study was conducted on 73 COPD patients infected by COVID-19 in Afzalipour Hospital, Iran. Peripheral blood samples were collected for hematological parameter testing, including eosinophil percentage, using Giemsa staining. Eosinophilia was defined as≥ 2% and non-eosinophilia as< 2%. The severity of pulmonary involvement was determined based on chest CT severity score (CT-SS) (based on the degree of involvement of the lung lobes, 0%: 0 points, 1-25%: 1 point, 26-50%: 2 points, 51-75%: 3 points, and 76-100%: 4 points). The CT-SS was the sum of the scores of the five lobes (range 0-20). RESULTS The average age of patients was 67.90±13.71 years, and most were male (54.8%). Non-eosinophilic COPD patients were associated with more severe COVID-19 (P= 0.01) and lower oxygen saturation (P= 0.001). In addition, the study revealed a significant difference in the chest CT severity score (CT-SS) between non-eosinophilic (9.76±0.7) and eosinophilic COPD patients (6.26±0.63) (P< 0.001). Although non-eosinophilic COPD patients had a higher mortality rate, this difference was not statistically significant (P= 0.16). CONCLUSIONS Our study demonstrated that reduced peripheral blood eosinophil levels in COPD patients with COVID-19 correlate with unfavorable outcomes. Understanding this association can help us identify high-risk COPD patients and take appropriate management strategies to improve their prognosis.
Collapse
Affiliation(s)
- Mitra Samareh Fekri
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Zohreh Najminejad
- Clinical Research Development Unit, Afzalipour Hospital' Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Karami Robati
- Clinical Research Development Unit, Afzalipour Hospital' Kerman University of Medical Sciences, Kerman, Iran
| | - Behnam Dalfardi
- Advanced Thoracic Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdiyeh Lashkarizadeh
- Department of Pathology and Stem Cell Research Center, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
21
|
Jenks JD, Hoenigl M, Thompson GR. Study protocol: A randomized, double-blind, placebo-controlled trial of isavuconazole prophylaxis for the prevention of covid-19-associated pulmonary aspergillosis. Contemp Clin Trials Commun 2024; 39:101310. [PMID: 38832095 PMCID: PMC11144754 DOI: 10.1016/j.conctc.2024.101310] [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: 01/05/2024] [Revised: 03/28/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024] Open
Abstract
Background During the early stages of the coronavirus disease 2019 (COVID-19) pandemic, those with severe COVID-19 infection were at risk for a number of opportunistic infections including COVID-19-associated pulmonary aspergillosis (CAPA). We initiated a randomized clinical trial to evaluate whether isavuconazole, a triazole antifungal, could prevent CAPA and improve survival in patients admitted to the ICU with severe COVID-19 infection. Methods We designed a phase III/IV randomized, double-blind, two-arm, placebo-controlled trial evaluating standard of care (SOC) plus isavuconazole versus SOC plus placebo and were to enroll participants admitted to the ICU with severe COVID-19 infection at three medical centers in California, United States. The projected sample size was 162 participants. Results Due to poor enrollment and the declining number of COVID-19 cases over time, the study was terminated after 7 participants were enrolled, all enrolled at one study site (UC San Diego Health). CAPA was suspected in two participants and they were started on open-label isavuconazole. One was withdrawn due to possible isavuconazole-related adverse side effects. Conclusion Enrollment was slower-than-expected due to multiple factors, including competing COVID-19-related studies and hesitancy from potential study participants or their families to join the study. Our experience highlights some of the difficulties in planning and running a clinical trial focused on fungal superinfections involving severely ill patients during the height of the COVID-19 pandemic. Lessons learned from this study will help in the design of proposed studies examining antifungal prophylaxis against aspergillosis following other severe respiratory viral infections.
Collapse
Affiliation(s)
- Jeffrey D. Jenks
- Durham County Department of Public Health, Durham, NC, USA
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, NC, USA
| | - Martin Hoenigl
- Division of Infectious Diseases, ECMM Excellence Center for Medical Mycology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- BioTechMed, Graz, Austria
| | - George R. Thompson
- University of California Davis Center for Valley Fever, Sacramento, CA, USA
- Department of Internal Medicine, Division of Infectious Diseases, University of California Davis Medical Center, Sacramento, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| |
Collapse
|
22
|
Clark JJ, Penrice-Randal R, Sharma P, Dong X, Pennington SH, Marriott AE, Colombo S, Davidson A, Kavanagh Williamson M, Matthews DA, Turtle L, Prince T, Hughes GL, Patterson EI, Shawli G, Mega DF, Subramaniam K, Sharp J, Turner JD, Biagini GA, Owen A, Kipar A, Hiscox JA, Stewart JP. Sequential Infection with Influenza A Virus Followed by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Leads to More Severe Disease and Encephalitis in a Mouse Model of COVID-19. Viruses 2024; 16:863. [PMID: 38932155 PMCID: PMC11209060 DOI: 10.3390/v16060863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
COVID-19 is a spectrum of clinical symptoms in humans caused by infection with SARS-CoV-2. The coalescence of SARS-CoV-2 with seasonal respiratory viruses, particularly influenza viruses, is a global health concern. To understand this, transgenic mice expressing the human ACE2 receptor (K18-hACE2) were infected with influenza A virus (IAV) followed by SARS-CoV-2 and the host response and effect on virus biology was compared to K18-hACE2 mice infected with IAV or SARS-CoV-2 alone. The sequentially infected mice showed reduced SARS-CoV-2 RNA synthesis, yet exhibited more rapid weight loss, more severe lung damage and a prolongation of the innate response compared to the singly infected or control mice. Sequential infection also exacerbated the extrapulmonary encephalitic manifestations associated with SARS-CoV-2 infection. Conversely, prior infection with a commercially available, multivalent live-attenuated influenza vaccine (Fluenz Tetra) elicited the same reduction in SARS-CoV-2 RNA synthesis, albeit without the associated increase in disease severity. This suggests that the innate immune response stimulated by IAV inhibits SARS-CoV-2. Interestingly, infection with an attenuated, apathogenic influenza vaccine does not result in an aberrant immune response and enhanced disease severity. Taken together, the data suggest coinfection ('twinfection') is deleterious and mitigation steps should be instituted as part of the comprehensive public health and management strategy of COVID-19.
Collapse
Affiliation(s)
- Jordan J. Clark
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK (R.P.-R.); (P.S.); (T.P.); (G.S.); (A.K.)
| | - Rebekah Penrice-Randal
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK (R.P.-R.); (P.S.); (T.P.); (G.S.); (A.K.)
| | - Parul Sharma
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK (R.P.-R.); (P.S.); (T.P.); (G.S.); (A.K.)
| | - Xiaofeng Dong
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK (R.P.-R.); (P.S.); (T.P.); (G.S.); (A.K.)
| | - Shaun H. Pennington
- Department of Tropical Disease Biology, Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK (J.D.T.)
| | - Amy E. Marriott
- Department of Tropical Disease Biology, Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK (J.D.T.)
| | - Stefano Colombo
- Department of Tropical Disease Biology, Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK (J.D.T.)
| | - Andrew Davidson
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol BS8 1QU, UK; (A.D.); (D.A.M.)
| | - Maia Kavanagh Williamson
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol BS8 1QU, UK; (A.D.); (D.A.M.)
| | - David A. Matthews
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol BS8 1QU, UK; (A.D.); (D.A.M.)
| | - Lance Turtle
- Department of Clinical Infection Microbiology and Immunology and NIHR Health Protection Research Unit for Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK
- Tropical & Infectious Disease Unit, Royal Liverpool University Hospital, Liverpool L7 8YE, UK
| | - Tessa Prince
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK (R.P.-R.); (P.S.); (T.P.); (G.S.); (A.K.)
| | - Grant L. Hughes
- Departments of Vector Biology and Tropical Disease Biology, Centre for Neglected Tropical Disease, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK; (G.L.H.)
| | - Edward I. Patterson
- Departments of Vector Biology and Tropical Disease Biology, Centre for Neglected Tropical Disease, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK; (G.L.H.)
| | - Ghada Shawli
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK (R.P.-R.); (P.S.); (T.P.); (G.S.); (A.K.)
| | - Daniele F. Mega
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK (R.P.-R.); (P.S.); (T.P.); (G.S.); (A.K.)
| | - Krishanthi Subramaniam
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK (R.P.-R.); (P.S.); (T.P.); (G.S.); (A.K.)
| | - Jo Sharp
- Department of Pharmacology and Therapeutics, Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool L69 3BX, UK; (J.S.); (A.O.)
| | - Joseph D. Turner
- Department of Tropical Disease Biology, Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK (J.D.T.)
| | - Giancarlo A. Biagini
- Department of Tropical Disease Biology, Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK (J.D.T.)
| | - Andrew Owen
- Department of Pharmacology and Therapeutics, Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool L69 3BX, UK; (J.S.); (A.O.)
| | - Anja Kipar
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK (R.P.-R.); (P.S.); (T.P.); (G.S.); (A.K.)
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland
| | - Julian A. Hiscox
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK (R.P.-R.); (P.S.); (T.P.); (G.S.); (A.K.)
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
- Infectious Diseases Horizontal Technology Centre (ID HTC), Agency for Science, Technology and Research (A*STAR), Singapore 138632, Singapore
| | - James P. Stewart
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK (R.P.-R.); (P.S.); (T.P.); (G.S.); (A.K.)
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
- Department of Infectious Disease, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
23
|
Qian X, Zuo Z, Xu D, He S, Zhou C, Wang Z, Xie S, Zhang Y, Wu F, Lyu F, Zhang L, Qian Z. Demystifying COVID-19 mortality causes with interpretable data mining. Sci Rep 2024; 14:10076. [PMID: 38698064 PMCID: PMC11066015 DOI: 10.1038/s41598-024-60841-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: 01/31/2024] [Accepted: 04/28/2024] [Indexed: 05/05/2024] Open
Abstract
While COVID-19 becomes periodical, old individuals remain vulnerable to severe disease with high mortality. Although there have been some studies on revealing different risk factors affecting the death of COVID-19 patients, researchers rarely provide a comprehensive analysis to reveal the relationships and interactive effects of the risk factors of COVID-19 mortality, especially in the elderly. Through retrospectively including 1917 COVID-19 patients (102 were dead) admitted to Xiangya Hospital from December 2022 to March 2023, we used the association rule mining method to identify the risk factors leading causes of death among the elderly. Firstly, we used the Affinity Propagation clustering to extract key features from the dataset. Then, we applied the Apriori Algorithm to obtain 6 groups of abnormal feature combinations with significant increments in mortality rate. The results showed a relationship between the number of abnormal feature combinations and mortality rates within different groups. Patients with "C-reactive protein > 8 mg/L", "neutrophils percentage > 75.0 %", "lymphocytes percentage < 20%", and "albumin < 40 g/L" have a 2 × mortality rate than the basic one. When the characteristics of "D-dimer > 0.5 mg/L" and "WBC > 9.5 × 10 9 /L" are continuously included in this foundation, the mortality rate can be increased to 3 × or 4 × . In addition, we also found that liver and kidney diseases significantly affect patient mortality, and the mortality rate can be as high as 100%. These findings can support auxiliary diagnosis and treatment to facilitate early intervention in patients, thereby reducing patient mortality.
Collapse
Affiliation(s)
- Xinyu Qian
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Zhihong Zuo
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Danni Xu
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Shanyun He
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Conghao Zhou
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada
| | - Zhanwen Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shucai Xie
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongmin Zhang
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Fan Wu
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China.
| | - Feng Lyu
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China.
| | - Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zhaoxin Qian
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
Jiang J, Shu H, Wang DW, Hui R, Li C, Ran X, Wang H, Zhang J, Nie S, Cui G, Xiang D, Shao Q, Xu S, Zhou N, Li Y, Gao W, Chen Y, Bian Y, Wang G, Xia L, Wang Y, Zhao C, Zhang Z, Zhao Y, Wang J, Chen S, Jiang H, Chen J, Du X, Chen M, Sun Y, Li S, Ding H, Ma X, Zeng H, Lin L, Zhou S, Ma L, Tao L, Chen J, Zhou Y, Guo X. Chinese Society of Cardiology guidelines on the diagnosis and treatment of adult fulminant myocarditis. SCIENCE CHINA. LIFE SCIENCES 2024; 67:913-939. [PMID: 38332216 DOI: 10.1007/s11427-023-2421-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/25/2023] [Indexed: 02/10/2024]
Abstract
Fulminant myocarditis is an acute diffuse inflammatory disease of myocardium. It is characterized by acute onset, rapid progress and high risk of death. Its pathogenesis involves excessive immune activation of the innate immune system and formation of inflammatory storm. According to China's practical experience, the adoption of the "life support-based comprehensive treatment regimen" (with mechanical circulation support and immunomodulation therapy as the core) can significantly improve the survival rate and long-term prognosis. Special emphasis is placed on very early identification,very early diagnosis,very early prediction and very early treatment.
Collapse
Affiliation(s)
- Jiangang Jiang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongyang Shu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dao Wen Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Rutai Hui
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Chenze Li
- Zhongnan Hospital of Wuhan University, Wuhan, 430062, China
| | - Xiao Ran
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hong Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Zhang
- Fuwai Huazhong Cardiovascular Hospital, Zhengzhou, 450003, China
| | - Shaoping Nie
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Guanglin Cui
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dingcheng Xiang
- Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, China
| | - Qun Shao
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Shengyong Xu
- Union Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ning Zhou
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuming Li
- Taida Hospital, Tianjin, 300457, China
| | - Wei Gao
- Peking University Third Hospital, Beijing, 100191, China
| | - Yuguo Chen
- Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yuan Bian
- Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Guoping Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Liming Xia
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chunxia Zhao
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiren Zhang
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yuhua Zhao
- Kanghua Hospital, Dongguan, Guangzhou, 523080, China
| | - Jianan Wang
- Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shaoliang Chen
- Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Hong Jiang
- Renmin Hospital of Wuhan University, Wuhan, 430060, Wuhan, China
| | - Jing Chen
- Renmin Hospital of Wuhan University, Wuhan, 430060, Wuhan, China
| | - Xianjin Du
- Renmin Hospital of Wuhan University, Wuhan, 430060, Wuhan, China
| | - Mao Chen
- West China Hospital, Sichuan University, Chengdu, 610044, China
| | - Yinxian Sun
- First Hospital of China Medical University, Shenyang, 110002, China
| | - Sheng Li
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hu Ding
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xueping Ma
- General Hospital of Ningxia Medical University, Yinchuan, 750003, China
| | - Hesong Zeng
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Lin
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shenghua Zhou
- The Second Xiangya Hospital, Central South University, Changsha, 410012, China
| | - Likun Ma
- The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230002, China
| | - Ling Tao
- The First Affiliated Hospital of Air Force Medical University, Xi'an, 710032, China
| | - Juan Chen
- Central Hospital of Wuhan City, Wuhan, 430014, China
| | - Yiwu Zhou
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaomei Guo
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
25
|
Aboras SI, Megahed AA, El-Yazbi F, Maher HM. Utility of sustainable ratio derivative spectrophotometry for the concurrent assay of synergistic repurposed drugs for COVID-19 infections; Insilico pharmacokinetics proof. BMC Chem 2024; 18:50. [PMID: 38454503 PMCID: PMC10921645 DOI: 10.1186/s13065-024-01147-w] [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: 10/12/2023] [Accepted: 02/19/2024] [Indexed: 03/09/2024] Open
Abstract
The cutting-edge combination of fluvoxamine (FVM) and ivermectin (IVM) has been presented as a proposed dosage form for the treatment of COVID-19 infections in early diagnosed patients. The main objective of this work is to develop simple, sensitive, and efficient methods for the synchronous quantification of FVM and IVM without any prior separation. Four green UV-methods were employed for the synchronous quantification, namely: Fourier functions convolution of absorption spectra, FFAS, Fourier functions convolution of derivative spectra of absorption curves, FFDS, Fourier function convolution of ratio spectra of absorption curves, FFRS and the dual-wavelength method, DWM. FFRS and DWM approaches can be able to reconcile the two components' significantly interfering spectrum presented in this commixture. Good linearity was checked in the range of 5-40, and 2.5-25 μg/mL for the FVM, and IVM, respectively. All approaches developed have been recommended in compliance with ICH principles. Furthermore, the approaches' greenness was predestined by "National Environmental Method Index" (NEMI), "Analytical GREEnness metric (AGREE)", the "Analytical Eco-Scale", and the "Green Analytical Procedure Index" (GAPI). In addition, spider diagram was utilized for the assessment of the greenness index of the solvent used. Beside greenness, the sustainability of our methods was investigated using the HEXAGON tool. Continuing the constant pursuit of greenness, drug-drug interactions (DDIs) between FVM & IVM were predicted by insilico tools to ensure the safety of the suggested mixture as a preliminary step before invitro and in vivo studies. Because they were deemed sustainable, affordable, and successful, the suggested UV-methods may be used for routine quality control investigations of the indicated formulations FVM & IVM.
Collapse
Affiliation(s)
- Sara I Aboras
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, University of Alexandria, Al-mesallah, Alexandria, 21521, Egypt.
| | - Ahmed A Megahed
- Al-Basra Health Unit, Alamriya Medical Area, Ministry of Health, Alexandria, Egypt
| | - Fawzy El-Yazbi
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, University of Alexandria, Al-mesallah, Alexandria, 21521, Egypt
| | - Hadir M Maher
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, University of Alexandria, Al-mesallah, Alexandria, 21521, Egypt
| |
Collapse
|
26
|
López-Herrero R, Sánchez-de Prada L, Tamayo-Velasco A, Heredia-Rodríguez M, Bardají Carrillo M, Jorge Monjas P, de la Varga-Martínez O, Resino S, Sarmentero-López de Quintana G, Gómez-Sánchez E, Tamayo E. Epidemiology of fungal infection in COVID 19 in Spain during 2020 and 2021: a nationwide study. Sci Rep 2024; 14:5203. [PMID: 38433130 PMCID: PMC10909879 DOI: 10.1038/s41598-024-54340-1] [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: 11/19/2023] [Accepted: 02/12/2024] [Indexed: 03/05/2024] Open
Abstract
We realize a nationwide population-based retrospective study to analyze the characteristics and risk factors of fungal co-infections in COVID-19 hospitalized patients as well as describe their causative agents in the Spanish population in 2020 and 2021. Data were obtained from records in the Minimum Basic Data Set of the National Surveillance System for Hospital Data in Spain, provided by the Ministry of Health, and annually published with two years lag. The assessment of the risk associated with the development of healthcare-associated fungal co-infections was assessed using an adjusted logistic regression model. The incidence of fungal co-infection in COVID-19 hospitalized patients was 1.41%. The main risk factors associated were surgery, sepsis, age, male gender, obesity, and COPD. Co-infection was associated with worse outcomes including higher in-hospital and in ICU mortality, and higher length of stay. Candida spp. and Aspergillus spp. were the microorganisms more frequent. This is the first study analyzing fungal coinfection at a national level in hospitalized patients with COVID-19 in Spanish population and one of the few studies available that demonstrate that surgery was an independent risk factor of Aspergillosis coinfection in COVID-19 patients.
Collapse
Affiliation(s)
- R López-Herrero
- BioCritic, Group for Biomedical Research in Critical Care Medicine, 47005, Valladolid, Spain
- Anesthesiology and Critical Care Department, Hospital Clínico Universitario de Valladolid, 47003, Valladolid, Spain
- Department of Surgery, Faculty of Medicine, Universidad de Valladolid, 47005, Valladolid, Spain
| | - L Sánchez-de Prada
- BioCritic, Group for Biomedical Research in Critical Care Medicine, 47005, Valladolid, Spain
- Microbiology Department, Hospital Universitario Río Hortega, 47012, Valladolid, Spain
| | - A Tamayo-Velasco
- BioCritic, Group for Biomedical Research in Critical Care Medicine, 47005, Valladolid, Spain.
- Haematology and Hemotherapy Department, Hospital Clínico Universitario de Valladolid, 47003, Valladolid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| | - M Heredia-Rodríguez
- BioCritic, Group for Biomedical Research in Critical Care Medicine, 47005, Valladolid, Spain
- Anesthesiology and Critical Care Department, Complejo Asistencial Universitario de Salamanca, 37007, Salamanca, Spain
| | - M Bardají Carrillo
- BioCritic, Group for Biomedical Research in Critical Care Medicine, 47005, Valladolid, Spain
- Anesthesiology and Critical Care Department, Hospital Clínico Universitario de Valladolid, 47003, Valladolid, Spain
| | - P Jorge Monjas
- BioCritic, Group for Biomedical Research in Critical Care Medicine, 47005, Valladolid, Spain
- Anesthesiology and Critical Care Department, Hospital Clínico Universitario de Valladolid, 47003, Valladolid, Spain
- Department of Surgery, Faculty of Medicine, Universidad de Valladolid, 47005, Valladolid, Spain
| | - O de la Varga-Martínez
- BioCritic, Group for Biomedical Research in Critical Care Medicine, 47005, Valladolid, Spain
- Department of Anesthesiology, Hospital Universitario Infanta Leonor, 28031, Madrid, Spain
| | - S Resino
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - G Sarmentero-López de Quintana
- BioCritic, Group for Biomedical Research in Critical Care Medicine, 47005, Valladolid, Spain
- Anesthesiology and Critical Care Department, Hospital Clínico Universitario de Valladolid, 47003, Valladolid, Spain
| | - E Gómez-Sánchez
- BioCritic, Group for Biomedical Research in Critical Care Medicine, 47005, Valladolid, Spain
- Anesthesiology and Critical Care Department, Hospital Clínico Universitario de Valladolid, 47003, Valladolid, Spain
- Department of Surgery, Faculty of Medicine, Universidad de Valladolid, 47005, Valladolid, Spain
| | - E Tamayo
- BioCritic, Group for Biomedical Research in Critical Care Medicine, 47005, Valladolid, Spain
- Anesthesiology and Critical Care Department, Hospital Clínico Universitario de Valladolid, 47003, Valladolid, Spain
- Department of Surgery, Faculty of Medicine, Universidad de Valladolid, 47005, Valladolid, Spain
| |
Collapse
|
27
|
de Azambuja Pias Weber A, Viero FT, Pillat MM, de Lima Gonçalves T. Changes in markers of inflammation and their correlation with death in patients with COVID-19 in the intensive care unit. Cytokine 2024; 175:156509. [PMID: 38241964 DOI: 10.1016/j.cyto.2024.156509] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/07/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
This study aimed to characterize the changes in serum inflammatory mediators in hospitalized patients with COVID-19 correlating with death. The study includes 58 participants: i) inpatients (n = 37): patients suffering from severe acute respiratory syndrome due to COVID-19, who were admitted at Intensive Care Unit (ICU) recovered and who died and ii) control group (n = 21): community volunteers. Inflammatory mediators evaluated interleukin-2 (IL-2), interleukin-4 (IL-4), interleukin-6 (IL-6), interleukin-10 (IL-10), interleukin-17 (IL-17), interferon-gamma (IFN-γ) and interferon-gamma protein levels (IFN-γ), as well as, Urea, LDH, D-dimer, TAP/INR, AST, ALT and lymphocytes. Our results suggest that high levels of inflammatory markers, such as pro-inflammatory cytokines, and laboratory parameters, such as low levels of lymphocytes and high levels of IL-6, are associated with disease severity, especially in individuals who died. Constant measurement and monitoring of these inflammatory parameters is an effective tool in clinical practice, and it can help choosing appropriate therapies during the course of the disease.
Collapse
Affiliation(s)
- Andressa de Azambuja Pias Weber
- Postgraduate Program in Pharmaceutical Sciences, Department of Clinical and Toxicology Analysis, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| | - Fernanda Tibolla Viero
- Postgraduate Program in Pharmacology, Department of Microbiology and Parasitology, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| | - Micheli Mainardi Pillat
- Postgraduate Program in Pharmacology, Department of Microbiology and Parasitology, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| | - Thissiane de Lima Gonçalves
- Postgraduate Program in Pharmaceutical Sciences, Department of Clinical and Toxicology Analysis, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| |
Collapse
|
28
|
Michels EHA, Appelman B, de Brabander J, van Amstel RBE, van Linge CCA, Chouchane O, Reijnders TDY, Schuurman AR, Sulzer TAL, Klarenbeek AM, Douma RA, Bos LDJ, Wiersinga WJ, Peters-Sengers H, van der Poll T. Host Response Changes and Their Association with Mortality in COVID-19 Patients with Lymphopenia. Am J Respir Crit Care Med 2024; 209:402-416. [PMID: 37948687 PMCID: PMC10878379 DOI: 10.1164/rccm.202305-0890oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/09/2023] [Indexed: 11/12/2023] Open
Abstract
Rationale: Lymphopenia in coronavirus disease (COVID-19) is associated with increased mortality. Objectives: To explore the association between lymphopenia, host response aberrations, and mortality in patients with lymphopenic COVID-19. Methods: We determined 43 plasma biomarkers reflective of four pathophysiological domains: endothelial cell and coagulation activation, inflammation and organ damage, cytokine release, and chemokine release. We explored if decreased concentrations of lymphocyte-derived proteins in patients with lymphopenia were associated with an increase in mortality. We sought to identify host response phenotypes in patients with lymphopenia by cluster analysis of plasma biomarkers. Measurements and Main Results: A total of 439 general ward patients with COVID-19 were stratified by baseline lymphocyte counts: normal (>1.0 × 109/L; n = 167), mild lymphopenia (>0.5 to ⩽1.0 × 109/L; n = 194), and severe lymphopenia (⩽0.5 × 109/L; n = 78). Lymphopenia was associated with alterations in each host response domain. Lymphopenia was associated with increased mortality. Moreover, in patients with lymphopenia (n = 272), decreased concentrations of several lymphocyte-derived proteins (e.g., CCL5, IL-4, IL-13, IL-17A) were associated with an increase in mortality (at P < 0.01 or stronger significance levels). A cluster analysis revealed three host response phenotypes in patients with lymphopenia: "hyporesponsive" (23.2%), "hypercytokinemic" (36.4%), and "inflammatory-injurious" (40.4%), with substantially differing mortality rates of 9.5%, 5.1%, and 26.4%, respectively. A 10-biomarker model accurately predicted these host response phenotypes in an external cohort with similar mortality distribution. The inflammatory-injurious phenotype showed a remarkable combination of relatively high inflammation and organ damage markers with high antiinflammatory cytokine levels yet low proinflammatory cytokine levels. Conclusions: Lymphopenia in COVID-19 signifies a heterogenous group of patients with distinct host response features. Specific host responses contribute to lymphopenia-associated mortality in COVID-19, including reduced CCL5 levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Renée A. Douma
- Department of Internal Medicine, Flevo Hospital, Almere, the Netherlands; and
| | | | - W. Joost Wiersinga
- Center for Experimental and Molecular Medicine
- Division of Infectious Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Hessel Peters-Sengers
- Center for Experimental and Molecular Medicine
- Department of Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine
- Division of Infectious Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
29
|
Hao X, Bao Z, Dai R, Wu X, Li X, Zhang M, Li H, Xu L, Qiao P, Liu X, Hu W, Zhang Z, Fang J, Zhou M, Wang W, Qu J. A pilot study on Paxlovid therapy for hemodialysis patients with severe acute respiratory syndrome coronavirus 2 infections. Front Med 2024; 18:169-179. [PMID: 37978164 DOI: 10.1007/s11684-023-1011-0] [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: 01/01/2023] [Accepted: 05/19/2023] [Indexed: 11/19/2023]
Abstract
We aimed to investigate the safety and efficacy of nirmatrelvir/ritonavir (Paxlovid) therapy for hemodialysis-dependent patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Thirteen hemodialysis patients infected with the Omicron variant of SARS-CoV-2 from April 3 to May 30, 2022, were recruited. Laboratory parameters and chest CT (computed tomography) imaging were analyzed. The treatment group included six patients who received 150 mg/100 mg of Paxlovid orally once daily for 5 days, whereas the control group included seven patients who received basic treatment. No serious adverse reactions or safety events were recorded. Four control patients progressed to moderate disease, and none in the treatment group showed progression of chest CT findings (P < 0.05). Paxlovid therapy tended toward early viral clearance and low viral load on Day 8. Moreover, 83.3% of the patients in the treatment group and 57.1% of the patients in the control group turned negative within 22 days. In the Paxlovid treatment group, we found significantly increased levels of lymphocytes (P=0.03) and eosinophils (P=0.02) and decreased levels of D-dimer on Day 8 compared with those on Day 1. Paxlovid therapy showed a potential therapeutic effect with good tolerance in hemodialysis patients. The optimal dose and effectiveness evaluation must be further investigated in a largeer cohort.
Collapse
Affiliation(s)
- Xu Hao
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhiyao Bao
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ranran Dai
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaojing Wu
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Li
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Muyin Zhang
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hao Li
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lili Xu
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Panpan Qiao
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xuefei Liu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Weiting Hu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ze Zhang
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Fang
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Min Zhou
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Weiming Wang
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
30
|
Kovács A, Hantosi D, Szabó N, Letoha A, Lengyel C, Földesi I, Burián K, Palkó A, Veréb D, Kincses ZT. D-dimer levels to exclude pulmonary embolism and reduce the need for CT angiography in COVID-19 in an outpatient population. PLoS One 2024; 19:e0297023. [PMID: 38232069 DOI: 10.1371/journal.pone.0297023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 12/26/2023] [Indexed: 01/19/2024] Open
Abstract
OBJECTIVES Emerging results indicate that, in COVID-19, thromboembolic complications contribute to the high mortality and morbidity. Previous research showed that the prevalence of pulmonary embolism (PE) is between 25-50% in COVID-19 patients, however, most of these reports are based on data from patients with severe pneumonia, treated in intensive care units. MATERIALS AND METHODS We conducted a retrospective, single-center, observational study to estimate the prevalence of PE in COVID-19 patients who underwent CT angiography and to identify the most important predictors. Adult outpatients with COVID-19, who presented at our COVID Outpatient Clinic between 1st and 31st of March in 2021 and underwent CTA examination were included in this study. Multiple linear regression analysis was used to identify predictors of PE in COVID-19 patients. The predictors were: age, gender, disease duration, CT severity index and log-transformed quantitative D-dimer (logQDDIM) value. RESULTS 843 COVID-19 patients were included into the study. 82.56% (693 patients) of the infected patients had a pulmonary CTA examination and D-dimer levels (mean age: 59.82 years ± 15.66). 7.61% (53 patients) of the patients had PE. 2.02% (14 patients) of the patients had main branch or lobar PE. The multiple regression analysis found that only logQDDIM was a significant predictor. A logQDDIM cut-off value of 0.0169 (1.0171 ug/ml serum D-dimer) predicted PE with 99% sensitivity (p<0.0001, degree-of-freedom = 570, AUC = 0.72). CONCLUSIONS We demonstrated in a large cohort of COVID-19 patients that a cut-off value of QDDIM of 1ug/ml can exclude pulmonary embolism in an outpatient setting, implicating that QDDIM might potentially supersede CTA as a screening approach in COVID-19 outpatient clinics.
Collapse
Affiliation(s)
- Anita Kovács
- Department of Radiology, Albert Szent-Györgyi Medical Center, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Dóra Hantosi
- Department of Radiology, Albert Szent-Györgyi Medical Center, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Nikoletta Szabó
- Department of Neurology, Albert Szent-Györgyi Medical Center, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Annamária Letoha
- Department of Internal Medicine, Albert Szent-Györgyi Medical Center, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Csaba Lengyel
- Department of Internal Medicine, Albert Szent-Györgyi Medical Center, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical Center, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Katalin Burián
- Department of Medical Microbiology, Albert Szent-Györgyi Medical Center, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - András Palkó
- Department of Radiology, Albert Szent-Györgyi Medical Center, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Dániel Veréb
- Department of Radiology, Albert Szent-Györgyi Medical Center, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Zsigmond Tamás Kincses
- Department of Radiology, Albert Szent-Györgyi Medical Center, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| |
Collapse
|
31
|
Chen L, Gong P, Su Y, Meng L, Wang M, Gao W, Liu Q. Angiotensin II type 2 receptor agonist attenuates LPS-induced acute lung injury through modulating THP-1-derived macrophage reprogramming. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:99-108. [PMID: 37368029 DOI: 10.1007/s00210-023-02589-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a devastating respiratory disorder, characterized by overwhelming inflammation in the alveoli without effective pharmacological treatment. We aimed to investigate the effect and mechanism of angiotensin II type 2 receptor (AT2R) agonist, Compound 21 (C21), on the lipopolysaccharide (LPS)-induced acute lung injury (ALI) model. The protective effect of C21 was evaluated via enzyme-linked immunosorbent assay (ELISA), Western blot (WB), real-time PCR, and fluorescence microscopy in LPS-challenged THP1-derived macrophages. Besides, the in vivo efficacy of C21 was assessed using cell counting, ELISA, protein quantification, hematoxylin-eosin (H&E) staining, and WB in an LPS-induced ALI mouse model. The results showed that C21 significantly inhibited the secretion of pro-inflammatory cytokines (CCL-2, IL-6), overproduction of intracellular ROS, and activation of inflammatory pathways (NF-κB/NLRP3, p38/MAPK) in THP-1 cell-derived macrophages stimulated by LPS. In in vivo study, intraperitoneal injection of C21 could reduce airway leukocytes accumulation and chemokine/cytokine (keratinocyte chemoattractant (KC), IL-6) generation, as well as alleviate diffuse alveolar damage induced by LPS. Conclusively, the AT2R agonist C21 significantly inhibited LPS-stimulated excess inflammatory responses and oxidative stress in macrophages. Meanwhile, C21 could effectively alleviate acute inflammation and tissue damage in the lungs of ALI mice challenged by LPS. The results of this study bring new hope for the early treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Liangzhi Chen
- Shandong University of Traditional Chinese Medicine, Shandong, 250002, People's Republic of China
| | - Ping Gong
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine Shandong University, Shandong, 250033, People's Republic of China
| | - Yue Su
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China
| | - Linlin Meng
- Shandong University of Traditional Chinese Medicine, Shandong, 250002, People's Republic of China
| | - Muyun Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China
| | - Wei Gao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China.
| | - Qinghua Liu
- Shandong University of Traditional Chinese Medicine, Shandong, 250002, People's Republic of China.
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China.
| |
Collapse
|
32
|
Kim SG, Han CH, Yu SB, Lee H, Kwon S, Kim Y, Lee J, Kim DK, Oh YK, Lim CS, Kim YS, Kim BG, Lee JP. Trajectory of AKI and hospital mortality among patients with COVID-19. Ren Fail 2023; 45:2177086. [PMID: 36876658 PMCID: PMC10013401 DOI: 10.1080/0886022x.2023.2177086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND Acute kidney injury (AKI) in COVID-19 patients is associated with poor prognosis. Characterization of AKI by timing and trajectory and early prediction of AKI progression is required for better preventive management and the prediction of patient outcomes. METHODS A total of 858 patients who were hospitalized due to coronavirus disease 2019 (COVID-19) were retrospectively enrolled from December 2020 to August 2021. The occurrence of AKI was evaluated throughout hospitalization. The hazard ratios (HRs) of mortality outcomes according to the trajectory of AKI were measured using Cox regression models after adjustment for multiple variables. RESULTS Among 858 patients, 226 (26.3%) presented AKI at admission, and 44 (5.1%) developed AKI during hospitalization. Patients with AKI at admission or hospital-acquired AKI had a higher risk of mortality than those without AKI, with HRs of 9.87 (2.81-34.67) and 13.74 (3.57-52.84), respectively. Of 226 patients with AKI at admission, 104 (46.0%) recovered within 48 hr, 83 (36.7%) had AKI beyond 48 hr and recovered in 7 days, and 39 (17.3%) showed no recovery from AKI on Day 7. Delayed recovery and persistent AKI were significantly associated with an increased risk of mortality, with HRs of 4.39 (1.06-18.24) and 24.33 (7.10-83.36), respectively. CONCLUSIONS The onset and progression of AKI was significantly associated with in-hospital mortality in patients with COVID-19. A thorough observation of the recovery trajectory of early AKI after infection is necessary.
Collapse
Affiliation(s)
- Seong Geun Kim
- Department of Internal Medicine-Nephrology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chung Hee Han
- Department of Obstetrics and Gynecology, Bagaehospital, Pyeongtaek, Gyunggi-Do, Republic of Korea
| | - Sung Bong Yu
- Department of Surgery, Bagaehospital, Pyeongtaek, Gyunggi-Do, Republic of Korea
| | - Hyeseung Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Soie Kwon
- Department of Internal Medicine-Nephrology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yerim Kim
- Department of Internal Medicine-Nephrology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Jeonghwan Lee
- Department of Internal Medicine-Nephrology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Ki Kim
- Department of Internal Medicine-Nephrology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun Kyu Oh
- Department of Internal Medicine-Nephrology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Chun Soo Lim
- Department of Internal Medicine-Nephrology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Yon Su Kim
- Department of Internal Medicine-Nephrology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Byung Gun Kim
- Department of Orthopedic Surgery, Bagaehospital, Pyeongtaek, Gyunggi-Do, Republic of Korea
| | - Jung Pyo Lee
- Department of Internal Medicine-Nephrology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| |
Collapse
|
33
|
Han Y, Yang Z, Fang S, Zhang M, Xie Z, Fan Y, Zhao T. Data-mining-based of ancient traditional Chinese medicine records from 475 BC to 1949 to potentially treat COVID-19. Anat Rec (Hoboken) 2023; 306:2984-2996. [PMID: 35263033 PMCID: PMC9082487 DOI: 10.1002/ar.24888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/09/2021] [Accepted: 12/01/2021] [Indexed: 12/17/2022]
Abstract
Traditional Chinese Medicine (TCM) plays a role in preventing and treating COVID-19 in China. Based on the manifestations and symptoms of COVID-19, our study used the data mining method to summarize related therapeutic experience left by predecessors who used TCM to treat epidemics in their eras. Initially, we collected abundant medical records with similar manifestations of COVID-19 in Chinese ancient times. The key words including wen (), yi (), li (), and zhang () were searched in ZhongyiZhiku (https://www.zk120.com/) from Warring States Period (475 BC-221 BC) to the Republic of China era (1912-1949) to locate ancient medical records according to inclusion criteria and exclusion criteria. Moreover, COVID-19-related manifestations and corresponding medications in those records were categorized. Eventually, Traditional Chinese Medicine Inheritance Support System version 2.5 was used to build a medical record database of TCM treating COVID-19. Our study collected 263 epidemic medical records comprising COVID-19 related manifestations and found that Chinese Materia Medica (CMM) combinations excavated from ancient medical records included Ren Shen Bai Du San, Wu Ling San, Xiao Chai Hu Tang, Da Cheng Qi Tang, Da Chai Hu Tang, Ling Gui Zhu Gan Tang, and Qing Wen Bai Du Yin. The recurrent CMMs with a high frequency for treating COVID-19 manifestations were Scutellariae Radix (Huang Qin), Paeoniae Alba Radix (Bai Shao), Poria (Fu Ling), and Bupleuri Radix (Chai Hu). Our study suggests that TCM might offer new therapeutic strategies for COVID-19.
Collapse
Affiliation(s)
- Yaxue Han
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Zi Yang
- The Third School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Shan Fang
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Mengqing Zhang
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Zhijun Xie
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Yongsheng Fan
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Ting Zhao
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| |
Collapse
|
34
|
Liu Y, Rajeevan H, Simonov M, Lee S, Wilson FP, Desir GV, Vinetz JM, Yan X, Wang Z, Clark BJ, Possick JD, Price C, Lutchmansingh DD, Ortega H, Zaeh S, Gomez JVL, Cohn L, Gautam S, Chupp GL. Differences in Mortality Among Patients With Asthma and COPD Hospitalized With COVID-19. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:3383-3390.e3. [PMID: 37454926 PMCID: PMC10787810 DOI: 10.1016/j.jaip.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/14/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND It remains unclear whether patients with asthma and/or chronic obstructive pulmonary disease (COPD) are at increased risk for severe coronavirus disease 2019 (COVID-19). OBJECTIVE Compare in-hospital COVID-19 outcomes among patients with asthma, COPD, and no airway disease. METHODS A retrospective cohort study was conducted on 8,395 patients admitted with COVID-19 between March 2020 and April 2021. Airway disease diagnoses were defined using International Classification of Diseases, 10th Revision codes. Mortality and sequential organ failure assessment (SOFA) scores were compared among groups. Logistic regression analysis was used to identify and adjust for confounding clinical features associated with mortality. RESULTS The median SOFA score in patients without airway disease was 0.32 and mortality was 11%. In comparison, asthma patients had lower SOFA scores (median 0.15; P < .01) and decreased mortality, even after adjusting for age, diabetes, and other confounders (odds ratio 0.65; P = .01). Patients with COPD had higher SOFA scores (median 0.86; P < .01) and increased adjusted odds of mortality (odds ratio 1.40; P < .01). Blood eosinophil count of 200 cells/μL or greater, a marker of type 2 inflammation, was associated with lower mortality across all groups. Importantly, patients with asthma showed improved outcomes even after adjusting for eosinophilia, indicating that noneosinophilic asthma was associated with protection as well. CONCLUSIONS COVID-19 severity was increased in patients with COPD and decreased in those with asthma, eosinophilia, and noneosinophilic asthma, independent of clinical confounders. These findings suggest that COVID-19 severity may be influenced by intrinsic immunological factors in patients with airway diseases, such as type 2 inflammation.
Collapse
Affiliation(s)
- Yunqing Liu
- Department of Biostatistics, Yale School of Public Health, New Haven, Conn
| | - Haseena Rajeevan
- Biomedical Informatics & Data Science, Yale School of Medicine, New Haven, Conn; Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Michael Simonov
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn; Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Seohyuk Lee
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - F Perry Wilson
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn; Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Gary V Desir
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Joseph M Vinetz
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Xiting Yan
- Department of Biostatistics, Yale School of Public Health, New Haven, Conn; Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, Conn
| | - Brian J Clark
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Jennifer D Possick
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Christina Price
- Section of Allergy and Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Denyse D Lutchmansingh
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Hector Ortega
- Clinical Development, Nexstone Immunology, San Diego, Calif
| | - Sandra Zaeh
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Jose Villa-Lobos Gomez
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Lauren Cohn
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Samir Gautam
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Geoffrey L Chupp
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn.
| |
Collapse
|
35
|
Mellett L, Amarasinghe G, Farnsworth CW, Khader SA. Elevated Level of Circulating but Not Urine S100A8/A9 Identifies Poor COVID-19 Outcomes. ACS Infect Dis 2023; 9:1815-1820. [PMID: 37787461 PMCID: PMC10580308 DOI: 10.1021/acsinfecdis.3c00249] [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: 05/30/2023] [Indexed: 10/04/2023]
Abstract
The alarmin calprotectin (S100A8/A9) is thought to drive a cytokine storm, a hallmark of severe COVID-19. Recent studies report circulating S100A8/A9 levels can distinguish COVID-19 severity but have only been conducted in non-U.S. cohorts and mainly focus on serum S100A8/A9 levels. Thus, we quantified S100A8/A9 in serum and urine samples from a hospital cohort in St. Louis, Missouri, to expand the understanding of S100A8/A9 as a prognostic biomarker for COVID-19. Elevated S100A8/A9 serum levels were observed in ICU patients (n = 49, p = 0.0370) and patients with fatal cases of COVID-19 (n = 76, p = 0.0018). We observed no correlation in the S100A8/A9 levels in matched serum and urine samples. Our results support the association of serum S100A8/A9 levels with COVID-19 severity and suggest that further investigation of urine S100A8/A9 as a COVID-19 biomarker is not warranted.
Collapse
Affiliation(s)
- Leah Mellett
- Department
of Molecular Microbiology, Washington University
in St. Louis, St. Louis, Missouri 63108, United States
- Department
of Pathology and Immunology, Washington
University School of Medicine, St. Louis, Missouri 63108, United States
| | - Gaya Amarasinghe
- Department
of Pathology and Immunology, Washington
University School of Medicine, St. Louis, Missouri 63108, United States
| | - Christopher W. Farnsworth
- Department
of Pathology and Immunology, Washington
University School of Medicine, St. Louis, Missouri 63108, United States
| | - Shabaana A. Khader
- Department
of Molecular Microbiology, Washington University
in St. Louis, St. Louis, Missouri 63108, United States
- Department
of Microbiology, University of Chicago, Chicago, Illinois 60637 United States
| |
Collapse
|
36
|
D'Carmo Sodré MM, Dos Santos UR, Povoas HP, Guzmán JL, Junqueira C, Trindade TO, Gadelha SR, Romano CC, da Conceição AO, Gross E, Silva A, Rezende RP, Fontana R, da Mata CPSM, Marin LJ, de Carvalho LD. Relationship between clinical-epidemiological parameters and outcomes of patients with COVID-19 admitted to the intensive care unit: a report from a Brazilian hospital. Front Public Health 2023; 11:1241444. [PMID: 37808991 PMCID: PMC10556466 DOI: 10.3389/fpubh.2023.1241444] [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: 06/16/2023] [Accepted: 08/28/2023] [Indexed: 10/10/2023] Open
Abstract
Background People in low-income countries, especially those with low socio-economic conditions, are likelier to test positive for SARS-CoV-2. The unequal conditions of public health systems also increase the infection rate and make early identification and treatment of at-risk patients difficult. Here, we aimed to characterize the epidemiological profile of COVID-19 patients in intensive care and identify laboratory and clinical markers associated with death. Materials and methods We conducted an observational, descriptive, and cross-sectional study in a reference hospital for COVID-19 treatment in the Southern Region of Bahia State, in Brazil, to evaluate the epidemiological, clinical, and laboratory characteristics of COVID-19 patients admitted to the intensive care unit (ICU). Additionally, we used the area under the curve (AUC) to classify survivors and non-survivors and a multivariate logistic regression analysis to assess factors associated with death. Data was collected from the hospital databases between April 2020 and July 2021. Results The use of bladder catheters (OR 79.30; p < 0.0001) and central venous catheters (OR, 45.12; p < 0.0001) were the main factors associated with death in ICU COVID-19 patients. Additionally, the number of non-survivors increased with age (p < 0.0001) and prolonged ICU stay (p < 0.0001). Besides, SAPS3 presents a higher sensibility (77.9%) and specificity (63.1%) to discriminate between survivors and non-survivor with an AUC of 0.79 (p < 0.0001). Conclusion We suggest that multi-laboratory parameters can predict patient prognosis and guide healthcare teams toward more assertive clinical management, better resource allocation, and improved survival of COVID-19 patients admitted to the ICU.
Collapse
Affiliation(s)
| | | | | | | | - Caroline Junqueira
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | | | - Sandra Rocha Gadelha
- Department of Biological Sciences, Santa Cruz State University, Ilhéus, Bahia, Brazil
| | - Carla Cristina Romano
- Department of Biological Sciences, Santa Cruz State University, Ilhéus, Bahia, Brazil
| | | | - Eduardo Gross
- Department of Biological Sciences, Santa Cruz State University, Ilhéus, Bahia, Brazil
| | - Aline Silva
- Department of Biological Sciences, Santa Cruz State University, Ilhéus, Bahia, Brazil
| | - Rachel Passos Rezende
- Department of Biological Sciences, Santa Cruz State University, Ilhéus, Bahia, Brazil
| | - Renato Fontana
- Department of Biological Sciences, Santa Cruz State University, Ilhéus, Bahia, Brazil
| | | | | | | |
Collapse
|
37
|
Wan EYF, Zhang R, Mathur S, Yan VKC, Lai FTT, Chui CSL, Li X, Wong CKH, Chan EWY, Lau CS, Wong ICK. Post-acute sequelae of COVID-19 in older persons: multi-organ complications and mortality. J Travel Med 2023; 30:taad082. [PMID: 37310901 DOI: 10.1093/jtm/taad082] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/29/2023] [Accepted: 06/10/2023] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Evidence on long-term associations between coronavirus disease 2019 (COVID-19) and risks of multi-organ complications and mortality in older population is limited. This study evaluates these associations. RESEARCH DESIGN AND METHODS The cohorts included patients aged ≥60 year diagnosed with COVID-19 infection (cases), between 16 March 2020 and 31 May 2021 from the UK Biobank; and between 01 April 2020 and 31 May 2022 from the electronic health records in Hong Kong. Each patient was randomly matched with individuals without COVID-19 infection based on year of birth and sex and were followed for up to 18 months until 31 August 2021 for UKB, and up to 28 months until 15 August 2022 for HK cohort. Patients with COVID-19 infection over 6 months after the date of last dose of vaccination and their corresponding controls were excluded from our study. Characteristics between cohorts were further adjusted with Inverse Probability Treatment Weighting. For evaluating long-term association of COVID-19 with multi-organ disease complications and mortality after 21-days of diagnosis, Cox regression was employed. RESULT 10,759 (UKB) and 165,259 (HK) older adults with COVID-19 infection with matched 291,077 (UKB) and 1,100,394 (HK) non-COVID-19-diagnosed older adults were recruited. Older adults with COVID-19 were associated with a significantly higher risk of cardiovascular outcomes [major cardiovascular disease (stroke, heart failure and coronary heart disease): hazard ratio(UKB): 1.4 (95% Confidence interval: 1.1,1.6), HK:1.2 (95% CI: 1.1,1.3)]; myocardial infarction: HR(UKB): 1.8 (95% CI: 1.3,2.4), HK:1.2 (95% CI: 1.0,1.4)]; respiratory outcomes [interstitial lung disease: HR(UKB: 3.4 (95% CI: 2.5,4.5), HK: 4.0 (95% CI: 1.3,12.8); chronic pulmonary disease: HR(UKB): 1.7 (95% CI: 1.3,2.2), HK:1.6 (95% CI: 1.3,2.1)]; neuropsychiatric outcomes [seizure: HR(UKB): 2.6 (95% CI: 1.7,4.1), HK: 1.6 (95% CI: 1.2,2.1)]; and renal outcomes [acute kidney disease: HR(UKB): 1.4 (95% CI: 1.1,1.6), HK:1.6 (95% CI: 1.3,2.1)]; and all-cause mortality [HR(UKB): 4.9 (95% CI: 4.4,5.4), HK:2.5 (95% CI: 2.5,2.6)]. CONCLUSION COVID-19 is associated with long-term risks of multi-organ complications in older adults (aged ≥ 60). Infected patients in this age-group may benefit from appropriate monitoring of signs/symptoms for developing these complications.
Collapse
Affiliation(s)
- Eric Yuk Fai Wan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ran Zhang
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sukriti Mathur
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Vincent Ka Chun Yan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Francisco Tsz Tsun Lai
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
| | - Celine Sze Ling Chui
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- School of Nursing, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xue Li
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Carlos King Ho Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Esther Wai Yin Chan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Pharmacy, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- The University of Hong Kong Shenzhen Institute of Research and Innovation, Shenzhen 518053, China
| | - Chak Sing Lau
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ian Chi Kei Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Aston Pharmacy School, Aston University, Birmingham B4 7ET, UK
| |
Collapse
|
38
|
Lentzen M, Linden T, Veeranki S, Madan S, Kramer D, Leodolter W, Frohlich H. A Transformer-Based Model Trained on Large Scale Claims Data for Prediction of Severe COVID-19 Disease Progression. IEEE J Biomed Health Inform 2023; 27:4548-4558. [PMID: 37347632 DOI: 10.1109/jbhi.2023.3288768] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
In situations like the COVID-19 pandemic, healthcare systems are under enormous pressure as they can rapidly collapse under the burden of the crisis. Machine learning (ML) based risk models could lift the burden by identifying patients with a high risk of severe disease progression. Electronic Health Records (EHRs) provide crucial sources of information to develop these models because they rely on routinely collected healthcare data. However, EHR data is challenging for training ML models because it contains irregularly timestamped diagnosis, prescription, and procedure codes. For such data, transformer-based models are promising. We extended the previously published Med-BERT model by including age, sex, medications, quantitative clinical measures, and state information. After pre-training on approximately 988 million EHRs from 3.5 million patients, we developed models to predict Acute Respiratory Manifestations (ARM) risk using the medical history of 80,211 COVID-19 patients. Compared to Random Forests, XGBoost, and RETAIN, our transformer-based models more accurately forecast the risk of developing ARM after COVID-19 infection. We used Integrated Gradients and Bayesian networks to understand the link between the essential features of our model. Finally, we evaluated adapting our model to Austrian in-patient data. Our study highlights the promise of predictive transformer-based models for precision medicine.
Collapse
|
39
|
Sarıoğlu E, Sarıaltın SY, Çoban T. Neurological complications and effects of COVID-19: Symptoms and conceivable mechanisms. BRAIN HEMORRHAGES 2023; 4:154-173. [PMID: 36789140 PMCID: PMC9911160 DOI: 10.1016/j.hest.2023.02.001] [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: 01/12/2023] [Revised: 02/04/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
A novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first identified in December 2019 in Wuhan, China. The new coronavirus disease (COVID-19) was declared a global pandemic by the World Health Organization (WHO) in March 2020. SARS-CoV-2 can invade the nervous system aside from infecting the respiratory system as its primary target. The most common nervous system symptoms of COVID-19 are stated as headache, myalgia, fatigue, nausea, vomiting, sudden and unexplained anosmia, and ageusia. More severe conditions such as encephalomyelitis, acute myelitis, thromboembolic events, ischemic stroke, intracerebral hemorrhage, Guillain-Barré-syndrome, Bell's palsy, rhabdomyolysis, and even coma have also been reported. Cohort studies revealed that neurological findings are associated with higher morbidity and mortality. The neurological symptoms and manifestations caused by SARS-CoV-2 and COVID-19 are examined and summarized in this article.
Collapse
Affiliation(s)
- Elif Sarıoğlu
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 06560 Ankara, Turkey
| | - Sezen Yılmaz Sarıaltın
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 06560 Ankara, Turkey
| | - Tülay Çoban
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 06560 Ankara, Turkey
| |
Collapse
|
40
|
Silva MDJ, de Andrade CM, Fiuza BSD, Pinheiro GP, Nova Santana CV, Costa RDS, Barnes K, Cruz ÁA, Figueiredo CA. Genetic variants associated with SARS-CoV-2 infection also affect lung function and asthma severity. Heliyon 2023; 9:e19235. [PMID: 37662742 PMCID: PMC10474403 DOI: 10.1016/j.heliyon.2023.e19235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/10/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Abstract
Background Host genetic factors may be associated with COVID-19 unfavourable outcomes. The first genome-wide association study (GWAS) conducted in individuals with respiratory failure due to COVID-19 revealed susceptibility loci close to six genes (SLC6A20, LZTFL1, CCR9, FYCO1, CXCR6 and XCR1) and the ABO blood-group gene. We aimed to investigate how polymorphisms in those genes could relate to lung function and severe asthma in a Brazilian population. Methods DNA samples of 784 individuals following the ProAR (Programa para Controle da Asma e Rinite Alérgica da Bahia) were genotyped by the Multi-Ethnic Global Array panel with ∼2 million polymorphisms (Illumina). Polymorphisms in SLC6A20, LZTFL1, CCR9, FYCO1, CXCR6, XCR1 and the ABO blood-group gene were evaluated. Logistic regression for severe asthma, airway obstruction and lack of FEV1 reversibility was performed using PLINK software 1.9, in the additive model and was adjusted for sex, age and PCA-1. Pairwise Linkage disequilibrium analyses were performed using Haploview 4.2. The haplotypes and gene score analyses were performed in the SNPstat tool. In silico functions of polymorphisms were analysed using rSNPbase and RegulomeDB plataforms. Results We identified the rs8176733 (G allele) and rs8176725 (A allele) in the ABO blood-group gene as risk factors for severe asthma, lower pulmonary obstruction and lack of FEV1 reversibility. Polymorphisms in CCR9 are risk factors for both severe asthma (A allele of rs34338823) and airway obstruction (A allele of rs6806802). The markers rs13079478 (A allele) and rs75817942 (A allele) in FYCO1 are related to more severe asthma and a lack of FEV1 reversibility, respectively. We identified the A allele of both rs35731912 and rs34338823 in LZTFL1 as risk factors for severe asthma. The marker rs6806802 (C allele) was associated with airway obstruction and rs7614952 (A allele), rs7625839 (G allele) and rs112509260 (A allele) are related to a lack of FEV1 reversibility. The A allele of rs2531747 in the SLC6A20 gene is also associated with severe asthma. Conversely, polymorphisms in XCR1 play a protective role in relation to severe asthma (A allele of rs2036295) and airway obstruction (A allele of rs2036295). Additionally, we found that individuals with a higher number of risk alleles have a greater risk of severe asthma, airway obstruction and FEV1 reversibility. Conclusion Our study suggests that polymorphisms in genes associated with respiratory failure in SARS-CoV-2-infected individuals are associated with greater susceptibility to severe asthma and reduced lung function in subjects with asthma.
Collapse
Affiliation(s)
| | | | | | | | | | - Ryan dos S. Costa
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Kathleen Barnes
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Álvaro A. Cruz
- Fundação ProAR and Faculdade de Medicina da Universidade Federal da Bahia, Brazil
| | | |
Collapse
|
41
|
Romero MJ, Yue Q, Singla B, Hamacher J, Sridhar S, Moseley AS, Song C, Mraheil MA, Fischer B, Zeitlinger M, Chakraborty T, Fulton D, Gan L, Annex BH, Csanyi G, Eaton DC, Lucas R. Direct endothelial ENaC activation mitigates vasculopathy induced by SARS-CoV2 spike protein. Front Immunol 2023; 14:1241448. [PMID: 37638055 PMCID: PMC10449264 DOI: 10.3389/fimmu.2023.1241448] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Although both COVID-19 and non-COVID-19 ARDS can be accompanied by significantly increased levels of circulating cytokines, the former significantly differs from the latter by its higher vasculopathy, characterized by increased oxidative stress and coagulopathy in lung capillaries. This points towards the existence of SARS-CoV2-specific factors and mechanisms that can sensitize the endothelium towards becoming dysfunctional. Although the virus is rarely detected within endothelial cells or in the circulation, the S1 subunit of its spike protein, which contains the receptor binding domain (RBD) for human ACE2 (hACE2), can be detected in plasma from COVID-19 patients and its levels correlate with disease severity. It remains obscure how the SARS-CoV2 RBD exerts its deleterious actions in lung endothelium and whether there are mechanisms to mitigate this. Methods In this study, we use a combination of in vitro studies in RBD-treated human lung microvascular endothelial cells (HL-MVEC), including electrophysiology, barrier function, oxidative stress and human ACE2 (hACE2) surface protein expression measurements with in vivo studies in transgenic mice globally expressing human ACE2 and injected with RBD. Results We show that SARS-CoV2 RBD impairs endothelial ENaC activity, reduces surface hACE2 expression and increases reactive oxygen species (ROS) and tissue factor (TF) generation in monolayers of HL-MVEC, as such promoting barrier dysfunction and coagulopathy. The TNF-derived TIP peptide (a.k.a. solnatide, AP301) -which directly activates ENaC upon binding to its a subunit- can override RBD-induced impairment of ENaC function and hACE2 expression, mitigates ROS and TF generation and restores barrier function in HL-MVEC monolayers. In correlation with the increased mortality observed in COVID-19 patients co-infected with S. pneumoniae, compared to subjects solely infected with SARS-CoV2, we observe that prior intraperitoneal RBD treatment in transgenic mice globally expressing hACE2 significantly increases fibrin deposition and capillary leak upon intratracheal instillation of S. pneumoniae and that this is mitigated by TIP peptide treatment.
Collapse
Affiliation(s)
- Maritza J. Romero
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Qian Yue
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Bhupesh Singla
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Jürg Hamacher
- Pneumology, Clinic for General Internal Medicine, Lindenhofspital, Bern, Switzerland
- Lungen-und Atmungsstiftung, Bern, Switzerland
- Medical Clinic V—Pneumology, Allergology, Intensive Care Medicine, and Environmental Medicine, Faculty of Medicine, Saarland University, University Medical Centre of the Saarland, Homburg, Germany
| | - Supriya Sridhar
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Auriel S. Moseley
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Chang Song
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Mobarak A. Mraheil
- Institute for Medical Microbiology, German Centre for Infection Giessen-Marburg-Langen Site, Faculty of Medicine, Justus-Liebig University, Giessen, Germany
| | | | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Trinad Chakraborty
- Institute for Medical Microbiology, German Centre for Infection Giessen-Marburg-Langen Site, Faculty of Medicine, Justus-Liebig University, Giessen, Germany
| | - David Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Lin Gan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Brian H. Annex
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Gabor Csanyi
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Douglas C. Eaton
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Division of Pulmonary and Critical Care Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
42
|
Valdetaro L, Thomasi B, Ricciardi MC, Santos KDM, Coelho-Aguiar JDM, Tavares-Gomes AL. Enteric nervous system as a target and source of SARS-CoV-2 and other viral infections. Am J Physiol Gastrointest Liver Physiol 2023; 325:G93-G108. [PMID: 37253656 PMCID: PMC10390051 DOI: 10.1152/ajpgi.00229.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 05/02/2023] [Accepted: 05/29/2023] [Indexed: 06/01/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has been demonstrated to affect several systems of the human body, including the gastrointestinal and nervous systems. The enteric nervous system (ENS) is a division of the autonomic nervous system that extends throughout the gut, regulates gastrointestinal function, and is therefore involved in most gut dysfunctions, including those resulting from many viral infections. Growing evidence highlights enteric neural cells and microbiota as important players in gut inflammation and dysfunction. Furthermore, the ENS and gastrointestinal immune system work together establishing relevant neuroimmune interactions during both health and disease. In recent years, gut-driven processes have also been implicated as players in systemic inflammation and in the initiation and propagation of several central nervous system pathologies, which seem to be hallmarks of COVID-19. In this review, we aim to describe evidence of the gastrointestinal and ENS infection with a focus on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We discuss here viral-induced mechanisms, neuroplasticity, and neuroinflammation to call attention to the enteric neuroglial network as a nervous system with a sensitive and crucial position to be not only a target of the new coronavirus but also a way in and trigger of COVID-19-related symptoms.
Collapse
Affiliation(s)
- Luisa Valdetaro
- Postgraduate Program in Neuroscience, Neurobiology Department, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, New York, United States
| | - Beatriz Thomasi
- Postgraduate Program in Neuroscience, Neurobiology Department, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
| | - Maria Carolina Ricciardi
- Postgraduate Program in Neuroscience, Neurobiology Department, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| | - Karoline de Melo Santos
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Ana Lúcia Tavares-Gomes
- Postgraduate Program in Neuroscience, Neurobiology Department, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Wu H, Ruan W, Wang J, Zheng D, Liu B, Geng Y, Chai X, Chen J, Li K, Li S, Helal S. Interpretable Machine Learning for COVID-19: An Empirical Study on Severity Prediction Task. IEEE TRANSACTIONS ON ARTIFICIAL INTELLIGENCE 2023; 4:764-777. [PMID: 37954545 PMCID: PMC10620962 DOI: 10.1109/tai.2021.3092698] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/07/2021] [Accepted: 06/08/2021] [Indexed: 11/14/2023]
Abstract
The black-box nature of machine learning models hinders the deployment of some high-accuracy medical diagnosis algorithms. It is risky to put one's life in the hands of models that medical researchers do not fully understand or trust. However, through model interpretation, black-box models can promptly reveal significant biomarkers that medical practitioners may have overlooked due to the surge of infected patients in the COVID-19 pandemic. This research leverages a database of 92 patients with confirmed SARS-CoV-2 laboratory tests between 18th January 2020 and 5th March 2020, in Zhuhai, China, to identify biomarkers indicative of infection severity prediction. Through the interpretation of four machine learning models, decision tree, random forests, gradient boosted trees, and neural networks using permutation feature importance, partial dependence plot, individual conditional expectation, accumulated local effects, local interpretable model-agnostic explanations, and Shapley additive explanation, we identify an increase in N-terminal pro-brain natriuretic peptide, C-reaction protein, and lactic dehydrogenase, a decrease in lymphocyte is associated with severe infection and an increased risk of death, which is consistent with recent medical research on COVID-19 and other research using dedicated models. We further validate our methods on a large open dataset with 5644 confirmed patients from the Hospital Israelita Albert Einstein, at São Paulo, Brazil from Kaggle, and unveil leukocytes, eosinophils, and platelets as three indicative biomarkers for COVID-19.
Collapse
Affiliation(s)
- Han Wu
- University of ExeterEX4 4PYExeterU.K.
| | | | | | | | - Bei Liu
- Department of Gastroenterology910 Hospital of PLABeijingChina
| | - Yayuan Geng
- Scientific Research Department BeijingHY Medical TechnologyBeijing100192China
| | - Xiangfei Chai
- Scientific Research Department BeijingHY Medical TechnologyBeijing100192China
| | - Jian Chen
- Department of RadiologyHospital of Sun Yat-sen UniversityZhuhai519000China
| | - Kunwei Li
- Department of RadiologyHospital of Sun Yat-sen UniversityZhuhai519000China
| | - Shaolin Li
- Department of Radiology, and Guangdong Provincial Key Laboratory of Biomedical ImagingHospital of Sun Yat-sen UniversityZhuhai519000China
| | - Sumi Helal
- University of FloridaGainesvilleFL32611USA
| |
Collapse
|
44
|
Yaluri N, Stančáková Yaluri A, Žeňuch P, Žeňuchová Z, Tóth Š, Kalanin P. Cardiac Biomarkers and Their Role in Identifying Increased Risk of Cardiovascular Complications in COVID-19 Patients. Diagnostics (Basel) 2023; 13:2508. [PMID: 37568870 PMCID: PMC10417576 DOI: 10.3390/diagnostics13152508] [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: 05/29/2023] [Revised: 07/03/2023] [Accepted: 07/17/2023] [Indexed: 08/13/2023] Open
Abstract
Cardiovascular disease (CVD) is a global health concern, causing significant morbidity and mortality. Both lifestyle and genetics influence the development of CVD. It is often diagnosed late, when the treatment options are limited. Early diagnosis of CVD with help of biomarkers is necessary to prevent adverse outcomes. SARS-CoV-2 infection can cause cardiovascular complications even in patients with no prior history of CVD. This review highlights cardiovascular biomarkers, including novel ones, and their applications as diagnostic and prognostic markers of cardiovascular complications related to SARS-CoV-2 infection. Patients with severe SARS-CoV-2 infection were shown to have elevated levels of cardiac biomarkers, namely N-terminal pro-brain natriuretic peptide (NT-pro-BNP), creatine kinase-myocardial band (CK-MB), and troponins, indicating acute myocardial damage. These biomarkers were also associated with higher mortality rates and therefore should be used throughout COVID-19 patient care to identify high-risk patients promptly to optimize their outcomes. Additionally, microRNAs (miRNAs) are also considered as potential biomarkers and predictors of cardiac and vascular damage in SARS-CoV-2 infection. Identifying molecular pathways contributing to cardiovascular manifestations in COVID-19 is essential for development of early biomarkers, identification of new therapeutic targets, and better prediction and management of cardiovascular outcomes.
Collapse
Affiliation(s)
- Nagendra Yaluri
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | | | - Pavol Žeňuch
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Zuzana Žeňuchová
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Štefan Tóth
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Peter Kalanin
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| |
Collapse
|
45
|
Mihajlović A, Ivanov D, Tapavički B, Marković M, Vukas D, Miljković A, Bajić D, Semnic I, Bogdan M, Karaba Jakovljević D, Nikolić S, Slavić D, Lendak D. Prognostic Value of Routine Biomarkers in the Early Stage of COVID-19. Healthcare (Basel) 2023; 11:2137. [PMID: 37570378 PMCID: PMC10418955 DOI: 10.3390/healthcare11152137] [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: 06/26/2023] [Revised: 07/18/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
Various biomarkers like certain complete blood cell count parameters and the derived ratios including neutrophil-lymphocyte ratio are commonly used to evaluate disease severity. Our study aimed to establish if baseline levels of complete blood cell count-derived biomarkers and CRP, measured before any treatment which can interfere with their values, could serve as a predictor of development of pneumonia and the need for hospitalization requiring oxygen therapy. We retrospectively analyzed the laboratory data of 200 consecutive patients without comorbidities, who denied usage of medications prior to blood analysis and visited a COVID-19 ambulance between October and December 2021. Multivariate regression analysis extracted older age, elevated CRP and lower eosinophil count as significant independent predictors of pneumonia (p = 0.003, p = 0.000, p = 0.046, respectively). Independent predictors of hospitalization were higher CRP (p = 0.000) and lower platelet count (p = 0.005). There was no significant difference in the neutrophil-lymphocyte and platelet-lymphocyte ratios between examined groups. Individual biomarkers such as platelet and eosinophil count might be better in predicting the severity of COVID-19 than the neutrophil-lymphocyte and platelet-lymphocyte ratios.
Collapse
Affiliation(s)
- Andrea Mihajlović
- Department of Physiology, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21137 Novi Sad, Serbia
| | - David Ivanov
- Department of Physiology, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21137 Novi Sad, Serbia
| | - Borislav Tapavički
- Department of Physiology, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21137 Novi Sad, Serbia
| | - Milica Marković
- Health Centre Novi Sad, Bulevar Cara Lazara 75, 21102 Novi Sad, Serbia
| | - Dragana Vukas
- Health Centre Novi Sad, Bulevar Cara Lazara 75, 21102 Novi Sad, Serbia
| | - Ana Miljković
- Health Centre Novi Sad, Bulevar Cara Lazara 75, 21102 Novi Sad, Serbia
- Department of General Medicine and Geriatrics, Faculty of Medicine Novi Sad, University of Novi Sad, Hajduk Veljkova 3, 21137 Novi Sad, Serbia
| | - Dejana Bajić
- Department of Biochemistry, Faculty of Medicine Novi Sad, University of Novi Sad, Hajduk Veljkova 3, 21137 Novi Sad, Serbia
| | - Isidora Semnic
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21137 Novi Sad, Serbia
- Clinic of Anesthesia and Intensive Care, University Clinical Center of Vojvodina, Hajduk Veljkova 1, 21137 Novi Sad, Serbia
| | - Maja Bogdan
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21137 Novi Sad, Serbia
- Institute for Pulmonary Diseases of Vojvodina, Put Dr Goldmana Street 4, 21204 Sremska Kamenica, Serbia
| | - Dea Karaba Jakovljević
- Department of Physiology, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21137 Novi Sad, Serbia
| | - Stanislava Nikolić
- Department of Pathophysiology and Laboratory Medicine, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21137 Novi Sad, Serbia
- Center of Laboratory Medicine, Clinical Center of Vojvodina, Hajduk Veljkova 1, 21137 Novi Sad, Serbia
| | - Danijel Slavić
- Department of Physiology, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21137 Novi Sad, Serbia
| | - Dajana Lendak
- Department of Infectious Diseases, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21137 Novi Sad, Serbia
- Clinic for Infectious Diseases, University Clinical Center of Vojvodina, Hajduk Veljkova 1, 21137 Novi Sad, Serbia
| |
Collapse
|
46
|
Pandak N, Khamis F, Al Balushi Z, Chhetri S, Al Lawati A, AbouElhamd H, Golchinheydari S, Sidrah AK, Al Jahwari SK, Al Dowaiki S. Low Rate of Bacterial Coinfections and Antibiotic Overprescribing During COVID-19 Pandemic: A Retrospective Study from Oman. Oman Med J 2023; 38:e525. [PMID: 37720342 PMCID: PMC10500094 DOI: 10.5001/omj.2023.83] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 12/04/2022] [Indexed: 09/19/2023] Open
Abstract
Objectives The recommended treatment for COVID-19 includes antiviral drugs, corticosteroids, immunomodulatory drugs, low molecular weight heparin, as well as antibiotics. Although COVID-19 is a viral disease, many studies indicate that antibiotics are prescribed frequently, mainly to treat suspected bacterial coinfection. At the same time, the prevalence of bacterial coinfections during COVID-19 is rather low indicating the significant antibiotic overuse in these patients. It is well known that this can trigger antibiotic bacterial resistance, and once it emerges the reversal of resistance is a complex and long-lasting process. The aims of this study were to estimate the prevalence of bacterial coinfections during the COVID-19 and to analyze the antibiotic treatment justification during this pandemic in Oman. Methods This retrospective analysis was conducted using the Royal Hospital COVID-19 Registry Database. The study analyzed demographic and clinical characteristics, as well as laboratory parameters and antibiotic treatment of hospitalized patients. Results During the study period, 584 patients were enrolled in the analysis. Coinfection was rare as it was confirmed in 0.9% of patients. Superinfections were present in 15.2% of patients. Gram-negative bacteria were isolated in 95 (69.9%) samples, gram-positive bacteria in 25 (18.4%) samples, while Candida spp. was found in 16 (11.8%) samples. On admission, empirical antibiotic treatment was started in 543 (93.0%) patients. Conclusions During COVID-19, coinfections are rarely seen and the overuse of antibiotics is not justified. The incidence of superinfections is the same as in other patients in healthcare settings caused by the same resistant microorganisms, which implies the use of even more.
Collapse
Affiliation(s)
- Nenad Pandak
- Infection Diseases Unit, Department of Medicine, Royal Hospital, Muscat, Oman
| | - Faryal Khamis
- Infection Diseases Unit, Department of Medicine, Royal Hospital, Muscat, Oman
| | - Zakariya Al Balushi
- Infection Diseases Unit, Department of Medicine, Royal Hospital, Muscat, Oman
| | - Shabnam Chhetri
- Infection Diseases Unit, Department of Medicine, Royal Hospital, Muscat, Oman
| | - Adil Al Lawati
- Acute Medicine Unit, Department of Medicine, Royal Hospital, Muscat, Oman
| | - Hend AbouElhamd
- Neurology Unit, Department of Medicine, Royal Hospital, Muscat, Oman
| | | | | | | | - Samata Al Dowaiki
- Infection Diseases Unit, Department of Medicine, Royal Hospital, Muscat, Oman
| |
Collapse
|
47
|
Zhang L, Wang Z, Lyu F, Liu C, Li C, Liu W, Ma X, Zhou J, Qian X, Qian Z, Lu Y. Characterizing distinct profiles of immune and inflammatory response with age to Omicron infection. Front Immunol 2023; 14:1189482. [PMID: 37457688 PMCID: PMC10348361 DOI: 10.3389/fimmu.2023.1189482] [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: 03/19/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Background Understanding inflammatory and immune responses to Omicron infection based on age is crucial when addressing this global health threat. However, the lacking of comprehensive elucidation hinders the development of distinct treatments tailored to different age populations. Methods 1299 cases of Omicron infection in Shanghai were enrolled between April 10, 2022 and June 3, 2022, dividing into three groups by ages: Adult group (18-59 years), Old group (60-79 years), and Elder group (≥ 80 years). Laboratory data including inflammatory cytokines, cellular, and humoral immunity were collected and analyzed. Results The mean age of Adult, Old, and Elder groups were 44.14, 69.98, and 89.35 years, respectively, with 40.9% being men. The Elder group patients exhibited higher white blood cell (WBC) counts and elevated levels of inflammatory cytokines, but their lymphocyte counts were relatively lower. In comparison to the Old group patients, the Elder group patients demonstrated significantly lower CD3+ T-cell counts, CD3+ T-cell proportion, CD4+ T-cell counts, CD8+ T-cell counts, and CD19+ B-cell counts, while the NK-cell counts were higher. Omicron negative patients displayed a higher proportion of CD19+ B-cells and higher levels of Complement-3 and IL-17 compared to the positive patients in the Old group. Omicron negative patients had lower WBC counts, CD3+CD8+ T-cells proportion, and the levels of serum amyloid A and IgA in the Elder group, but the CD4+/CD8+ ratio was higher. Conclusions Our study identified the distinct profiles of inflammatory and immune responses to Omicron infection varying with age and highlighted the diverse correlations between the levels of various biomarkers and Omicron infected/convalescent patients.
Collapse
Affiliation(s)
- Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhanwen Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Lyu
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Chun Liu
- Respiratory and Critical Care Medicine Department, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Chunhui Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinhua Ma
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jieyu Zhou
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Xinyu Qian
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Zhaoxin Qian
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Lu
- Department of Radiology, Ruijin Hospital Luwan Branch, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
48
|
Shirure VS, Yechikov S, Shergill BS, Dehghani T, Block AV, Sodhi H, Panitch A, George SC. Mitigating neutrophil trafficking and cardiotoxicity with DS-IkL in a microphysiological system of a cytokine storm. LAB ON A CHIP 2023; 23:3050-3061. [PMID: 37278194 PMCID: PMC10330849 DOI: 10.1039/d2lc01070d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A feature of severe COVID-19 is the onset of an acute and intense systemic inflammatory response referred to as the "cytokine storm". The cytokine storm is characterized by high serum levels of inflammatory cytokines and the subsequent transport of inflammatory cells to damaging levels in vital organs (e.g., myocarditis). Immune trafficking and its effect on underlying tissues (e.g., myocardium) are challenging to observe at a high spatial and temporal resolution in mouse models. In this study, we created a vascularized organ-on-a-chip system to mimic cytokine storm-like conditions and tested the effectiveness of a novel multivalent selectin-targeting carbohydrate conjugate (composed of DS - dermatan sulfate and IkL - a selectin-binding peptide, termed DS-IkL) in blocking infiltration of polymorphonuclear leukocytes (PMN). Our data shows that cytokine storm-like conditions induce endothelial cells to produce additional inflammatory cytokines and facilitate infiltration of PMNs into tissue. Treatment of tissues with DS-IkL (60 μM) reduced PMN accumulation in the tissue by >50%. We then created cytokine storm-like conditions in a vascularized cardiac tissue-chip and found that PMN infiltration increases the spontaneous beating rate of the cardiac tissue, and this effect is eliminated by treatment with DS-IkL (60 μM). In summary, we demonstrate the utility of an organ-on-a-chip platform to mimic COVID-19 related cytokine storm and that blocking leukocyte infiltration with DS-IkL could be a viable strategy to mitigate associated cardiac complications.
Collapse
Affiliation(s)
- Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Sergey Yechikov
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Bhupinder S Shergill
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Tima Dehghani
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Anton V Block
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Harkanwalpreet Sodhi
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| |
Collapse
|
49
|
Shegay P, Leontyev A, Baranovskii D, Davydov G, Poluektova M, Grivtsova L, Petriev V, Stepanenko V, Gulidov I, Krylov V, Osadchaya S, Petrov V, Sedova M, Vekilyan M, Krasilnikova O, Morozov S, Ivanov S, Klabukov I, Kaprin A. World's First Experience of the Low-Dose Radionuclide Inhalation Therapy in the Treatment of COVID-19-Associated Viral Pneumonia: Phase 1/2 Clinical Trial. Curr Radiopharm 2023; 16:243-252. [PMID: 36880188 PMCID: PMC11851150 DOI: 10.2174/1874471016666230307113045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/13/2022] [Accepted: 01/04/2023] [Indexed: 03/08/2023]
Abstract
OBJECTIVE Previously, low-dose radiation therapy was used for pneumonia treatment. We aimed to investigate the safety and effectiveness of carbon nanoparticles labeled with Technetium isotope (99mTc) in a form of ultradispersed aerosol in combination with standard COVID-19 therapy. The study was a randomized phase 1 and phase 2 clinical trial of low-dose radionuclide inhalation therapy for patients with COVID-19 related pneumonia. METHODS We enrolled 47 patients with confirmed COVID-19 infection and early laboratory signs of cytokine storm and randomized them into the Treatment and Control groups. We analyzed blood parameters reflecting the COVID-19 severity and inflammatory response. RESULTS Low-dose 99mTc-labeled inhalation showed a minimal accumulation of radionuclide in lungs in healthy volunteers. We observed no significant differences between the groups before treatment in WBC-count, D-dimer, CRP, Ferritin or LDH levels. We found that Ferritin and LDH levels significantly raised after the 7th day follow-up only in the Control group (p < 0.0001 and p = 0.0005, respectively), while mean values of the same indicators did not change in patients in the Treatment group after the radionuclide treatment. D-dimer values also lowered in the radionuclide treated group, however, this effect was not statistically significant. Furthermore, we observed a significant decrease in CD19+ cell counts in patients of the radionuclide-treated group. CONCLUSION Inhalation low-dose radionuclide therapy of 99mTc aerosol affects the major prognostic indicators of COVID-19- related pneumonia restraining inflammatory response. Overall, we identified no evidence of major adverse events in the group receiving radionuclide.
Collapse
Affiliation(s)
- Peter Shegay
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Alexey Leontyev
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Denis Baranovskii
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Internal Medicine Department, 24 Moscow City State Hospital, Moscow, Russia
- Department of Urology and Operative Nephrology, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - German Davydov
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Marina Poluektova
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Lyudmila Grivtsova
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Vasily Petriev
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Valeriy Stepanenko
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Igor Gulidov
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Valeriy Krylov
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Svetlana Osadchaya
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Vladimir Petrov
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Maria Sedova
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Mikhail Vekilyan
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Olga Krasilnikova
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Sergey Morozov
- Research and Practical Center of Medical Radiology, Department of Health Care of Moscow, Moscow, Russia
| | - Sergey Ivanov
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Ilya Klabukov
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Department of Urology and Operative Nephrology, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
- Obninsk Institute for Nuclear Power Engineering, National Research Nuclear University MEPhI, Obninsk, Russia
| | - Andrey Kaprin
- Center of Innovative Radiological and Regenerative Technologies, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Department of Urology and Operative Nephrology, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
50
|
Rimini M, Franco P, Bertolini F, Berardino DB, Giulia ZM, Stefano V, Andrikou K, Arcadipane F, Napolitano M, Buno LV, Alessandra GM, Olivero F, Ferreri F, Ricardi U, Cascinu S, Casadei-Gardini A. The Prognostic Role of Baseline Eosinophils in HPV-Related Cancers: a Multi-institutional Analysis of Anal SCC and OPC Patients Treated with Radical CT-RT. J Gastrointest Cancer 2023; 54:662-671. [PMID: 35915202 PMCID: PMC9342937 DOI: 10.1007/s12029-022-00850-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND AND AIM Anal squamous cell carcinoma (SCC) and oropharyngeal cancer (OPC) are rare tumors associated with HPV infection. Bioumoral predictors of response to chemoradiation (CT-RT) are lacking in these settings. With the aim to find new biomarkers, we investigated the role of eosinophils in both HPV-positive anal SCC and HPV-related oropharyngeal cancer (OPC). METHODS We retrieved clinical and laboratory data of patients with HPV-positive anal SCC treated with CT-RT in 5 institutions, and patients with locally advanced OPC SCC treated with CT-RT in 2 institutions. We examined the association between baseline eosinophil count (the best cutoff has been evaluated by ROC curve analysis: 100 × 10^9/L) and disease-free survival (DFS). Unadjusted and adjusted hazard ratios by baseline characteristics were calculated using the Cox proportional hazards model. RESULTS Three hundred four patients with HPV-positive anal SCCs and 168 patients with OPCs (122 HPV-positive, 46 HPV-negative diseases) were analyzed. In anal SCC, low eosinophil count (< 100 × 10^9/L) correlates to a better DFS (HR = 0.59; p = 0.0392); likewise, in HPV-positive OPC, low eosinophil count correlates to a better DFS (HR = 0.50; p = 0.0428). In HPV-negative OPC, low eosinophil count confers worse DFS compared to high eosinophil count (HR = 3.53; p = 0.0098). After adjustment for age and sex, eosinophils were confirmed to be independent prognostic factors for DFS (HR = 4.55; p = 0.0139). CONCLUSION Eosinophil count could be used as a prognostic factor in anal HPV-positive SCC. The worse prognosis showed in HPV-positive patients with high eosinophil count is likely to derive from an unfavorable interaction between the HPV-induced immunomodulation and eosinophils, which may hamper the curative effect of RT.
Collapse
Affiliation(s)
- Margherita Rimini
- Oncologic Department, IRCCS San Raffaele Scientific Institute Hospital, 20019, Milan, Italy
| | - Pierfrancesco Franco
- Department of Oncology - Radiation Oncology, University of Turin School of Medicine, Via Genova 3, 10126, Turin, Italy.
| | - Federica Bertolini
- Department of Oncology and Hematology, Division of Oncology, University Hospital Modena, Modena, Italy
| | - De Bari Berardino
- Radiation Oncology, Centre Hospitalier Universitaire de Besançon, 25000, Besançon cedex, France
- Radiation Oncology, Réseau Hospitalier Neuchâtelois, CH-2300, La Chaux-de-Fonds, Switzerland
| | - Zampino Maria Giulia
- Division of Radiation Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Vegge Stefano
- Radiation Oncology Department, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Kalliopi Andrikou
- Oncologic Department, Istituto Scientifico Romagnolo per lo Studio e la Cura Dei Tumori, IRCCS, Meldola (Forlì), Italy
| | - Francesca Arcadipane
- Department of Oncology - Radiation Oncology, University of Turin School of Medicine, Via Genova 3, 10126, Turin, Italy
| | - Martina Napolitano
- Department of Oncology and Hematology, Division of Oncology, University Hospital Modena, Modena, Italy
| | - Lavajo Vieira Buno
- Radiation Oncology, Centre Hospitalier Universitaire de Besançon, 25000, Besançon cedex, France
| | | | - Francesco Olivero
- Department of Oncology - Radiation Oncology, University of Turin School of Medicine, Via Genova 3, 10126, Turin, Italy
| | - Filippo Ferreri
- Department of Oncology - Radiation Oncology, University of Turin School of Medicine, Via Genova 3, 10126, Turin, Italy
| | - Umberto Ricardi
- Department of Oncology - Radiation Oncology, University of Turin School of Medicine, Via Genova 3, 10126, Turin, Italy
| | - Stefano Cascinu
- Oncologic Department, IRCCS San Raffaele Scientific Institute Hospital, 20019, Milan, Italy
| | - Andrea Casadei-Gardini
- Oncologic Department, IRCCS San Raffaele Scientific Institute Hospital, 20019, Milan, Italy
| |
Collapse
|