1
|
Zhou J, Fang S, Zhu J, Su Y, Zhou H. Multiple Organ Dysfunction Syndrome Associated With Pneumonia: A Retrospective Study With a Focus on Respiratory Failure. Br J Hosp Med (Lond) 2025; 86:1-13. [PMID: 40405862 DOI: 10.12968/hmed.2024.0851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Aims/Background Pneumonia is a complex condition with rapid progression, often leading to multiple organ dysfunction syndrome (MODS), which significantly impacts patient quality of life and prognosis. This study aims to identify risk factors closely associated with the development of MODS by conducting a retrospective analysis of clinical data from 150 patients with pneumonia, providing insights for informing the prevention and treatment of pneumonia. Methods We recruited 150 patients with pneumonia admitted to Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University between 2019 and 2022. The patients were divided into two groups according to whether they had developed MODS: the non-MODS group (n = 100) and the MODS group (n = 50). Patients' data, such as their demographic information, clinical characteristics, laboratory test results, treatment course and outcomes, were collected. Logistic regression analysis was conducted to determine the independent predictors of MODS occurrence. Results After propensity score matching, the baseline data of the two groups of patients presented no significant difference (p > 0.05). The predominant clinical and laboratory manifestations of both groups of patients were fever, cough, decreased arterial partial pressure of oxygen (PaO2), as well as elevated white blood cell (WBC) count, C-reactive protein (CRP) level, and procalcitonin (PCT) level, but inter-group differences in these metrics were not statistically significant (p > 0.05). However, the clinical manifestation of dyspnea was significantly more prevalent in the MODS group (p < 0.01). The MODS group also had a significantly higher likelihood of receiving mechanical ventilation and renal replacement therapy (p < 0.001), as well as experiencing respiratory failure, cardiac failure, and death (p < 0.01). Univariate analysis indicated that factors including age ≥65 years, WBC count ≥15 × 109/L, PCT ≥2 ng/mL, PaO2 ≤60 mmHg, arterial partial pressure of carbon dioxide (PaCO2) ≥50 mmHg, severe pneumonia, and dyspnea were associated with a significantly increased risk of MODS (p < 0.05). These factors, except for the PCT, were validated as independent risk factors for MODS in the multivariate logistic regression analysis (p < 0.05). Conclusion Age ≥65 years, WBC count ≥15 × 109/L, PaO2 ≤60 mmHg, PaCO2 ≥50 mmHg, severe pneumonia, and dyspnea are independent risk factors for developing MODS in patients with pneumonia. Patients with MODS presenting respiratory failure as the main manifestation have a high mortality rate, and timely mechanical ventilation and supportive treatment are essential to improve the prognosis.
Collapse
Affiliation(s)
- Jie Zhou
- Intensive Care Unit, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Shunjin Fang
- Intensive Care Unit, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Jianfeng Zhu
- Intensive Care Unit, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Yao Su
- Intensive Care Unit, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Huifei Zhou
- Intensive Care Unit, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| |
Collapse
|
2
|
Ignacio-Mejía I, Bandala C, González-Zamora JF, Chavez-Galan L, Buendia-Roldan I, Pérez-Torres K, Rodríguez-Díaz MZ, Pacheco-Tobón DX, Quintero-Fabián S, Vargas-Hernández MA, Carrasco-Vargas H, Falfán-Valencia R, Pérez-Rubio G, Hernández-Lara KA, Gómez-Manzo S, Ortega-Cuellar D, Ignacio-Mejía F, Cárdenas-Rodríguez N. Association of Vitamin D Supplementation with Glutathione Peroxidase (GPx) Activity, Interleukine-6 (IL-6) Levels, and Anxiety and Depression Scores in Patients with Post-COVID-19 Condition. Int J Mol Sci 2025; 26:4582. [PMID: 40429727 PMCID: PMC12110956 DOI: 10.3390/ijms26104582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19) presents with various symptoms, and some patients develop post-COVID-19 condition (PCC). Vitamin D has shown therapeutic potential in COVID-19 and may offer benefits for PCC. The aim of this study was to evaluate the differences associated with two supplementation strategies (bolus and daily) on interleukin-6 (IL-6) levels, glutathione peroxidase (GPx) activity, and clinical outcomes in PCC patients, regardless of whether target 25 (OH) D levels reached the ideal range. We conducted a self-controlled study in which 54 participants with PCC were supplemented with vitamin D3 (n = 28 bolus and n = 26 daily) for 2 months. Blood samples were collected to measure IL-6 levels and GPx activity using spectrophotometric methods. The Hospital Anxiety and Depression Scale (HADS) was used to assess mental function. Both bolus and daily vitamin D supplementation were significantly associated with increased GPx activity and decreased IL-6 levels. Daily supplementation was additionally associated with a significant reduction in anxiety and depression scores. However, neither regimen was associated with improvements in cough, dyspnea, or fatigue. These findings suggest a potential association between vitamin D supplementation and improvements in antioxidant and neuropsychiatric parameters in PCC, possibly mediated by its immunomodulatory and antioxidant properties. Further placebo-controlled trials are warranted to determine whether these observed associations reflect causal relationships.
Collapse
Affiliation(s)
- Iván Ignacio-Mejía
- Laboratorio de Medicina Traslacional, Escuela Militar de Graduados de Sanidad, Universidad del Ejército y Fuerza Aérea, Mexico City 11200, Mexico; (I.I.-M.); (S.Q.-F.)
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Cindy Bandala
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | | | - Leslie Chavez-Galan
- Laboratorio de Inmunología Integrativa, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico;
| | - Ivette Buendia-Roldan
- Laboratorio de Investigación Traslacional en Envejecimiento y Enfermedades Fibróticas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.B.-R.); (K.P.-T.); (M.Z.R.-D.); (D.X.P.-T.)
| | - Karina Pérez-Torres
- Laboratorio de Investigación Traslacional en Envejecimiento y Enfermedades Fibróticas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.B.-R.); (K.P.-T.); (M.Z.R.-D.); (D.X.P.-T.)
| | - María Zobeida Rodríguez-Díaz
- Laboratorio de Investigación Traslacional en Envejecimiento y Enfermedades Fibróticas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.B.-R.); (K.P.-T.); (M.Z.R.-D.); (D.X.P.-T.)
| | - Denilson Xipe Pacheco-Tobón
- Laboratorio de Investigación Traslacional en Envejecimiento y Enfermedades Fibróticas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.B.-R.); (K.P.-T.); (M.Z.R.-D.); (D.X.P.-T.)
| | - Saray Quintero-Fabián
- Laboratorio de Medicina Traslacional, Escuela Militar de Graduados de Sanidad, Universidad del Ejército y Fuerza Aérea, Mexico City 11200, Mexico; (I.I.-M.); (S.Q.-F.)
| | - Marco Antonio Vargas-Hernández
- Subdirección de Investigación, Escuela Militar de Graduados de Sanidad, Universidad del Ejército y Fuerza Aérea, Mexico City 11200, Mexico;
| | - Humberto Carrasco-Vargas
- Dirección de la Escuela Militar de Medicina, Universidad del Ejército y Fuerza Aérea, Mexico City 11200, Mexico;
| | - Ramcés Falfán-Valencia
- Laboratorio de HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (R.F.-V.)
| | - Gloria Pérez-Rubio
- Laboratorio de HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (R.F.-V.)
| | - Kevin Alexis Hernández-Lara
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| | - Daniel Ortega-Cuellar
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| | | | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| |
Collapse
|
3
|
Muñoz-Carrillo JL, Palomeque-Molina PI, Villacis-Valencia MS, Gutiérrez-Coronado O, Chávez-Ruvalcaba F, Vázquez-Alcaraz SJ, Villalobos-Gutiérrez PT, Palomeque-Molina J. Relationship between periodontitis, type 2 diabetes mellitus and COVID-19 disease: a narrative review. Front Cell Infect Microbiol 2025; 15:1527217. [PMID: 40406515 PMCID: PMC12095153 DOI: 10.3389/fcimb.2025.1527217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
Inflammation plays a fundamental role in the development and bidirectional association of di-verse diseases, such as periodontitis and type 2 diabetes mellitus (T2DM), which generates important clinical complications, where chronic exposure to high levels of blood glucose affects the repair process of periodontal tissues. Likewise, it has been observed that comorbidity, between these two diseases, influences the development of the COVID-19 disease towards a more severe course. However, there is currently very little scientific evidence on the relationship between periodontitis, T2DM and COVID-19 disease. This narrative review aims to provide an understanding of the current and most relevant aspects of the relationship between periodontitis, T2DM and COVID-19 disease. A narrative review was performed through a systematic search of published studies, without date restrictions, indexed in the electronic databases of PubMed, for the inclusion of articles in English, and LILACS for the inclusion of articles in Spanish. This review included different articles, which addressed the most important aspects to present a current perspective on the relationship and influence between periodontitis, T2DM and COVID-19 disease. Comorbidity between periodontitis and T2DM represents a greater risk of developing a more severe course of COVID-19 disease, because these three diseases share three important axes: a clinicopathological axis; an axis associated with glycemia, and an immunological axis associated with inflammation.
Collapse
Affiliation(s)
- José Luis Muñoz-Carrillo
- Laboratorio de Inmunología, Centro Universitario de los Lagos, Universidad de Guadalajara, Lagos de Moreno, Jalisco, Mexico
- Escuela de Odontología, Global University, Aguascalientes, Aguascalientes, Mexico
| | | | | | - Oscar Gutiérrez-Coronado
- Laboratorio de Inmunología, Centro Universitario de los Lagos, Universidad de Guadalajara, Lagos de Moreno, Jalisco, Mexico
| | | | | | | | | |
Collapse
|
4
|
Carollo C, Benfante A, Sorce A, Montalbano K, Cirafici E, Calandra L, Geraci G, Mulè G, Scichilone N. Predictive Biomarkers of Acute Kidney Injury in COVID-19: Distinct Inflammatory Pathways in Patients with and Without Pre-Existing Chronic Kidney Disease. Life (Basel) 2025; 15:720. [PMID: 40430148 PMCID: PMC12113585 DOI: 10.3390/life15050720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Acute kidney injury (AKI) has emerged as a significant complication in patients with coronavirus disease 2019 (COVID-19). The pathophysiology of COVID-19-associated AKI is multifactorial, involving both direct viral effects on renal cells and indirect mechanisms such as systemic inflammation and cytokine storms. This highlights the critical need for early detection and effective management strategies to mitigate kidney injury and improve patient outcomes. The aim of our study is to assess the potential predictive role of inflammatory biomarkers in determining the risk of developing COVID-19-associated AKI in patients with and without pre-existing CKD. METHODS This study included 84 patients stratified by pre-existing chronic kidney disease (CKD) status. Demographic, clinical, and laboratory data were collected, including vital signs, hematological profiles, renal function markers, inflammatory biomarkers, coagulation parameters, and treatments. Outcomes such as acute kidney injury (AKI) and in-hospital mortality were documented. RESULTS In patients with pre-existing CKD, IL-6 and NLR demonstrated high predictive accuracy for AKI onset. In patients without pre-existing CKD, white blood cell (WBC) count emerged as a significant predictor of AKI onset. CONCLUSIONS The differential roles of IL-6, NLR, and WBC in predicting AKI onset highlight distinct physiopathological pathways influenced by COVID-19. In CKD+ patients, chronic inflammation and immune dysregulation are key drivers of AKI, with IL-6 and NLR serving as robust markers of this inflammatory state. In contrast, in CKD- patients, AKI may be more influenced by acute inflammatory responses and infectious factors, as reflected by WBC count.
Collapse
Affiliation(s)
- Caterina Carollo
- Unit of Nephrology and Dialysis, Hypertension Excellence Centre, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy; (A.S.); (E.C.); (L.C.); (G.M.)
| | - Alida Benfante
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy; (A.B.); (N.S.)
| | - Alessandra Sorce
- Unit of Nephrology and Dialysis, Hypertension Excellence Centre, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy; (A.S.); (E.C.); (L.C.); (G.M.)
| | - Katia Montalbano
- Nephrology and Dialysis Unit, Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, 90146 Palermo, Italy
| | - Emanuele Cirafici
- Unit of Nephrology and Dialysis, Hypertension Excellence Centre, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy; (A.S.); (E.C.); (L.C.); (G.M.)
| | - Leonardo Calandra
- Unit of Nephrology and Dialysis, Hypertension Excellence Centre, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy; (A.S.); (E.C.); (L.C.); (G.M.)
| | - Giulio Geraci
- Department of Medicine and Surgery, “Kore” University of Enna, 94100 Enna, Italy;
| | - Giuseppe Mulè
- Unit of Nephrology and Dialysis, Hypertension Excellence Centre, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy; (A.S.); (E.C.); (L.C.); (G.M.)
| | - Nicola Scichilone
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy; (A.B.); (N.S.)
| |
Collapse
|
5
|
Takeuchi Y, Ishikawa E, Sato T, Shinkai M, Takahashi Y, Lu X, Yamasaki S. Analysis of antigen specificity of Treg clonotypes expanded upon SARS-CoV-2 infection. Int Immunol 2025; 37:287-293. [PMID: 39665863 DOI: 10.1093/intimm/dxae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/11/2024] [Indexed: 12/13/2024] Open
Abstract
The pandemic outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has threatened human health worldwide. Among protective immune reactions, T cell responses are diverse among individuals, which is related to the differences in severity. A T cell subset, regulatory T (Treg) cells, is crucial for limiting excessive immune responses. If SARS-CoV-2-specific Tregs are developed during infection, they may counteract antiviral immunity and cause severe symptoms. To address this possibility, we conducted single-cell TCR-RNA-sequencing of peripheral blood mononuclear cells from convalescent Coronavirus disease 2019 (COVID-19) patients. Among 13 donors, one with severe symptoms had substantially more FOXP3-expressing Treg clonotypes activated in the presence of SARS-CoV-2 virion or other major antigen proteins. To define the reactivity of these Treg clonotypes, 15 highly expanded Treg clonotypes were reconstituted into reporter cells and stimulated with 27 distinct peptide pools that cover all SARS-CoV-2 proteins. However, none of these clonotypes react to any SARS-CoV-2 antigens. Instead, the reporter cells expressing one TCR clonotype (23599) were activated in the presence of Epstein-Barr virus-transformed B cells without adding exogenous antigens. Furthermore, 23599 TCR-expressing cells were activated by non-transformed naïve syngeneic B cells in a DQA1*03:03-DQB1*04:01-dependent manner, suggesting that clonotype 23599 may be autoreactive. This Treg clonotype, 23599, was also detected in a public TCR database and significantly expanded in COVID-19 patients compared to healthy donors. These results suggest that SARS-CoV-2 is not the dominant antigen inducing Treg cells during infection.
Collapse
Affiliation(s)
- Yukiko Takeuchi
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Eri Ishikawa
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Takashi Sato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, 390-8621, Japan
- Tokyo Shinagawa Hospital, Tokyo, 140-8522, Japan
| | | | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Xiuyuan Lu
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Sho Yamasaki
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
6
|
Chen X, Lin X, Cheng F, Zheng S, Zhang Q, Wu T, Jiang X, Shi J. Effectiveness and Safety of Simnotrelvir/Ritonavir and Nirmatrelvir/Ritonavir in the Treatment of Moderate to Severe COVID-19. Immun Inflamm Dis 2025; 13:e70174. [PMID: 40226977 PMCID: PMC11995419 DOI: 10.1002/iid3.70174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 01/18/2025] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND AND AIM Simnotrelvir/ritonavir and nirmatrelvir/ritonavir are major treatments for COVID-19, but their comparative efficacy and safety, especially in patients with moderate to severe COVID-19, remain unclear. METHODS This was a retrospective cohort study using electronic medical record data. From May 30, 2023, to October 8, 2023, 115 patients with moderate to severe COVID-19 were retrospectively collected from Wenzhou Central Hospital. They were treated with simnotrelvir/ritonavir or nirmatrelvir/ritonavir. The clinical effectiveness and adverse reactions were analyzed and compared between the two groups. RESULTS A total of 115 hospitalized patients were included in the study. They were 65 (56.5%) men and 50 (43.5%) women, with a mean age of 61 years. 58 (50.4%) were treated with simnotrelvir/ritonavir and 57 (49.6%) with nirmatrelvir/ritonavir. There was a similar rate of composite disease progression (10.3% vs. 7.0%, χ2 = 0.401, p = 0.527) and mortality (5.2% vs. 3.5%, χ2 = 0.191, p = 0.662) between the two groups. The progression rate from moderate COVID-19 to severe COVID-19 was not significantly different between the two groups (4.5% vs. 6.4%, χ2 = 0.148, p = 0.701). Median time for hospitalization was 7.0 (6.0, 8.0) days and 9.0 (8.0, 10.0) days (p = 0.338), and time for SARS-CoV-2 negative conversion was 6.0 (6.0, 7.0) days and 7.0 (6.0, 7.0) days (p = 0.934) in the simnotrelvir/ritonavir group and nirmatrelvir/ritonavir group, respectively. Among moderate patients, time for hospitalization was shorter in the simnotrelvir/ritonavir group [6.0 (6.0, 7.0) vs. 8.0 (8.0, 10.0) days, log-rank p = 0.004, HR = 1.838 (95% CI 1.199-2.815)]. And 5 (8.6%) had adverse drug reactions (ADRs) in the simnotrelvir/ritonavir group and 6 (10.5%) had ADRs in the nirmatrelvir/ritonavir group. CONCLUSION This is the first study comparing the effectiveness of simnotrelvir/ritonavir and nirmatrelvir/ritonavir in moderate and severe COVID-19 patients. Patients who received simnotrelvir/ritonavir exhibited shorter hospitalization. Disease progression, viral clearance times, and symptom resolution time were similar between the two groups.
Collapse
Affiliation(s)
- Xin Chen
- Department of Infectious DiseaseWenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Wenzhou Sixth People's HospitalZhejiangChina
| | - Xiao‐Qing Lin
- Department of Infectious DiseaseWenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Wenzhou Sixth People's HospitalZhejiangChina
| | - Fang Cheng
- Department of Infectious DiseaseWenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Wenzhou Sixth People's HospitalZhejiangChina
| | - Shi‐Lin Zheng
- Department of Infectious DiseaseWenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Wenzhou Sixth People's HospitalZhejiangChina
| | - Qiang Zhang
- Department of Infectious DiseaseWenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Wenzhou Sixth People's HospitalZhejiangChina
| | - Te Wu
- Department of Infectious DiseaseWenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Wenzhou Sixth People's HospitalZhejiangChina
| | - Xian‐Gao Jiang
- Department of Infectious DiseaseWenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Wenzhou Sixth People's HospitalZhejiangChina
| | - Ji‐Chan Shi
- Department of Infectious DiseaseWenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Wenzhou Sixth People's HospitalZhejiangChina
| |
Collapse
|
7
|
Lin MW, Lin CH, Chang JR, Chiang HH, Wu TH, Lin CS. The influence of PM2.5 exposure on SARS-CoV-2 infection via modulating the expression of angiotensin converting enzyme II. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136887. [PMID: 39700942 DOI: 10.1016/j.jhazmat.2024.136887] [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: 10/08/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Particulate matter 2.5 (PM2.5) pollution and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic are the greatest environmental health issues worldwide. Several statistics revealed the significant positive correlation between the morbidity of coronavirus disease-19 (COVID-19) and the levels of air pollution. Nevertheless, there is no direct experimental evidence to indicate the effect of PM2.5 exposure on SARS-CoV-2 infection. The objective of this study was to evaluate whether the infection of SARS-CoV-2 affected by PM2.5 through angiotensin-converting enzyme II (ACE2) expression enhances and investigate the function of ACE2 in lung injury induced by PM2.5. An animal model of PM2.5-induced lung injury was established using wild-type (WT, C57BL/6), human ACE2 transgenic (K18-hACE2 TG), and murine ACE2 gene knockout (mACE2 KO) mice. The results indicate that PM2.5 exposure facilitates SARS-CoV-2 infection through inducing ACE2 expression in vitro (10 μg/mL) and in vivo (6.25 mg/kg/day in 50 μL saline). The levels of ACE, inflammatory cytokines, and mitogen-activated protein kinase (MAPK) proteins in WT, K18-hACE TG and mACE2 KO mice were significantly increased after PM2.5 instillation. The severest PM2.5-induced lung damage was observed in mACE2 KO mice. In summary, ACE2 plays a double-edged sword role in lung injury, PM2.5 exposure contributed to SARS-CoV-2 infection through inducing ACE2 expression, but ACE2 also protected pulmonary inflammation from PM2.5 challenge.
Collapse
Affiliation(s)
- Meng-Wei Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Cheng-Han Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Jia-Rong Chang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Hua-Hsin Chiang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Ting-Hsuan Wu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Chih-Sheng Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| |
Collapse
|
8
|
Adilović M, Hromić-Jahjefendić A, Mahmutović L, Šutković J, Rubio-Casillas A, Redwan EM, Uversky VN. Intrinsic Factors Behind the Long-COVID: V. Immunometabolic Disorders. J Cell Biochem 2025; 126:e30683. [PMID: 39639607 DOI: 10.1002/jcb.30683] [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: 07/07/2024] [Revised: 11/02/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
The complex link between COVID-19 and immunometabolic diseases demonstrates the important interaction between metabolic dysfunction and immunological response during viral infections. Severe COVID-19, defined by a hyperinflammatory state, is greatly impacted by underlying chronic illnesses aggravating the cytokine storm caused by increased levels of Pro-inflammatory cytokines. Metabolic reprogramming, including increased glycolysis and altered mitochondrial function, promotes viral replication and stimulates inflammatory cytokine production, contributing to illness severity. Mitochondrial metabolism abnormalities, strongly linked to various systemic illnesses, worsen metabolic dysfunction during and after the pandemic, increasing cardiovascular consequences. Long COVID-19, defined by chronic inflammation and immune dysregulation, poses continuous problems, highlighting the need for comprehensive therapy solutions that address both immunological and metabolic aspects. Understanding these relationships shows promise for effectively managing COVID-19 and its long-term repercussions, which is the focus of this review paper.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Lejla Mahmutović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Jasmin Šutković
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan, Mexico
- Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Mexico
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
9
|
Pawlik MT, Rinneberg G, Koch A, Meyringer H, Loew TH, Kjellberg A. Is there a rationale for hyperbaric oxygen therapy in the patients with Post COVID syndrome? : A critical review. Eur Arch Psychiatry Clin Neurosci 2024; 274:1797-1817. [PMID: 39545965 PMCID: PMC11579208 DOI: 10.1007/s00406-024-01911-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 09/16/2024] [Indexed: 11/17/2024]
Abstract
The SARS-CoV-2 pandemic has resulted in 762 million infections worldwide from 2020 to date, of which approximately ten percent are suffering from the effects after infection in 2019 (COVID-19) [1, 40]. In Germany, it is now assumed that at least one million people suffer from post-COVID condition with long-term consequences. These have been previously reported in diseases like Myalgic Encephalomyelitis (ME) and Chronic Fatigue Syndrome (CFS). Symptoms show a changing variability and recent surveys in the COVID context indicate that 10-30 % of outpatients, 50 to 70% of hospitalised patients suffer from sequelae. Recent data suggest that only 13% of all ill people were completely free of symptoms after recovery [3, 9]. Current hypotheses consider chronic inflammation, mitochondrial dysfunction, latent viral persistence, autoimmunity, changes of the human microbiome or multilocular sequelae in various organ system after infection. Hyperbaric oxygen therapy (HBOT) is applied since 1957 for heart surgery, scuba dive accidents, CO intoxication, air embolisms and infections with anaerobic pathogens. Under hyperbaric pressure, oxygen is physically dissolved in the blood in higher concentrations and reaches levels four times higher than under normobaric oxygen application. Moreover, the alternation of hyperoxia and normoxia induces a variety of processes at the cellular level, which improves oxygen supply in areas of locoregional hypoxia. Numerous target gene effects on new vessel formation, anti-inflammatory and anti-oedematous effects have been demonstrated [74]. The provision of intermittently high, local oxygen concentrations increases repair and regeneration processes and normalises the predominance of hyperinflammation. At present time only one prospective, randomized and placebo-controlled study exists with positive effects on global cognitive function, attention and executive function, psychiatric symptoms and pain interference. In conclusion, up to this date HBO is the only scientifically proven treatment in a prospective randomized controlled trial to be effective for cognitive improvement, regeneration of brain network and improvement of cardiac function. HBOT may have not only theoretical but also potential impact on targets of current pathophysiology of Post COVID condition, which warrants further scientific studies in patients.
Collapse
Affiliation(s)
- M T Pawlik
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany.
- Institute of Experimental Medicine, Christian-Albrechts-University of Kiel c/o German Naval Medical Institute, Kronshagen, Germany.
| | - G Rinneberg
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany
| | - A Koch
- Institute of Experimental Medicine, Christian-Albrechts-University of Kiel c/o German Naval Medical Institute, Kronshagen, Germany
| | - H Meyringer
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany
| | - T H Loew
- Department of Psychosomatic Medicine, University Hospital Regensburg, Regensburg, Germany
| | - A Kjellberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
- Perioperative Medicine and Intensive Care, Medical Unit Intensive Care and Thoracic surgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
10
|
Wang L, Zhang A, Hu Y, Yang W, Zhong L, Shi J, Wang Z, Tao Q, Liang Q, Yao X. Landscape of multiple tissues' gene expression pattern associated with severe sepsis: Genetic insights from Mendelian randomization and trans-omics analysis. Life Sci 2024; 358:123181. [PMID: 39471899 DOI: 10.1016/j.lfs.2024.123181] [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: 06/25/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024]
Abstract
BACKGROUND Sepsis, a systemic syndrome often culminating in multiple organ failure (MOF), poses a substantial global health threat. However, the gene expression pattern of various tissues associated with severe sepsis remains elusive. METHODS Applying the summary data-based Mendelian randomization (SMR) method, we integrated sepsis genome-wide association study (GWAS) data and expression quantitative trait loci (eQTLs) summaries. This facilitated the investigation of gene causality across 12 tissue types within 26 cohorts linked to adverse sepsis outcomes, including critical care and 28-day mortality. Additionally, trans-omics analyses, including blood transcriptome and single-cell RNA sequencing, were conducted to examine cellular origins and gene functions. The effects of ST7L on sepsis were validated in vivo and in vitro. RESULTS We identified 127 genes associated with severe sepsis across diverse tissues. Cross-tissue analysis highlighted ST7L as a significant pan-tissue risk factor for severe sepsis, displaying significance across 11 tissues for both critical care sepsis (meta OR 1.19, 95 % CI: 1.14-1.25, meta p < 0.0001) and 28-day-death sepsis (meta OR: 1.22, 95 % CI: 1.17-1.27, meta p < 0.0001). Notably, independent blood single-cell RNA sequencing data showed specific expression of ST7L in dendritic cells (DCs). ST7L+ DCs were elevated in non-surviving sepsis patients and exhibited an augmented inflammatory molecular pattern compared to ST7L- DCs. Both transcription and translation level of ST7L in DCs exhibited a dose-dependent pattern with LPS. Knocking down ST7L by siRNA was sufficient to alleviate the inflammation phenotype of DCs, including inhibiting p65/NF-kB pathway and inflammatory factors. CONCLUSION Our findings underscore ST7L as a pan-tissue risk factor for severe sepsis, specifically manifested in DCs and associated with an inflammatory phenotype. These results offer essential insights into the gene expression profiles across multiple tissues in severe sepsis, potentially identifying therapeutic targets for effective sepsis management.
Collapse
Affiliation(s)
- Lei Wang
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Aiping Zhang
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China; Critical Care Medicine Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Yehong Hu
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China; Critical Care Medicine Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Wanwei Yang
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Li Zhong
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Jianfeng Shi
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Zhiguo Wang
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Qing Tao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, Jiangsu, China
| | - Qiao Liang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, Jiangsu, China.
| | - Xiaoming Yao
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| |
Collapse
|
11
|
Guarienti FA, Gonçalves JIB, Gonçalves JB, Antônio Costa Xavier F, Marinowic D, Machado DC. COVID-19: a multi-organ perspective. Front Cell Infect Microbiol 2024; 14:1425547. [PMID: 39492990 PMCID: PMC11527788 DOI: 10.3389/fcimb.2024.1425547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/18/2024] [Indexed: 11/05/2024] Open
Abstract
In this mini review, we explore the complex network of inflammatory reactions incited by SARS-CoV-2 infection, which extends its reach well beyond the respiratory domain to influence various organ systems. Synthesizing existing literature, it elucidates how the hyperinflammation observed in COVID-19 patients affects multiple organ systems leading to physiological impairments that can persist over long after the resolution of infection. By exploring the systemic manifestations of this inflammatory cascade, from acute respiratory distress syndrome (ARDS) to renal impairment and neurological sequelae, the review highlights the profound interplay between inflammation and organ dysfunction. By synthesizing recent research and clinical observations, this mini review aims to provide an overview of the systemic interactions and complications associated with COVID-19, underscoring the need for an integrated approach to treatment and management. Understanding these systemic effects is crucial for improving patient outcomes and preparing for future public health challenges.
Collapse
Affiliation(s)
- Fabiana Amaral Guarienti
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - João Ismael Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Júlia Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Fernando Antônio Costa Xavier
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Daniel Marinowic
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Denise Cantarelli Machado
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| |
Collapse
|
12
|
Guo ZF, Tongmuang N, Li C, Zhang C, Hu L, Capreri D, Zuo MX, Summer R, Sun J. Inhibiting endothelial cell Mst1 attenuates acute lung injury in mice. JCI Insight 2024; 9:e178208. [PMID: 39253972 PMCID: PMC11385092 DOI: 10.1172/jci.insight.178208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 07/25/2024] [Indexed: 09/11/2024] Open
Abstract
Lung endothelium plays a pivotal role in the orchestration of inflammatory responses to acute pulmonary insults. Mammalian sterile 20-like kinase 1 (Mst1) is a serine/threonine kinase that has been shown to play an important role in the regulation of apoptosis, stress responses, and organ growth. This study investigated the role of Mst1 in lung endothelial activation and acute lung injury (ALI). We found that Mst1 was significantly activated in inflamed lung endothelial cells (ECs) and mouse lung tissues. Overexpression of Mst1 promoted nuclear factor κ-B (NF-κB) activation through promoting JNK and p38 activation in lung ECs. Inhibition of Mst1 by either its dominant negative form (DN-Mst1) or its pharmacological inhibitor markedly attenuated cytokine-induced expression of cytokines, chemokines, and adhesion molecules in lung ECs. Importantly, in a mouse model of lipopolysaccharide-induced (LPS-induced) ALI, both deletion of Mst1 in lung endothelium and treatment of WT mice with a pharmacological Mst1 inhibitor significantly protected mice from LPS-induced ALI. Together, our findings identified Mst1 kinase as a key regulator in controlling lung EC activation and suggest that therapeutic strategies aimed at inhibiting Mst1 activation might be effective in the prevention and treatment of inflammatory lung diseases.
Collapse
Affiliation(s)
- Zhi-Fu Guo
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai
| | - Nopprarat Tongmuang
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Chao Li
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Chen Zhang
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Louis Hu
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Daniel Capreri
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Mei-Xing Zuo
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ross Summer
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jianxin Sun
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Iwasaki M, Yamamoto M, Tomihari M, Ishikawa M. Ropivacaine Administration Suppressed A549 Lung Adenocarcinoma Cell Proliferation and Migration via ACE2 Upregulation and Inhibition of the Wnt1 Pathway. Int J Mol Sci 2024; 25:9334. [PMID: 39273283 PMCID: PMC11395614 DOI: 10.3390/ijms25179334] [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: 07/13/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Previous studies have suggested that perioperative anesthesia could have direct impacts on cancer cell biology. The present study investigated the effects of ropivacaine administration on lung adenocarcinoma cells. METHODS Ropivacaine was administered to A549 cells at concentrations of 0.1, 1, and 6 mM for 2 h. Angiotensin-converting enzyme 2 (ACE2) small interfering RNA (siRNA) transfection was performed 6 h prior to ropivacaine administration. Cell proliferation and migration were assessed with cell counting kit 8 (CCK-8) and a wound healing assay at 0 and 24 h after anesthesia exposure. PCR arrays were performed, followed by PCR validation. RESULTS Ropivacaine administration inhibited A549 cell proliferation and migration in a concentration-dependent manner, with ACE2 upregulation and HIF1α (hypoxia-inducible factor 1α) downregulation. The anticancer effect of ropivacaine was canceled out via ACE2 siRNA transfection. PCR arrays showed specific gene change patterns in the ropivacaine and respective ACE2-knockdown groups. EGFR (epidermal growth factor receptor), BAX (Bcl-2-associated X protein) and BCL2 (B-cell/CLL lymphoma 2) were suppressed with ropivacaine administration; these effects were reversed via ACE2 siRNA induction. CONCLUSION Ropivacaine administration inhibited A549 cell biology in conjunction with ACE2 upregulation via the inhibition of the Wnt1 (wingless/Integrated 1) pathway.
Collapse
Affiliation(s)
- Masae Iwasaki
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan; (M.Y.); (M.T.); (M.I.)
| | | | | | | |
Collapse
|
14
|
Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function. Int J Mol Sci 2024; 25:7553. [PMID: 39062796 PMCID: PMC11277036 DOI: 10.3390/ijms25147553] [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: 05/27/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Proteases are produced and released in the mucosal cells of the respiratory tract and have important physiological functions, for example, maintaining airway humidification to allow proper gas exchange. The infectious mechanism of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), takes advantage of host proteases in two ways: to change the spatial conformation of the spike (S) protein via endoproteolysis (e.g., transmembrane serine protease type 2 (TMPRSS2)) and as a target to anchor to epithelial cells (e.g., angiotensin-converting enzyme 2 (ACE2)). This infectious process leads to an imbalance in the mucosa between the release and action of proteases versus regulation by anti-proteases, which contributes to the exacerbation of the inflammatory and prothrombotic response in COVID-19. In this article, we describe the most important proteases that are affected in COVID-19, and how their overactivation affects the three main physiological systems in which they participate: the complement system and the kinin-kallikrein system (KKS), which both form part of the contact system of innate immunity, and the renin-angiotensin-aldosterone system (RAAS). We aim to elucidate the pathophysiological bases of COVID-19 in the context of the imbalance between the action of proteases and anti-proteases to understand the mechanism of aprotinin action (a panprotease inhibitor). In a second-part review, titled "Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions", we explain in depth the pharmacodynamics, pharmacokinetics, toxicity, and use of aprotinin as an antiviral drug.
Collapse
Affiliation(s)
- Juan Fernando Padín
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
| | - José Manuel Pérez-Ortiz
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco Javier Redondo-Calvo
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
- Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
- Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
| |
Collapse
|
15
|
Borowczak J, Gąsiorek-Kwiatkowska A, Szczerbowski K, Maniewski M, Zdrenka M, Szadurska-Noga M, Gostomczyk K, Rutkiewicz P, Olejnik K, Cnota W, Karpów-Greiner M, Knypiński W, Sekielska-Domanowska M, Ludwikowski G, Dubiel M, Szylberg Ł, Bodnar M. SARS-CoV-2 Infection during Delivery Causes Histopathological Changes in the Placenta. Diseases 2024; 12:142. [PMID: 39057113 PMCID: PMC11276080 DOI: 10.3390/diseases12070142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND SARS-CoV-2 can damage human placentas, leading to pregnancy complications, such as preeclampsia and premature birth. This study investigates the histopathological changes found in COVID-19-affected placentas. MATERIALS AND METHODS This study included 23 placentas from patients with active COVID-19 during delivery and 22 samples from patients without COVID-19 infection in their medical history. The samples underwent histopathological examination for pathology, such as trophoblast necrosis, signs of vessel damage, or fetal vascular malperfusion. RESULTS Newborns from the research group have lower weights and Apgar scores than healthy newborns. In the COVID-19 group, calcifications and collapsed intervillous space were more frequent, and inflammation was more severe than in the healthy group. At the same time, the placenta of SARS-CoV-2-positive patients showed signs of accelerated vascular maturation. Trophoblast necrosis was found only in the placentas of the research group. The expression of CD68+ was elevated in the COVID-19 cohort, suggesting that macrophages constituted a significant part of the inflammatory infiltrate. The increase in lymphocyte B markers was associated with placental infarctions, while high levels of CD3+, specific for cytotoxic T lymphocytes, correlated with vascular injury. CONCLUSIONS SARS-CoV-2 is associated with pathological changes in the placenta, including trophoblast necrosis, calcification, and accelerated villous maturation. Those changes appear to be driven by T cells and macrophages, whose increased expression reflects ongoing histiocytic intervillositis in the placenta.
Collapse
Affiliation(s)
- Jędrzej Borowczak
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
| | - Agnieszka Gąsiorek-Kwiatkowska
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Krzysztof Szczerbowski
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
| | - Mateusz Maniewski
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
- Doctoral School of Medical and Health Sciences, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland
| | - Marek Zdrenka
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
| | - Marta Szadurska-Noga
- Department of Pathomorphology and Forensic Medicine, Faculty of Medical Sciences, University of Warmia and Mazury, 10-561 Olsztyn, Poland;
| | - Karol Gostomczyk
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Paula Rutkiewicz
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| | - Katarzyna Olejnik
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| | - Wojciech Cnota
- Chair and Department of Gynaecology and Obstetrics, Faculty of Health Sciences in Katowice, Medical University of Silesia, 41-703 Ruda Śląska, Poland
| | - Magdalena Karpów-Greiner
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Wojciech Knypiński
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Marta Sekielska-Domanowska
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Grzegorz Ludwikowski
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Mariusz Dubiel
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Łukasz Szylberg
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| | - Magdalena Bodnar
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| |
Collapse
|
16
|
Cannarella R, Marino M, Crafa A, Bagnara V, La Vignera S, Condorelli RA, Calogero AE. Impact of COVID-19 on testicular function: a systematic review and meta-analysis. Endocrine 2024; 85:44-66. [PMID: 38345682 PMCID: PMC11246276 DOI: 10.1007/s12020-024-03705-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 01/17/2024] [Indexed: 07/14/2024]
Abstract
INTRODUCTION Studies investigating the effects of SARS-CoV-2 on male reproductive function are few and heterogeneous, and results are often conflicting. This systematic review and meta-analysis was carried out on studies conducted in men with active or anamnestic SARS-CoV-2 infection to evaluate its consequences on the male sex hormone profile and semen parameters. MATERIALS AND METHOD This meta-analysis follows the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) protocols. PubMed, Scopus, Cochrane, and Embase databases were searched to identify relevant studies. We originally selected 3553 articles. After the eligibility phase, 16 articles met our inclusion criteria encompassing 11 case-control studies and 5 cohort studies (2 prospective and 3 retrospective studies). We performed the quantitative analysis with Comprehensive Meta-Analysis Software. Cochran-Q and heterogeneity (I2) indexes were used to assess statistical heterogeneity. Sensitivity analysis and publication bias tests were also performed. RESULTS Overall, 1250 patients with active or recent (up to 80 days before) COVID-19 infection and 1232 matched healthy controls were included. Sperm concentration, total sperm count, and total motility were significantly lower in patients compared with controls. Patients also showed lower levels of total testosterone and follicle-stimulating hormone, and higher levels of luteinizing hormone, 17β-estradiol, and prolactin compared with healthy controls. None of the included studies found the presence of SARS-CoV-2 mRNA in the semen of infected patients. CONCLUSION The present systematic review and meta-analysis suggests the presence of an association between SARS-CoV-2 infection and primary testicular damage manifested with a picture of altered steroidogenesis and worsening spermatogenesis. The absence of the virus in the seminal fluid indicates a low possibility of sexual transmission of the infection to partners and offspring. However, our findings mostly show short-term follow-up, while few studies have considered the long-term consequences of the viral infection, thus further studies are needed to evaluate the long-term consequences on male reproductive health.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Marta Marino
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Vincenzo Bagnara
- Pediatric Surgery Unit, Policlinic G.B. Morgagni, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
17
|
Shen J, Li J, Lei Y, Chen Z, Wu L, Lin C. Frontiers and hotspots evolution in cytokine storm: A bibliometric analysis from 2004 to 2022. Heliyon 2024; 10:e30955. [PMID: 38774317 PMCID: PMC11107250 DOI: 10.1016/j.heliyon.2024.e30955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/24/2024] Open
Abstract
Background As a fatal disease, cytokine storm has garnered research attention in recent years. Nonetheless, as the body of related studies expands, a thorough and impartial evaluation of the current status of research on cytokine storms remains absent. Consequently, this study aimed to thoroughly explore the research landscape and evolution of cytokine storm utilizing bibliometric and knowledge graph approaches. Methods Research articles and reviews centered on cytokine storms were retrieved from the Web of Science Core Collection database. For bibliometric analysis, tools such as Excel 365, CiteSpace, VOSviewer, and the Bibliometrix R package were utilized. Results This bibliometric analysis encompassed 6647 articles published between 2004 and 2022. The quantity of pertinent articles and citation frequency exhibited a yearly upward trend, with a sharp increase starting in 2020. Network analysis of collaborations reveals that the United States holds a dominant position in this area, boasting the largest publication count and leading institutions. Frontiers in Immunology ranks as the leading journal for the largest publication count in this area. Stephan A. Grupp, a prominent researcher in this area, has authored the largest publication count and has the second-highest citation frequency. Research trends and keyword evaluations show that the connection between cytokine storm and COVID-19, as well as cytokine storm treatment, are hot topics in research. Furthermore, research on cytokine storm and COVID-19 sits at the forefront in this area. Conclusion This study employed bibliometric analysis to create a visual representation of cytokine storm research, revealing current trends and burgeoning topics in this area for the first time. It will provide valuable insights, helping scholars pinpoint critical research areas and potential collaborators.
Collapse
Affiliation(s)
- Junyi Shen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiaming Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yuqi Lei
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhengrui Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Lingling Wu
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Chunyan Lin
- Department of Teaching and Research Section of Internal Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
18
|
Fabrizi F, Nardelli L, Regalia A, Zanoni F, Castellano G. Are Kidneys Affected by SARS-CoV-2 Infection? An Updated Review on COVID-19-Associated AKI. Pathogens 2024; 13:325. [PMID: 38668280 PMCID: PMC11054118 DOI: 10.3390/pathogens13040325] [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: 03/11/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Human kidneys are an important target of SARS-CoV-2 infection, and many renal abnormalities have been found in patients with SARS-CoV-2 infection, including proteinuria, hematuria, and acute kidney injury. Acute kidney injury is now considered a common complication of COVID-19, and the epidemiology of AKI in SARS-CoV-2-infected patients continues to be controversial. AIM AND METHODS We have carried out a narrative review to evaluate the frequency and risk factors for AKI among patients hospitalized due to COVID-19, and the latest surveys on this topic have been included. The mechanisms by which AKI occurs in COVID-19 patients have also been reviewed. RESULTS Multiple risk factors for the development of AKI in patients with SARS-CoV-2 infection have been identified; these have been classified in various groups (management and background factors, among others). SARS-CoV-2 targets the kidneys by indirect activity, but SARS-CoV-2 infects tubular epithelial cells and podocytes. We retrieved 24 reports (n = 502,593 unique patients with SARS-CoV-2 infection) and found an incidence of AKI of 31.8% (range, 0.5% to 56.9%). Only a minority (n = 2) of studies had a prospective design. We found that the AKI risk was greater in SARS-CoV-2 patients who underwent in-hospital deaths vs. those who survived; the summary estimate of the unadjusted RR of AKI was 2.63 (95% CI, 2.37; 2.93) (random-effects model). A stratified analysis showed that the incidence of AKI was greater in those reports where the frequency of COVID-19-positive patients having comorbidities (diabetes mellitus, arterial hypertension, and advanced age) was high. The unadjusted relative risk (aRR) of AKI was greater in SARS-CoV-2 patients who underwent ICU admission vs. those who did not; the pooled estimate of AKI risk was 2.64 (95% CI, 1.96; 3.56) according to the random-effects model. CONCLUSIONS AKI is a common complication of hospitalized SARS-CoV-2-infected patients, and some comorbidities are important risk factors for it. The direct activity of the virus on the kidneys has been mentioned in the pathogenesis of AKI in SARS-CoV-2 patients. Further studies are ongoing in order to identify the mechanisms underlying the kidney injury in this population. The role of AKI on survival in SARS-CoV-2-infected patients is another area of active investigation.
Collapse
Affiliation(s)
- Fabrizio Fabrizi
- Division of Nephrology, Dialysis and Kidney Transplant, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.N.); (A.R.); (F.Z.); (G.C.)
| | - Luca Nardelli
- Division of Nephrology, Dialysis and Kidney Transplant, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.N.); (A.R.); (F.Z.); (G.C.)
- Department of Clinical Sciences and Community Health, University School of Medicine, 20122 Milan, Italy
| | - Anna Regalia
- Division of Nephrology, Dialysis and Kidney Transplant, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.N.); (A.R.); (F.Z.); (G.C.)
| | - Francesca Zanoni
- Division of Nephrology, Dialysis and Kidney Transplant, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.N.); (A.R.); (F.Z.); (G.C.)
| | - Giuseppe Castellano
- Division of Nephrology, Dialysis and Kidney Transplant, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.N.); (A.R.); (F.Z.); (G.C.)
- Department of Clinical Sciences and Community Health, University School of Medicine, 20122 Milan, Italy
| |
Collapse
|
19
|
Zou H, Zhang K, Chen X, Sha S. Vogt-Koyanagi-Harada disease after SARS-CoV-2 infection: Case report and literature review. Immun Inflamm Dis 2024; 12:e1250. [PMID: 38661242 PMCID: PMC11044218 DOI: 10.1002/iid3.1250] [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: 07/28/2023] [Revised: 03/20/2024] [Accepted: 03/31/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for coronavirus disease 2019 (COVID-19), a complex and multifaceted illness. COVID-19 is associated with various ocular manifestations including conjunctivitis, retinal vein occlusion and optic neuritis. However, the case of Vogt-Koyanagi-Harada (VKH) disease associated with SARS-CoV-2 is infrequent, and the specific association is still unclear. CASE PRESENTATION In the present study, a 35-year-old female patient without any significant medical history presented with 1 week of bilateral blurred vision, occurring 2 weeks after a clinical course of COVID-19. Upon examination, both eyes exhibited bullous serous retinal detachments. She was diagnosed with incomplete VKH disease. Early diagnosis and treatment of VKH disease are essential for the visual prognosis of this aggressive disease. In this particular patient, ocular inflammatory signs and visual acuity improved via corticosteroid therapy. It is worth noting that the occurrence of VKH disease associated with SARS-CoV-2 is uncommon, and the specific connection between the two remains unknown. We review and summarize the clinical characteristics of VKH disease following SARS-CoV-2 infection, and discuss the potential mechanisms that may explain this phenomenon, based on similar studies previously reported. CONCLUSION Despite the unclear causality, it is important for ophthalmologists and physicians to be recognizant of the possible association between VKH disease and COVID-19. SARS-CoV-2 may play a potential immunological triggering role in VKH disease. However, further in-depth research is necessary to investigate the clinical and epidemiological features, as well as the underlying mechanisms of this association.
Collapse
Affiliation(s)
- Hui Zou
- Department of OphthalmologyHunan Provincial People's Hospital (The First Hospital of Hunan Normal University)ChangshaHunanChina
| | - Ke Zhang
- Department of OphthalmologyHunan Provincial People's Hospital (The First Hospital of Hunan Normal University)ChangshaHunanChina
| | - Xuan Chen
- Department of OphthalmologyHunan Provincial People's Hospital (The First Hospital of Hunan Normal University)ChangshaHunanChina
| | - Sha Sha
- Department of OphthalmologyHunan Provincial People's Hospital (The First Hospital of Hunan Normal University)ChangshaHunanChina
| |
Collapse
|
20
|
Zheng G, Qiu G, Qian H, Shu Q, Xu J. Multifaceted role of SARS-CoV-2 structural proteins in lung injury. Front Immunol 2024; 15:1332440. [PMID: 38375473 PMCID: PMC10875085 DOI: 10.3389/fimmu.2024.1332440] [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: 11/02/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the third human coronavirus to cause acute respiratory distress syndrome (ARDS) and contains four structural proteins: spike, envelope, membrane, and nucleocapsid. An increasing number of studies have demonstrated that all four structural proteins of SARS-CoV-2 are capable of causing lung injury, even without the presence of intact virus. Therefore, the topic of SARS-CoV-2 structural protein-evoked lung injury warrants more attention. In the current article, we first synopsize the structural features of SARS-CoV-2 structural proteins. Second, we discuss the mechanisms for structural protein-induced inflammatory responses in vitro. Finally, we list the findings that indicate structural proteins themselves are toxic and sufficient to induce lung injury in vivo. Recognizing mechanisms of lung injury triggered by SARS-CoV-2 structural proteins may facilitate the development of targeted modalities in treating COVID-19.
Collapse
Affiliation(s)
| | - Guanguan Qiu
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Huifeng Qian
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Qiang Shu
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Jianguo Xu
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| |
Collapse
|
21
|
Obeng-Gyasi E. How will Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) and lead (Pb) exposure interact in the cardiovascular system? INTERNATIONAL PUBLIC HEALTH JOURNAL 2024; 16:3-10. [PMID: 39139795 PMCID: PMC11321597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
The combined effects of lead exposure and the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and how they cause cardiovascular dysfunction are examined in this viewpoint manuscript. Lead was picked as a representative environmental contaminant because it has been studied extensively for more than 40 years, while SARS-CoV-2 illustrates the unpredictability of a new infectious agent that can quickly increase the environmental burden on communities. Given that this interaction represents a plausible combined exposure for many people, it is crucial to discuss how it might happen at a mechanistic level. SARS-CoV-2 causes the coronavirus disease 2019 (COVID-19), which can spread by contact and respiratory droplets. One may be exposed to lead and SARS-CoV-2 simultaneously depending on their living situation. Uncertainty exists regarding the consequences of this exposure on cardiovascular disease and its underlying processes. This paper aims to investigate these mechanisms and provide potential causes behind them. The interaction of lead with SARS-CoV-2 and its effects on the cardiovascular system are likely caused by direct damage, the promotion of reactive oxygen species (ROS) production, oxidative stress, and inflammation. In particular, the stimulation of nuclear factor kappa B (NF-B), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF) were all identified as potentially important pathways. Blood clotting, cytokine storm cycle development, and increased hypoxia may also be the results of this. In summary, the effects on the immune system, coagulation, the renin-angiotensin-aldosterone system, and cardiac contractility were the four primary areas identified.
Collapse
Affiliation(s)
- Emmanuel Obeng-Gyasi
- Department of Built Environment and Environmental Health and Disease Laboratory, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, USA
| |
Collapse
|
22
|
Zhang Z, Wu S, Wang Z, Wang Y, Chen H, Wu C, Xiong L. Long-term oral ACEI/ARB therapy is associated with disease severity in elderly COVID-19 omicron BA.2 patients with hypertension. BMC Infect Dis 2023; 23:882. [PMID: 38110869 PMCID: PMC10726588 DOI: 10.1186/s12879-023-08913-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 12/14/2023] [Indexed: 12/20/2023] Open
Abstract
OBJECTIVE To explore the effects of long-term oral ACEIs/ARBs on the incidence of exacerbation and in-hospital mortality in elderly COVID-19 Omicron BA.2 patients with hypertension, especially patients aged 80 years or older. MATERIALS AND METHODS In this retrospective study, patients suffering mild and rcommon COVID-19 with hypertension who were hospitalized in the Shanghai Fourth People's Hospital between April 2022 and June 2022 were enrolled. Primary outcomes included the incidence of exacerbation and in-hospital mortality. Secondary outcomes included the incidence of respiratory failure of patients, use of mechanical ventilation, nucleic acid conversion time (NCT), hospitalization costs, and the temporal trend of the incidence of exacerbations and in-hospital mortality in different age groups. The data were analysed using propensity score weighting (PSW). RESULTS In the entire cohort, there were 298 ACEI/ARB users and 465 non-ACEI/ARB users. The ACEI/ARB group showed a lower incidence of exacerbation (OR = 0.64, 95% CI for OR: 0.46-0.89, P = 0.0082) and lower in-hospital mortality (OR = 0.49, 95% CI for OR: 0.27-0.89, P = 0.0201) after PSW. Sensitivity analysis obtained the same results. The results of the subgroup of patients aged 80 years and older obtained a similar conclusion as the whole cohort. Most of the study indicators did not differ statistically significantly in the subgroup of patients aged 60 to 79 years except for rates of mechanical ventilation and respiratory failure. CONCLUSION Antihypertensive therapy with ACEIs/ARBs might reduce the incidence of exacerbation and in-hospital mortality. The findings of this study support the use of ACEIs/ARBs in COVID-19 patients infected by Omicron BA.2, especially in patients aged 80 years or older with hypertension.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Anesthesiology and Perioperative medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Shengyong Wu
- Department of Military Health Statistics, Naval Medical University, Shanghai, 200433, China
| | - Zhiyong Wang
- Department of Information Management, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, 200434, China
| | - Yue Wang
- Department of Anesthesiology and Perioperative medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Hui Chen
- Department of Anesthesiology and Perioperative medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Cheng Wu
- Department of Military Health Statistics, Naval Medical University, Shanghai, 200433, China.
| | - Lize Xiong
- Department of Anesthesiology and Perioperative medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
| |
Collapse
|
23
|
Lu P, Leslie F, Wang H, Sodhi A, Choi CY, Pekosz A, Cui H, Jia H. Discovery, validation, and prodrug design of an ACE2 activator for treating bacterial infection-induced lung inflammation. J Control Release 2023; 364:1-11. [PMID: 37858626 PMCID: PMC10872764 DOI: 10.1016/j.jconrel.2023.10.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
Exacerbated inflammatory responses can be detrimental and pose fatal threats to the host, as exemplified by the global impact of the COVID-19 pandemic, resulting in millions of fatalities. Developing novel drugs to combat the damaging effects of inflammation is essential for both preventive measures and therapeutic interventions. Accumulating evidence suggests that Angiotensin Converting Enzyme 2 (ACE2) possesses the ability to optimize inflammatory responses. However, the clinical applicability of this potential is limited due to the lack of dependable ACE2 activators. In this study, we conducted a screening of an FDA-approved drug library and successfully identified a novel ACE2 activator, termed H4. The activator demonstrated the capability to mitigate lung inflammation caused by bacterial lung infections, effectively modulating neutrophil infiltration. Importantly, to improve the clinical applicability of the poorly water-soluble H4, we developed a prodrug variant with significantly enhanced water solubility while maintaining a similar level of efficacy as H4 in attenuating inflammatory responses in the lungs of mice exposed to bacterial infections. This finding highlights the potential of formulated H4 as a promising candidate for the treatment and prevention of inflammatory diseases, including lung-related conditions.
Collapse
Affiliation(s)
- Peng Lu
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Faith Leslie
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Han Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anjali Sodhi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chang-Yong Choi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hongpeng Jia
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
24
|
Zhang Y, Li J, Feng L, Luo Y, Pang W, Qiu K, Mao M, Song Y, Cheng D, Rao Y, Wang X, Hu Y, Ying Z, Pu X, Lin S, Huang S, Liu G, Zhang W, Xu W, Zhao Y, Ren J. A Population-Based Outcome-Wide Association Study of the Comorbidities and Sequelae Following COVID-19 Infection. J Epidemiol Glob Health 2023; 13:870-885. [PMID: 37889436 PMCID: PMC10686900 DOI: 10.1007/s44197-023-00161-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: 05/03/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Immense attention has been given to the outcome of COVID-19 infection. However, comprehensive studies based on large populational cohort with long-term follow-up are still lacking. This study aimed to investigate the risk of various short-term comorbidities (within one month) and long-term sequelae (above one month) after COVID-19 infection. METHODS In this large prospective cohort study with 14 months follow-up information based on UK biobank, we included 16,776 COVID-19-positive participants and 58,281 COVID-19-negative participants matched for comparison. The risk of each comorbidity and sequela was evaluated by multivariable logistic regression analysis and presented as hazard ratio (HR) and 95% confidence interval (95% CI). RESULTS COVID-19-positive individuals had a higher risk of 47 types of comorbidities within one month following COVID-19 infection, especially those who were older, male, overweight/obese, ever-smoked, with more pre-existing comorbidities and hospitalized. About 70.37% of COVID-19 patients with comorbidities had more than one co-occurring comorbidities. Additionally, only 6 high-risk sequelae were observed after one month of COVID-19 infection, and the incidence was relatively low (< 1%). CONCLUSION In addition to long-term sequelae following COVID-19 infection, plenty of comorbidities were observed, especially in patients with older age, male gender, overweight/obese, more pre-existing comorbidities and severe COVID-19, indicating that more attention should be given to these susceptible persons within this period.
Collapse
Affiliation(s)
- Yuyang Zhang
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Junhong Li
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Feng
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yaxin Luo
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Wendu Pang
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ke Qiu
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Minzi Mao
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Song
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Danni Cheng
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yufang Rao
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyi Wang
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Hu
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiye Ying
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaobin Pu
- Department of Oto-Rhino-Laryngology, Langzhong People's Hospital, Langzhong, China
| | - Shuyan Lin
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shaohui Huang
- Department of Radiation Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, Canada
| | - Geoffrey Liu
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, Canada
| | - Wei Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Xu
- Department of Biostatistics, Princess Margaret Cancer Centre and Dalla Lana School of Public Health, 10-511, 610 University Avenue Toronto, Toronto, ON, Canada.
| | - Yu Zhao
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Jianjun Ren
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China.
- Department of Oto-Rhino-Laryngology, Langzhong People's Hospital, Langzhong, China.
| |
Collapse
|
25
|
Baral B, Saini V, Tandon A, Singh S, Rele S, Dixit AK, Parmar HS, Meena AK, Jha HC. SARS-CoV-2 envelope protein induces necroptosis and mediates inflammatory response in lung and colon cells through receptor interacting protein kinase 1. Apoptosis 2023; 28:1596-1617. [PMID: 37658919 DOI: 10.1007/s10495-023-01883-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
SARS-CoV-2 Envelope protein (E) is one of the crucial components in virus assembly and pathogenesis. The current study investigated its role in the SARS-CoV-2-mediated cell death and inflammation in lung and gastrointestinal epithelium and its effect on the gastrointestinal-lung axis. We observed that transfection of E protein increases the lysosomal pH and induces inflammation in the cell. The study utilizing Ethidium bromide/Acridine orange and Hoechst/Propidium iodide staining demonstrated necrotic cell death in E protein transfected cells. Our study revealed the role of the necroptotic marker RIPK1 in cell death. Additionally, inhibition of RIPK1 by its specific inhibitor Nec-1s exhibits recovery from cell death and inflammation manifested by reduced phosphorylation of NFκB. The E-transfected cells' conditioned media induced inflammation with differential expression of inflammatory markers compared to direct transfection in the gastrointestinal-lung axis. In conclusion, SARS-CoV-2 E mediates inflammation and necroptosis through RIPK1, and the E-expressing cells' secretion can modulate the gastrointestinal-lung axis. Based on the data of the present study, we believe that during severe COVID-19, necroptosis is an alternate mechanism of cell death besides ferroptosis, especially when the disease is not associated with drastic increase in serum ferritin.
Collapse
Affiliation(s)
- Budhadev Baral
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Vaishali Saini
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Akrati Tandon
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Siddharth Singh
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Samiksha Rele
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Amit Kumar Dixit
- Central Ayurveda Research Institute, 4-CN Block, Sector-V, Bidhannagar, Kolkata, 700091, India
| | - Hamendra Singh Parmar
- School of Biotechnology, Devi Ahilya Vishwavidyalaya, Takshashila Campus, Indore, Madhya Pradesh, 452001, India
| | - Ajay Kumar Meena
- Regional Ayurveda Research Institute, Amkhoh, Gwalior, Madhya Pradesh, 474001, India
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India.
| |
Collapse
|
26
|
Choi YJ, Seo YB, Seo JW, Lee J, Nham E, Seong H, Yoon JG, Noh JY, Cheong HJ, Kim WJ, Kim EJ, Song JY. Effectiveness of Antiviral Therapy on Long COVID: A Systematic Review and Meta-Analysis. J Clin Med 2023; 12:7375. [PMID: 38068427 PMCID: PMC10707593 DOI: 10.3390/jcm12237375] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/13/2023] [Accepted: 11/25/2023] [Indexed: 09/26/2024] Open
Abstract
Antiviral treatment reduces the severity and mortality of SARS-CoV-2 infection; however, its effectiveness against long COVID-19 is unclear. This study aimed to evaluate the effectiveness of antiviral drugs in preventing long COVID and related hospitalizations/deaths. Scientific and medical databases were searched from 1 January 2020 to 30 June 2023. We included observational cohort studies comparing individuals receiving early antiviral therapy for COVID-19 and those receiving supportive treatment. A fixed-effects model was used to merge the effects reported in two or more studies. The risk of post-acute sequelae of COVID-19 (PASC) was combined as an odds ratio (OR). Six studies were selected, including a total of 3,352,235 participants. The occurrence of PASC was 27.5% lower in patients who received antiviral drugs during the early stages of SARS-CoV-2 infection (OR = 0.725; 95% confidence interval [CI] = 0.409-0.747) than in the supportive treatment group. Moreover, the risk of PASC-associated hospitalization and mortality was 29.7% lower in patients receiving early antiviral therapy than in the supportive treatment group (OR = 0.721; 95% CI = 0.697-0.794). Early antiviral therapy was associated with a reduced risk of PASC and related hospitalization or death. Thus, early antiviral therapy is recommended for at-risk individuals.
Collapse
Affiliation(s)
- Yu Jung Choi
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (Y.J.C.); (E.N.); (H.S.); (J.G.Y.); (J.Y.N.); (H.J.C.); (W.J.K.)
- Vaccine Innovation Center-KU Medicine, Seoul 02841, Republic of Korea
| | - Yu Bin Seo
- Division of Infectious Disease, Department of Internal Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07442, Republic of Korea; (Y.B.S.); (J.L.)
| | - Jun-Won Seo
- Departments of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea;
| | - Jacob Lee
- Division of Infectious Disease, Department of Internal Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07442, Republic of Korea; (Y.B.S.); (J.L.)
| | - Eliel Nham
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (Y.J.C.); (E.N.); (H.S.); (J.G.Y.); (J.Y.N.); (H.J.C.); (W.J.K.)
- Vaccine Innovation Center-KU Medicine, Seoul 02841, Republic of Korea
| | - Hye Seong
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (Y.J.C.); (E.N.); (H.S.); (J.G.Y.); (J.Y.N.); (H.J.C.); (W.J.K.)
- Vaccine Innovation Center-KU Medicine, Seoul 02841, Republic of Korea
| | - Jin Gu Yoon
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (Y.J.C.); (E.N.); (H.S.); (J.G.Y.); (J.Y.N.); (H.J.C.); (W.J.K.)
- Vaccine Innovation Center-KU Medicine, Seoul 02841, Republic of Korea
| | - Ji Yun Noh
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (Y.J.C.); (E.N.); (H.S.); (J.G.Y.); (J.Y.N.); (H.J.C.); (W.J.K.)
- Vaccine Innovation Center-KU Medicine, Seoul 02841, Republic of Korea
| | - Hee Jin Cheong
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (Y.J.C.); (E.N.); (H.S.); (J.G.Y.); (J.Y.N.); (H.J.C.); (W.J.K.)
- Vaccine Innovation Center-KU Medicine, Seoul 02841, Republic of Korea
| | - Woo Joo Kim
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (Y.J.C.); (E.N.); (H.S.); (J.G.Y.); (J.Y.N.); (H.J.C.); (W.J.K.)
| | - Eun Jung Kim
- Health, Welfare, Family and Gender Equality Team, National Assembly Research Service, Seoul 07233, Republic of Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (Y.J.C.); (E.N.); (H.S.); (J.G.Y.); (J.Y.N.); (H.J.C.); (W.J.K.)
- Vaccine Innovation Center-KU Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
27
|
Kumar M. Hydrogen sulfide: From a toxic gas to a potential therapy for COVID-19 and inflammatory disorders. Nitric Oxide 2023; 140-141:8-15. [PMID: 37648016 DOI: 10.1016/j.niox.2023.08.002] [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: 05/15/2023] [Revised: 07/06/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023]
Abstract
COVID-19 has been shown to induce inflammatory disorders and CNS manifestations. Swift and efficient treatment strategies are urgently warranted for the management of COVID, inflammatory and neurological disorders. Hydrogen sulfide (H2S) has been associated with several clinical disorders due to its potential to influence a broad range of biological signalling pathways. According to recent clinical studies, COVID patients with lower physiological H2S had higher fatality rates. These findings clearly demonstrate an inverse correlation between H2S levels and the severity of COVID-19. H2S has been proposed as a protective molecule because of its antioxidant, anti-inflammatory, and antiviral properties. Various H2S-releasing prodrugs, hybrids and natural compounds have been tested for their therapeutic efficacy in viral infections and inflammatory disorders. In this review, I am highlighting the rationale for using H2S-based interventions for the management of COVID-19 and post-infection inflammatory disorders including neuroinflammation. I am also proposing therepurposing of existing H2S-releasing prodrugs, developing new NO-H2S-hybrids, targeting H2S metabolic pathways, and using H2S-producing dietary supplements as viable defensive strategies against SARS-CoV-2 infection and COVID-19 pathologies.
Collapse
Affiliation(s)
- Mohit Kumar
- Centre for Excellence in Functional Foods, Food and Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Punjab, 140306, India.
| |
Collapse
|
28
|
Wang Z, Zhao C, Li C, Liu S, Ding J, He C, Liu J, Dong B, Yang Z, Liu Q, Zhu H, Liu Y. Molecular PET/CT mapping of rhACE2 distribution and quantification in organs to aid in SARS-CoV-2 targeted therapy. J Med Virol 2023; 95:e29221. [PMID: 38009705 DOI: 10.1002/jmv.29221] [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: 05/06/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023]
Abstract
Coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, poses a significant threat to public health. Angiotensin-converting enzyme 2 (ACE2) is a key receptor for SARS-CoV-2 infection. Recombinant human ACE2 (RhACE2), as a soluble supplement for human ACE2, can competitively block SARS-CoV-2 infection. In this study, a mouse organ in situ rhACE2 high aggregation model was constructed for the first time, and in vivo real-time positron emission tomography (PET) imaging of rhACE2 in the mouse model was performed using an ACE2-specific agent 68 Ga-HZ20. This radiotracer exhibits reliable radiochemical properties in vitro and maintains a high affinity for rhACE2 in vivo. In terms of probe uptake, 68 Ga-HZ20 showed a good target-to-nontarget ratio and was rapidly cleared from the circulatory system and excreted by the kidneys and urinary system. PET imaging with this radiotracer can noninvasively and accurately monitor the content and distribution of rhACE2 in the body, which clarifies that rhACE2 can aggregate in multiple organs, suggesting the preventive and therapeutic potential of rhACE2 for SARS-CoV-2 and COVID-19.
Collapse
Affiliation(s)
- Zilei Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanke Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuangui Li
- Department of Nuclear Medicine, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Song Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jin Ding
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chengxue He
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiayue Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Bin Dong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
- Department of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Qi Liu
- Department of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
- International Cancer Center, Department of medicine, Shenzhen University, Shenzhen, Guangdong, China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
- Department of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Youping Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
29
|
Guo ZF, Tongmuang N, Li C, Zhang C, Hu L, Capreri D, Zuo MX, Summer R, Sun J. Inhibiting endothelial cell Mst1 attenuates acute lung injury in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559864. [PMID: 37808846 PMCID: PMC10557750 DOI: 10.1101/2023.09.27.559864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Background Lung endothelium plays a pivotal role in the orchestration of inflammatory and injury responses to acute pulmonary insults. Mammalian sterile 20-like kinase 1 (Mst1), a mammalian homolog of Hippo, is a serine/threonine kinase that is ubiquitously expressed in many tissues and has been shown to play an important role in the regulation of apoptosis, inflammation, stress responses, and organ growth. While Mst1 exhibits high expression in the lung, its involvement in the endothelial response to pulmonary insults remains largely unexplored. Methods Mst1 activity was assessed in lung endothelium by western blot. Mst1 endothelial specific knockout mice and a pharmacological inhibitor were employed to assess the effects of Mst1 on homeostatic and lipopolysaccharide (LPS)-induced endothelial responses. Readouts for these studies included various assays, including NF-κB activation and levels of various inflammatory cytokines and adhesion molecules. The role of Mst1 in lung injury was evaluated in a LPS-induced murine model of acute lung injury (ALI). Results Mst1 phosphorylation was significantly increased in lung endothelial cells after exposure to tumor necrosis factor (TNF)-alpha (TNF-α) and mouse lung tissues after LPS exposure. Overexpression of full length Mst1 or its kinase domain promoted nuclear factor kappaB (NF-κB) activation through promoting JNK and p38 activation, whereas dominant negative forms of Mst1 (DN-Mst1) attenuated endothelial responses to TNF-α and interleukin-1β. Consistent with this, targeted deletion of Mst1 in lung endothelium reduced lung injury to LPS in mice. Similarly, wild-type mice were protected from LPS-induced lung injury following treatment with a pharmacological inhibitor of Mst1/2. Conclusions Our findings identified Mst1 kinase as a key regulator in the control of lung EC activation and suggest that therapeutic strategies aimed at inhibiting Mst1 activation might be effective in the prevention and treatment of lung injury to inflammatory insults.
Collapse
|
30
|
Yuan C, Ma Z, Xie J, Li W, Su L, Zhang G, Xu J, Wu Y, Zhang M, Liu W. The role of cell death in SARS-CoV-2 infection. Signal Transduct Target Ther 2023; 8:357. [PMID: 37726282 PMCID: PMC10509267 DOI: 10.1038/s41392-023-01580-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/09/2023] [Accepted: 07/31/2023] [Indexed: 09/21/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), showing high infectiousness, resulted in an ongoing pandemic termed coronavirus disease 2019 (COVID-19). COVID-19 cases often experience acute respiratory distress syndrome, which has caused millions of deaths. Apart from triggering inflammatory and immune responses, many viral infections can cause programmed cell death in infected cells. Cell death mechanisms have a vital role in maintaining a suitable environment to achieve normal cell functionality. Nonetheless, these processes are dysregulated, potentially contributing to disease pathogenesis. Over the past decades, multiple cell death pathways are becoming better understood. Growing evidence suggests that the induction of cell death by the coronavirus may significantly contributes to viral infection and pathogenicity. However, the interaction of SARS-CoV-2 with cell death, together with its associated mechanisms, is yet to be elucidated. In this review, we summarize the existing evidence concerning the molecular modulation of cell death in SARS-CoV-2 infection as well as viral-host interactions, which may shed new light on antiviral therapy against SARS-CoV-2.
Collapse
Affiliation(s)
- Cui Yuan
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Zhenling Ma
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Jiufeng Xie
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Wenqing Li
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Lijuan Su
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Guozhi Zhang
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Jun Xu
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Yaru Wu
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Min Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Wei Liu
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China.
| |
Collapse
|
31
|
Do MH, Li H, Cho SY, Oh S, Jeong JH, Song MS, Jeong JM. Animal efficacy study of a plant extract complex (BEN815) as a potential treatment for COVID-19. PLoS One 2023; 18:e0291537. [PMID: 37708114 PMCID: PMC10501575 DOI: 10.1371/journal.pone.0291537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/22/2023] [Indexed: 09/16/2023] Open
Abstract
In a short time, several types of injectable and oral therapeutics have been developed and used to effectively manage patients with coronavirus disease 2019 (COVID-19). BEN815 is an improved mixture of three extracts (Psidium guajava, Camellia sinensis, and Rosa hybrida) recognized by the Ministry of Food and Drug Safety of Korea as a health food ingredient that alleviates allergic rhinitis. The current animal efficacy study was performed to assess its probability of improving COVID-19 symptoms. BEN815 treatment significantly increased the survival of K18-hACE2 transgenic mice and reduced viral titers in the lungs at 5 days post infection (DPI). Furthermore, the lungs of the treated mice showed mild tissue damage at 5 DPI and nearly complete recovery from COVID-19 at 14 DPI. BEN815 appears to be an effective and minimally toxic anti-SARS-CoV-2 agent in mice and has potential for clinical applications.
Collapse
Affiliation(s)
- Moon Ho Do
- Biotechnology Research Center, Ben’s Lab Co., Ltd., Anyang-si, Gyeonggi-do, Republic of Korea
| | - Hua Li
- Biotechnology Research Center, Ben’s Lab Co., Ltd., Anyang-si, Gyeonggi-do, Republic of Korea
| | - Su Yeon Cho
- Biotechnology Research Center, Ben’s Lab Co., Ltd., Anyang-si, Gyeonggi-do, Republic of Korea
| | - Subin Oh
- Biotechnology Research Center, Ben’s Lab Co., Ltd., Anyang-si, Gyeonggi-do, Republic of Korea
| | - Ju Hwan Jeong
- Department of Microbiology, Chungbuk National University College of Medicine and Medical Research Institute, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Min-Suk Song
- Department of Microbiology, Chungbuk National University College of Medicine and Medical Research Institute, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Jong-Moon Jeong
- Biotechnology Research Center, Ben’s Lab Co., Ltd., Anyang-si, Gyeonggi-do, Republic of Korea
- Department of Bioscience, The University of Suwon, Hwasung-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
32
|
Gasparello J, Marzaro G, Papi C, Gentili V, Rizzo R, Zurlo M, Scapoli C, Finotti A, Gambari R. Effects of Sulforaphane on SARS‑CoV‑2 infection and NF‑κB dependent expression of genes involved in the COVID‑19 'cytokine storm'. Int J Mol Med 2023; 52:76. [PMID: 37477130 PMCID: PMC10555481 DOI: 10.3892/ijmm.2023.5279] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 06/09/2023] [Indexed: 07/22/2023] Open
Abstract
Since its spread at the beginning of 2020, the coronavirus disease 2019 (COVID‑19) pandemic represents one of the major health problems. Despite the approval, testing, and worldwide distribution of anti‑severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) vaccines, the development of specific antiviral agents targeting the SARS‑CoV‑2 life cycle with high efficiency, and/or interfering with the associated 'cytokine storm', is highly required. A recent study, conducted by the authors' group indicated that sulforaphane (SFN) inhibits the expression of IL‑6 and IL‑8 genes induced by the treatment of IB3‑1 bronchial cells with a recombinant spike protein of SARS‑CoV‑2. In the present study, the ability of SFN to inhibit SARS‑CoV‑2 replication and the expression of pro‑inflammatory genes encoding proteins of the COVID‑19 'cytokine storm' was evaluated. SARS‑CoV‑2 replication was assessed in bronchial epithelial Calu‑3 cells. Moreover, SARS‑CoV‑2 replication and expression of pro‑inflammatory genes was evaluated by reverse transcription quantitative droplet digital PCR. The effects on the expression levels of NF‑κB were assessed by western blotting. Molecular dynamics simulations of NF‑kB/SFN interactions were conducted with Gromacs 2021.1 software under the Martini 2 CG force field. Computational studies indicated that i) SFN was stably bound with the NF‑κB monomer; ii) a ternary NF‑kB/SFN/DNA complex was formed; iii) the SFN interacted with both the protein and the nucleic acid molecules modifying the binding mode of the latter, and impairing the full interaction between the NF‑κB protein and the DNA molecule. This finally stabilized the inactive complex. Molecular studies demonstrated that SFN i) inhibits the SARS‑CoV‑2 replication in infected Calu‑3 cells, decreasing the production of the N‑protein coding RNA sequences, ii) decreased NF‑κB content in SARS‑CoV‑2 infected cells and inhibited the expression of NF‑kB‑dependent IL‑1β and IL‑8 gene expression. The data obtained in the present study demonstrated inhibitory effects of SFN on the SARS‑CoV‑2 life cycle and on the expression levels of the pro‑inflammatory genes, sustaining the possible use of SFN in the management of patients with COVID‑19.
Collapse
Affiliation(s)
- Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, I-35131 Padova
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Valentina Gentili
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, I-44121 Ferrara, Italy
| | - Roberta Rizzo
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, I-44121 Ferrara, Italy
| | - Matteo Zurlo
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Chiara Scapoli
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| |
Collapse
|
33
|
Moghaddam RR, Khorasanchi Z, Noor AR, Moghadam MSF, Esfahani AJ, Alyakobi AKM, Alboresha ML, Sharifan P, Bahari A, Rezvani R, Aghasizade M, Heshmati M, Darban RA, Ferns G, Mobarhan MG. High-dose vitamin D supplementation is related to an improvement in serum alkaline phosphatase in COVID-19 patients; a randomized double-blinded clinical trial. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2023; 42:71. [PMID: 37491318 PMCID: PMC10369932 DOI: 10.1186/s41043-023-00409-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023]
Abstract
BACKGROUND The benefits and harms of vitamin D supplementation in the treatment of COVID-19 have not yet been fully documented. In this study, we aimed to evaluate the effects of high-dose vitamin D supplementation on liver function tests in COVID-19. METHOD This double-blinded randomized clinical trial was conducted on 140 hospitalized patients aged > 30 years. Patients were randomly allocated to receive either intervention group (n = 70 receiving 50,000 IU of vitamin D capsules orally as a single dose and then 10,000 IU syrup daily from the second day of admission for 30 days) and the control group (n = 70 receiving 1000 IU vitamin D syrup orally per day). Liver function tests (LFT), including alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), gamma-glutamyl transferase (GGT), and Lactate Dehydrogenase (LDH) were evaluated at baseline and at the end of the intervention. Decision tree analysis was performed to identify the predictors for change in liver enzymes. RESULTS Among COVID-19 patients, a significant decrease was observed in serum level of ALP between intervention and placebo groups (p = 0.04). In addition, decision tree analysis revealed that GGT, temperature, serum magnesium level at baseline and gender were the most important predictors of ALT changes in COVID-19 patients. CONCLUSION High-dose vitamin D supplementation improved ALP markers among COVID-19 patients. More randomized controlled trials with longer follow-up times will be required.
Collapse
Affiliation(s)
- Reza Rezvani Moghaddam
- Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Khorasanchi
- Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ayad Rasool Noor
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | | | | | | | - Payam Sharifan
- Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Bahari
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Rezvani
- Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Aghasizade
- International UNESCO Center for Health Related Basic Sciences and Human Nutrition, Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Heshmati
- Department of Clinical Care Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Assaran Darban
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Gordon Ferns
- Brighton and Sussex Medical School, Division of Medical Education, Brighton, UK
| | - Majid Ghayour Mobarhan
- International UNESCO Center for Health Related Basic Sciences and Human Nutrition, Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
34
|
Tomkinson S, Triscott C, Schenk E, Foey A. The Potential of Probiotics as Ingestible Adjuvants and Immune Modulators for Antiviral Immunity and Management of SARS-CoV-2 Infection and COVID-19. Pathogens 2023; 12:928. [PMID: 37513775 PMCID: PMC10384479 DOI: 10.3390/pathogens12070928] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Probiotic bacteria are able to modulate general antiviral responsiveness, including barrier functionality and innate and adaptive immune responses. The COVID-19 pandemic, resulting from SARS-CoV-2 infection, has created a need to control and treat this viral infection and its ensuing immunopathology with a variety of approaches; one such approach may involve the administration of probiotic bacteria. As with most viral infections, its pathological responses are not fully driven by the virus, but are significantly contributed to by the host's immune response to viral infection. The potential adoption of probiotics in the treatment of COVID-19 will have to appreciate the fine line between inducing antiviral immunity without over-provoking immune inflammatory responses resulting in host-derived immunopathological tissue damage. Additionally, the effect exerted on the immune system by SARS-CoV-2 evasion strategies will also have to be considered when developing a robust response to this virus. This review will introduce the immunopathology of COVID-19 and the immunomodulatory effects of probiotic strains, and through their effects on a range of respiratory pathogens (IAV, SARS-CoV, RSV), as well as SARS-CoV-2, will culminate in a focus on how these bacteria can potentially manipulate both infectivity and immune responsiveness via barrier functionality and both innate and adaptive immunity. In conclusion, the harnessing of induction and augmentation of antiviral immunity via probiotics may not only act as an ingestible adjuvant, boosting immune responsiveness to SARS-CoV-2 infection at the level of barrier integrity and innate and adaptive immunity, but also act prophylactically to prevent infection and enhance protection afforded by current vaccine regimens.
Collapse
Affiliation(s)
- Sophie Tomkinson
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - Cloe Triscott
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - Emily Schenk
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
- Peninsula Medical School, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - Andrew Foey
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| |
Collapse
|
35
|
Raque M, Raev SA, Guo Y, Kick MK, Saif LJ, Vlasova AN. Host Cell Response to Rotavirus Infection with Emphasis on Virus-Glycan Interactions, Cholesterol Metabolism, and Innate Immunity. Viruses 2023; 15:1406. [PMID: 37515094 PMCID: PMC10385841 DOI: 10.3390/v15071406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Although rotavirus A (RVA) is the primary cause of acute viral gastroenteritis in children and young animals, mechanisms of its replication and pathogenesis remain poorly understood. We previously demonstrated that the neuraminidase-mediated removal of terminal sialic acids (SAs) significantly enhanced RVA-G9P[13] replication, while inhibiting RVA-G5P[7] replication. In this study, we compared the transcriptome responses of porcine ileal enteroids (PIEs) to G5P[7] vs. G9P[13] infections, with emphasis on the genes associated with immune response, cholesterol metabolism, and host cell attachment. The analysis demonstrated that G9P[13] infection led to a robust modulation of gene expression (4093 significantly modulated genes vs. 488 genes modulated by G5P[7]) and a significant modulation of glycosyltransferase-encoding genes. The two strains differentially affected signaling pathways related to immune response, with G9P[13] mostly upregulating and G5P[7] inhibiting them. Both RVAs modulated the expression of genes encoding for cholesterol transporters. G9P[13], but not G5P[7], significantly affected the ceramide synthesis pathway known to affect both cholesterol and glycan metabolism. Thus, our results highlight the unique mechanisms regulating cellular response to infection caused by emerging/re-emerging and historical RVA strains relevant to RVA-receptor interactions, metabolic pathways, and immune signaling pathways that are critical in the design of effective control strategies.
Collapse
Affiliation(s)
- Molly Raque
- Center for Food Animal Health Research Program, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 43210, USA
| | - Sergei A Raev
- Center for Food Animal Health Research Program, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 43210, USA
| | - Yusheng Guo
- Center for Food Animal Health Research Program, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 43210, USA
| | - Maryssa K Kick
- Center for Food Animal Health Research Program, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 43210, USA
| | - Linda J Saif
- Center for Food Animal Health Research Program, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 43210, USA
| | - Anastasia N Vlasova
- Center for Food Animal Health Research Program, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 43210, USA
| |
Collapse
|
36
|
Wu WY, Jiao X, Song WX, Wu P, Xiao PQ, Huang XF, Wang K, Zhan SF. Network pharmacology and bioinformatics analysis identifies potential therapeutic targets of Naringenin against COVID-19/LUSC. Front Endocrinol (Lausanne) 2023; 14:1187882. [PMID: 37347115 PMCID: PMC10281056 DOI: 10.3389/fendo.2023.1187882] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) is a highly contagious respiratory disease that has posed a serious threat to people's daily lives and caused an unprecedented challenge to public health and people's health worldwide. Lung squamous cell carcinoma (LUSC) is a common type of lung malignancy with a highly aggressive nature and poor prognosis. Patients with LUSC could be at risk for COVID-19, We conducted this study to examine the potential for naringenin to develop into an ideal medicine and investigate the underlying action mechanisms of naringenin in COVID-19 and LUSC due to the anti-viral, anti-tumor, and anti-inflammatory activities of naringenin. Methods LUSC related genes were obtained from TCGA, PharmGKB, TTD,GeneCards and NCBI, and then the transcriptome data for COVID-19 was downloaded from GEO, DisGeNET, CTD, DrugBank, PubChem, TTD, NCBI Gene, OMIM. The drug targets of Naringenin were revealed through CTD, BATMAN, TCMIP, SymMap, Chemical Association Networks, SwissTargetPrediction, PharmMapper, ECTM, and DGIdb. The genes related to susceptibility to COVID-19 in LUSC patients were obtained through differential analysis. The interaction of COVID-19/LUSC related genes was evaluated and demonstrated using STRING to develop a a COX risk regression model to screen and evaluate the association of genes with clinical characteristics. To investigate the related functional and pathway analysis of the common targets of COVID-19/LUSC and Naringenin, KEGG and GO enrichment analysis were employed to perform the functional analysis of the target genes. Finally, The Hub Gene was screened and visualized using Cytoscape, and molecular docking between the drug and the target was performed using Autodock. Results We discovered numerous COVID-19/LUSC target genes and examined their prognostic value in LUSC patients utilizing a variety of bioinformatics and network pharmacology methods. Furthermore, a risk score model with strong predictive performance was developed based on these target genes to assess the prognosis of LUSC patients with COVID-19. We intersected the therapeutic target genes of naringenin with the LUSC, COVID-19-related targets, and identified 354 common targets, which could be used as potential target genes for naringenin to treat COVID-19/LUSC. The treatment of COVID-19/LUSC with naringenin may involve oxidative stress, anti-inflammatory, antiviral, antiviral, apoptosis, immunological, and multiple pathways containing PI3K-Akt, HIF-1, and VEGF, according to the results of the GO and KEGG enrichment analysis of these 354 common targets. By constructing a PPI network, we ascertained AKT1, TP53, SRC, MAPK1, MAPK3, and HSP90AA1 as possible hub targets of naringenin for the treatment of COVID-19/LUSC. Last but not least, molecular docking investigations showed that naringenin has strong binding activity in COVID-19/LUSC. Conclusion We revealed for the first time the pharmacological targets and potential molecular processes of naringenin for the treatment of COVID-19/LUSC. However, these results need to be confirmed by additional research and validation in real LUSC patients with COVID-19.
Collapse
Affiliation(s)
- Wen-yu Wu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Jiao
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wen-xin Song
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Wu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pei-qi Xiao
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiu-fang Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kai Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shao-feng Zhan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
37
|
Huang X, Fan W, Sun J, Yang J, Zhang Y, Wang Q, Li P, Zhang Y, Zhang S, Li H, Wang J, Feng L, Zhao J, Chen L, Linbing Q. SARS-CoV-2 induces cardiomyocyte apoptosis and inflammation but can be ameliorated by ACE inhibitor Captopril. Antiviral Res 2023; 215:105636. [PMID: 37207821 DOI: 10.1016/j.antiviral.2023.105636] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/21/2023]
Abstract
Although the clinical manifestation of COVID-19 is mainly respiratory symptoms, approximately 20% of patients suffer from cardiac complications. COVID-19 patients with cardiovascular disease have higher severity of myocardial injury and poor outcomes. The underlying mechanism of myocardial injury caused by SARS-CoV-2 infection remains unclear. Using a non-transgenic mouse model infected with Beta variant (B.1.351), we found that the viral RNA could be detected in lungs and hearts of infected mice. Pathological analysis showed thinner ventricular wall, disorganized and ruptured myocardial fiber, mild inflammatory infiltration, and mild epicardia or interstitial fibrosis in hearts of infected mice. We also found that SARS-CoV-2 could infect cardiomyocytes and produce infectious progeny viruses in human pluripotent stem cell-derived cardiomyocyte-like cells (hPSC-CMs). SARS-CoV-2 infection caused apoptosis, reduction of mitochondrial integrity and quantity, and cessation of beating in hPSC-CMs. In order to dissect the mechanism of myocardial injury caused by SARS-CoV-2 infection, we employed transcriptome sequencing of hPSC-CMs at different time points after viral infection. Transcriptome analysis showed robust induction of inflammatory cytokines and chemokines, up-regulation of MHC class I molecules, activation of apoptosis signaling and cell cycle arresting. These may cause aggravate inflammation, immune cell infiltration, and cell death. Furthermore, we found that Captopril (hypotensive drugs targeting ACE) treatment could alleviate SARS-CoV-2 induced inflammatory response and apoptosis in cardiomyocytes via inactivating TNF signaling pathways, suggesting Captopril may be beneficial for reducing COVID-19 associated cardiomyopathy. These findings preliminarily explain the molecular mechanism of pathological cardiac injury caused by SARS-CoV-2 infection, providing new perspectives for the discovery of antiviral therapeutics.
Collapse
Affiliation(s)
- Xiaohan Huang
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Wenxia Fan
- Guangzhou Laboratory, Guangzhou, 510320, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Jiaqing Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Guangzhou Laboratory, Guangzhou, 510320, China
| | - Yanjun Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Qian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Pingchao Li
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yudi Zhang
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Shengnan Zhang
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Heying Li
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jianhua Wang
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Liqiang Feng
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Guangzhou Laboratory, Guangzhou, 510320, China.
| | - Qu Linbing
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
38
|
Ecarnot F, Boccardi V, Calcagno A, Franceschi C, Fülop T, Itzhaki RF, Michel JP, Panza F, Rainero I, Solfrizzi V, Ticinesi A, Veronese N, Maggi S. Dementia, infections and vaccines: 30 years of controversy. Aging Clin Exp Res 2023; 35:1145-1160. [PMID: 37160649 PMCID: PMC10169152 DOI: 10.1007/s40520-023-02409-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 03/21/2023] [Indexed: 05/11/2023]
Abstract
This paper reports the proceedings of a virtual meeting convened by the European Interdisciplinary Council on Ageing (EICA), to discuss the involvement of infectious disorders in the pathogenesis of dementia and neurological disorders leading to dementia. We recap how our view of the infectious etiology of dementia has changed over the last 30 years in light of emerging evidence, and we present evidence in support of the implication of infection in dementia, notably Alzheimer's disease (AD). The bacteria and viruses thought to be responsible for neuroinflammation and neurological damage are reviewed. We then review the genetic basis for neuroinflammation and dementia, highlighting the genes that are currently the focus of investigation as potential targets for therapy. Next, we describe the antimicrobial hypothesis of dementia, notably the intriguing possibility that amyloid beta may itself possess antimicrobial properties. We further describe the clinical relevance of the gut-brain axis in dementia, the mechanisms by which infection can move from the intestine to the brain, and recent findings regarding dysbiosis patterns in patients with AD. We review the involvement of specific pathogens in neurological disorders, i.e. SARS-CoV-2, human immunodeficiency virus (HIV), herpes simplex virus type 1 (HSV1), and influenza. Finally, we look at the role of vaccination to prevent dementia. In conclusion, there is a large body of evidence supporting the involvement of various infectious pathogens in the pathogenesis of dementia, but large-scale studies with long-term follow-up are needed to elucidate the role that infection may play, especially before subclinical or clinical disease is present.
Collapse
Affiliation(s)
- Fiona Ecarnot
- EA3920, University of Franche-Comté, 25000, Besancon, France
- Department of Cardiology, University Hospital Besancon, 3-8 Boulevard Fleming, 25000, Besancon, France
| | - Virginia Boccardi
- Institute of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Santa Maria Della Misericordia Hospital, Piazzale Gambuli 1, 06132, Perugia, Italy
| | - Andrea Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Claudio Franceschi
- Laboratory of Systems Medicine of Healthy Aging, Institute of Biology and Biomedicine and Institute of Information Technology, Mathematics and Mechanics, Department of Applied Mathematics, N. I. Lobachevsky State University, Nizhny Novgorod, Russia
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Tamas Fülop
- Department of Medicine, Geriatrics Division, Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, J1H 5N4, Canada
| | - Ruth F Itzhaki
- Institute of Population Ageing, University of Oxford and Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | - Francesco Panza
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Innocenzo Rainero
- Dementia Center, Department of Neuroscience "Rita Levi Montalcini", University of Torino, Turin, Italy
| | - Vincenzo Solfrizzi
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Andrea Ticinesi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Geriatric-Rehabilitation Department, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Nicola Veronese
- Geriatrics Section, Department of Internal Medicine, University of Palermo, Palermo, Italy.
| | - Stefania Maggi
- National Research Council, Neuroscience Institute, Aging Branch, Padua, Italy
| |
Collapse
|
39
|
Sharbatdar Y, Mousavian R, Noorbakhsh Varnosfaderani SM, Aziziyan F, Liaghat M, Baziyar P, Yousefi Rad A, Tavakol C, Moeini AM, Nabi-Afjadi M, Zalpoor H, Kazemi-Lomedasht F. Diabetes as one of the long-term COVID-19 complications: from the potential reason of more diabetic patients' susceptibility to COVID-19 to the possible caution of future global diabetes tsunami. Inflammopharmacology 2023; 31:1029-1052. [PMID: 37079169 PMCID: PMC10116486 DOI: 10.1007/s10787-023-01215-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 03/27/2023] [Indexed: 04/21/2023]
Abstract
According to recent researches, people with diabetes mellitus (type 1 and 2) have a higher incidence of coronavirus disease 2019 (COVID-19), which is caused by a SARS-CoV-2 infection. In this regard, COVID-19 may make diabetic patients more sensitive to hyperglycemia by modifying the immunological and inflammatory responses and increasing reactive oxygen species (ROS) predisposing the patients to severe COVID-19 and potentially lethal results. Actually, in addition to COVID-19, diabetic patients have been demonstrated to have abnormally high levels of inflammatory cytokines, increased virus entrance, and decreased immune response. On the other hand, during the severe stage of COVID-19, the SARS-CoV-2-infected patients have lymphopenia and inflammatory cytokine storms that cause damage to several body organs such as β cells of the pancreas which may make them as future diabetic candidates. In this line, the nuclear factor kappa B (NF-κB) pathway, which is activated by a number of mediators, plays a substantial part in cytokine storms through various pathways. In this pathway, some polymorphisms also make the individuals more competent to diabetes via infection with SARS-CoV-2. On the other hand, during hospitalization of SARS-CoV-2-infected patients, the use of some drugs may unintentionally lead to diabetes in the future via increasing inflammation and stress oxidative. Thus, in this review, we will first explain why diabetic patients are more susceptible to COVID-19. Second, we will warn about a future global diabetes tsunami via the SARS-CoV-2 as one of its long-term complications.
Collapse
Affiliation(s)
- Yasamin Sharbatdar
- Department of Anesthesiology, School of Allied Medical Sciences, Ahvaz Jundishapur, University of Medical Sciences, Ahvaz, Iran
| | - Ronak Mousavian
- Department of Clinical Biochemistry, School of Medicine, Cellular and Molecular Research Center, Medical Basic Science Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Fatemeh Aziziyan
- Department of Biochemistry, Faculty of Biological Sciences, University of Tarbiat Modares, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahsa Liaghat
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Islamic Azad University, Kazerun Branch, Kazerun, Iran
| | - Payam Baziyar
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Ali Yousefi Rad
- Department of Biochemistry, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Chanour Tavakol
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Mansour Moeini
- Department of Internal Medicine, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, University of Tarbiat Modares, Tehran, Iran.
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
40
|
Agirbasli M, Korkmaz R, Isman FK. Association of Low Levels of Soluble Low-Density Lipoprotein Receptor-Related Protein-1 and COVID-19 Outcomes During the First Wave of Pandemic. Cureus 2023; 15:e37254. [PMID: 37168200 PMCID: PMC10166571 DOI: 10.7759/cureus.37254] [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: 04/06/2023] [Indexed: 05/13/2023] Open
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP1) is an endocytosis receptor that clears inflammatory proteins from circulation. LRP1 has anti-inflammatory effects that bind pro-inflammatory cytokines or ligands. LRP1 has a soluble form (sLRP1) which can be measured in serum. We report sLRP1 levels in hospitalized patients with COVID-19. The first objective of this study is to compare the sLRP1 levels between COVID-19 patients and healthy controls. The second objective is to examine the association between sLRP1 and the clinical outcome of COVID-19. All patients (20-80 years of age) were evaluated in a hospital using a positive PCR test for SARS‑CoV‑2 between April 1, 2020, and June 1, 2020. Controls (n=59) were selected from healthy subjects. sLRP1 levels were measured in patients from the emergency department (ED), inpatient service (IS), and the intensive care unit (ICU). The study included 180 cases. COVID-19 patients showed significantly lower sLRP1 levels compared to controls (1.43 (1.86) versus 2.27 (1.68) μg/mL, respectively, p<0.001). sLRP1 levels were 1.26 (1.81), 1.37 (1.65), and 1.74 (1.98) μg/mL in patients from ED, IS, and ICU, respectively (p=0.022). Patients who were admitted from ED displayed lower sLRP1 levels compared to those who were discharged (median sLRP1 levels were 0.86 versus 1.7 μg/mL, p=0.045). COVID-19 patients display significantly lower sLRP1 levels compared to the healthy controls. sLRP1 levels do not show any association with the clinical outcome of COVID-19. This study demonstrates that LRP1 displays a bidirectional course in COVID-19. A low sLRP1 level is a potential risk factor for susceptibility and hospital admission due to COVID-19. Further studies with larger sample sizes and longer follow-ups are needed to understand the long-term effects of novel biomarkers such as sLRP1 on the outcome of COVID-19.
Collapse
Affiliation(s)
- Mehmet Agirbasli
- Department of Cardiology, İstanbul Medeniyet University, Faculty of Medicine, Istanbul, TUR
| | - Rabia Korkmaz
- Department of Medical Biochemistry, Istanbul Medeniyet University, School of Medicine, Istanbul, TUR
| | - Ferruh K Isman
- Department of Medical Biochemistry, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, TUR
| |
Collapse
|
41
|
Lai M, Wang K, Ding C, Yin Y, Lin X, Xu C, Hu Z, Peng Z. Impact of inactivated COVID-19 vaccines on lung injury in B.1.617.2 (Delta) variant-infected patients. Ann Clin Microbiol Antimicrob 2023; 22:22. [PMID: 36944961 PMCID: PMC10029781 DOI: 10.1186/s12941-023-00569-z] [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: 08/27/2022] [Accepted: 02/19/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Chest computerized tomography (CT) scan is an important strategy that quantifies the severity of COVID-19 pneumonia. To what extent inactivated COVID-19 vaccines could impact the COVID-19 pneumonia on chest CT is not clear. METHODS This study recruited 357 SARS-COV-2 B.1.617.2 (Delta) variant-infected patients admitted to the Second Hospital of Nanjing from July to August 2021. An artificial intelligence-assisted CT imaging system was used to quantify the severity of COVID-19 pneumonia. We compared the volume of infection (VOI), percentage of infection (POI) and chest CT scores among patients with different vaccination statuses. RESULTS Of the 357 Delta variant-infected patients included for analysis, 105 were unvaccinated, 72 were partially vaccinated and 180 were fully vaccinated. Fully vaccination had the least lung injuries when quantified by VOI (median VOI of 222.4 cm3, 126.6 cm3 and 39.9 cm3 in unvaccinated, partially vaccinated and fully vaccinated, respectively; p < 0.001), POI (median POI of 7.60%, 3.55% and 1.20% in unvaccinated, partially vaccinated and fully vaccinated, respectively; p < 0.001) and chest CT scores (median CT score of 8.00, 6.00 and 4.00 in unvaccinated, partially vaccinated and fully vaccinated, respectively; p < 0.001). After adjustment for age, sex, comorbidity, time from illness onset to hospitalization and viral load, fully vaccination but not partial vaccination was significantly associated with less lung injuries quantified by VOI {adjust coefficient[95%CI] for "full vaccination": - 106.10(- 167.30,44.89); p < 0.001}, POI {adjust coefficient[95%CI] for "full vaccination": - 3.88(- 5.96, - 1.79); p = 0.001} and chest CT scores {adjust coefficient[95%CI] for "full vaccination": - 1.81(- 2.72, - 0.91); p < 0.001}. The extent of reduction of pulmonary injuries was more profound in fully vaccinated patients with older age, having underlying diseases, and being female sex, as demonstrated by relatively larger absolute values of adjusted coefficients. Finally, even within the non-severe COVID-19 population, fully vaccinated patients were found to have less lung injuries. CONCLUSION Fully vaccination but not partially vaccination could significantly protect lung injury manifested on chest CT. Our study provides additional evidence to encourage a full course of vaccination.
Collapse
Affiliation(s)
- Miao Lai
- School of Public Health, Nanjing Medical University, 101 Longmian Ave, Nanjing, 211166, China
| | - Kai Wang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Chengyuan Ding
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yi Yin
- School of Public Health, Nanjing Medical University, 101 Longmian Ave, Nanjing, 211166, China
| | - Xiaoling Lin
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chuanjun Xu
- Department of Radiology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, China.
| | - Zhiliang Hu
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, China.
| | - Zhihang Peng
- School of Public Health, Nanjing Medical University, 101 Longmian Ave, Nanjing, 211166, China.
| |
Collapse
|
42
|
The potential impact of COVID-19 disease caused multi-organ injuries on patients' surgical outcomes. ANESTHESIOLOGY AND PERIOPERATIVE SCIENCE 2023. [PMCID: PMC9998254 DOI: 10.1007/s44254-023-00004-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Abstract
Purpose
To provide an expert commentary on the impact of prior COVID-19 infection on patient’s surgical outcomes and postoperative recovery. To highlight the need for greater focus on peri-operative care of patients who have recovered from COVID-19.
Methods
A narrative review of the literature was conducted by searching Pubmed and EMBASE for relevant articles using keywords such as “COVID-19”, “Coronavirus”, “surgery” and “peri-operative infection”.
Results
Post-COVID-19 condition also known as long COVID has an estimated incidence of between 3.0 to 11.7%. COVID-19 has been shown to cause a series of short and long-term sequelae including cardiopulmonary complications, renal impairment, chronic fatigue and muscular deconditioning. Peri-operative infection with COVID-19 is associated with increased peri-operative mortality. Elective surgery patients who developed COVID-19 were 26 times more likely to die whilst in hospital compared to controls without COVID-19 infection, and for emergency surgery patients with COVID-19 infection were six times more likely to die. A large international prospective cohort study identified that patients who had surgery delayed over 7 weeks from the date of COVID-19 infection had no increased 30-day postoperative mortality, except those with ongoing symptoms.
Conclusions
COVID-19 infection and its complications have been shown to adversely affect surgical outcomes. Further research is required to better characterise long COVID and the long-term sequelae that develop, which should be used to guide comprehensive peri-operative assessment of patients.
Graphical Abstract
Collapse
|
43
|
Güven YZ, Kıratlı K, Kahraman HG, Akay F, Yurdakul ES. Evaluation of acute effects of pulmonary involvement and hypoxia on retina and choroid in coronavirus disease 2019: An optic coherence tomography study. Photodiagnosis Photodyn Ther 2023; 41:103265. [PMID: 36592784 PMCID: PMC9801694 DOI: 10.1016/j.pdpdt.2022.103265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/07/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022]
Abstract
PURPOSE We investigated the acute subclinical choroidal and retinal changes caused by Coronavirus Disease 2019 (COVID-19) in patients with and without pulmonary involvement, using spectral domain optic coherence tomography. METHODS This prospective case-control study included COVID-19 patients: 50 with pulmonary involvement and 118 with non-pulmonary involvement. All patients were examined 1 month after recovering from COVID-19. The changes were followed using optic coherence tomography parameters such as choroidal and macular thickness and retinal nerve fibre layer and ganglion cell complex measurements. RESULTS All choroidal thicknesses in the pulmonary involvement group were lower than in the non-pulmonary involvement group and the subfoveal choroidal thickness differed significantly (p=0.036). Although there were no significant differences between the central and average macular thicknesses in the two groups, they were slightly thicker in the pulmonary involvement group (p=0.152 and p=0.180, respectively). A significant decrease was detected in the pulmonary involvement group in all ganglion cell complex segments, except for the outer nasal inferior segment (p<0.05). In addition, a thinning tendency was observed in all retinal nerve fibre layer quadrants in the pulmonary involvement group compared to the non-pulmonary involvement group. CONCLUSION In COVID-19 patients with pulmonary involvement, subclinical choroidal and retinal changes may occur due to hypoxia and ischemia in the acute period. These patients may be predisposed to ischemic retinal and optic nerve diseases in the future. Therefore, COVID-19 patients with pulmonary involvement should be followed for ophthalmological diseases.
Collapse
Affiliation(s)
- Yusuf Ziya Güven
- İzmir Katip Çelebi University Atatürk Educating and Research Hospital, Department of Ophthalmology, İzmir 35200, Turkey.
| | - Kazım Kıratlı
- İzmir Katip Çelebi University Atatürk Educating and Research Hospital, Department of Infectious Diseases, İzmir, Turkey
| | - Hazan Gül Kahraman
- İzmir Katip Çelebi University Atatürk Educating and Research Hospital, Department of Ophthalmology, İzmir 35200, Turkey
| | - Fahrettin Akay
- University of Health Sciences, Gulhane School of Medicine, Department of Ophthalmology, Ankara, Turkey
| | - Eray Serdar Yurdakul
- University of Health Sciences, Gulhane School of Medicine, Department of Medical History and Bioethics, Ankara, Turkey
| |
Collapse
|
44
|
Zaaqoq A, Sallam T, Merley C, Galloway LA, Desale S, Varghese J, Alnababteh M, Kriner E, Kitahara H, Shupp J, Dalton H, Molina E. The interplay of inflammation and coagulation in COVID-19 Patients receiving extracorporeal membrane oxygenation support. Perfusion 2023; 38:384-392. [PMID: 35000466 PMCID: PMC9931882 DOI: 10.1177/02676591211057506] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Bleeding and thrombosis are common complications during Extracorporeal Membrane Oxygenation (ECMO) support for COVID-19 patients. We sought to examine the relationship between inflammatory status, coagulation effects, and observed bleeding and thrombosis in patients receiving venovenous (VV) ECMO for COVID-19 respiratory failure. STUDY DESIGN Cross-sectional cohort study. SETTINGS Quaternary care institution. PATIENTS The study period from April 1, 2020, to January 1, 2021, we included all patients with confirmed COVID-19 who received VV ECMO support. INTERVENTION None. MEASUREMENTS AND MAIN RESULTS Thirty-two patients were supported with VV ECMO during the study period, and 17 patients (53%) survived to hospital discharge. The ECMO nonsurvivors mean lactate dehydrogenase (LDH) levels were markedly elevated in comparison to survivors (1046 u/L [IQR = 509, 1305] vs 489 u/L [385 658], p = 0.003). Platelet/fibrinogen dysfunction, as reflected by the low Maximum Amplitude (MA) on viscoelastic testing, was worse in nonsurvivors (65.25 mm [60.68, 67.67] vs 74.80 mm [73.10, 78.40], p = 0.01). Time-group interaction for the first seven days of ECMO support, showed significantly lower platelet count in the nonsurvivors (140 k/ul [103, 170] vs 189.5 k/ul [ 146, 315], p < 0.001) and higher D-dimer in (21 μg/mL [13, 21] vs 14 μg/mL [3, 21], p < 0.001) in comparison to the survivors. Finally, we found profound statistically significant correlations between the clinical markers of inflammation and markers of coagulation in the nonsurvivors group. The ECMO nonsurvivors experienced higher rate of bleeding (73.3% vs 35.3%, p = 0.03), digital ischemia (46.7% vs 11.8%, p = 0.02), acute renal failure (60% vs 11.8%, p = 0.01) and bloodstream infection (60% vs 23.5%, p = 0.03). CONCLUSION The correlation between inflammation and coagulation in the nonsurvivors supported with VV ECMO could indicate dysregulated inflammatory response and worse clinical outcomes.
Collapse
Affiliation(s)
- Akram Zaaqoq
- Department of Critical Care
Medicine, MedStar Washington Hospital
Center, Georgetown University, Washington, DC, USA,Department of Medicine, MedStar Washington Hospital
Center, Georgetown University, Washington, DC, USA,Akram Zaaqoq, MD Department of Critical
Care Medicine Georgetown University 110 Irving St NW, suite 4B65 Washington, DC
20010, USA. E-mail:
| | - Tariq Sallam
- Department of Medicine, MedStar Washington Hospital
Center, Georgetown University, Washington, DC, USA
| | - Caitlin Merley
- Department of Medicine, University of
Pennsylvania, Philadelphia, PA, USA
| | - Lan Anh Galloway
- Department of Urology, Vanderbilt University, Nashville, TN, USA
| | - Sameer Desale
- MedStar Health Research
Institute, Hyattsville, MD, USA
| | - Jobin Varghese
- Department of Medicine, MedStar Washington Hospital
Center, Georgetown University, Washington, DC, USA
| | - Muhtadi Alnababteh
- Department of Medicine, MedStar Washington Hospital
Center, Georgetown University, Washington, DC, USA
| | - Eric Kriner
- Department of Critical Care
Medicine, MedStar Washington Hospital
Center, Georgetown University, Washington, DC, USA
| | - Hiroto Kitahara
- Department of Cardiac Surgery, MedStar Washington Hospital
Center, Georgetown University, Washington, DC, USA
| | - Jeffrey Shupp
- Department of Biochemistry and
Molecular & Cellular Biology, MedStar Washington Hospital
Center, Georgetown University Medical Center, Washington, DC,
USA,Department of Surgery, MedStar Washington Hospital Center
and MedStar Georgetown University Hospital, Washington, DC, USA
| | - Heidi Dalton
- Department of Pediatrics, Inova Fairfax Hospital, Virginia, USA, USA
| | - Ezequiel Molina
- Department of Cardiac Surgery, MedStar Washington Hospital
Center, Georgetown University, Washington, DC, USA
| |
Collapse
|
45
|
Yasmeen N, Selvaraj H, Lakhawat SS, Datta M, Sharma PK, Jain A, Khanna R, Srinivasan J, Kumar V. Possibility of averting cytokine storm in SARS-COV 2 patients using specialized pro-resolving lipid mediators. Biochem Pharmacol 2023; 209:115437. [PMID: 36731803 PMCID: PMC9884647 DOI: 10.1016/j.bcp.2023.115437] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023]
Abstract
Fatal "cytokine storms (CS)" observed in critically ill COVID-19 patients are consequences of dysregulated host immune system and over-exuberant inflammatory response. Acute respiratory distress syndrome (ARDS), multi-system organ failure, and eventual death are distinctive symptoms, attributed to higher morbidity and mortality rates among these patients. Consequent efforts to save critical COVID-19 patients via the usage of several novel therapeutic options are put in force. Strategically, drugs being used in such patients are dexamethasone, remdesivir, hydroxychloroquine, etc. along with the approved vaccines. Moreover, it is certain that activation of the resolution process is important for the prevention of chronic diseases. Until recently Inflammation resolution was considered a passive process, rather it's an active biochemical process that can be achieved by the use of specialized pro-resolving mediators (SPMs). These endogenous mediators are an array of atypical lipid metabolites that include Resolvins, lipoxins, maresins, protectins, considered as immunoresolvents, but their role in COVID-19 is ambiguous. Recent evidence from studies such as the randomized clinical trial, in which omega 3 fatty acid was used as supplement to resolve inflammation in COVID-19, suggests that direct supplementation of SPMs or the use of synthetic SPM mimetics (which are still being explored) could enhance the process of resolution by regulating the aberrant inflammatory process and can be useful in pain relief and tissue remodeling. Here we discussed the biosynthesis of SPMs, & their mechanistic pathways contributing to inflammation resolution along with sequence of events leading to CS in COVID-19, with a focus on therapeutic potential of SPMs.
Collapse
Affiliation(s)
- Nusrath Yasmeen
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Harikrishnan Selvaraj
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Sudarshan S Lakhawat
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Manali Datta
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Pushpender K Sharma
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Ajay Jain
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Rakhi Khanna
- Rajasthan State Regional Forensic Science Laboratory, Kota, Rajasthan, India
| | | | - Vikram Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India.
| |
Collapse
|
46
|
Symptomatic Popliteal Artery Aneurysms in Recently SARS-CoV-2-Infected Patients: The Microangiopathic Thrombosis That Undermines Treatment. Diagnostics (Basel) 2023; 13:diagnostics13040647. [PMID: 36832134 PMCID: PMC9955742 DOI: 10.3390/diagnostics13040647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/15/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Arterial and venous thrombosis are complications in SARS-CoV-2-infected patients. The microangiopathic thrombosis in affected patients can compromise results in urgent limb revascularizations. Aim of our study is to report on the incidence of symptoms development in patients affected by popliteal artery aneurysm (PAA) and to analyze the effect of COVID-19 infection on outcomes. METHODS Data on patients surgically treated for PAA from the massive widespread of COVID-19 vaccine (March 2021) to March 2022 were prospectively collected. Factors considered for analysis were: presence of symptoms, aneurysm diameter and length, time from symptom onset and hospital referral, ongoing or recently COVID-19 infection. Outcomes measures were: death, amputation, and neurological deficit. RESULTS Between March 2021 and March 2022, 35 patients were surgically treated for PAA. Among them 15 referred to our hospital for symptomatic PAA and were urgently treated. Urgent treatments included both endovascular procedures and open surgeries. Nine out of 15 symptomatic patients had an ongoing or recently recovered COVID-19 infection. COVID-19 infection was strongly associated to symptoms development in patients affected by PAA and to surgical failure in those patients (OR 40, 95% CI 2.01-794.31, p = 0.005). CONCLUSION In our series, presence of COVID-19 infection was strongly associated to ischemic symptoms onset and to complications after urgent treatment in symptomatic patients.
Collapse
|
47
|
Bowyer RCE, Huggins C, Toms R, Shaw RJ, Hou B, Thompson EJ, Kwong ASF, Williams DM, Kibble M, Ploubidis GB, Timpson NJ, Sterne JAC, Chaturvedi N, Steves CJ, Tilling K, Silverwood RJ. Characterising patterns of COVID-19 and long COVID symptoms: evidence from nine UK longitudinal studies. Eur J Epidemiol 2023; 38:199-210. [PMID: 36680646 PMCID: PMC9860244 DOI: 10.1007/s10654-022-00962-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 12/26/2022] [Indexed: 01/22/2023]
Abstract
Multiple studies across global populations have established the primary symptoms characterising Coronavirus Disease 2019 (COVID-19) and long COVID. However, as symptoms may also occur in the absence of COVID-19, a lack of appropriate controls has often meant that specificity of symptoms to acute COVID-19 or long COVID, and the extent and length of time for which they are elevated after COVID-19, could not be examined. We analysed individual symptom prevalences and characterised patterns of COVID-19 and long COVID symptoms across nine UK longitudinal studies, totalling over 42,000 participants. Conducting latent class analyses separately in three groups ('no COVID-19', 'COVID-19 in last 12 weeks', 'COVID-19 > 12 weeks ago'), the data did not support the presence of more than two distinct symptom patterns, representing high and low symptom burden, in each group. Comparing the high symptom burden classes between the 'COVID-19 in last 12 weeks' and 'no COVID-19' groups we identified symptoms characteristic of acute COVID-19, including loss of taste and smell, fatigue, cough, shortness of breath and muscle pains or aches. Comparing the high symptom burden classes between the 'COVID-19 > 12 weeks ago' and 'no COVID-19' groups we identified symptoms characteristic of long COVID, including fatigue, shortness of breath, muscle pain or aches, difficulty concentrating and chest tightness. The identified symptom patterns among individuals with COVID-19 > 12 weeks ago were strongly associated with self-reported length of time unable to function as normal due to COVID-19 symptoms, suggesting that the symptom pattern identified corresponds to long COVID. Building the evidence base regarding typical long COVID symptoms will improve diagnosis of this condition and the ability to elicit underlying biological mechanisms, leading to better patient access to treatment and services.
Collapse
Affiliation(s)
- Ruth C E Bowyer
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, UK
- The Alan Turing Institute, London, UK
| | - Charlotte Huggins
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Renin Toms
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Population Wellbeing, School of Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - Richard J Shaw
- MRC/CSO Social and Public Health Sciences Unit, University of Glasgow, Glasgow, UK
| | - Bo Hou
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Ellen J Thompson
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, UK
| | - Alex S F Kwong
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Dylan M Williams
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Milla Kibble
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, UK
| | - George B Ploubidis
- Centre for Longitudinal Studies, UCL Social Research Institute, University College London, London, UK
| | - Nicholas J Timpson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
| | - Jonathan A C Sterne
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, Bristol, UK
- Health Data Research UK South West, Bristol, UK
| | - Nishi Chaturvedi
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - Claire J Steves
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, UK
- Department of Ageing and Health, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Kate Tilling
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
| | - Richard J Silverwood
- Centre for Longitudinal Studies, UCL Social Research Institute, University College London, London, UK.
| |
Collapse
|
48
|
Su B, Wang Y, Jian S, Tang H, Deng H, Zhu L, Zhao X, Liu J, Cheng H, Zhang L, Hu Y, Xu Z. In vitro and in vivo antiviral activity of monolaurin against Seneca Valley virus. Front Vet Sci 2023; 10:980187. [PMID: 36777661 PMCID: PMC9911909 DOI: 10.3389/fvets.2023.980187] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
Introduction Surveillance of the Seneca Valley virus (SVV) shows a disproportionately higher incidence on Chinese pig farms. Currently, there are no vaccines or drugs to treat SVV infection effectively and effective treatment options are urgently needed. Methods In this study, we evaluated the antiviral activity of the following medium-chain fatty acids (MCFAs) or triglycerides (MCTs) against SVV: caprylic acid, caprylic monoglyceride, capric monoglyceride, and monolaurin. Results In vitro experiments showed that monolaurin inhibited viral replication by up to 80%, while in vivo studies showed that monolaurin reduced clinical manifestations, viral load, and organ damage in SVV-infected piglets. Monolaurin significantly reduced the release of inflammatory cytokines and promoted the release of interferon-γ, which enhanced the viral clearance activity of this type of MCFA. Discussion Therefore, monolaurin is a potentially effective candidate for the treatment of SVV infection in pigs.
Collapse
Affiliation(s)
- Bo Su
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Yingjie Wang
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Shanqiu Jian
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Huaqiao Tang
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Huidan Deng
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Ling Zhu
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Xiaonan Zhao
- Innovation Center of Guangdong Nuacid Biotechnology Co., Ltd., Qingyuan, China
| | - Jian Liu
- Innovation Center of Guangdong Nuacid Biotechnology Co., Ltd., Qingyuan, China
| | - Huangzuo Cheng
- Innovation Center of Guangdong Nuacid Biotechnology Co., Ltd., Qingyuan, China
| | - Lina Zhang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Youjun Hu
- Innovation Center of Guangdong Nuacid Biotechnology Co., Ltd., Qingyuan, China,*Correspondence: Youjun Hu ✉
| | - Zhiwen Xu
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China,Zhiwen Xu ✉
| |
Collapse
|
49
|
Donniacuo M, De Angelis A, Rafaniello C, Cianflone E, Paolisso P, Torella D, Sibilio G, Paolisso G, Castaldo G, Urbanek K, Rossi F, Berrino L, Cappetta D. COVID-19 and atrial fibrillation: Intercepting lines. Front Cardiovasc Med 2023; 10:1093053. [PMID: 36755799 PMCID: PMC9899905 DOI: 10.3389/fcvm.2023.1093053] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Almost 20% of COVID-19 patients have a history of atrial fibrillation (AF), but also a new-onset AF represents a frequent complication in COVID-19. Clinical evidence demonstrates that COVID-19, by promoting the evolution of a prothrombotic state, increases the susceptibility to arrhythmic events during the infective stages and presumably during post-recovery. AF itself is the most frequent form of arrhythmia and is associated with substantial morbidity and mortality. One of the molecular factors involved in COVID-19-related AF episodes is the angiotensin-converting enzyme (ACE) 2 availability. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses ACE2 to enter and infect multiple cells. Atrial ACE2 internalization after binding to SARS-CoV-2 results in a raise of angiotensin (Ang) II, and in a suppression of cardioprotective Ang(1-7) formation, and thereby promoting cardiac hypertrophy, fibrosis and oxidative stress. Furthermore, several pharmacological agents used in COVID-19 patients may have a higher risk of inducing electrophysiological changes and cardiac dysfunction. Azithromycin, lopinavir/ritonavir, ibrutinib, and remdesivir, used in the treatment of COVID-19, may predispose to an increased risk of cardiac arrhythmia. In this review, putative mechanisms involved in COVID-19-related AF episodes and the cardiovascular safety profile of drugs used for the treatment of COVID-19 are summarized.
Collapse
Affiliation(s)
- Maria Donniacuo
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Concetta Rafaniello
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Pasquale Paolisso
- Cardiovascular Center Aalst, OLV Hospital, Aalst, Belgium
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | | | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giuseppe Castaldo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| |
Collapse
|
50
|
Nejat R, Torshizi MF, Najafi DJ. S Protein, ACE2 and Host Cell Proteases in SARS-CoV-2 Cell Entry and Infectivity; Is Soluble ACE2 a Two Blade Sword? A Narrative Review. Vaccines (Basel) 2023; 11:204. [PMID: 36851081 PMCID: PMC9968219 DOI: 10.3390/vaccines11020204] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/07/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Since the spread of the deadly virus SARS-CoV-2 in late 2019, researchers have restlessly sought to unravel how the virus enters the host cells. Some proteins on each side of the interaction between the virus and the host cells are involved as the major contributors to this process: (1) the nano-machine spike protein on behalf of the virus, (2) angiotensin converting enzyme II, the mono-carboxypeptidase and the key component of renin angiotensin system on behalf of the host cell, (3) some host proteases and proteins exploited by SARS-CoV-2. In this review, the complex process of SARS-CoV-2 entrance into the host cells with the contribution of the involved host proteins as well as the sequential conformational changes in the spike protein tending to increase the probability of complexification of the latter with angiotensin converting enzyme II, the receptor of the virus on the host cells, are discussed. Moreover, the release of the catalytic ectodomain of angiotensin converting enzyme II as its soluble form in the extracellular space and its positive or negative impact on the infectivity of the virus are considered.
Collapse
Affiliation(s)
- Reza Nejat
- Department of Anesthesiology and Critical Care Medicine, Laleh Hospital, Tehran 1467684595, Iran
| | | | | |
Collapse
|