1
|
Long JP, Prakash R, Edelkamp P, Knafl M, Lionel AC, Nair R, Ahmed S, Strati P, Castillo LEM, Al-Zaki A, Chien K, Chihara D, Westin J, Khawaja F, Nastoupil LJ, Mulanovich V, Futreal A, Woodman SE, Daver NG, Flowers CR, Neelapu S, Manzano JG, Iyer SP. Cytokine Storms in COVID-19, Hemophagocytic Lymphohistiocytosis, and CAR-T Therapy. JAMA Netw Open 2025; 8:e253455. [PMID: 40193078 PMCID: PMC11976493 DOI: 10.1001/jamanetworkopen.2025.3455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/27/2024] [Indexed: 04/10/2025] Open
Abstract
Importance Cytokine storm (CS) is a hyperinflammatory syndrome causing multiorgan dysfunction and high mortality, especially in patients with malignant hematologic neoplasms. Triggers include malignant neoplasm-associated hemophagocytic lymphohistiocytosis (MN-HLH), cytokine release syndrome from chimeric antigen receptor T-cell therapy (CAR-T CRS), and COVID-19, but the underlying mechanisms of inflammation and their impact on outcomes are poorly understood. Objective To delineate the inflammatory patterns characterizing different CS etiologies and their association with clinical outcomes. Design, Setting, and Participants This retrospective cohort study was conducted at the MD Anderson Cancer Center in Houston, Texas, between March 1, 2020, and November 20, 2022, using the software-as-a-service Syntropy Foundry Platform. Participants were patients with malignant hematologic neoplasms who developed CS from COVID-19 (COVID-CS), MN-HLH, or CAR-T CRS. Exposure Diagnostic criteria for COVID-CS were developed based on surging inflammatory markers (interleukin-6, C-reactive protein, and ferritin), while diagnosis of MN-HLH and CAR-T CRS followed established guidelines. Main Outcomes and Measures The study compared cytokine levels, clinical characteristics, and survival outcomes across the 3 cohorts and focused on inflammatory markers, survival times, and key factors associated with survival identified through univariate and multivariable analyses. Results A total of 671 patients met the inclusion criteria. Of those, 220 (33%) had CAR-T CRS, 227 (34%) had COVID-CS, and 224 (33%) had MN-HLH. Patients were predominantly male (435 [65%]), and 461 (69%) were White, with significant differences in median age (CAR-T CRS, 63 [IQR, 54-71] years; COVID-CS, 63 [IQR, 52-72] years; MN-HLH, 55 [IQR, 41-65] years; P < .001) as well as number of admission days and underlying cancer type across cohorts. Marked variations in cytokine levels and survival outcomes were observed, with the MN-HLH cohort exhibiting the highest levels of inflammatory markers (eg, median TNF-α, 105 pg/mL [IQR, 38-201 pg/mL] for MN-HLH vs 23 pg/mL [IQR, 17-42 pg/mL] for COVID-CS) and lowest fibrinogen and albumin levels. The cohort with CAR-T CRS showed substantially longer survival times compared with the cohort with COVID-CS (hazard ratio [HR], 2.93; 95% CI, 1.95-4.41) and the cohort with MN-HLH (HR, 8.12; 95% CI, 5.51-12.00). Clustering analysis showed overlapping patterns between COVID-CS and CAR-T CRS, while MN-HLH formed a distinct cluster. Conclusions and Relevance This study of CS syndromes found distinct immune responses within each cohort. The distinct clinical patterns and outcomes associated with different CS etiologies emphasize the importance of early diagnosis and timely intervention.
Collapse
Affiliation(s)
- James P. Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Rishab Prakash
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Paul Edelkamp
- Department of Enterprise Data Engineering & Analytics, The University of Texas MD Anderson Cancer Center, Houston
| | - Mark Knafl
- Department of Enterprise Data Engineering & Analytics, The University of Texas MD Anderson Cancer Center, Houston
| | - Anath C. Lionel
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Ranjit Nair
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Sairah Ahmed
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Paolo Strati
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Luis E. Malpica Castillo
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Ajlan Al-Zaki
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Kelly Chien
- Department of Leukemia, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Dai Chihara
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Jason Westin
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Fareed Khawaja
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston
| | - Loretta J. Nastoupil
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Victor Mulanovich
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Scott E. Woodman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Naval G. Daver
- Department of Leukemia, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Christopher R. Flowers
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Sattva Neelapu
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Joanna-Grace Manzano
- Department of Hospital Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Swaminathan P. Iyer
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
2
|
Begue F, Veeren B, Rondeau P, Florence AM, Jamard S, Montravers P, Tanaka S, Meilhac O. HDL proteome and apolipoproteins concentrations in severe ICU COVID-19 patients. Lipids Health Dis 2025; 24:32. [PMID: 39891286 PMCID: PMC11783863 DOI: 10.1186/s12944-024-02381-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: 08/26/2024] [Accepted: 11/18/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND SARS-CoV-2 infection affects both lipid metabolism and lung function. The severity of the disease has been associated with reduced levels of both high-density lipoprotein (HDL) and low-density lipoprotein cholesterol. Despite the crucial role that these nanoparticles play in SARS-CoV-2 infection, few studies have examined their structure during COVID-19 beyond HDL quantity. The study aimed to assess apolipoprotein levels in COVID-19 patients who either survived or died following ICU admission. In addition, ICU survivors and non-survivors were compared for HDL particle size and proteome. METHODS Between February and April 2020, our study enrolled 37 COVID-19 patients upon their intensive care unit admission. Among them, 18 survived the disease, while 19 succumbed to it. We used mass spectrometry to assess plasma levels of 14 apolipoproteins and LCAT. Additionally, we analyzed HDL subpopulation distribution by utilizing native polyacrylamide gel electrophoresis. HDL particles were isolated from both surviving and non-surviving patients using ultracentrifugation, followed by characterization of their proteomes with NanoLC-MS/MS. RESULTS Plasma apolipoproteins, including Apo A-II, Apo Cs (I, II, III), Apo H, Apo J, Apo M, and LCAT, were decreased in patients who did not survive COVID-19. However, no alterations were noted in the distribution of HDL subpopulations in relation to mortality. HDL composition was further altered based on mortality, displaying a decline in Apo H and paraoxonase 3. CONCLUSION In conclusion, we have shown an alteration in plasma apolipoproteins and HDL composition between surviving COVID-19 patients and non-survivors. Some markers, such as Apo H, are more predictive than baseline lipid concentrations such as HDL-C. These markers appear to provide a more accurate indication of mortality during COVID-19 compared with baseline lipid concentrations such as HDL-C.
Collapse
Affiliation(s)
- Floran Begue
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), Saint-Pierre de La Réunion, 97410, France
- Délégation de la Recherche Clinique et de l'Innovation, USMD, CHU Réunion, Saint-Pierre, 97448, France
| | - Bryan Veeren
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), Saint-Pierre de La Réunion, 97410, France
| | - Philippe Rondeau
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), Saint-Pierre de La Réunion, 97410, France
| | - Aline-Marie Florence
- INSERM IAME UMR 1137, Université Paris Cité, Paris, France
- Department of Epidemiology Biostatistics and Clinical Research, Assistance Publique-Hopitaux de Paris (AP-HP) Nord, Hopital Bichat, Paris, France
| | - Simon Jamard
- Department of Infectious Disease, University Hospital of Tours, Tours, France
| | - Philippe Montravers
- Department of Anesthesiology and Critical Care Medicine, Assistance, AP-HP, Bichat-Claude Bernard Hospital, Paris, France
- University of Paris, UFR Denis Diderot, Paris, France
- PHERE, Physiopathology and Epidemiology of Respiratory Diseases, INSERM U1152, Paris, France
| | - Sébastien Tanaka
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), Saint-Pierre de La Réunion, 97410, France
- Department of Anesthesiology and Critical Care Medicine, Assistance, AP-HP, Bichat-Claude Bernard Hospital, Paris, France
| | - Olivier Meilhac
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), Saint-Pierre de La Réunion, 97410, France.
- CHU de La Réunion, Saint-Pierre, 97410, France.
| |
Collapse
|
3
|
Corona A, Simoncini S, Richini G, Gatti I, Santorsola C, Patroni A, Tomasini G, Capone A, Zendra E, Shuman M. Ig-M and Ig-A Enriched Ig-G Infusion as Adjuvant Therapy in the Critically ill Patients Experiencing SARS-CoV-2 Severe Infection. J Intensive Care Med 2024:8850666241301689. [PMID: 39648609 DOI: 10.1177/08850666241301689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
INTRODUCTION SARS-CoV-2 in patients who need Intensive Care (ICU) is associated with a mortality rate ranging from 10 to 40%-45%, with an increase in morbidity and mortality in presence of sepsis. METHODS We assumed that immunoglobulin (Ig) M and IgA enriched IgG (IGAM) therapy may support SARS COV-2 sepsis-related phase improving patient outcome. We conducted a retrospective case-control study on all the patients admitted to our ICU during the three pandemic waves between February 2020 and April 2021. Upon ICU admission, patients received anticoagulants with the standard supportive treatment (ST) ± IGAM therapy. After matching for the baseline characteristics and treatments, the patients receiving IGAM therapy too (group A), were compared with those undergoing ST (group B) only. RESULTS 85 patients were enrolled in group A, whereas 111 in group B. The mortality resulted lower in group A [37.6% versus 55.8%, OR: 0.7 (02-08), P = .01)]. A logistic regression analysis identified IGAM treatment as a survival predictor [OR: 0.35 (95%CI, 0.2-0.8)], whereas experiencing a super-infection [OR: 1.88 (95%CI, 1.5-4.9)] and a septic shock [OR: 1.92 (95%CI, 1.4-4.3)] as predictors of death. On day 7, the probability of dying was 3 times higher in patients treated with ST only. Variable life adjustment display (VLAD) was equal to 2.4 in group A, while - 2.2 group B (in terms of lives saved in relation with those expected, in according with Simplified Acute Physiology Score II (SAPS II) score. CONCLUSION The treatment based on IGAM infusion seems to give an advantage chance of survival in SARS-CoV-2 severe infection. Further prospective studies are warranted.
Collapse
Affiliation(s)
- Alberto Corona
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Sara Simoncini
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Giuseppe Richini
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Ivan Gatti
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Clemente Santorsola
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Andrea Patroni
- Medical Directorate, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Giacomina Tomasini
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Alice Capone
- ICU, Anaesthesia and Emergency Department, ASST Spedali Civili di Brescia, Brescia (BS), Italy
| | - Elena Zendra
- ICU, Anaesthesia and Emergency Department, ASST Spedali Civili di Brescia, Brescia (BS), Italy
| | - Myriam Shuman
- Department of Anaesthesiology, Pain Medicine and Perioperative Care, University of Washington, Seattle, Washington, USA
| |
Collapse
|
4
|
Bi Q, Zhu J, Zheng J, Xu Q, Chen J, Zhang L, Mu X. Blood Inflammatory Markers and Cytokines in COVID-19 Patients With Bacterial Coinfections. Immun Inflamm Dis 2024; 12:e70105. [PMID: 39692539 PMCID: PMC11653711 DOI: 10.1002/iid3.70105] [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: 03/05/2024] [Revised: 11/16/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Bacterial coinfection in patients with SARS-CoV-2 infection is an important risk factor for death. This study investigated whether there were differences in levels of serum inflammatory markers in COVID-19 patients with bacterial coinfections compared with those without bacterial infection. METHODS A total of 235 inpatients with SARS-CoV-2 infection admitted to Qingdao Central Hospital from December 7, 2022, to August 7, 2024, were included. Patients were divided into a bacteria-positive group (115 cases) and a bacteria-negative group (120 cases) according to whether they had bacterial coinfections. PCT, CRP, and 12 kinds of cytokines were compared between groups, and the distribution of bacterial species in the positive group was statistically analyzed. RESULTS The serum levels of CRP (Z = 8.94, p < 0.001), PCT (Z = 5.59, p < 0.001), IL-1β (t = 4.863, p < 0.001), IL-2 (t = 5.810, p < 0.001), IL-5 (t = 3.837, p < 0.001), IL-6 (t = 4.910, p < 0.001), IL-8 (t = 3.325, p < 0.001), ILIL-12p70 (t = 4.722, p < 0.001), IL-17 (t = 3.315, p = 0.001) and TNF-α (t = 4.251, p < 0.001) between the two groups were significantly different. IL-4, IL-10, IFN-α, and IFN-γ were not statistically significant (p > 0.05). Among the 115 bacteria-positive patients, 56 patients were positive for one species and 59 patients were multiple infections. Acinetobacter baumannii, Klebsiella pneumoniae, Pseudomonas aeruginosa, Staphylococcus aureus, and Haemophilus influenzae were common species. CONCLUSIONS Serum PCT and CRP levels in COVID-19 patients with bacterial coinfection are higher than those without bacterial infection. Cytokines such as IL-1β, IL-2, IL-5, IL-6, IL-8, IL-12p70, IL-17, and TNF-α may be involved in the progression of COVID-19 combined with bacterial infection. They can be used as potential markers to evaluate the disease condition and prognosis.
Collapse
Affiliation(s)
- Qingqing Bi
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| | - Jie Zhu
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| | - Jinju Zheng
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| | - Qingyun Xu
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
- Department of Clinical LaboratoryPeking University First HospitalBeijingChina
| | - Juan Chen
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| | - Lei Zhang
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| | - Xiaofeng Mu
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| |
Collapse
|
5
|
Durán-Sotuela A, Vázquez-García J, Relaño-Fernández S, Balboa-Barreiro V, Fernández-Tajes J, Blanco FJ, Rego-Pérez I. An exploratory analysis of associations of genetic variation with the efficacy of tocilizumab in severe COVID-19 patients. A pharmacogenetic study based on next-generation sequencing. Front Pharmacol 2024; 15:1426826. [PMID: 39346556 PMCID: PMC11428153 DOI: 10.3389/fphar.2024.1426826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/26/2024] [Indexed: 10/01/2024] Open
Abstract
Background In the context of the cytokine storm the takes place in severe COVID-19 patients, the Interleukin 6 (IL6) pathway emerges as one of the key pathways involved in the pathogenesis of this hyperinflammatory state. The strategy of blocking the inflammatory storm by targeting the IL6 is a promising therapy to mitigate mortality. The use of Tocilizumab was recommended by the World Health Organization (WHO) to treat severe COVID-19 patients. However, the efficacy of Tocilizumab is variable. We hypothesize that the genetic background could be behind the efficacy of Tocilizumab in terms of mortality. Methods We performed a targeted-next generation sequencing of 287 genes, of which 264 belong to a community panel of ThermoFisher for the study of genetic causes of primary immunodeficiency disorders, and 23 additional genes mostly related to inflammation, not included in the original community panel. This panel was sequenced in an initial cohort of 425 COVID-19 patients, of which 232 were treated with Tocilizumab and standard therapy, and 193 with standard therapy only. Selected genetic variants were genotyped by single base extension in additional 245 patients (95 treated with Tocilizumab and 150 non-treated with Tocilizumab). Appropriate statistical analyses and internal validation, including logistic regression models, with the interaction between Tocilizumab and genetic variants, were applied to assess the impact of these genetic variants in the efficacy of Tocilizumab in terms of mortality. Results Age (p < 0.001) and cardiovascular disease (p < 0.001) are risk factors for mortality in COVID-19 patients. The presence of GG and TT genotypes at IL10Rβ (rs2834167) and IL1β (rs1143633) genes significantly associates with a reduced risk of mortality in patients treated with Tocilizumab (OR = 0.111; 95%CI = 0.015-0.829; p = 0.010 and OR = 0.378; 95%CI = 0.154-0.924; p = 0.028 respectively). The presence of CC genotype at IL1RN (rs2234679) significantly associates with an increased risk of mortality, but only in patients not treated with Tocilizumab (OR = 3.200; 95%CI = 1.512-6.771; p = 0.002). Exhaustive internal validation using a bootstrap method (B = 500 replicates) validated the accuracy of the predictive models. Conclusion We developed a series of predictive models based on three genotypes in genes with a strong implication in the etiopathogenesis of COVID-19 disease capable of predicting the risk of mortality in patients treated with Tocilizumab.
Collapse
Affiliation(s)
- Alejandro Durán-Sotuela
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC) Sergas, Universidade da Coruña (UDC), A Coruña, Spain
| | - Jorge Vázquez-García
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC) Sergas, Universidade da Coruña (UDC), A Coruña, Spain
| | - Sara Relaño-Fernández
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC) Sergas, Universidade da Coruña (UDC), A Coruña, Spain
| | - Vanesa Balboa-Barreiro
- Unidad de Apoyo a La Investigación, Grupo de Investigación en Enfermería y Cuidados en Salud, Grupo de Investigación en Reumatología y Salud (GIR-S), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC) Sergas, Universidade da Coruña (UDC), A Coruña, Spain
| | - Juan Fernández-Tajes
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC) Sergas, Universidade da Coruña (UDC), A Coruña, Spain
| | - Francisco J. Blanco
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC) Sergas, Universidade da Coruña (UDC), A Coruña, Spain
- Universidade da Coruña (UDC), Centro de Investigación de Ciencias Avanzadas (CICA), Grupo de Investigación en Reumatología y Salud (GIR-S), Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Campus de Oza, A Coruña, Spain
| | - Ignacio Rego-Pérez
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC) Sergas, Universidade da Coruña (UDC), A Coruña, Spain
| |
Collapse
|
6
|
Qu Y, Chu B, Li J, Deng H, Niu T, Qian Z. Macrophage-Biomimetic Nanoplatform-Based Therapy for Inflammation-Associated Diseases. SMALL METHODS 2024; 8:e2301178. [PMID: 38037521 DOI: 10.1002/smtd.202301178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/23/2023] [Indexed: 12/02/2023]
Abstract
Inflammation-associated diseases are very common clinically with a high incidence; however, there is still a lack of effective treatments. Cell-biomimetic nanoplatforms have led to many breakthroughs in the field of biomedicine, significantly improving the efficiency of drug delivery and its therapeutic implications especially for inflammation-associated diseases. Macrophages are an important component of immune cells and play a critical role in the occurrence and progression of inflammation-associated diseases while simultaneously maintaining homeostasis and modulating immune responses. Therefore, macrophage-biomimetic nanoplatforms not only inherit the functions of macrophages including the inflammation tropism effect for targeted delivery of drugs and the neutralization effect of pro-inflammatory cytokines and toxins via membrane surface receptors or proteins, but also maintain the functions of the inner nanoparticles. Macrophage-biomimetic nanoplatforms are shown to have remarkable therapeutic efficacy and excellent application potential in inflammation-associated diseases. In this review, inflammation-associated diseases, the physiological functions of macrophages, and the classification and construction of macrophage-biomimetic nanoplatforms are first introduced. Next, the latest applications of different macrophage-biomimetic nanoplatforms for the treatment of inflammation-associated diseases are summarized. Finally, challenges and opportunities for future biomedical applications are discussed. It is hoped that the review will provide new ideas for the further development of macrophage-biomimetic nanoplatforms.
Collapse
Affiliation(s)
- Ying Qu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bingyang Chu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianan Li
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hanzhi Deng
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
7
|
Shohan M, Mahmoudian-Sani MR, Saeedi-Boroujeni A, Iranparast S, Nashibi R, Abolnezhadian F, Yousefi F, Alavi SM, Cheraghian B, Khodadadi A. The Effects of Convalescent Plasma Transfusion on Serum Levels of Macrophage-Associated Inflammatory Biomarkers in Patients with Severe COVID-19. J Interferon Cytokine Res 2024; 44:316-324. [PMID: 38738802 DOI: 10.1089/jir.2024.0018] [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: 05/14/2024] Open
Abstract
As an antibody-based therapy, plasma therapy has been used as an emergency therapeutic strategy against severe acute respiratory syndrome coronavirus type 2 infection. Due to the critical role of macrophages in coronavirus disease-19 (COVID-19)-associated hyperinflammation, the main objective of this study was to assess the effect of plasma transfusion on the expression levels of the inflammatory biomarkers involved in activation and pulmonary infiltration of macrophages. The target population included 50 severe hospitalized COVID-19 patients who were randomly assigned into 2 groups, including intervention and control. Serum levels of chemokine (C-C motif) ligand (CCL)-2, CCL-3, tumor necrosis factor (TNF)-α, and interleukin (IL)-6 were measured by enzyme-linked immunosorbent assay. Moreover, quantitative real-time polymerase chain reaction (PCR) was carried out to assess the relative expression of nuclear factor (NF)-κB1, NF-κB2, nuclear factor erythroid 2 p45-related factor 2 (NRF-2), and thioredoxin-interacting protein genes. Sampling was done at baseline and 72 h after receiving plasma. The intervention group demonstrated significantly lower serum levels of IL-6, TNF-α, and CCL-3. In addition, real-time PCR data analyses showed that the relative expression of NF-κB2 was significantly declined in the patients who received plasma. The use of convalescent plasma probably has a significant inhibitory effect on the cytokines, chemokines, and inflammatory genes related to macrophage activation, which are closely associated with the worsening of clinical outcomes in severe COVID-19.
Collapse
Affiliation(s)
- Mojtaba Shohan
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Reza Mahmoudian-Sani
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Saeedi-Boroujeni
- Department of Basic Medical Sciences, Faculty of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| | - Sara Iranparast
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Roohangiz Nashibi
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farhad Abolnezhadian
- Department of Pediatrics, Abuzar children's hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farid Yousefi
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Mohammad Alavi
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bahman Cheraghian
- Department of Biostatistics and Epidemiology, School of Public Health, Alimentary Tract Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Khodadadi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cancer, Petroleum, and Environmental pollutants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
8
|
Iancu IV, Diaconu CC, Plesa A, Fudulu A, Albulescu A, Neagu AI, Pitica IM, Dragu LD, Bleotu C, Chivu‐Economescu M, Matei L, Mambet C, Nedeianu S, Popescu CP, Sultana C, Ruta SM, Botezatu A. LncRNAs expression profile in a family household cluster of COVID-19 patients. J Cell Mol Med 2024; 28:e18226. [PMID: 38501860 PMCID: PMC10949602 DOI: 10.1111/jcmm.18226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/19/2024] [Accepted: 02/24/2024] [Indexed: 03/20/2024] Open
Abstract
More than 3 years after the start of SARS-CoV-2 pandemic, the molecular mechanisms behind the viral pathogenesis are still not completely understood. Long non-coding RNAs (lncRNAs), well-known players in viral infections, can represent prime candidates for patients' risk stratification. The purpose of the current study was to investigate the lncRNA profile in a family cluster of COVID-19 cases with different disease progression, during the initial wave of the pandemic and to evaluate their potential as biomarkers for COVID-19 evolution. LncRNA expression was investigated in nasopharyngeal swabs routinely collected for diagnosis. Distinct expression patterns of five lncRNAs (HOTAIR, HOTAIRM1, TMEVPG1, NDM29 and snaR) were identified in all the investigated cases, and they were associated with disease severity. Additionally, a significant increase in the expression of GAS5-family and ZFAS1 lncRNAs, which target factors involved in the inflammatory response, was observed in the sample collected from the patient with the most severe disease progression. An lncRNA prognostic signature was defined, opening up novel research avenues in understanding the interactions between lncRNAs and SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Adriana Plesa
- Stefan S Nicolau Institute of VirologyBucharestRomania
| | - Alina Fudulu
- Stefan S Nicolau Institute of VirologyBucharestRomania
| | - Adrian Albulescu
- Stefan S Nicolau Institute of VirologyBucharestRomania
- Department of PharmacologyNational Institute for Chemical Pharmaceutical Research and DevelopmentBucharestRomania
| | - Ana Iulia Neagu
- Stefan S Nicolau Institute of VirologyBucharestRomania
- Carol Davila University of Medicine and PharmacyBucharestRomania
| | | | | | | | | | - Lilia Matei
- Stefan S Nicolau Institute of VirologyBucharestRomania
| | | | | | - Corneliu Petru Popescu
- Carol Davila University of Medicine and PharmacyBucharestRomania
- Dr Victor Babes Infectious and Tropical Diseases Clinical HospitalBucharestRomania
| | - Camelia Sultana
- Stefan S Nicolau Institute of VirologyBucharestRomania
- Carol Davila University of Medicine and PharmacyBucharestRomania
| | - Simona Maria Ruta
- Stefan S Nicolau Institute of VirologyBucharestRomania
- Carol Davila University of Medicine and PharmacyBucharestRomania
| | - Anca Botezatu
- Stefan S Nicolau Institute of VirologyBucharestRomania
| |
Collapse
|
9
|
Zheng Y, Li Y, Li M, Wang R, Jiang Y, Zhao M, Lu J, Li R, Li X, Shi S. COVID-19 cooling: Nanostrategies targeting cytokine storm for controlling severe and critical symptoms. Med Res Rev 2024; 44:738-811. [PMID: 37990647 DOI: 10.1002/med.21997] [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: 06/04/2022] [Revised: 08/16/2023] [Accepted: 10/29/2023] [Indexed: 11/23/2023]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants continue to wreak havoc worldwide, the "Cytokine Storm" (CS, also known as the inflammatory storm) or Cytokine Release Syndrome has reemerged in the public consciousness. CS is a significant contributor to the deterioration of infected individuals. Therefore, CS control is of great significance for the treatment of critically ill patients and the reduction of mortality rates. With the occurrence of variants, concerns regarding the efficacy of vaccines and antiviral drugs with a broad spectrum have grown. We should make an effort to modernize treatment strategies to address the challenges posed by mutations. Thus, in addition to the requirement for additional clinical data to monitor the long-term effects of vaccines and broad-spectrum antiviral drugs, we can use CS as an entry point and therapeutic target to alleviate the severity of the disease in patients. To effectively combat the mutation, new technologies for neutralizing or controlling CS must be developed. In recent years, nanotechnology has been widely applied in the biomedical field, opening up a plethora of opportunities for CS. Here, we put forward the view of cytokine storm as a therapeutic target can be used to treat critically ill patients by expounding the relationship between coronavirus disease 2019 (COVID-19) and CS and the mechanisms associated with CS. We pay special attention to the representative strategies of nanomaterials in current neutral and CS research, as well as their potential chemical design and principles. We hope that the nanostrategies described in this review provide attractive treatment options for severe and critical COVID-19 caused by CS.
Collapse
Affiliation(s)
- Yu Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mao Li
- Health Management Centre, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Rujing Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Su P, Jiang C, Zhang Y. The implication of infection with respiratory syncytial virus in pediatric recurrent wheezing and asthma: knowledge expanded post-COVID-19 era. Eur J Clin Microbiol Infect Dis 2024; 43:403-416. [PMID: 38153660 DOI: 10.1007/s10096-023-04744-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/21/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection has been identified to serve as the primary cause of acute lower respiratory infectious diseases in children under the age of one and a significant risk factor for the emergence and development of pediatric recurrent wheezing and asthma, though the exact mechanism is still unknown. METHODS AND RESULTS In this study, we discuss the key routes that lead to recurrent wheezing and bronchial asthma following RSV infection. It is interesting to note that following the coronavirus disease 2019 (COVID-19) epidemic, the prevalence of RSV changes significantly. This presents us with a rare opportunity to better understand the associated mechanism for RSV infection, its effects on the respiratory system, and the immunological response to RSV following the COVID-19 epidemic. To better understand the associated mechanisms in the occurrence and progression of pediatric asthma, we thoroughly described how the RSV infection directly destroys the physical barrier of airway epithelial tissue, promotes inflammatory responses, enhances airway hyper-responsiveness, and ultimately causes the airway remodeling. More critically, extensive discussion was also conducted regarding the potential impact of RSV infection on host pulmonary immune response. CONCLUSION In conclusion, this study offers a comprehensive perspective to better understand how the RSV infection interacts in the control of the host's pulmonary immune system, causing recurrent wheezing and the development of asthma, and it sheds fresh light on potential avenues for pharmaceutical therapy in the future.
Collapse
Affiliation(s)
- Peipei Su
- Xi'an Medical University, Xi'an, 710068, Shaanxi, China
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, National Regional Children's Medical Centre (Northwest), Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
| | - Congshan Jiang
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, National Regional Children's Medical Centre (Northwest), Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
| | - Yanmin Zhang
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, National Regional Children's Medical Centre (Northwest), Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China.
- Department of Cardiology, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China.
| |
Collapse
|
11
|
Kahrizi MS, Nasiri K, Ebrahimzadeh F, Yaseri AF, Ghodratizadeh S, Gholamrezaei M, Rahat Dahmardeh A, Adili A, Amjidifar R, Hemmatzadeh M, Arabi M, Maghsoudi MR, Mohammadi H. Lymphopenia associated with survivin and its downstream pathway in COVID-19 serving as a potential route in COVID-19 pathogenesis. Adv Med Sci 2024; 69:190-197. [PMID: 38521459 DOI: 10.1016/j.advms.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/16/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
PURPOSE Starting in 2019, coronavirus disease 2019 (COVID-19) caused an epidemic that was growing rapidly and has harmed millions of people globally. It has been demonstrated that survivin regulates lymphocyte survival, a main route involved in COVID-19 pathogenesis. Survivin belongs to the inhibitor of apoptosis protein (IAP) family, and its primary functions comprise regulating mitosis and inhibiting apoptosis. Since lower survivin expression has been shown to increase the sensitivity of lymphocytes to apoptotic induction, we looked into the function of survivin and its corresponding pathways in COVID-19 pathogenesis. MATERIALS AND METHODS The expression of survivin, X-linked inhibitor of apoptosis protein (XIAP), caspases 3, 7, 9, and poly (ADP-ribose) polymerase (PARP) was evaluated at both mRNA and protein levels in peripheral blood mononuclear cells (PBMCs) derived from healthy donors and patients with severe and moderate COVID-19 by qRT-PCR and Western blotting, respectively. Then, we enforced apoptosis to COVID-19 patient-derived lymphocytes, and the percent was assessed by flow cytometry. RESULTS Survivin and XIAP were less expressed in PBMCs derived from COVID-19 patients as apoptosis inhibitors than PARP, cleaved-PARP, caspase 9, and cleaved caspases 3 and 7, according to the results of real-time PCR and Western blot analysis. Additionally, according to the flow cytometry results, the down-regulation of survivin served as a potential factor in the lymphocyte depletion observed in patients with COVID-19. CONCLUSION The role of survivin and its related pathway was first discovered in the development of COVID-19 and may serve as a potential prognostic and therapeutic target.
Collapse
Affiliation(s)
| | - Kamyar Nasiri
- Department of Dentistry, Islamic Azad University, Tehran, Iran
| | - Farnoosh Ebrahimzadeh
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Soroush Ghodratizadeh
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mostafa Gholamrezaei
- Department of Parasitology and Mycology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alireza Rahat Dahmardeh
- Department of Anesthesiology and Critical Care, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ali Adili
- Department of Oncology, Tabriz University of Medical Sciences, Tabriz, Iran; Senior Adult Oncology Department, Moffitt Cancer Center, University of South, Florida, USA
| | - Rosita Amjidifar
- Department of Microbiology, Islamic Azad University of Iran, Ahar, Iran
| | - Maryam Hemmatzadeh
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Arabi
- Department of Physiology, Pharmacology and Medical Physics, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Reza Maghsoudi
- Faculty of Emergency Medicine & Toxicology, Emergency Department, Alborz University of Medical Sciences, Karaj, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
12
|
George RS, Goodey H, Russo MA, Tula R, Ghezzi P. Use of immunology in news and YouTube videos in the context of COVID-19: politicisation and information bubbles. Front Public Health 2024; 12:1327704. [PMID: 38435297 PMCID: PMC10906096 DOI: 10.3389/fpubh.2024.1327704] [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: 10/26/2023] [Accepted: 01/18/2024] [Indexed: 03/05/2024] Open
Abstract
Background The COVID-19 pandemic propelled immunology into global news and social media, resulting in the potential for misinterpreting and misusing complex scientific concepts. Objective To study the extent to which immunology is discussed in news articles and YouTube videos in English and Italian, and if related scientific concepts are used to support specific political or ideological narratives in the context of COVID-19. Methods In English and Italian we searched the period 11/09/2019 to 11/09/2022 on YouTube, using the software Mozdeh, for videos mentioning COVID-19 and one of nine immunological concepts: antibody-dependent enhancement, anergy, cytokine storm, herd immunity, hygiene hypothesis, immunity debt, original antigenic sin, oxidative stress and viral interference. We repeated this using MediaCloud for news articles.Four samples of 200 articles/videos were obtained from the randomised data gathered and analysed for mentions of concepts, stance on vaccines, masks, lockdown, social distancing, and political signifiers. Results Vaccine-negative information was higher in videos than news (8-fold in English, 6-fold in Italian) and higher in Italian than English (4-fold in news, 3-fold in videos). We also observed the existence of information bubbles, where a negative stance towards one intervention was associated with a negative stance to other linked ideas. Some immunological concepts (immunity debt, viral interference, anergy and original antigenic sin) were associated with anti-vaccine or anti-NPI (non-pharmacological intervention) views. Videos in English mentioned politics more frequently than those in Italian and, in all media and languages, politics was more frequently mentioned in anti-guidelines and anti-vaccine media by a factor of 3 in video and of 3-5 in news. Conclusion There is evidence that some immunological concepts are used to provide credibility to specific narratives and ideological views. The existence of information bubbles supports the concept of the "rabbit hole" effect, where interest in unconventional views/media leads to ever more extreme algorithmic recommendations.
Collapse
Affiliation(s)
| | - Hannah Goodey
- Brighton and Sussex Medical School, Brighton, United Kingdom
| | | | - Rovena Tula
- Department of Biomolecular Sciences, University of Urbino, Urbino, Italy
| | - Pietro Ghezzi
- Brighton and Sussex Medical School, Brighton, United Kingdom
- Department of Biomolecular Sciences, University of Urbino, Urbino, Italy
| |
Collapse
|
13
|
Huang P, Tan YY, Chen B, Shang HF, Wang LJ, Liu CF, Chen L, Chang Y, Wang H, Wang XL, Lei XG, Yao LF, Yu Y, Ye Z, Chen HB, Chen SD. Life and disease status of patients with Parkinson's disease during and after zero-COVID in China: an online survey. Transl Neurodegener 2024; 13:8. [PMID: 38317265 PMCID: PMC10845503 DOI: 10.1186/s40035-024-00399-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/14/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Little is known about the impact of the COVID-19 pandemic on patients with Parkinson's disease (PD) at different stages of the pandemic. This study aims to assess the lives and disease status of PD patients during the zero-COVID policy period and after ending the zero-COVID policy. METHODS This multicenter cross-sectional study included two online surveys among PD patients in China, from May 30 to June 30 in 2022 and from January 1 to February 28 in 2023, respectively. The survey questionnaires contained four sections: (1) status of COVID-19 infection; (2) impact on motor and non-motor symptoms; (3) impact on daily and social lives; and (4) impact on PD disease management. RESULTS A total of 1764 PD patients participated in the first online survey, with 200 patients having lockdown experience and 3 being COVID-19-positive (0.17%). In addition, 537 patients participated in the second online survey, with 467 patients having COVID-19 infection (86.96%). (1) During zero-COVID, all of the COVID-19-positive patients had mild symptoms of COVID-19 and no death was reported. After zero-COVID, 83.51% of the COVID-19-positive patients had mild symptoms. The overall death rate and inpatient mortality rate of COVID-19-positive PD patients were 3.21% and 30.00%, respectively. (2) During zero-COVID, 49.43% of PD patients reported worsening of PD-related symptoms (lockdown vs. unlockdown, 60.50% vs. 48.02%, P = 0.0009). After zero-COVID, 54.93% of PD patients reported worsening of PD-related symptoms (COVID-19 positive vs. COVID-19 negative, 59.31% vs. 25.71%, P < 0.0001). (3) During zero-COVID, 62.36% of patients felt worried, and 'limited outdoor activities' (55.39%) was the top reason for mental health problems. After zero-COVID, 59.03% of patients felt worried, with 'poor health' (58.10%) being the top reason. The PD patients tended to change their daily activities from offline to online, and their economic and caregiver burdens increased both during and after zero-COVID. (4) Most PD patients would like to choose online rehabilitation during (69.56%) and after zero-COVID (69.27%). The demand for online medication purchasing also increased during (47.00%) and after zero-COVID (26.63%). CONCLUSIONS The COVID-19 pandemic aggravated the motor and non-motor symptoms of PD patients either during or after the zero-COVID policy period. The PD patients also experienced prominent mental health problems, changes in daily activities, and increases in economic and caregiver burdens. The COVID-19 pandemic has changed ways of PD management with increasing demands for online medication purchasing and rehabilitation.
Collapse
Affiliation(s)
- Pei Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu-Yan Tan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Biao Chen
- Department of Neurology, Xuan Wu Hospital Affiliated to Capital Medical University, Beijing, 100053, China
| | - Hui-Fang Shang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li-Juan Wang
- Department of Neurology, Guangdong General Hospital, Guangzhou, 510080, China
| | - Chun-Feng Liu
- Department of Neurology, Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Ling Chen
- Department of Neurology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Ying Chang
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, 130031, China
| | - Han Wang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xue-Lian Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiao-Guang Lei
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Li-Fen Yao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yang Yu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Zheng Ye
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hai-Bo Chen
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China.
| | - Sheng-Di Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, 201210, China.
| |
Collapse
|
14
|
Arab FL, Hoseinzadeh A, Mohammadi FS, Rajabian A, Faridzadeh A, Mahmoudi M. Immunoregulatory effects of nanocurcumin in inflammatory milieu: Focus on COVID-19. Biomed Pharmacother 2024; 171:116131. [PMID: 38198954 DOI: 10.1016/j.biopha.2024.116131] [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: 10/12/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
The use of natural compounds, such as curcumin, to treat infections caused by bacteria, viruses, fungi, parasites, inflammatory diseases, and various types of cancer is an active and dynamic area of research. Curcumin has a long history of use in the food industry, and there is currently a growing interest in its therapeutic applications. Numerous clinical trials have consistently shown that curcumin, a polyphenolic compound, is safe and well-tolerated even at high doses. There is no toxicity limit. However, the clinical efficacy of curcumin has been limited by its constraints. However, scientific evidence indicates that the use of adjuvants and carriers, such as nanoparticles, exosomes, micelles, and liposomes, can help overcome this limitation. The properties, functions, and human benefits of using nanocurcumin are well-supported by scientific research. Recent evidence suggests that nanocurcumin may be a beneficial therapeutic modality due to its potential to decrease gene expression and secretion of specific inflammatory biomarkers involved in the cytokinestorm seen in severe COVID-19, as well as increase lymphocyte counts. Nanocurcumin has demonstrated the ability to improve clinical manifestations and modulate immune response and inflammation in various autoinflammatory diseases. Additionally, its efficacy, affordability, and safety make it a promising replacement for residual cancer cells after tumor removal. However, further studies are necessary to evaluate the safety and efficacy of nanocurcumin as a new therapeutic in clinical trials, including appropriate dosage, frequency, and duration.
Collapse
Affiliation(s)
- Fahimeh Lavi Arab
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Akram Hoseinzadeh
- Immunology Research Center, Bu‑Ali Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Sadat Mohammadi
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Rajabian
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
|
16
|
Dittmar JW, Teplensky MH, Evangelopoulos M, Qin L, Zhang B, Mirkin CA. Tuning DNA Dissociation from Spherical Nucleic Acids for Enhanced Immunostimulation. ACS NANO 2023; 17:17996-18007. [PMID: 37713675 PMCID: PMC10801821 DOI: 10.1021/acsnano.3c04333] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
The stability of the core can significantly impact the therapeutic effectiveness of liposome-based drugs. While the spherical nucleic acid (SNA) architecture has elevated liposomal stability to increase therapeutic efficacy, the chemistry used to anchor the DNA to the liposome core is an underexplored design parameter with a potentially widespread biological impact. Herein, we explore the impact of SNA anchoring chemistry on immunotherapeutic function by systematically studying the importance of hydrophobic dodecane anchoring groups in attaching DNA strands to the liposome core. By deliberately modulating the size of the oligomer that defines the anchor, a library of structures has been established. These structures, combined with in vitro and in vivo immune stimulation analyses, elucidate the relationships between and importance of anchoring strength and dissociation of DNA from the SNA shell on its biological properties. Importantly, the most stable dodecane anchor, (C12)9, is superior to the n = 4-8 and 10 structures and quadruples immune stimulation compared to conventional cholesterol-anchored SNAs. When the OVA1 peptide antigen is encapsulated by the (C12)9 SNA and used as a therapeutic vaccine in an E.G7-OVA tumor model, 50% of the mice survived the initial tumor, and all of those survived tumor rechallenge. Importantly, the strong innate immune stimulation does not cause a cytokine storm compared to linear immunostimulatory DNA. Moreover, a (C12)9 SNA that encapsulates a peptide targeting SARS-CoV-2 generates a robust T cell response; T cells raised from SNA treatment kill >40% of target cells pulsed with the same peptide and ca. 45% of target cells expressing the entire spike protein. This work highlights the importance of using anchor chemistry to elevate SNA stability to achieve more potent and safer immunotherapeutics in the context of both cancer and infectious disease.
Collapse
Affiliation(s)
- Jasper W Dittmar
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Michelle H Teplensky
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Michael Evangelopoulos
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Lei Qin
- Department of Medicine, Division of Hematology and Oncology, Northwestern University, 420 E Superior Street, Chicago, Illinois 60611, United States
| | - Bin Zhang
- Department of Medicine, Division of Hematology and Oncology, Northwestern University, 420 E Superior Street, Chicago, Illinois 60611, United States
| | - Chad A Mirkin
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
17
|
Hajjar J, Dziegielewski C, Dickson S, Simpson A, Kyeremanteng K. The role of low-carbohydrate diets in the intensive care unit. Nutr Health 2023; 29:377-381. [PMID: 36591890 DOI: 10.1177/02601060221149088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Low-carbohydrate, high-fat (LCHF) nutrition therapy is characterized by carbohydrates comprising <26% of the daily caloric intake and a higher proportion of fat. LCHF therapies reduce exogenous glucose load, improve glycemic control, decrease inflammation, and improve clinical outcomes such as respiratory function. Given the altered metabolism in critically ill patients, LCHF nutrition therapy may be especially beneficial as it enables the conservation of protein and glucose for metabolic roles beyond energy use. In critical illness, LCHF diets have the potential to reduce hyperglycemia, improve ventilation, decrease hospital length of stay and reduce hospital costs. The purpose of this commentary piece is to describe LCHF nutrition therapy, summarize its impact on health outcomes, and discuss its role in the intensive care unit (ICU). Additional research on the effects of LCHF nutrition therapy on critically ill patients is warranted, including a focus on COVID-19.
Collapse
Affiliation(s)
- Julia Hajjar
- Institut du Savoir Montfort, Ottawa, ON, Canada
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | - Sarah Dickson
- Institut du Savoir Montfort, Ottawa, ON, Canada
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Allison Simpson
- Department of Critical Care, The Ottawa Hospital, Ottawa, ON, Canada
| | - Kwadwo Kyeremanteng
- Institut du Savoir Montfort, Ottawa, ON, Canada
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Critical Care, The Ottawa Hospital, Ottawa, ON, Canada
| |
Collapse
|
18
|
Yu F, He H, Huang T, Zhou Y. Study on the cytokines related to SARS-Cov-2 in testicular cells and the interaction network between cells based on scRNA-seq data. Open Life Sci 2023; 18:20220661. [PMID: 37589002 PMCID: PMC10426268 DOI: 10.1515/biol-2022-0661] [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: 03/09/2023] [Revised: 06/04/2023] [Accepted: 06/23/2023] [Indexed: 08/18/2023] Open
Abstract
Inflammatory cytokine storms (CS) in COVID-19 patients are associated with elevated levels of 13 specific cytokines, potentially impacting male fertility by causing testicular cell damage and disrupting the immune microenvironment. Some patients present with scrotal discomfort and orchitis. However, few studies have explored cytokine expression in testicular cells and their role in cell-to-cell communication. In this study, we integrated single-cell sequencing data sets of testicular cells, annotating 20 cell clusters using marker genes and the Human Cell Landscape database. We constructed cell pseudo-chronological trajectories, hub genes, and analyzed the cytokine interaction network between sperm cells using CellChat. Our findings identified 12 types of testicular cells, with four cytokines (IL8, CCL2, CCL3, and TNF) potentially involved in immune processes. Pseudo-chronological trajectory analysis indicated IL8 and CCL3's essential roles in testicular macrophages and endothelial cell development, affecting the immune microenvironment. We determined eight key cytokines (IL1, IL2, IL4, IL6, CCL, CSF3, TNF, and IFN-II) functions in cell interaction networks. Network analysis of exogenous cytokines directly acting on testicular cells showed IL2 potentially affecting all testicular cells, suggesting a vital role in cell communication. This research offers valuable insights into CSs effects on testicular cells and their potential impact on male fertility during COVID-19 infection.
Collapse
Affiliation(s)
- Fan Yu
- Center of Clinical Laboratory Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 518000, China
| | - Haihong He
- Center of Clinical Laboratory Medicine, Shenzhen Hospital, Southern Medical University, No. 1333
Xinhu Road, Shenzhen, Guangdong, 518000, China
| | - Tingting Huang
- Center of Clinical Laboratory Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 518000, China
| | - Yiwen Zhou
- Center of Clinical Laboratory Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 518000, China
| |
Collapse
|
19
|
Regolo M, Sorce A, Vaccaro M, Colaci M, Stancanelli B, Natoli G, Motta M, Isaia I, Castelletti F, Giangreco F, Fichera D, Aparo P, Lanzafame A, Russo M, Santangelo N, Noto P, Malatino L. Assessing Humoral Immuno-Inflammatory Pathways Associated with Respiratory Failure in COVID-19 Patients. J Clin Med 2023; 12:4057. [PMID: 37373750 DOI: 10.3390/jcm12124057] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
All severe cases of SARS-CoV-2 infections are characterized by a high risk of disease progression towards ARDS, leading to a bad outcome. Respiratory symptoms in COVID-19 patients often do not correspond to disease's worsening. In our sample, median age was 74 years (72-75) and 54% were men. The median period of hospitalization was 9 days. Firstly, we observed a significant asynchronous trend of neutrophil-to-lymphocyte ratio (NLR) and C-reactive protein (CRP) in 764 selected among 963 patients, who were consecutively recruited in two hospitals (Cannizzaro, S. Marco) in Catania, Italy. NLR values in deceased patients showed an increase from baseline over time. By contrast, CRP tended to fall from baseline to median day of hospitalization in all three subgroups, but steeply increased at the end of hospitalization only in ICU-admitted patients. Then, we evaluated the relationships between NLR and CRP as continuous variables with PaO2/FiO2 ratio (P/F). NLR was an independent predictor of mortality (HR: 1.77, p < 0.0001), while ICU admission was more significantly associated with CRP (HR: 1.70, p < 0.0001). Finally, age, neutrophils, CRP, and lymphocytes are significantly and directly linked to P/F, while the influence of inflammation on P/F, reflected by CRP, was also mediated by neutrophils.
Collapse
Affiliation(s)
- Matteo Regolo
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Alessandra Sorce
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, "G. D'Alessandro" (PROMISE), Unit of Nephrology and Hypertension, European Society of Hypertension Excellence Centre, University of Palermo, 90133 Palermo, Italy
| | - Mauro Vaccaro
- Department of Emergency Medicine, San Marco-Polyclinic Academic Hospital, 95121 Catania, Italy
| | - Michele Colaci
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Benedetta Stancanelli
- Unit of Internal Medicine, San Marco-Polyclinic Academic Hospital, 95121 Catania, Italy
| | - Giuseppe Natoli
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Massimo Motta
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Ivan Isaia
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Federica Castelletti
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Federica Giangreco
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Daniela Fichera
- Department of Emergency Medicine, San Marco-Polyclinic Academic Hospital, 95121 Catania, Italy
| | - Paola Aparo
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Alessandra Lanzafame
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Mario Russo
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Nicola Santangelo
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Paola Noto
- Department of Emergency Medicine, San Marco-Polyclinic Academic Hospital, 95121 Catania, Italy
| | - Lorenzo Malatino
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| |
Collapse
|
20
|
Akkiz H. Unraveling the Molecular and Cellular Pathogenesis of COVID-19-Associated Liver Injury. Viruses 2023; 15:1287. [PMID: 37376587 DOI: 10.3390/v15061287] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 06/29/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2) continues to cause substantial morbidity and mortality. Most infections are mild; however, some patients experience severe and potentially fatal systemic inflammation, tissue damage, cytokine storm, and acute respiratory distress syndrome. Patients with chronic liver disease have been frequently affected, experiencing high morbidity and mortality. In addition, elevated liver enzymes may be a risk factor for disease progression, even in the absence of underlying liver disease. While the respiratory tract is a primary target of SARS-CoV-2, it has become evident that COVID-19 is a multisystemic infectious disease. The hepatobiliary system might be influenced during COVID-19 infection, ranging from a mild elevation of aminotransferases to the development of autoimmune hepatitis and secondary sclerosing cholangitis. Furthermore, the virus can promote existing chronic liver diseases to liver failure and activate the autoimmune liver disease. Whether the direct cytopathic effects of the virus, host reaction, hypoxia, drugs, vaccination, or all these risk factors cause liver injury has not been clarified to a large extent in COVID-19. This review article discussed the molecular and cellular mechanisms involved in the pathogenesis of SARS-CoV-2 virus-associated liver injury and highlighted the emerging role of liver sinusoidal epithelial cells (LSECs) in virus-related liver damage.
Collapse
Affiliation(s)
- Hikmet Akkiz
- Department of Gastroenterology and Hepatology, Medical Faculty, Bahçeşehir University, Istanbul 34349, Turkey
| |
Collapse
|
21
|
Talukdar D, Bandopadhyay P, Ray Y, Paul SR, Sarif J, D'Rozario R, Lahiri A, Das S, Bhowmick D, Chatterjee S, Das B, Ganguly D. Association of gut microbial dysbiosis with disease severity, response to therapy and disease outcomes in Indian patients with COVID-19. Gut Pathog 2023; 15:22. [PMID: 37161621 PMCID: PMC10170741 DOI: 10.1186/s13099-023-00546-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/13/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Severe coronavirus disease 2019 (COVID-19) is associated with systemic hyper-inflammation. An adaptive interaction between gut microbiota and host immune systems is important for intestinal homeostasis and systemic immune regulation. The association of gut microbial composition and functions with COVID-19 disease severity is sparse, especially in India. We analysed faecal microbial diversity and abundances in a cohort of Indian COVID-19 patients to identify key signatures in the gut microbial ecology in patients with severe COVID-19 disease as well as in response to different therapies. The composition of the gut microbiome was characterized using 16Sr RNA gene sequences of genomic DNA extracted from faecal samples of 52 COVID-19 patients. Metabolic pathways across the groups were predicted using PICRUSt2. All statistical analyses were done using Vegan in the R environment. Plasma cytokine abundance at recruitment was measured in a multiplex assay. RESULTS The gut microbiome composition of mild and severe patients was found to be significantly different. Immunomodulatory commensals, viz. Lachnospiraceae family members and Bifidobacteria producing butyrate and short-chain fatty acids (SCFAs), were under represented in patients with severe COVID-19, with an increased abundance of opportunistic pathogens like Eggerthella. The higher abundance of Lachnoclostridium in severe disease was reduced in response to convalescent plasma therapy. Specific microbial genera showed distinctive trends in enriched metabolic pathways, strong correlations with blood plasma cytokine levels, and associative link to disease outcomes. CONCLUSION Our study indicates that, along with SARS-CoV-2, a dysbiotic gut microbial community may also play an important role in COVID-19 severity through modulation of host immune responses.
Collapse
Affiliation(s)
- Daizee Talukdar
- Functional Genomics Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Purbita Bandopadhyay
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Yogiraj Ray
- Department of Medicine, Infectious Diseases and Beleghata General Hospital, Kolkata, India
- Department of Infectious Disease, SSKM Hospital, Kolkata, India
| | - Shekhar Ranjan Paul
- Department of Medicine, Infectious Diseases and Beleghata General Hospital, Kolkata, India
| | - Jafar Sarif
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Ranit D'Rozario
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Abhishake Lahiri
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Santanu Das
- Functional Genomics Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Debaleena Bhowmick
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Shilpak Chatterjee
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Bhabatosh Das
- Functional Genomics Laboratory, Translational Health Science and Technology Institute, Faridabad, India.
| | - Dipyaman Ganguly
- CSIR-Indian Institute of Chemical Biology, Kolkata, India.
- Academy of Scientific and Innovative Research, Ghaziabad, India.
| |
Collapse
|
22
|
Wu C, Peng W, Cheng D, Gu H, Liu F, Peng S, Fu L. Efficacy and Economic Evaluation of Nonbiological Artificial Liver Therapy in Acute-on-chronic Hepatitis B Liver Failure. J Clin Transl Hepatol 2023; 11:433-440. [PMID: 36643036 PMCID: PMC9817044 DOI: 10.14218/jcth.2022.00106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/27/2022] [Accepted: 06/12/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND AND AIMS Nonbiological artificial liver (NBAL) is frequently used as a first-line treatment for hepatitis B virus-associated acute-on-chronic liver failure (HBV-ACLF). This study aimed to compare the therapeutic efficacy and cost-effectiveness ratio (CER) of comprehensive medical treatment, plasma exchange (PE), and double plasma molecular adsorption system (DPMAS) plus half-dose PE (DPMAS+PE) in patients with HBV-ACLF. METHODS A total of 186 patients with HBV-ACLF randomly received comprehensive medical treatment, PE, or DPMAS+PE and were prospectively evaluated. Patients were divided into four subgroups based on the pretreatment prothrombin activity (PTA): Group I (PTA>40%), group II (PTA 30-40%), group III (PTA 20-30%), and group IV (PTA<20%). The main outcome measures were 28 day effectiveness; 90 day liver transplantation-free survival; change of biochemical parameters; and CER. RESULTS DPMAS+PE treatment was associated with significantly higher 28 day effectiveness and 90 day liver transplantation-free survival compared with PE treatment in patients with group I liver failure. Clearance of serum total bilirubin (TBIL), AST, and creatinine (Cr) were significantly higher in the DPMAS+PE group than in the PE group. For subjects with group I liver failure, DPMAS+PE treatment had advantages of lower CER values and better cost-effectiveness. CONCLUSIONS Compared with comprehensive medical treatment and PE alone, DPMAS with half-dose sequential PE treatment more effectively improved TBIL, AST, and Cr in HBV-ACLF patients, improved 28 day effectiveness and 90 day survival rates in patients with group I liver failure, and was more cost effective. DPMAS+PE is a viable NBAL approach for treatment of HBV-ACLF.
Collapse
Affiliation(s)
- Cichun Wu
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Wenting Peng
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Da Cheng
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Huimin Gu
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Fei Liu
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Shifang Peng
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Lei Fu
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China
- Correspondence to: Lei Fu, Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China. ORCID: https://orcid.org/0000-0001-7550-1254. Tel: +86-731-89753067, Fax: +86-731-4327332, E-mail:
| |
Collapse
|
23
|
Nie Y, Mou L, Long Q, Deng D, Hu R, Cheng J, Wu J. SARS-CoV-2 ORF3a positively regulates NF-κB activity by enhancing IKKβ-NEMO interaction. Virus Res 2023; 328:199086. [PMID: 36894068 PMCID: PMC10009424 DOI: 10.1016/j.virusres.2023.199086] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/22/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a global pandemic caused by SARS-CoV-2 infection. Patients with severe COVID-19 exhibit robust induction of proinflammatory cytokines, which are closely associated with the development of acute respiratory distress syndrome. However, the underlying mechanisms of the NF-κB activation mediated by SARS-CoV-2 infection remain poorly understood. Here, we screened SARS-CoV-2 genes and found that ORF3a induces proinflammatory cytokines by activating the NF-κB pathway. Moreover, we found that ORF3a interacts with IKKβ and NEMO and enhances the interaction of IKKβ-NEMO, thereby positively regulating NF-κB activity. Together, these results suggest ORF3a may play pivotal roles in the pathogenesis of SARS-CoV-2 and provide novel insights into the interaction between host immune responses and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ying Nie
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; These authors contributed equally: Ying Nie, Lumin Mou
| | - Lumin Mou
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China; These authors contributed equally: Ying Nie, Lumin Mou
| | - Qizhou Long
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Dongqing Deng
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Rongying Hu
- The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Jinzhi Cheng
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Jiahong Wu
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China.
| |
Collapse
|
24
|
Farooqui AA, Farooqui T, Sun GY, Lin TN, Teh DBL, Ong WY. COVID-19, Blood Lipid Changes, and Thrombosis. Biomedicines 2023; 11:biomedicines11041181. [PMID: 37189799 DOI: 10.3390/biomedicines11041181] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Although there is increasing evidence that oxidative stress and inflammation induced by COVID-19 may contribute to increased risk and severity of thromboses, the underlying mechanism(s) remain to be understood. The purpose of this review is to highlight the role of blood lipids in association with thrombosis events observed in COVID-19 patients. Among different types of phospholipases A2 that target cell membrane phospholipids, there is increasing focus on the inflammatory secretory phospholipase A2 IIA (sPLA2-IIA), which is associated with the severity of COVID-19. Analysis indicates increased sPLA2-IIA levels together with eicosanoids in the sera of COVID patients. sPLA2 could metabolise phospholipids in platelets, erythrocytes, and endothelial cells to produce arachidonic acid (ARA) and lysophospholipids. Arachidonic acid in platelets is metabolised to prostaglandin H2 and thromboxane A2, known for their pro-coagulation and vasoconstrictive properties. Lysophospholipids, such as lysophosphatidylcholine, could be metabolised by autotaxin (ATX) and further converted to lysophosphatidic acid (LPA). Increased ATX has been found in the serum of patients with COVID-19, and LPA has recently been found to induce NETosis, a clotting mechanism triggered by the release of extracellular fibres from neutrophils and a key feature of the COVID-19 hypercoagulable state. PLA2 could also catalyse the formation of platelet activating factor (PAF) from membrane ether phospholipids. Many of the above lipid mediators are increased in the blood of patients with COVID-19. Together, findings from analyses of blood lipids in COVID-19 patients suggest an important role for metabolites of sPLA2-IIA in COVID-19-associated coagulopathy (CAC).
Collapse
Affiliation(s)
- Akhlaq A Farooqui
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
| | - Tahira Farooqui
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Teng-Nan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11929, Taiwan
| | - Daniel B L Teh
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119260, Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119260, Singapore
- Neurobiology Research Programme, Life Sciences Institute, National University of Singapore, Singapore 119260, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119260, Singapore
- Neurobiology Research Programme, Life Sciences Institute, National University of Singapore, Singapore 119260, Singapore
| |
Collapse
|
25
|
Alomair BM, Al‐Kuraishy HM, Al‐Gareeb AI, Al‐Buhadily AK, Alexiou A, Papadakis M, Alshammari MA, Saad HM, Batiha GE. Mixed storm in SARS-CoV-2 infection: A narrative review and new term in the Covid-19 era. Immun Inflamm Dis 2023; 11:e838. [PMID: 37102645 PMCID: PMC10132185 DOI: 10.1002/iid3.838] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/28/2023] Open
Abstract
Coronavirus disease 2019 (Covid-19) is caused by a novel severe acute respiratory syndrome coronavirus virus type 2 (SARS-CoV-2) leading to the global pandemic worldwide. Systemic complications in Covid-19 are mainly related to the direct SARS-CoV-2 cytopathic effects, associated hyperinflammation, hypercytokinemia, and the development of cytokine storm (CS). As well, Covid-19 complications are developed due to the propagation of oxidative and thrombotic events which may progress to a severe state called oxidative storm and thrombotic storm (TS), respectively. In addition, inflammatory and lipid storms are also developed in Covid-19 due to the activation of inflammatory cells and the release of bioactive lipids correspondingly. Therefore, the present narrative review aimed to elucidate the interrelated relationship between different storm types in Covid-19 and the development of the mixed storm (MS). In conclusion, SARS-CoV-2 infection induces various storm types including CS, inflammatory storm, lipid storm, TS and oxidative storm. These storms are not developing alone since there is a close relationship between them. Therefore, the MS seems to be more appropriate to be related to severe Covid-19 than CS, since it develops in Covid-19 due to the intricate interface between reactive oxygen species, proinflammatory cytokines, complement activation, coagulation disorders, and activated inflammatory signaling pathway.
Collapse
Affiliation(s)
- Basil Mohammed Alomair
- Department of Medicine, College of Medicine, Internal Medicine and EndocrinologyJouf UniversityAl‐JoufSaudi Arabia
| | - Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali K. Al‐Buhadily
- Department of Clinical Pharmacology, Medicine, and Therapeutic, Medical Faculty, College of MedicineAl‐Mustansiriyah UniversityBaghdadIraq
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- AFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐HerdeckeUniversity of Witten‐HerdeckeWuppertalGermany
| | - Majed Ayed Alshammari
- Department of MedicinePrince Mohammed Bin Abdulaziz Medical CitySakakaAl‐JoufSaudi Arabia
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMarsaMatruhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|
26
|
Narang J, Jatana S, Ponti AK, Musich R, Gallop J, Wei AH, Seck S, Johnson J, Kokoczka L, Nowacki AS, McBride JD, Mireles-Cabodevila E, Gordon S, Cooper K, Fernandez AP, McDonald C. Abnormal thrombosis and neutrophil activation increase hospital-acquired sacral pressure injuries and morbidity in COVID-19 patients. Front Immunol 2023; 14:1031336. [PMID: 37026002 PMCID: PMC10070761 DOI: 10.3389/fimmu.2023.1031336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 03/08/2023] [Indexed: 04/08/2023] Open
Abstract
Hospitalized patients have an increased risk of developing hospital-acquired sacral pressure injury (HASPI). However, it is unknown whether SARS-CoV-2 infection affects HASPI development. To explore the role of SARS-CoV-2 infection in HASPI development, we conducted a single institution, multi-hospital, retrospective study of all patients hospitalized for ≥5 days from March 1, 2020 to December 31, 2020. Patient demographics, hospitalization information, ulcer characteristics, and 30-day-related morbidity were collected for all patients with HASPIs, and intact skin was collected from HASPI borders in a patient subset. We determined the incidence, disease course, and short-term morbidity of HASPIs in COVID-19(+) patients, and characterized the skin histopathology and tissue gene signatures associated with HASPIs in COVID-19 disease. COVID-19(+) patients had a 63% increased HASPI incidence rate, HASPIs of more severe ulcer stage (OR 2.0, p<0.001), and HASPIs more likely to require debridement (OR 3.1, p=0.04) compared to COVID-19(-) patients. Furthermore, COVID-19(+) patients with HASPIs had 2.2x increased odds of a more severe hospitalization course compared to COVID-19(+) patients without HASPIs. HASPI skin histology from COVID-19(+) patients predominantly showed thrombotic vasculopathy, with the number of thrombosed vessels being significantly greater than HASPIs from COVID-19(-) patients. Transcriptional signatures of a COVID-19(+) sample subset were enriched for innate immune responses, thrombosis, and neutrophil activation genes. Overall, our results suggest that immunologic dysregulation secondary to SARS-CoV-2 infection, including neutrophil dysfunction and abnormal thrombosis, may play a pathogenic role in development of HASPIs in patients with severe COVID-19.
Collapse
Affiliation(s)
- Jatin Narang
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Samreen Jatana
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - András K. Ponti
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Ryan Musich
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Joshua Gallop
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Angela H. Wei
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sokhna Seck
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jessica Johnson
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Lynne Kokoczka
- Medical Intensive Care Unit, Cleveland Clinic, Cleveland, OH, United States
| | - Amy S. Nowacki
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Jeffrey D. McBride
- Department of Dermatology, The University of Oklahoma College of Medicine, Oklahoma City, OK, United States
| | | | - Steven Gordon
- Department of Infectious Disease, Cleveland Clinic, Cleveland, OH, United States
| | - Kevin Cooper
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Anthony P. Fernandez
- Department of Dermatology, Dermatology and Plastic Surgery Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Anatomic Pathology, Pathology and Lab Medicine, Cleveland Clinic, Cleveland, OH, United States
| | - Christine McDonald
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
27
|
Neutrophil Extracellular Traps in Airway Diseases: Pathological Roles and Therapeutic Implications. Int J Mol Sci 2023; 24:ijms24055034. [PMID: 36902466 PMCID: PMC10003347 DOI: 10.3390/ijms24055034] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Neutrophils are important effector cells of the innate immune response that fight pathogens by phagocytosis and degranulation. Neutrophil extracellular traps (NETs) are released into the extracellular space to defend against invading pathogens. Although NETs play a defensive role against pathogens, excessive NETs can contribute to the pathogenesis of airway diseases. NETs are known to be directly cytotoxic to the lung epithelium and endothelium, highly involved in acute lung injury, and implicated in disease severity and exacerbation. This review describes the role of NET formation in airway diseases, including chronic rhinosinusitis, and suggests that targeting NETs could be a therapeutic strategy for airway diseases.
Collapse
|
28
|
The Landscape of Expressed Chimeric Transcripts in the Blood of Severe COVID-19 Infected Patients. Viruses 2023; 15:v15020433. [PMID: 36851647 PMCID: PMC9958880 DOI: 10.3390/v15020433] [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: 12/28/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
The ongoing COVID-19 pandemic caused by SARS-CoV-2 infections has quickly developed into a global public health threat. COVID-19 patients show distinct clinical features, and in some cases, during the severe stage of the condition, the disease severity leads to an acute respiratory disorder. In spite of several pieces of research in this area, the molecular mechanisms behind the development of disease severity are still not clearly understood. Recent studies demonstrated that SARS-CoV-2 alters the host cell splicing and transcriptional response to overcome the host immune response that provides the virus with favorable conditions to replicate efficiently within the host cells. In several disease conditions, aberrant splicing could lead to the development of novel chimeric transcripts that could promote the functional alternations of the cell. As severe SARS-CoV-2 infection was reported to cause abnormal splicing in the infected cells, we could expect the generation and expression of novel chimeric transcripts. However, no study so far has attempted to check whether novel chimeric transcripts are expressed in severe SARS-CoV-2 infections. In this study, we analyzed several publicly available blood transcriptome datasets of severe COVID-19, mild COVID-19, other severe respiratory viral infected patients, and healthy individuals. We identified 424 severe COVID-19 -specific chimeric transcripts, 42 of which were recurrent. Further, we detected 189 chimeric transcripts common to severe COVID-19 and multiple severe respiratory viral infections. Pathway and gene enrichment analysis of the parental genes of these two subsets of chimeric transcripts reveals that these are potentially involved in immune-related processes, interferon signaling, and inflammatory responses, which signify their potential association with immune dysfunction leading to the development of disease severity. Our study provides the first detailed expression landscape of chimeric transcripts in severe COVID-19 and other severe respiratory viral infections.
Collapse
|
29
|
Zafarani A, Razizadeh MH, Pashangzadeh S, Amirzargar MR, Taghavi-Farahabadi M, Mahmoudi M. Natural killer cells in COVID-19: from infection, to vaccination and therapy. Future Virol 2023:10.2217/fvl-2022-0040. [PMID: 36936055 PMCID: PMC10013930 DOI: 10.2217/fvl-2022-0040] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 01/31/2023] [Indexed: 03/15/2023]
Abstract
Natural killer (NK) cells are among the most important innate immunity members, which are the first cells that fight against infected cells. The function of these cells is impaired in patients with COVID-19 and they are not able to prevent the spread of the disease or destroy the infected cells. Few studies have evaluated the effects of COVID-19 vaccines on NK cells, though it has been demonstrated that DNA vaccines and BNT162b2 can affect NK cell response. In the present paper, the effects of SARS-CoV-2 on the NK cells during infection, the effect of vaccination on NK cells, and the NK cell-based therapies were reviewed.
Collapse
Affiliation(s)
- Alireza Zafarani
- 1Department of Hematology & Blood Banking, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Salar Pashangzadeh
- 3Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
- 4Immunology Today, Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mohammad Reza Amirzargar
- 1Department of Hematology & Blood Banking, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Taghavi-Farahabadi
- 5Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahmoudi
- 6Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
- Author for correspondence: Tel.: +98 936 002 0731;
| |
Collapse
|
30
|
Huang P, Zhang LY, Tan YY, Chen SD. Links between COVID-19 and Parkinson's disease/Alzheimer's disease: reciprocal impacts, medical care strategies and underlying mechanisms. Transl Neurodegener 2023; 12:5. [PMID: 36717892 PMCID: PMC9885419 DOI: 10.1186/s40035-023-00337-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
The impact of coronavirus disease 2019 (COVID-19) pandemic on patients with neurodegenerative diseases and the specific neurological manifestations of COVID-19 have aroused great interest. However, there are still many issues of concern to be clarified. Therefore, we review the current literature on the complex relationship between COVID-19 and neurodegenerative diseases with an emphasis on Parkinson's disease (PD) and Alzheimer's disease (AD). We summarize the impact of COVID-19 infection on symptom severity, disease progression, and mortality rate of PD and AD, and discuss whether COVID-19 infection could trigger PD and AD. In addition, the susceptibility to and the prognosis of COVID-19 in PD patients and AD patients are also included. In order to achieve better management of PD and AD patients, modifications of care strategies, specific drug therapies, and vaccines during the pandemic are also listed. At last, mechanisms underlying the link of COVID-19 with PD and AD are reviewed.
Collapse
Affiliation(s)
- Pei Huang
- grid.16821.3c0000 0004 0368 8293Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Lin-Yuan Zhang
- grid.412478.c0000 0004 1760 4628Department of Neurology, Shanghai General Hospital, Shanghai, 200080 China
| | - Yu-Yan Tan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Sheng-Di Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, 201210, China.
| |
Collapse
|
31
|
Jiang C, Jiang K, Li X, Zhang N, Zhu W, Meng L, Zhang Y, Lu S. Evaluation of immunoprotection against coronavirus disease 2019: Novel variants, vaccine inoculation, and complications. J Pharm Anal 2023; 13:1-10. [PMID: 36317070 PMCID: PMC9605787 DOI: 10.1016/j.jpha.2022.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022] Open
Abstract
The strikingly rapidly mutating nature of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome has been a constant challenge during the coronavirus disease 2019 (COVID-19) pandemic. In this study, various techniques, including reverse transcription-quantitative polymerase chain reaction, antigen-detection rapid diagnostic tests, and high-throughput sequencing were analyzed under different scenarios and spectra for the etiological diagnosis of COVID-19 at the population scale. This study aimed to summarize the latest research progress and provide up-to-date understanding of the methodology used for the evaluation of the immunoprotection conditions against future variants of SARS-CoV-2. Our novel work reviewed the current methods for the evaluation of the immunoprotection status of a specific population (endogenous antibodies) before and after vaccine inoculation (administered with biopharmaceutical antibody products). The present knowledge of the immunoprotection status regarding the COVID-19 complications was also discussed. Knowledge on the immunoprotection status of specific populations can help guide the design of pharmaceutical antibody products, inform practice guidelines, and develop national regulations with respect to the timing of and need for extra rounds of vaccine boosters.
Collapse
Affiliation(s)
- Congshan Jiang
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, China
| | - Kaichong Jiang
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, China
| | - Xiaowei Li
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ning Zhang
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Wenhua Zhu
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Liesu Meng
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yanmin Zhang
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, China
| | - Shemin Lu
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
32
|
Ng WH, Tang PCH, Mahalingam S, Liu X. Repurposing of drugs targeting the cytokine storm induced by SARS-CoV-2. Br J Pharmacol 2023; 180:133-143. [PMID: 36394425 PMCID: PMC10953344 DOI: 10.1111/bph.15987] [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: 06/06/2022] [Revised: 09/26/2022] [Accepted: 11/02/2022] [Indexed: 11/19/2022] Open
Abstract
A cytokine storm is one of the leading causes of acute respiratory distress syndrome (ARDS) and sepsis-associated multiple organ failure in many respiratory viral infections, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The coronavirus disease 2019 (COVID-19) pandemic has caused millions of deaths worldwide, resulting in an urgent need for effective therapeutic interventions. Repurposing immunosuppressive drugs that target cytokines with immunomodulatory properties is a promising approach to counteract SARS-CoV-2-induced ARDS at the infective and post-infective stages. In this minireview, we examine drugs targeting IL-1β, IL-4/IL-13, IL-6 and TNF-α tested in COVID-19 patients.
Collapse
Affiliation(s)
- Wern Hann Ng
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute QueenslandGriffith UniversityGold CoastQLDAustralia
- Global Virus Network (GVN) Centre of Excellence in ArbovirusesGriffith UniversityGold CoastQLDAustralia
- School of Pharmacy and Medical SciencesGriffith UniversityGold CoastQLDAustralia
| | - Patrick Chun Hean Tang
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute QueenslandGriffith UniversityGold CoastQLDAustralia
- Global Virus Network (GVN) Centre of Excellence in ArbovirusesGriffith UniversityGold CoastQLDAustralia
- School of Pharmacy and Medical SciencesGriffith UniversityGold CoastQLDAustralia
| | - Suresh Mahalingam
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute QueenslandGriffith UniversityGold CoastQLDAustralia
- Global Virus Network (GVN) Centre of Excellence in ArbovirusesGriffith UniversityGold CoastQLDAustralia
- School of Pharmacy and Medical SciencesGriffith UniversityGold CoastQLDAustralia
| | - Xiang Liu
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute QueenslandGriffith UniversityGold CoastQLDAustralia
- Global Virus Network (GVN) Centre of Excellence in ArbovirusesGriffith UniversityGold CoastQLDAustralia
- School of Pharmacy and Medical SciencesGriffith UniversityGold CoastQLDAustralia
| |
Collapse
|
33
|
Cardiac MRI in patients with COVID-19 infection. Eur Radiol 2022; 33:3867-3877. [PMID: 36512043 PMCID: PMC9745285 DOI: 10.1007/s00330-022-09325-x] [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: 05/09/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022]
Abstract
Objective COVID-19 infection is a systemic disease with various cardiovascular symptoms and complications. Cardiac MRI with late gadolinium enhancement is the modality of choice for the assessment of myocardial involvement. T1 and T2 mapping can increase diagnostic accuracy and improve further management. Our study aimed to evaluate the different aspects of myocardial damage in cases of COVID-19 infection using cardiac MRI. Methods This descriptive retrospective study included 86 cases, with a history of COVID-19 infection confirmed by positive RT-PCR, who met the inclusion criteria. Patients had progressive chest pain or dyspnoea with a suspected underlying cardiac cause, either by an abnormal electrocardiogram or elevated troponin levels. Cardiac MRI was performed with late contrast-enhanced (LGE) imaging, followed by T1 and T2 mapping. Results Twenty-four patients have elevated hsTnT with a median hsTnT value of 133 ng/L (IQR: 102 to 159 ng/L); normal value < 14 ng/L. Other sixty-two patients showed elevated hsTnI with a median hsTnI value of 1637 ng/L (IQR: 1340 to 2540 ng/L); normal value < 40 ng/L. CMR showed 52 patients with acute myocarditis, 23 with Takotsubo cardiomyopathy, and 11 with myocardial infarction. Invasive coronary angiography was performed only in selected patients. Conclusion Different COVID-19-related cardiac injuries may cause similar clinical symptoms. Cardiac MRI is the modality of choice to differentiate between the different types of myocardial injury such as Takotsubo cardiomyopathy and infection-related cardiomyopathy or even acute coronary syndrome secondary to vasculitis or oxygen-demand mismatch. Key Points • It is essential to detect early COVID-related cardiac injury using different cardiac biomarkers and cardiac imaging, as it has a significant impact on patient management and outcome. • Cardiac MRI is the modality of choice to differentiate between the different aspects of COVID-related myocardial injury.
Collapse
|
34
|
Maleksabet H, Rezaee E, Tabatabai SA. Host-Cell Surface Binding Targets in SARS-CoV-2 for Drug Design. Curr Pharm Des 2022; 28:3583-3591. [PMID: 36420875 DOI: 10.2174/1381612829666221123111849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/20/2022] [Accepted: 08/31/2022] [Indexed: 11/27/2022]
Abstract
The ongoing pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) became a major public health threat to all countries worldwide. SARS-CoV-2 interactions with its receptor are the first step in the invasion of the host cell. The coronavirus spike protein (S) is crucial in binding to receptors on host cells. Additionally, targeting the SARS-CoV-2 viral receptors is considered a therapeutic option in this regard. In this review of literature, we summarized five potential host cell receptors, as host-cell surface bindings, including angiotensin-converting enzyme 2 (ACE2), neuropilin 1 (NRP-1), dipeptidyl peptidase 4 (DPP4), glucose regulated protein-78 (GRP78), and cluster of differentiation 147 (CD147) related to the SARS-CoV-2 infection. Among these targets, ACE2 was recognized as the main SARS-CoV-2 receptor, expressed at a low/moderate level in the human respiratory system, which is also involved in SARS-CoV-2 entrance, so the virus may utilize other secondary receptors. Besides ACE2, CD147 was discovered as a novel SARS-CoV-2 receptor, CD147 appears to be an alternate receptor for SARSCoV- 2 infection. NRP-1, as a single-transmembrane glycoprotein, has been recently found to operate as an entrance factor and enhance SARS Coronavirus 2 (SARS-CoV-2) infection under in-vitro. DPP4, which was discovered as the first gene clustered with ACE2, may serve as a potential SARS-CoV-2 spike protein binding target. GRP78 could be recognized as a secondary receptor for SARS-CoV-2 because it is widely expressed at substantially greater levels, rather than ACE2, in bronchial epithelial cells and the respiratory mucosa. This review highlights recent literature on this topic.
Collapse
Affiliation(s)
- Hanieh Maleksabet
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Rezaee
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sayyed Abbas Tabatabai
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Karabulut Uzunçakmak S. SARS-CoV-2 Infection and Candidate Biomarkers. Eurasian J Med 2022; 54:16-22. [PMID: 36655440 PMCID: PMC11163343 DOI: 10.5152/eurasianjmed.2022.22305] [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: 10/15/2022] [Accepted: 11/27/2022] [Indexed: 01/19/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 is a virus that can still infect individuals and whose deadly effects continue despite the current vaccines and drugs. Since 2019, many studies on the pathogenesis of the disease have been completed and continue to be done. In addition to the diagnosis and treatment of the disease, many molecules that can be markers of the disease have been investigated. In the early stages of the pandemic, many nonspecific and infection-related laboratory findings and chest computed tomography were used to obtain information about the diagnosis of the disease. The more individual molecules became associated with the disease yet. The purpose of this review is to summarize the impact and role of many molecules associated with coronavirus disease-2019 infection that have been previously used and newly revealed. Numerous studies are summarized in this review. The obtained data show that previously used laboratory findings and new potential biomarkers are not specific to the disease. New potential biomarkers have been associated with the severity of the disease itself, as can be seen with lung imaging and even with routine laboratory findings. One of the important points that are seen frequently in studies is that the effectiveness of these molecules has been shown not only in coronavirus disease-2019 infection but also in many other diseases. This removes the pathogenesis of the disease from being a unique mechanism created by the Severe acute respiratory syndrome coronavirus 2 and provides a general perspective formed by viral or bacterial infections. However, there are still many molecular changes that need to be investigated. Future studies will continue to update themselves with the mutations of the virus.
Collapse
|
36
|
McCann N, Castellino FJ. Cell Entry and Unusual Replication of SARS-CoV-2. Curr Drug Targets 2022; 23:1539-1554. [PMID: 36239725 DOI: 10.2174/1389450124666221014102927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND SARS-CoV-2 is the causative virus for the CoVID-19 pandemic that has frequently mutated to continue to infect and resist available vaccines. Emerging new variants of the virus have complicated notions of immunity conferred by vaccines versus immunity that results from infection. While we continue to progress from epidemic to endemic as a result of this collective immunity, the pandemic remains a morbid and mortal problem. OBJECTIVE The SARS-CoV-2 virus has a very complex manner of replication. The spike protein, one of the four structural proteins of the encapsulated virus, is central to the ability of the virus to penetrate cells to replicate. The objective of this review is to summarize these complex features of viral replication. METHODS A review of the recent literature was performed on the biology of SARS-CoV-2 infection from published work from PubMed and works reported to preprint servers, e.g., bioRxiv and medRxiv. RESULTS AND CONCLUSION The complex molecular and cellular biology involved in SARS-CoV-2 replication and the origination of >30 proteins from a single open reading frame (ORF) have been summarized, as well as the structural biology of spike protein, a critical factor in the cellular entry of the virus, which is a necessary feature for it to replicate and cause disease.
Collapse
Affiliation(s)
- Nathan McCann
- Department of Chemistry and Biochemistry and W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46530, USA
| | - Francis J Castellino
- Department of Chemistry and Biochemistry and W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46530, USA
| |
Collapse
|
37
|
Abstract
Purpose: COVID-19, a novel infection, presented with several complications, including socioeconomical and reproductive health challenges such as erectile dysfunction (ED). The present review summarizes the available shreds of evidence on the impact of COVID-19 on ED.Materials and methods: All published peer-reviewed articles from the onset of the COVID-19 outbreak to date, relating to ED, were reviewed. Results: Available pieces of evidence that ED is a consequence of COVID-19 are convincing. COVID-19 and ED share common risk factors such as disruption of vascular integrity, cardiovascular disease (CVD), cytokine storm, diabetes, obesity, and chronic kidney disease (CKD). COVID-19 also induces impaired pulmonary haemodynamics, increased ang II, testicular damage and low serum testosterone, and reduced arginine-dependent NO bioavailability that promotes reactive oxygen species (ROS) generation and endothelial dysfunction, resulting in ED. In addition, COVID-19 triggers psychological/mental stress and suppresses testosterone-dependent dopamine concentration, which contributes to incident ED.Conclusions: In conclusion, COVID-19 exerts a detrimental effect on male reproductive function, including erectile function. This involves a cascade of events from multiple pathways. As the pandemic dwindles, identifying the long-term effects of COVID-19-induced ED, and proffering adequate and effective measures in militating against COVID-19-induced ED remains pertinent.
Collapse
Affiliation(s)
- D H Adeyemi
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Osun State University, Nigeria
| | - A F Odetayo
- Department of Physiology, University of Ilorin, Ilorin, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
| | - M A Hamed
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- The Brainwill Laboratories, Osogbo, Nigeria
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - R E Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| |
Collapse
|
38
|
Dambha-Miller H, Hinton W, Wilcox CR, Joy M, Feher M, de Lusignan S. Mortality in COVID-19 among women on hormone replacement therapy: a retrospective cohort study. Fam Pract 2022; 39:1049-1055. [PMID: 35577349 PMCID: PMC9129218 DOI: 10.1093/fampra/cmac041] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Limited recent observational data have suggested that there may be a protective effect of oestrogen on the severity of COVID-19 disease. Our aim was to investigate the association between hormone replacement therapy (HRT) or combined oral contraceptive pill (COCP) use and the likelihood of death in women with COVID-19. METHODS We undertook a retrospective cohort study using routinely collected computerized medical records from the Oxford-Royal College of General Practitioners (RCGP) Research and Surveillance Centre (RSC) primary care database. We identified a cohort of 1,863,478 women over 18 years of age from 465 general practices in England. Mixed-effects logistic regression models were used to quantify the association between HRT or COCP use and all-cause mortality among women diagnosed with confirmed or suspected COVID-19 in unadjusted and adjusted models. RESULTS There were 5,451 COVID-19 cases within the cohort. HRT was associated with a reduction in all-cause mortality in COVID-19 (adjusted OR 0.22, 95% CI 0.05 to 0.94). There were no reported events for all-cause mortality in women prescribed COCPs. This prevented further examination of the impact of COCP. CONCLUSIONS We found that HRT prescription within 6 months of a recorded diagnosis of COVID-19 infection was associated with a reduction in all-cause mortality. Further work is needed in larger cohorts to examine the association of COCP in COVID-19, and to further investigate the hypothesis that oestrogens may contribute a protective effect against COVID-19 severity.
Collapse
Affiliation(s)
| | - William Hinton
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | | | - Mark Joy
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Michael Feher
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Simon de Lusignan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK.,Royal College of General Practitioners (RCGP) Research and Surveillance Centre (RSC), London, UK
| |
Collapse
|
39
|
Zhu Q, Xu Y, Wang T, Xie F. Innate and adaptive immune response in SARS-CoV-2 infection-Current perspectives. Front Immunol 2022; 13:1053437. [PMID: 36505489 PMCID: PMC9727711 DOI: 10.3389/fimmu.2022.1053437] [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: 09/25/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been a global pandemic, caused by a novel coronavirus strain with strong infectivity, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). With the in-depth research, the close relationship between COVID-19 and immune system has been dug out. During the infection, macrophages, dendritic cells, natural killer cells, CD8+ T cells, Th1, Th17, Tfh cells and effector B cells are all involved in the anti-SARS-CoV-2 responses, however, the dysfunctional immune responses will ultimately lead to the excessive inflammation, acute lung injury, even other organ failure. Thus, a detailed understanding of pertinent immune response during COVID-19 will provide insights in predicting disease outcomes and developing appropriate therapeutic approaches. In this review, we mainly clarify the role of immune cells in COVID-19 and the target-vaccine development and treatment.
Collapse
Affiliation(s)
- Qiugang Zhu
- Department of Laboratory Medicine, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Yan Xu
- Department of Respiratory Medicine, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Ting Wang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Feiting Xie
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,*Correspondence: Feiting Xie,
| |
Collapse
|
40
|
Ferraro JJ, Reynolds A, Edoigiawerie S, Seu MY, Horen SR, Aminzada A, Hamidian Jahromi A. Impact of gender-affirming hormone therapy on the development of COVID-19 infections and associated complications: A systematic review. World J Methodol 2022; 12:465-475. [PMID: 36479311 PMCID: PMC9720351 DOI: 10.5662/wjm.v12.i6.465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/14/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can produce a wide range of clinical manifestations from asymptomatic to life-threatening. Various researchers have worked to elucidate the pathogenic mechanisms underlying these variable presentations. Differences in individual responses to systemic inflammation and coagulopathy appear to be modulated by several factors, including sex steroid hormones. Transgender men or non-binary individuals who undergo gender-affirming hormone therapy (GAHT) are a unique population of interest for exploring the androgen-mediated coronavirus disease 2019 (COVID-19) hypothesis. As the search for reliable and effective COVID-19 treatments continues, understanding the risks and benefits of GAHT may mitigate COVID-19 related morbidity and mortality in this patient population. AIM To investigate the potential role of GAHT in the development of COVID-19 infections and complications. METHODS This systematic review implemented an algorithmic approach using PRISMA guidelines. PubMed, Scopus, Google Scholar top 100 results, and archives of Plastic and Reconstructive Surgery was on January 12, 2022 using the key words "gender" AND "hormone" AND "therapy" AND "COVID-19" as well as associated terms. Non-English articles, articles published prior to 2019 (prior to COVID-19), and manuscripts in the form of reviews, commentaries, or letters were excluded. References of the selected publications were screened as well. RESULTS The database search resulted in the final inclusion of 14 studies related to GAHT COVID-19. Of the included studies, only two studies directly involved and reported on COVID-19 in transgender patients. Several clinical trials looked at the relationship between testosterone, estrogen, and progesterone in COVID-19 infected cis-gender men and women. It has been proposed that androgens may facilitate initial COVID-19 infection, however, once this occurs, testosterone may have a protective effect. Multiple clinical studies have shown that low baseline testosterone levels in men with COVID-19 are associated with worsening outcomes. The role of female sex hormones, including estrogen and progesterone have also been proposed as potential protective factors in COVID-19 infection. This was exemplified in multiple studies investigating different outcomes in pre- and post-menopausal women as well as those taking hormone replacement therapy. Two studies related specifically to transgender patients and GAHT found that estrogen and progesterone could help protect men against COVID-19, and that testosterone hormone therapy may increase the risk of contracting COVID-19. CONCLUSION Few studies were found related to the role of GAHT in COVID-19 infections. Additional research is necessary to enhance our understanding of this relationship and provide better care for transgender patients.
Collapse
Affiliation(s)
- Jennifer J Ferraro
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, United States
| | - Allie Reynolds
- Undergraduate Studies, Princeton University, Princeton, NJ 08544, United States
| | - Sylvia Edoigiawerie
- Medical School, The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, United States
| | - Michelle Y Seu
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, United States
| | - Sydney R Horen
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, United States
| | - Amir Aminzada
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, United States
| | - Alireza Hamidian Jahromi
- Division of Plastic and Reconstructive Surgery, Temple University Health System, Philadelphia, PA 19140, United States
| |
Collapse
|
41
|
Optic Perineuritis in an Asymptomatic SARS-CoV-2 Infection. Neurologist 2022:00127893-990000000-00044. [PMID: 36730679 DOI: 10.1097/nrl.0000000000000472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Optic perineuritis (OPN) is a previously undescribed sequela of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Here we present a case of OPN that developed several weeks after initial confirmation of the presence of novel coronavirus RNA in the nasopharynx by polymerase chain reaction assay and subsequent confirmation of SARS-CoV-2 IgG seropositivity in the absence of other systemic inflammatory or infectious markers. CASE REPORT An asymptomatic 71-year-old man with noninsulin-dependent diabetes mellitus (NIDDM) tested RNA positive for SARS-CoV-2 during a routine screening of patients at a skilled nursing facility. ~3 weeks after the positive SARS-CoV-2 polymerase chain reaction test, the patient developed subacute ophthalmoparesis of the left eye, horizontal diplopia, retro-orbital pain, and frontal headache. An urgent magnetic resonance imaging of the head and orbits suggested OPN. Cerebrospinal fluid studies were without evidence of other infectious, inflammatory, neoplastic, or paraneoplastic processes. He was started on a 5-day course of high-dose intravenous steroids and later transitioned to oral steroid therapy. Sixteen days after the initiation of steroid therapy, the patient had no headache or retro-orbital pain and demonstrated a marked improvement in horizontal gaze. CONCLUSION SARS-CoV-2-associated neurological sequelae have been increasingly recognized during the current coronavirus disease 2019 pandemic. The present case suggests that patients with confirmed SARS-CoV-2 positivity, even without pulmonary or other classic manifestations of active infection, may manifest diverse clinical presentations including postinfectious OPN that could be related to an underlying autoimmune reactive inflammatory response.
Collapse
|
42
|
Azapira N, Pourjafar S, Habibi A, Tayebi L, Keshtkar S, Kaviani M. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Promising Treatment for COVID-19 Pandemic. EXP CLIN TRANSPLANT 2022; 20:980-983. [PMID: 33622217 DOI: 10.6002/ect.2020.0296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The pandemic of severe acute respiratory syndrome coronavirus-2 infection has prompted the urgent need for novel therapeutic approaches, especially for patients in critically severe conditions. To date, the pathogenesis of COVID-19 is not completely understood, and finding an effective new drug is still inconclusive. Mesenchymal stromal cell-derived extracellular vesicles contain large amounts of proteins, messenger RNA, and microRNAs that act as vehicles that transfer the cargo between cells. These nanotherapeutic materials exert anti-inflammatory effects on the immune system, which are necessary for subsidence of acute inflammation and promotion of tissue repair and regeneration. Therefore, the consideration of mesenchymal stromal cell-derived extracellular vesicles as a new, safe, and effective therapeutic approach in the treatment of COVID-19 pneumonia is suggested.
Collapse
Affiliation(s)
- Negar Azapira
- From the Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | | | | | |
Collapse
|
43
|
Upadhyay M, Gupta S. Endoplasmic reticulum secretory pathway: Potential target against SARS-CoV-2. Virus Res 2022; 320:198897. [PMID: 35988898 PMCID: PMC9387115 DOI: 10.1016/j.virusres.2022.198897] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has recently emerged throughout the world, resulting in more than 400 million cases and over 6 million deaths worldwide as of January 2022. Coronaviruses subvert or use certain aspects of the unfolded protein response in the endoplasmic reticulum to overcome protein translation shutdown to benefit their replication. New virions use the ER-Golgi intermediate compartment to assemble and gain transportation to the cell membrane. Extensive remodeling of the ER has been demonstrated during SARS-CoV-2 infection. In this review article, we discuss the role of the endoplasmic reticulum secretory pathway in the replication cycle of SARS-CoV-2. Currently, there is a dearth of therapeutic options for intervening with SARS-CoV-2 infection. To accelerate drug development, efforts around the globe have been focusing on repurposing drugs that have already been approved for clinical use by regulatory agencies. Targeting the ERS pathway is reasonable, as prior work has shown that SARS-CoV-2 egress is dependent on this pathway. Here we discuss the feasibility of off-patent, FDA-approved, pharmacological inhibitors of the ERS pathway to suppress the SARS-CoV-2 replication cycle, a promising approach that warrants investigation.
Collapse
Affiliation(s)
- Maarisha Upadhyay
- Discipline of Pathology, Cancer Progression and Treatment Research Group, Lambe Institute for Translational Research, School of Medicine, National University of Ireland-Galway, Galway, Ireland
| | - Sanjeev Gupta
- Discipline of Pathology, Cancer Progression and Treatment Research Group, Lambe Institute for Translational Research, School of Medicine, National University of Ireland-Galway, Galway, Ireland.
| |
Collapse
|
44
|
A Review of Routine Laboratory Biomarkers for the Detection of Severe COVID-19 Disease. Int J Anal Chem 2022; 2022:9006487. [PMID: 36267156 PMCID: PMC9578918 DOI: 10.1155/2022/9006487] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/07/2022] [Indexed: 01/08/2023] Open
Abstract
As the COVID-19 pandemic continues, there is an urgent need to identify clinical and laboratory predictors of disease severity and prognosis. Once the coronavirus enters the cell, it triggers additional events via different signaling pathways. Cellular and molecular deregulation evoked by coronavirus infection can manifest as changes in laboratory findings. Understanding the relationship between laboratory biomarkers and COVID-19 outcomes would help in developing a risk-stratified approach to the treatment of patients with this disease. The purpose of this review is to investigate the role of hematological (white blood cell (WBC), lymphocyte, and neutrophil count, neutrophil-to-lymphocyte ratio (NLR), platelet, and red blood cell (RBC) count), inflammatory (C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), and lactate dehydrogenase (LDH)), and biochemical (Albumin, aspartate aminotransferase (AST) and alanine aminotransferase (ALT), blood urea nitrogen (BUN), creatinine, D-dimer, total Cholesterol, low-density lipoprotein (LDL), and high-density lipoprotein (HDL)) biomarkers in the pathogenesis of COVID-19 disease and how their levels vary according to disease severity.
Collapse
|
45
|
Natesan Pushparaj P, Damiati LA, Denetiu I, Bakhashab S, Asif M, Hussain A, Ahmed S, Hamdard MH, Rasool M. Deciphering SARS CoV-2-associated pathways from RNA sequencing data of COVID-19-infected A549 cells and potential therapeutics using in silico methods. Medicine (Baltimore) 2022; 101:e29554. [PMID: 36107502 PMCID: PMC9439635 DOI: 10.1097/md.0000000000029554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Coronavirus (CoV) disease (COVID-19) identified in Wuhan, China, in 2019, is mainly characterized by atypical pneumonia and severe acute respiratory syndrome (SARS) and is caused by SARS CoV-2, which belongs to the Coronaviridae family. Determining the underlying disease mechanisms is central to the identification and development of COVID-19-specific drugs for effective treatment and prevention of human-to-human transmission, disease complications, and deaths. METHODS Here, next-generation RNA sequencing (RNA Seq) data were obtained using Illumina Next Seq 500 from SARS CoV-infected A549 cells and mock-treated A549 cells from the Gene Expression Omnibus (GEO) (GSE147507), and quality control (QC) was assessed before RNA Seq analysis using CLC Genomics Workbench 20.0. Differentially expressed genes (DEGs) were imported into BioJupies to decipher COVID-19 induced signaling pathways and small molecules derived from chemical synthesis or natural sources to mimic or reverse COVID -19 specific gene signatures. In addition, iPathwayGuide was used to identify COVID-19-specific signaling pathways, as well as drugs and natural products with anti-COVID-19 potential. RESULTS Here, we identified the potential activation of upstream regulators such as signal transducer and activator of transcription 2 (STAT2), interferon regulatory factor 9 (IRF9), and interferon beta (IFNβ), interleukin-1 beta (IL-1β), and interferon regulatory factor 3 (IRF3). COVID-19 infection activated key infectious disease-specific immune-related signaling pathways such as influenza A, viral protein interaction with cytokine and cytokine receptors, measles, Epstein-Barr virus infection, and IL-17 signaling pathway. Besides, we identified drugs such as prednisolone, methylprednisolone, diclofenac, compound JQ1, and natural products such as Withaferin-A and JinFuKang as candidates for further experimental validation of COVID-19 therapy. CONCLUSIONS In conclusion, we have used the in silico next-generation knowledge discovery (NGKD) methods to discover COVID-19-associated pathways and specific therapeutics that have the potential to ameliorate the disease pathologies associated with COVID-19.
Collapse
Affiliation(s)
- Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- * Correspondence: Peter Natesan Pushparaj, Department of Medical Laboratory Technology, Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia (e-mail: )
| | | | - Iuliana Denetiu
- King Fahad Medical Research Center, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sherin Bakhashab
- Department of Biochemistry, Faculty of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad Asif
- Department of Biotechnology, BUITEMS, Quetta, Pakistan
- Office of Research Innovation and Commercialization, BUITEMS, Quetta, Pakistan
| | - Abrar Hussain
- Department of Biotechnology, BUITEMS, Quetta, Pakistan
| | - Sagheer Ahmed
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University Islamabad, Pakistan
| | | | - Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
46
|
Gonzalez A, Abrigo J, Achiardi O, Simon F, Cabello-Verrugio C. Intensive care unit-acquired weakness: From molecular mechanisms to its impact in COVID-2019. Eur J Transl Myol 2022; 32. [PMID: 36036350 PMCID: PMC9580540 DOI: 10.4081/ejtm.2022.10511] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/08/2022] [Indexed: 01/08/2023] Open
Abstract
Intensive Care Unit-Acquired Weakness (ICU-AW) is a generalized and symmetric neuromuscular dysfunction associated with critical illness and its treatments. Its incidence is approximately 80% in intensive care unit patients, and it manifests as critical illness polyneuropathy, critical illness myopathy, and muscle atrophy. Intensive care unit patients can lose an elevated percentage of their muscle mass in the first days after admission, producing short- and long-term sequelae that affect patients’ quality of life, physical health, and mental health. In 2019, the world was faced with coronavirus disease 2019 (COVID-19), caused by the acute respiratory syndrome coronavirus 2. COVID-19 produces severe respiratory disorders, such as acute respiratory distress syndrome, which increases the risk of developing ICU-AW. COVID-19 patients treated in intensive care units have shown early diffuse and symmetrical muscle weakness, polyneuropathy, and myalgia, coinciding with the clinical presentation of ICU-AW. Besides, these patients require prolonged intensive care unit stays, invasive mechanical ventilation, and intensive care unit pharmacological therapy, which are risk factors for ICU-AW. Thus, the purposes of this review are to discuss the features of ICU-AW and its effects on skeletal muscle. Further, we will describe the mechanisms involved in the probable development of ICU-AW in severe COVID-19 patients.
Collapse
|
47
|
Chang CC, Vlad G, Vasilescu ER, Li P, Husain SA, Silvia EA, Cohen DJ, Ratner LE, Sun WZ, Mohan S, Suciu-Foca N. Previous SARS-CoV-2 infection or a third dose of vaccine elicited cross-variant neutralising antibodies in vaccinated solid-organ transplant recipients. Clin Transl Immunology 2022; 11:e1411. [PMID: 35979345 PMCID: PMC9371857 DOI: 10.1002/cti2.1411] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 12/24/2022] Open
Abstract
Objectives The SARS‐CoV‐2 pandemic poses a great threat to global health, particularly in solid organ transplant recipients (SOTRs). A 3‐dose mRNA vaccination protocol has been implemented for the majority of SOTRs, yet their immune responses are less effective compared to healthy controls (HCs). Methods We analyzed the humoral immune responses against the vaccine strain and variants of concern (VOC), including the highly mutated‐omicron variant in 113 SOTRs, of whom 44 had recovered from COVID‐19 (recovered‐SOTRs) and 69 had not contracted the virus (COVID‐naïve). In addition, 30 HCs, 8 of whom had recovered from COVID‐19, were also studied. Results Here, we report that three doses of the mRNA vaccine had only a modest effect in eliciting anti‐viral antibodies against all viral strains in the fully vaccinated COVID‐naive SOTRs (n = 47). Only 34.0% of this group of patients demonstrated both detectable anti‐RBD IgG with neutralization activities against alpha, beta, and delta variants, and only 8.5% of them showed additional omicron neutralizing capacities. In contrast, 79.5% of the recovered‐SOTRs who received two doses of vaccine demonstrated both higher anti‐RBD IgG levels and neutralizing activities against all VOC, including omicron. Conclusion These findings illustrate a significant impact of previous infection on the development of anti‐SARS‐CoV‐2 immune responses in vaccinated SOTRs and highlight the need for alternative strategies to protect a subset of a lesser‐vaccine responsive population.
Collapse
Affiliation(s)
- Chih-Chao Chang
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| | - George Vlad
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| | - Elena Rodica Vasilescu
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| | - Ping Li
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| | - Syed A Husain
- Division of Nephrology, Department of Medicine Columbia University Irving Medical Center New York NY USA.,The Columbia University Renal Epidemiology (CURE) Group New York NY USA
| | - Elaine A Silvia
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| | - David J Cohen
- Division of Nephrology, Department of Medicine Columbia University Irving Medical Center New York NY USA.,The Columbia University Renal Epidemiology (CURE) Group New York NY USA
| | - Lloyd E Ratner
- Department of Surgery Columbia University Irving Medical Center New York NY USA
| | - Wei-Zen Sun
- Department of Anesthesiology National Taiwan University Hospital Taipei Taiwan
| | - Sumit Mohan
- Division of Nephrology, Department of Medicine Columbia University Irving Medical Center New York NY USA.,The Columbia University Renal Epidemiology (CURE) Group New York NY USA
| | - Nicole Suciu-Foca
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| |
Collapse
|
48
|
Shin J, Toyoda S, Nishitani S, Onodera T, Fukuda S, Kita S, Fukuhara A, Shimomura I. SARS-CoV-2 infection impairs the insulin/IGF signaling pathway in the lung, liver, adipose tissue, and pancreatic cells via IRF1. Metabolism 2022; 133:155236. [PMID: 35688210 PMCID: PMC9173833 DOI: 10.1016/j.metabol.2022.155236] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/11/2022] [Accepted: 06/01/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND COVID-19 can cause multiple organ damages as well as metabolic abnormalities such as hyperglycemia, insulin resistance, and new onset of diabetes. The insulin/IGF signaling pathway plays an important role in regulating energy metabolism and cell survival, but little is known about the impact of SARS-CoV-2 infection. The aim of this work was to investigate whether SARS-CoV-2 infection impairs the insulin/IGF signaling pathway in the host cell/tissue, and if so, the potential mechanism and association with COVID-19 pathology. METHODS To determine the impact of SARS-CoV-2 on insulin/IGF signaling pathway, we utilized transcriptome datasets of SARS-CoV-2 infected cells and tissues from public repositories for a wide range of high-throughput gene expression data: autopsy lungs from COVID-19 patients compared to the control from non-COVID-19 patients; lungs from a human ACE2 transgenic mouse infected with SARS-CoV-2 compared to the control infected with mock; human pluripotent stem cell (hPSC)-derived liver organoids infected with SARS-CoV-2; adipose tissues from a mouse model of COVID-19 overexpressing human ACE2 via adeno-associated virus serotype 9 (AAV9) compared to the control GFP after SARS-CoV-2 infection; iPS-derived human pancreatic cells infected with SARS-CoV-2 compared to the mock control. Gain and loss of IRF1 function models were established in HEK293T and/or Calu3 cells to evaluate the impact on insulin signaling. To understand the mechanistic regulation and relevance with COVID-19 risk factors, such as older age, male sex, obesity, and diabetes, several transcriptomes of human respiratory, metabolic, and endocrine cells and tissue were analyzed. To estimate the association with COVID-19 severity, whole blood transcriptomes of critical patients with COVID-19 compared to those of hospitalized noncritical patients with COVID-19. RESULTS We found that SARS-CoV-2 infection impaired insulin/IGF signaling pathway genes, such as IRS, PI3K, AKT, mTOR, and MAPK, in the host lung, liver, adipose tissue, and pancreatic cells. The impairments were attributed to interferon regulatory factor 1 (IRF1), and its gene expression was highly relevant to risk factors for severe COVID-19; increased with aging in the lung, specifically in men; augmented by obese and diabetic conditions in liver, adipose tissue, and pancreatic islets. IRF1 activation was significantly associated with the impaired insulin signaling in human cells. IRF1 intron variant rs17622656-A, which was previously reported to be associated with COVID-19 prevalence, increased the IRF1 gene expression in human tissue and was frequently found in American and European population. Critical patients with COVID-19 exhibited higher IRF1 and lower insulin/IGF signaling pathway genes in the whole blood compared to hospitalized noncritical patients. Hormonal interventions, such as dihydrotestosterone and dexamethasone, ameliorated the pathological traits in SARS-CoV-2 infectable cells and tissues. CONCLUSIONS The present study provides the first scientific evidence that SARS-CoV-2 infection impairs the insulin/IGF signaling pathway in respiratory, metabolic, and endocrine cells and tissues. This feature likely contributes to COVID-19 severity with cell/tissue damage and metabolic abnormalities, which may be exacerbated in older, male, obese, or diabetic patients.
Collapse
Affiliation(s)
- Jihoon Shin
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Diabetes Care Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Shinichiro Toyoda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shigeki Nishitani
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Toshiharu Onodera
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shiro Fukuda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shunbun Kita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Adipose Management, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Atsunori Fukuhara
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Adipose Management, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
49
|
Dizaji Asl K, Mazloumi Z, Majidi G, Kalarestaghi H, Sabetkam S, Rafat A. NK cell dysfunction is linked with disease severity in SARS-CoV-2 patients. Cell Biochem Funct 2022; 40:559-568. [PMID: 35833321 PMCID: PMC9350078 DOI: 10.1002/cbf.3725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/16/2022] [Accepted: 06/02/2022] [Indexed: 12/13/2022]
Abstract
SARS-CoV-2 first raised from Wuhan City, Hubei Province in November 2019. The respiratory disorder, cough, weakness, fever are the main clinical symptoms of coronavirus disease 2019 (COVID-19) patients. Natural Killer (NK) cells as a first defense barrier of innate immune system have an essential role in early defense against pulmonary virus. They kill the infected cells by inducing apoptosis or the degranulation of perforin and granzymes. Collectively, NK cells function are coordinated by the transmitted signals from activating and inhibitory receptors. It is clear that the cytotoxic function of NK cells is disrupted in COVID-19 patients due to the dysregulation of activating and inhibitory receptors. Therefore, better understanding of the activating and inhibitory receptors mechanism could facilitate the treatment strategy in clinic. To improve the efficacy of immunotherapy in COVID-19 patients, the functional detail of NK cell and manipulation of their key checkpoints are gathered in current review.
Collapse
Affiliation(s)
- Khadijeh Dizaji Asl
- Clinical Research Development Unit of Tabriz Valiasr HospitalTabriz University of Medical SciencesTabrizIran
- Department of Histopathology and Anatomy, Faculty of Medicine, Tabriz BranchIslamic Azad UniversityTabrizIran
| | - Zeinab Mazloumi
- Department of Medical Applied Cell Sciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Ghazal Majidi
- Faculty of MedicineTabriz University of Medical SciencesTabrizIran
| | - Hossein Kalarestaghi
- Research Laboratory for Embryology and Stem Cell, Department of Anatomical Sciences, School of MedicineArdabil University of Medical SciencesArdabilIran
| | - Shahnaz Sabetkam
- Department of Histopathology and Anatomy, Faculty of Medicine, Tabriz BranchIslamic Azad UniversityTabrizIran
| | - Ali Rafat
- Department of Anatomical SciencesTabriz University of Medical SciencesTabrizIran
- Anatomical Sciences Research CenterKashan University of Medical SciencesKashanIran
| |
Collapse
|
50
|
COVID-19, Mucormycosis and Cancer: The Triple Threat-Hypothesis or Reality? J Pers Med 2022; 12:jpm12071119. [PMID: 35887616 PMCID: PMC9320339 DOI: 10.3390/jpm12071119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/04/2022] [Accepted: 07/08/2022] [Indexed: 01/09/2023] Open
Abstract
COVID-19 has been responsible for widespread morbidity and mortality worldwide. Invasive mucormycosis has death rates scaling 80%. India, one of the countries hit worst by the pandemic, is also a hotbed with the highest death rates for mucormycosis. Cancer, a ubiquitously present menace, also contributes to higher case fatality rates. All three entities studied here are individual, massive healthcare threats. The danger of one disease predisposing to the other, the poor performance status of patients with all three diseases, the impact of therapeutics for one disease on the pathology and therapy of the others all warrant physicians having a better understanding of the interplay. This is imperative so as to effectively establish control over the individual patient and population health. It is important to understand the interactions to effectively manage all three entities together to reduce overall morbidity. In this review article, we search for an inter-relationship between the COVID-19 pandemic, emerging mucormycosis, and the global giant, cancer.
Collapse
|