151
|
Baltu D, Kurt-Sukur ED, Tastemel Ozturk T, Gulhan B, Ozaltin F, Duzova A, Topaloglu R. COVID-19 in Children with Chronic Kidney Disease; Does it Differ Much? KLINISCHE PADIATRIE 2024. [PMID: 38224686 DOI: 10.1055/a-2207-3153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
BACKGROUND COVID-19 is known to have a mild course in children, however more data on pediatric chronic kidney disease (CKD) is needed. We aimed to assess the incidence and severity of COVID-19 in pediatric CKD patients. METHODS A questionnaire including demographics, COVID-19 history, symptoms, and vaccination status was applied to patients with CKD. We also retrospectively reviewed the presentation and outcomes of SARS-CoV-2 infection in this patient group from March 2020 to December 2021. RESULTS 220 patients were included, 48 were found to have experienced COVID-19. There was no significant difference regarding age, gender, underlying kidney disease, CKD stage, dialysis status, type or number of immunosuppressive medications, and glomerular filtration rate between patients with and without COVID-19. Most were infected by a household member (43.8%) and during outpatient or inpatient care (18.8%). Four (8.3%) were asymptomatic, and 43 (89.6%) had mild infection. Severe COVID-19 was observed in only one patient. Eleven (22.9%) patients with COVID-19 were previously vaccinated. Acute kidney injury was detected in 4 (8.3%); as stage 1 in all. Median follow-up after COVID-19 was 4.6 months. All patients fully recovered, and no renal disease flare or death was observed. CONCLUSIONS Although the vaccination rate was low in our cohort, the majority of the children with COVID-19 showed a mild course. Along with the vaccination, general precautions seemed to be successful for this population.
Collapse
Affiliation(s)
- Demet Baltu
- Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | | | | | - Bora Gulhan
- Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Fatih Ozaltin
- Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ali Duzova
- Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Rezan Topaloglu
- Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
152
|
Misyurina EN, Baryakh EA, Frolova NF, Kotenko ON, Mutovina ZY, Andreev SS, Tolstykh TN, Yatskov KV, Karimova EA, Makeshova AB, Rukavitsyn OA, Misyurin AV, Polyakov YY, Mingalimov MA, Chudnova TS, Gagloeva DE, Ivanova DD, Koneva AI, Kochneva OL, Zotina EN, Grishina EY, Shimanovskaya LT, Yakimets VN, Zhelnova EI. Basic therapeutic approaches to the management of hematology/oncology patients with new coronavirus infection (COVID-19). ONCOHEMATOLOGY 2024; 18:10-39. [DOI: 10.17650/1818-8346-2023-18-4(suppl)-10-39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
In December 2019, cases of severe respiratory infection were reported in Wuhan, China. The disease was caused by a new, previously undescribed coronavirus, structurally similar to the then known SARS-CoV virus. The World Health Organization has named the new virus SARS-CoV-2 and the disease it causes COVID-19. The problem of COVID-19 is exacerbated by the rapid spread of the SARS-CoV-2 virus and the development of life-threatening complications, the main of which is pneumonia. Due to the severity of the condition, from 5 to 10 % of patients are treated in intensive care units.SARS-CoV-2 initially attacks the respiratory system and causes symptoms such as fever, vomiting, headache, dizziness, general weakness, and diarrhea. Then these symptoms intensify in different directions, and the disease can often lead to death.Initially, only a few methods of symptomatic treatment were available and clinical trials of drugs that had previously shown their effectiveness against infection with the MERS-CoV and SARS-CoV viruses began. Temporary recommendations have appeared suggesting the use of some drugs both in monotherapy and in combination.In patients with hematologic malignancies, the immune response to the SARS-CoV-2 coronavirus is significantly reduced, which explains the high mortality rate (up to 38 %) of these patients hospitalized for SARS-CoV-2 infection. Recently, antiviral drugs and monoclonal antibodies have become available for pre- or post-exposure prophylaxis, as well as for early treatment of COVID-19. These treatments should be offered to patients at high risk of severe COVID-19 and to those who have not responded to vaccination. However, as changes in the genetic structure of the virus accumulate, some treatments may lose their clinical effectiveness against new variants.The combination of hematological malignancies and new coronavirus infection causes a more severe course of COVID-19 compared to the population and high mortality. Factors for an unfavorable prognosis for new coronavirus infection in patients with hematological malignancies include age over 60 years, a high comorbidity index, diagnoses such as acute leukemia, especially acute myeloid leukemia and myelodysplastic syndrome, disease status (relapse, progression, as well as newly diagnosed acute leukemia), severe COVID-19, agranulocytosis (myelotoxic or tumor).
Collapse
Affiliation(s)
- E. N. Misyurina
- City Clinical Hospital No. 52, Moscow Healthcare Department; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)
| | - E. A. Baryakh
- City Clinical Hospital No. 52, Moscow Healthcare Department; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University); Russian Medical Academy of Continuing Professional Education, Ministry of Health of Russia; N.I. Pirogov Russian National Research Medical University, Ministry of Health of Russia
| | - N. F. Frolova
- City Clinical Hospital No. 52, Moscow Healthcare Department; 5A.I. EvdokimovMoscow State University of Medicine and Dentistry, Ministry of Health of Russia
| | - O. N. Kotenko
- City Clinical Hospital No. 52, Moscow Healthcare Department
| | | | - S. S. Andreev
- City Clinical Hospital No. 52, Moscow Healthcare Department
| | - T. N. Tolstykh
- City Clinical Hospital No. 52, Moscow Healthcare Department; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)
| | - K. V. Yatskov
- City Clinical Hospital No. 52, Moscow Healthcare Department
| | - E. A. Karimova
- City Clinical Hospital No. 52, Moscow Healthcare Department
| | - A. B. Makeshova
- City Clinical Hospital No. 52, Moscow Healthcare Department; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)
| | - O. A. Rukavitsyn
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)
| | - A. V. Misyurin
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)
| | - Yu. Yu. Polyakov
- City Clinical Hospital No. 52, Moscow Healthcare Department; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)
| | - M. A. Mingalimov
- City Clinical Hospital No. 52, Moscow Healthcare Department; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)
| | - T. S. Chudnova
- City Clinical Hospital No. 52, Moscow Healthcare Department; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)
| | - D. E. Gagloeva
- City Clinical Hospital No. 52, Moscow Healthcare Department; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)
| | - D. D. Ivanova
- City Clinical Hospital No. 52, Moscow Healthcare Department
| | - A. I. Koneva
- City Clinical Hospital No. 52, Moscow Healthcare Department
| | - O. L. Kochneva
- City Clinical Hospital No. 52, Moscow Healthcare Department
| | - E. N. Zotina
- City Clinical Hospital No. 52, Moscow Healthcare Department; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)
| | | | | | - V. N. Yakimets
- City Clinical Hospital No. 52, Moscow Healthcare Department
| | - E. I. Zhelnova
- City Clinical Hospital No. 52, Moscow Healthcare Department; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)
| |
Collapse
|
153
|
Panagiotides NG, Poledniczek M, Andreas M, Hülsmann M, Kocher AA, Kopp CW, Piechota-Polanczyk A, Weidenhammer A, Pavo N, Wadowski PP. Myocardial Oedema as a Consequence of Viral Infection and Persistence-A Narrative Review with Focus on COVID-19 and Post COVID Sequelae. Viruses 2024; 16:121. [PMID: 38257821 PMCID: PMC10818479 DOI: 10.3390/v16010121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Microvascular integrity is a critical factor in myocardial fluid homeostasis. The subtle equilibrium between capillary filtration and lymphatic fluid removal is disturbed during pathological processes leading to inflammation, but also in hypoxia or due to alterations in vascular perfusion and coagulability. The degradation of the glycocalyx as the main component of the endothelial filtration barrier as well as pericyte disintegration results in the accumulation of interstitial and intracellular water. Moreover, lymphatic dysfunction evokes an increase in metabolic waste products, cytokines and inflammatory cells in the interstitial space contributing to myocardial oedema formation. This leads to myocardial stiffness and impaired contractility, eventually resulting in cardiomyocyte apoptosis, myocardial remodelling and fibrosis. The following article reviews pathophysiological inflammatory processes leading to myocardial oedema including myocarditis, ischaemia-reperfusion injury and viral infections with a special focus on the pathomechanisms evoked by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In addition, clinical implications including potential long-term effects due to viral persistence (long COVID), as well as treatment options, are discussed.
Collapse
Affiliation(s)
- Noel G. Panagiotides
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Michael Poledniczek
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | - Martin Andreas
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Martin Hülsmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Alfred A. Kocher
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | | | - Annika Weidenhammer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Noemi Pavo
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
154
|
Pociūtė A, Kriaučiūnaitė K, Kaušylė A, Zablockienė B, Alčauskas T, Jelinskaitė A, Rudėnaitė A, Jančorienė L, Ročka S, Verkhratsky A, Pivoriūnas A. Plasma of COVID-19 Patients Does Not Alter Electrical Resistance of Human Endothelial Blood-Brain Barrier In Vitro. FUNCTION 2024; 5:zqae002. [PMID: 38486975 PMCID: PMC10935481 DOI: 10.1093/function/zqae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 03/17/2024] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 instigated the most serious global health crisis. Clinical presentation of COVID-19 frequently includes severe neurological and neuropsychiatric symptoms. However, it is presently unknown whether and to which extent pathological impairment of blood-brain barrier (BBB) contributes to the development of neuropathology during COVID-19 progression. In the present study, we used human induced pluripotent stem cells-derived brain endothelial cells (iBECs) to study the effects of blood plasma derived from COVID-19 patients on the BBB integrity in vitro. We also performed a comprehensive analysis of the cytokine and chemokine profiles in the plasma of COVID-19 patients, healthy and recovered individuals. We found significantly increased levels of interferon γ-induced protein 10 kDa, hepatocyte growth factor, and interleukin-18 in the plasma of COVID-19 patients. However, blood plasma from COVID-19 patients did not affect transendothelial electrical resistance in iBEC monolayers. Our results demonstrate that COVID-19-associated blood plasma inflammatory factors do not affect BBB paracellular pathway directly and suggest that pathological remodeling (if any) of BBB during COVID-19 may occur through indirect or yet unknown mechanisms.
Collapse
Affiliation(s)
- Agnė Pociūtė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
| | - Karolina Kriaučiūnaitė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
| | - Aida Kaušylė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
| | - Birutė Zablockienė
- Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
- Centre of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, LT-08406 Vilnius, Lithuania
| | - Tadas Alčauskas
- Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
| | - Augustė Jelinskaitė
- Centre of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, LT-08406 Vilnius, Lithuania
| | - Akvilė Rudėnaitė
- Centre of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, LT-08406 Vilnius, Lithuania
| | - Ligita Jančorienė
- Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
- Centre of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, LT-08406 Vilnius, Lithuania
| | - Saulius Ročka
- Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
- Center of Neurosurgery, Vilnius University Hospital Santaros Klinikos, LT-08661 Vilnius, Lithuania
| | - Alexei Verkhratsky
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, 110052, China
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
| |
Collapse
|
155
|
Radwan E, Abdelaziz A, Mandour MAM, Meki ARMA, El-Kholy MM, Mohamed MN. MBOAT7 expression is associated with disease progression in COVID-19 patients. Mol Biol Rep 2024; 51:79. [PMID: 38183501 PMCID: PMC10771377 DOI: 10.1007/s11033-023-09009-9] [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: 09/06/2023] [Accepted: 10/09/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND AND AIM The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in late 2019 caused a pandemic of acute respiratory disease, named coronavirus disease 2019 (COVID-19). COVID-19 became one of the most challenging health emergencies, hence the necessity to find different prognostic factors for disease progression, and severity. Membrane bound O-acyltransferase domain containing 7 (MBOAT7) demonstrates anti-inflammatory effects through acting as a fine-tune regulator of the amount of cellular free arachidonic acid. We aimed in this study to evaluate MBOAT7 expression in COVID-19 patients and to correlate it with disease severity and outcomes. METHODS This case-control study included 56 patients with confirmed SARS-CoV-2 diagnosis and 28 control subjects. Patients were further classified into moderate (n = 28) and severe (n = 28) cases. MBOAT7, tumor necrosis factor-α (TNF-α), and interleukin-1ß (IL-1ß) mRNA levels were evaluated in peripheral blood mononuclear cells (PBMC) samples isolated from patients and control subjects by real time quantitative polymerase chain reaction (RT-qPCR). In addition, circulating MBOAT7 protein levels were assayed by enzyme-linked immunosorbent assay (ELISA). RESULTS Significant lower levels of circulating MBOAT7 mRNA and protein were observed in COVID-19 patients compared to control subjects with severe COVID-19 cases showing significant lower levels compared to moderate cases. Moreover, severe cases showed a significant upregulation of TNF-α and IL-1ß mRNA. MBOAT7 mRNA and protein levels were significantly correlated with inflammatory markers (TNF-α, IL-1ß, C-reactive protein (CRP), and ferritin), liver enzymes, severity, and oxygen saturation levels. CONCLUSION COVID-19 is associated with downregulation of MBAOT7, which correlates with disease severity.
Collapse
Affiliation(s)
- Eman Radwan
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt.
- Department of Biochemistry, Sphinx University, New Assiut City, Assiut 10, Egypt.
| | - Ahmed Abdelaziz
- Department of Biochemistry, Faculty of Pharmacy, Assiut University, Assiut, 71515, Egypt
| | - Manal A M Mandour
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Abdel-Raheim M A Meki
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
- Department of Biochemistry, Sphinx University, New Assiut City, Assiut 10, Egypt
| | - Maha M El-Kholy
- Department of Chest diseases and Tuberculosis, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Marwan N Mohamed
- Department of Chest diseases and Tuberculosis, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
156
|
Caturano A, Galiero R, Vetrano E, Medicamento G, Alfano M, Beccia D, Brin C, Colantuoni S, Di Salvo J, Epifani R, Nevola R, Marfella R, Sardu C, Coppola C, Scarano F, Maggi P, Calabrese C, De Lucia Sposito P, Rescigno C, Sbreglia C, Fraganza F, Parrella R, Romano A, Calabria G, Polverino B, Pagano A, Numis FG, Bologna C, Nunziata M, Esposito V, Coppola N, Maturo N, Nasti R, Di Micco P, Perrella A, Adinolfi LE, Di Domenico M, Monda M, Russo V, Ruggiero R, Docimo G, Rinaldi L, Sasso FC. Impact of Acute Kidney Injury on the COVID-19 In-Hospital Mortality in Octogenarian Patients: Insights from the COVOCA Study. Life (Basel) 2024; 14:86. [PMID: 38255701 PMCID: PMC10817510 DOI: 10.3390/life14010086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/01/2024] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND AND AIMS The COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, has fundamentally reshaped the landscape of global public health, with some people suffering more adverse clinical outcomes than others. The aim of this study is to deepen our understanding of the specific impact of acute kidney injury (AKI) on the in-hospital mortality in octogenarian patients with COVID-19. METHODS This is a prospective observational cohort study, which involved 23 COVID-19 hospital units in the Campania Region, Italy. Exposure variables were collected during hospital admission and at discharge. Only patients aged ≥80 years were deemed eligible for the study. RESULTS 197 patients were included in the study (median age 83.0 [82.0-87.0] years; 51.5% men), with a median duration of hospitalization of 15.0 [8.0-25.0] days. From the multivariable Cox regression analysis, after the application of Šidák correction, only the respiratory rate (HR 1.09, 95% CI: 1.04 to 1.14; p < 0.001) and AKI development (HR: 3.40, 95% CI: 1.80 to 6.40; p < 0.001) were independently associated with the primary outcome. Moreover, the Kaplan-Meier analysis showed a significantly different risk of in-hospital mortality between patients with and without AKI (log-rank: <0.0001). CONCLUSIONS In our investigation, we identified a significant association between AKI and mortality rates among octogenarian patients admitted for COVID-19. These findings raise notable concerns and emphasize the imperative for vigilant monitoring of this demographic cohort.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Giulia Medicamento
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Maria Alfano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Domenico Beccia
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Chiara Brin
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Sara Colantuoni
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Jessica Di Salvo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Raffaella Epifani
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Riccardo Nevola
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
- Ospedale Evangelico Betania, 80147 Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Carmine Coppola
- Hepatology Unit, Internal Medicine, Area Stabiese Hospital, 80053 Naples, Italy;
| | - Ferdinando Scarano
- COVID Center “S. Anna e SS. Madonna della Neve” Hospital, 80042 Boscotrecase, Italy;
| | - Paolo Maggi
- U.O.C. Infectious and Tropical Diseases, S. Anna e S. Sebastiano Hospital, 81100 Caserta, Italy;
| | - Cecilia Calabrese
- U.O.C. Pneumologia Vanvitelli, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy;
| | | | - Carolina Rescigno
- U.O.C. Infectious Diseases and Neurology, Cotugno Hospital, 80131 Naples, Italy;
| | - Costanza Sbreglia
- U.O.C. Infectious Diseases of the Elderly, Cotugno Hospital, 80131 Naples, Italy;
| | - Fiorentino Fraganza
- U.O.C. Anestesia and Intensive Care Unit, Cotugno Hospital, 80131 Naples, Italy;
| | - Roberto Parrella
- U.O.C. Respiratory Infectious Diseases, Cotugno Hospital, 80131 Naples, Italy;
| | | | - Giosuele Calabria
- IXth Division of Infectious Diseases and Interventional Ultrasound, Cotugno Hospital, 80131 Naples, Italy;
| | | | - Antonio Pagano
- Emergency and Acceptance Unit, “Santa Maria delle Grazie” Hospital, 80078 Pozzuoli, Italy; (A.P.); (F.G.N.)
| | - Fabio Giuliano Numis
- Emergency and Acceptance Unit, “Santa Maria delle Grazie” Hospital, 80078 Pozzuoli, Italy; (A.P.); (F.G.N.)
| | | | | | - Vincenzo Esposito
- IVth Division of Immunodeficiency and Gender Infectious Diseases, Cotugno Hospital, 80131 Naples, Italy;
| | - Nicola Coppola
- COVID Center, Department of Mental Health and Public Medicine, A.O.U. Vanvitelli, 80131 Naples, Italy;
| | - Nicola Maturo
- U.O.S.D. Infectious Diseases Emergency and Acceptance, Cotugno Hospital, 80131 Naples, Italy;
| | - Rodolfo Nasti
- Emergency Division, A.O.R.N. “Antonio Cardarelli”, 80131 Naples, Italy;
| | - Pierpaolo Di Micco
- Department of Internal Medicine, Fatebenefratelli Hospital of Naples, 80123 Naples, Italy;
| | | | - Luigi Elio Adinolfi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| | - Marina Di Domenico
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy;
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Vincenzo Russo
- Department of Biology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA;
- Division of Cardiology, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Roberto Ruggiero
- Division of General, Oncological, Mini-Invasive and Obesity Surgery, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy;
| | - Giovanni Docimo
- Unit of Thyroid Surgery, Department of Medical and Advanced Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Luca Rinaldi
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy; (R.G.); (E.V.); (G.M.); (M.A.); (D.B.); (C.B.); (S.C.); (J.D.S.); (R.E.); (R.N.); (R.M.); (C.S.); (L.E.A.); (F.C.S.)
| |
Collapse
|
157
|
Yang B, Pan M, Feng K, Wu X, Yang F, Yang P. Identification of the feature genes involved in cytokine release syndrome in COVID-19. PLoS One 2024; 19:e0296030. [PMID: 38165854 PMCID: PMC10760774 DOI: 10.1371/journal.pone.0296030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/04/2023] [Indexed: 01/04/2024] Open
Abstract
OBJECTIVE Screening of feature genes involved in cytokine release syndrome (CRS) from the coronavirus disease 19 (COVID-19). METHODS The data sets related to COVID-19 were retrieved using Gene Expression Omnibus (GEO) database, the differentially expressed genes (DEGs) related to CRS were analyzed with R software and Venn diagram, and the biological processes and signaling pathways involved in DEGs were analyzed with GO and KEGG enrichment. Core genes were screened using Betweenness and MCC algorithms. GSE164805 and GSE171110 dataset were used to verify the expression level of core genes. Immunoinfiltration analysis was performed by ssGSEA algorithm in the GSVA package. The DrugBank database was used to analyze the feature genes for potential therapeutic drugs. RESULTS This study obtained 6950 DEGs, of which 971 corresponded with CRS disease genes (common genes). GO and KEGG enrichment showed that multiple biological processes and signaling pathways associated with common genes were closely related to the inflammatory response. Furthermore, the analysis revealed that transcription factors that regulate these common genes are also involved in inflammatory response. Betweenness and MCC algorithms were used for common gene screening, yielding seven key genes. GSE164805 and GSE171110 dataset validation revealed significant differences between the COVID-19 and normal controls in four core genes (feature genes), namely IL6R, TLR4, TLR2, and IFNG. The upregulated IL6R, TLR4, and TLR2 genes were mainly involved in the Toll-like receptor signaling pathway of the inflammatory pathway, while the downregulated IFNG genes primarily participated in the necroptosis and JAK-STAT signaling pathways. Moreover, immune infiltration analysis indicated that higher expression of these genes was associated with immune cell infiltration that mediates inflammatory response. In addition, potential therapeutic drugs for these four feature genes were identified via the DrugBank database. CONCLUSION IL6R, TLR4, TLR2, and IFNG may be potential pathogenic genes and therapeutic targets for the CRS associated with COVID-19.
Collapse
Affiliation(s)
- Bing Yang
- The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Meijun Pan
- The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Kai Feng
- The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xue Wu
- The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Fang Yang
- The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Peng Yang
- The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
158
|
Ide Y, Urushibata N, Takayama W, Hondo K, Aiboshi J, Otomo Y. Clinical characteristics of pneumothorax and pneumomediastinum in mechanical ventilated patients with coronavirus disease 2019: a case series. J Med Case Rep 2024; 18:7. [PMID: 38166996 PMCID: PMC10759624 DOI: 10.1186/s13256-023-04281-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/21/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Pneumothorax (PTX) and pneumomediastinum (PM) have been reported as potential complications in patients with coronavirus disease 2019 (COVID-19); however, their risk factors and etiology remain unknown. Herein, we investigated the clinical characteristics of mechanically ventilated patients with COVID-19 with PTX or PM. METHODS We examined patients with severe COVID-19 requiring mechanical ventilation who were admitted to the intensive care unit of a tertiary-level emergency medical center in Tokyo, Japan between April 1, 2020. and October 31, 2021. We collected and analyzed the clinical characteristics of the patients who presented with either PTX or PM during mechanical ventilation. RESULTS During the study period, a total of 165 patients required mechanical ventilation, and 15 patients with PTX/PM during mechanical ventilation were selected. Three patients with obvious causes were excluded, and the remaining 12 patients were analyzed (7.3%). The mortality rate in these patients was as high as 50%, demonstrating the difficulty of treatment in the presence of PTX/PM. PTX/PM occurred 14.5 days after intubation. A peak pressure of > 30 cmH2O was only apparent in one patient, suggesting that high positive pressure ventilation may be less involved than mentioned in the literature. In addition, the inspiratory effort was not strong in our group of patients. (P0.1 was 2.1 cm H2O [1.0-3.8]). CONCLUSION Various factors are associated with the development of PTX/PM in patients on mechanical ventilation for COVID-19. We did not find a strong correlation between PTM/PM and barotrauma or strong inspiratory efforts, which have been identified as potential causes in previous studies.
Collapse
Affiliation(s)
- Yohei Ide
- Trauma and Acute Critical Center, Tokyo Medical and Dental University Hospital of Medicine, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-0034, Japan
| | - Nao Urushibata
- Trauma and Acute Critical Center, Tokyo Medical and Dental University Hospital of Medicine, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-0034, Japan.
| | - Wataru Takayama
- Trauma and Acute Critical Center, Tokyo Medical and Dental University Hospital of Medicine, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-0034, Japan
| | - Kenichi Hondo
- Trauma and Acute Critical Center, Tokyo Medical and Dental University Hospital of Medicine, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-0034, Japan
| | - Junichi Aiboshi
- Trauma and Acute Critical Center, Tokyo Medical and Dental University Hospital of Medicine, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-0034, Japan
| | - Yasuhiro Otomo
- Trauma and Acute Critical Center, Tokyo Medical and Dental University Hospital of Medicine, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-0034, Japan
| |
Collapse
|
159
|
Sivagurunathan N, Calivarathan L. SARS-CoV-2 Infection to Premature Neuronal Aging and Neurodegenerative Diseases: Is there any Connection with Hypoxia? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:431-448. [PMID: 37073650 DOI: 10.2174/1871527322666230418114446] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 01/28/2023] [Accepted: 02/09/2023] [Indexed: 04/20/2023]
Abstract
The pandemic of coronavirus disease-2019 (COVID-19), caused by SARS-CoV-2, has become a global concern as it leads to a spectrum of mild to severe symptoms and increases death tolls around the world. Severe COVID-19 results in acute respiratory distress syndrome, hypoxia, and multi- organ dysfunction. However, the long-term effects of post-COVID-19 infection are still unknown. Based on the emerging evidence, there is a high possibility that COVID-19 infection accelerates premature neuronal aging and increases the risk of age-related neurodegenerative diseases in mild to severely infected patients during the post-COVID period. Several studies correlate COVID-19 infection with neuronal effects, though the mechanism through which they contribute to the aggravation of neuroinflammation and neurodegeneration is still under investigation. SARS-CoV-2 predominantly targets pulmonary tissues and interferes with gas exchange, leading to systemic hypoxia. The neurons in the brain require a constant supply of oxygen for their proper functioning, suggesting that they are more vulnerable to any alteration in oxygen saturation level that results in neuronal injury with or without neuroinflammation. We hypothesize that hypoxia is one of the major clinical manifestations of severe SARS-CoV-2 infection; it directly or indirectly contributes to premature neuronal aging, neuroinflammation, and neurodegeneration by altering the expression of various genes responsible for the survival of the cells. This review focuses on the interplay between COVID-19 infection, hypoxia, premature neuronal aging, and neurodegenerative diseases and provides a novel insight into the molecular mechanisms of neurodegeneration.
Collapse
Affiliation(s)
- Narmadhaa Sivagurunathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur - 610005, Tamil Nadu, India
| | - Latchoumycandane Calivarathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur - 610005, Tamil Nadu, India
| |
Collapse
|
160
|
Engin AB, Engin ED, Engin A. Macrophage Activation Syndrome in Coinciding Pandemics of Obesity and COVID-19: Worse than Bad. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:919-954. [PMID: 39287877 DOI: 10.1007/978-3-031-63657-8_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Epigenetic changes have long-lasting impacts, which influence the epigenome and are maintained during cell division. Thus, human genome changes have required a very long timescale to become a major contributor to the current obesity pandemic. Whereas bidirectional effects of coronavirus disease 2019 (COVID-19) and obesity pandemics have given the opportunity to explore, how the viral microribonucleic acids (miRNAs) use the human's transcriptional machinery that regulate gene expression at a posttranscriptional level. Obesity and its related comorbidity, type 2 diabetes (T2D), and new-onset diabetes due to severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) are additional risk factors, which increase the severity of COVID-19 and its related mortality. The higher mortality rate of these patients is dependent on severe cytokine storm, which is the sum of the additional cytokine production by concomitant comorbidities and own cytokine synthesis of COVID-19. Patients with obesity facilitate the SARS-CoV-2 entry to host cell via increasing the host's cell receptor expression and modifying the host cell proteases. After entering the host cells, the SARS-CoV-2 genome directly functions as a messenger ribonucleic acid (mRNA) and encodes a set of nonstructural proteins via processing by the own proteases, main protease (Mpro), and papain-like protease (PLpro) to initiate viral genome replication and transcription. Following viral invasion, SARS-CoV-2 infection reduces insulin secretion via either inducing β-cell apoptosis or reducing intensity of angiotensin-converting enzyme 2 (ACE2) receptors and leads to new-onset diabetes. Since both T2D and severity of COVID-19 are associated with the increased serum levels of pro-inflammatory cytokines, high glucose levels in T2D aggravate SARS-CoV-2 infection. Elevated neopterin (NPT) value due to persistent interferon gamma (IFN-γ)-mediated monocyte-macrophage activation is an indicator of hyperactivated pro-inflammatory phenotype M1 macrophages. Thus, NPT could be a reliable biomarker for the simultaneously occurring COVID-19-, obesity- and T2D-induced cytokine storm. While host miRNAs attack viral RNAs, viral miRNAs target host transcripts. Eventually, the expression rate and type of miRNAs also are different in COVID-19 patients with different viral loads. It is concluded that specific miRNA signatures in macrophage activation phase may provide an opportunity to become aware of the severity of COVID-19 in patients with obesity and obesity-related T2D.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey
| | - Evren Doruk Engin
- Biotechnology Institute, Ankara University, Gumusdere Campus, Gumusdere, Ankara, Turkey
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
161
|
Fisicaro F, Lanza G, Concerto C, Rodolico A, Di Napoli M, Mansueto G, Cortese K, Mogavero MP, Ferri R, Bella R, Pennisi M. COVID-19 and Mental Health: A "Pandemic Within a Pandemic". ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1458:1-18. [PMID: 39102186 DOI: 10.1007/978-3-031-61943-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
The COVID-19 pandemic has brought significant changes in daily life for humanity and has had a profound impact on mental health. As widely acknowledged, the pandemic has led to notable increases in rates of anxiety, depression, distress, and other mental health-related issues, affecting both infected patients and non-infected individuals. COVID-19 patients and survivors face heightened risks for various neurological and psychiatric disorders and complications. Vulnerable populations, including those with pre-existing mental health conditions and individuals living in poverty or frailty, may encounter additional challenges. Tragically, suicide rates have also risen, particularly among young people, due to factors such as unemployment, financial crises, domestic violence, substance abuse, and social isolation. Efforts are underway to address these mental health issues, with healthcare professionals urged to regularly screen both COVID-19 and post-COVID-19 patients and survivors for psychological distress, ensuring rapid and appropriate interventions. Ongoing periodic follow-up and multidimensional, interdisciplinary approaches are essential for individuals experiencing long-term psychiatric sequelae. Preventive strategies must be developed to mitigate mental health problems during both the acute and recovery phases of COVID-19 infection. Vaccination efforts continue to prioritize vulnerable populations, including those with mental health conditions, to prevent future complications. Given the profound implications of mental health problems, including shorter life expectancy, diminished quality of life, heightened distress among caregivers, and substantial economic burden, it is imperative that political and health authorities prioritize the mental well-being of all individuals affected by COVID-19, including infected individuals, non-infected individuals, survivors, and caregivers.
Collapse
Affiliation(s)
- Francesco Fisicaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123, Catania, Italy
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgery Specialties, University of Catania, Via Santa Sofia 78, 95123, Catania, Italy.
- Clinical Neurophysiology Research Unit, Oasi Research Institute-IRCCS, Via Conte Ruggero 78, 94018, Troina, Italy.
| | - Carmen Concerto
- Department of Clinical and Experimental Medicine, Psychiatry Unit, University of Catania, Via Santa Sofia 89, 95123, Catania, Italy
| | - Alessandro Rodolico
- Department of Clinical and Experimental Medicine, Psychiatry Unit, University of Catania, Via Santa Sofia 89, 95123, Catania, Italy
| | - Mario Di Napoli
- Neurological Service, SS Annunziata Hospital, Viale Mazzini 100, 67039, Sulmona, L'Aquila, Italy
| | - Gelsomina Mansueto
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Piazza L. Miraglia 2, 80138, Naples, Italy
- Clinical Department of Laboratory Services and Public Health-Legal Medicine Unit, University of Campania "Luigi Vanvitelli", Via Luciano Armanni 5, 80138, Naples, Italy
| | - Klizia Cortese
- Department of Educational Sciences, University of Catania, Via Teatro Greco 84, 95124, Catania, Italy
| | - Maria P Mogavero
- Sleep Disorders Center, Division of Neuroscience, San Raffaele Scientific Institute, Via Stamira d'Ancona 20, 20127, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Raffaele Ferri
- Clinical Neurophysiology Research Unit, Oasi Research Institute-IRCCS, Via Conte Ruggero 78, 94018, Troina, Italy
| | - Rita Bella
- Department of Medical and Surgical Sciences and Advanced Technologies, University of Catania, Via Santa Sofia 87, 95123, Catania, Italy
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123, Catania, Italy
| |
Collapse
|
162
|
Cui A, Huang T, Li S, Ma A, Pérez JL, Sander C, Keskin DB, Wu CJ, Fraenkel E, Hacohen N. Dictionary of immune responses to cytokines at single-cell resolution. Nature 2024; 625:377-384. [PMID: 38057668 PMCID: PMC10781646 DOI: 10.1038/s41586-023-06816-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 11/01/2023] [Indexed: 12/08/2023]
Abstract
Cytokines mediate cell-cell communication in the immune system and represent important therapeutic targets1-3. A myriad of studies have highlighted their central role in immune function4-13, yet we lack a global view of the cellular responses of each immune cell type to each cytokine. To address this gap, we created the Immune Dictionary, a compendium of single-cell transcriptomic profiles of more than 17 immune cell types in response to each of 86 cytokines (>1,400 cytokine-cell type combinations) in mouse lymph nodes in vivo. A cytokine-centric view of the dictionary revealed that most cytokines induce highly cell-type-specific responses. For example, the inflammatory cytokine interleukin-1β induces distinct gene programmes in almost every cell type. A cell-type-centric view of the dictionary identified more than 66 cytokine-driven cellular polarization states across immune cell types, including previously uncharacterized states such as an interleukin-18-induced polyfunctional natural killer cell state. Based on this dictionary, we developed companion software, Immune Response Enrichment Analysis, for assessing cytokine activities and immune cell polarization from gene expression data, and applied it to reveal cytokine networks in tumours following immune checkpoint blockade therapy. Our dictionary generates new hypotheses for cytokine functions, illuminates pleiotropic effects of cytokines, expands our knowledge of activation states of each immune cell type, and provides a framework to deduce the roles of specific cytokines and cell-cell communication networks in any immune response.
Collapse
Affiliation(s)
- Ang Cui
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Faculty of Medicine, Harvard University, Boston, MA, USA.
| | - Teddy Huang
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shuqiang Li
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Aileen Ma
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jorge L Pérez
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Chris Sander
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- cBio Center, Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Derin B Keskin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Catherine J Wu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ernest Fraenkel
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
163
|
Aciole MR, Gonçales JP, Neves PAF, Soares CRP, de Oliveira MI, de Melo HRL, de Lima Neto RG, Moura LCRV, Araújo PSR, de Lorena VMB. Levels of soluble TNF receptors (sTNFR1 and sTNFR2) increase with clinical worsening of patients and are related to COVID-19 mortality. Immunobiology 2024; 229:152748. [PMID: 38128238 DOI: 10.1016/j.imbio.2023.152748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/01/2023] [Accepted: 09/13/2023] [Indexed: 12/23/2023]
Abstract
The present study aimed to inspect the serum levels of the soluble receptors, sTNFR1 and sTNFR2, in patients with COVID-19. The large production of inflammatory cytokines is an essential process in the pathogenesis of COVID-19. TNF is a multifaceted proinflammatory cytokine which has soluble and membrane receptors. Thus, knowing the role of these receptors will help better understand this disease's immunopathogenesis. We included 131 patients confirmed for SARS-CoV-2, separated into three groups: ward patients without O2 support, group A (n = 14); ward patients with O2 support, group B (n = 85), and patients in an intensive care unit (ICU), group C (n = 32), making up the receptors dosed by flow cytometry. The results showed that sTNFR1 and sTNFR2 are associated with disease severity, being higher in group C when compared to group A. As for the levels of receptors and their relationship with the degree of lung involvement, we found higher values of sTNFR1 in patients in group 1 (pulmonary involvement < 25%), suggesting that inflammatory processes related to TNF are not necessarily associated with the primary site of infection. When we analysed the patients who passed away compared to those who recovered, both receptors significantly increased the mortality numbers. These findings suggest a relevant influence of soluble receptors in the inflammatory processes involved in the pathogenesis of COVID-19. Wherefore, we suggest using these receptors as biomarkers of severity and mortality of the disease.
Collapse
Affiliation(s)
- Melayne Rocha Aciole
- Department of Immunology, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Pernambuco, Brazil; Federal University of Pernambuco - Postgraduate in Tropical Medicine, Recife, Pernambuco, Brazil; Ser Educational Group - Recife, Pernambuco, Brazil
| | - Juliana Prado Gonçales
- Department of Immunology, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Pernambuco, Brazil; Ser Educational Group - Recife, Pernambuco, Brazil
| | - Patrícia Areias Feitosa Neves
- Department of Immunology, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Pernambuco, Brazil; Federal University of Pernambuco - Postgraduate in Tropical Medicine, Recife, Pernambuco, Brazil
| | | | - Marta Iglis de Oliveira
- Federal University of Pernambuco - Postgraduate in Tropical Medicine, Recife, Pernambuco, Brazil
| | | | | | | | | | | |
Collapse
|
164
|
Qu D, Liu J, Zhou L, Yang Y, Wu C, Xu X, Zhu Q, Wang C, Zhao X. Association of serum amyloid A and prognosis in people with diabetes and COVID-19: A retrospective cohort study. J Diabetes Investig 2024; 15:44-51. [PMID: 38031656 PMCID: PMC10759722 DOI: 10.1111/jdi.14118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/12/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023] Open
Abstract
AIMS/INTRODUCTION Serum amyloid A (SAA) is an acute phase reactive protein that plays a vital role in the early diagnosis, risk prediction, efficacy observation and prognosis evaluation of infectious diseases. This study aimed to assess the association between SAA levels and the prognosis of patients with coronavirus disease 2019 (COVID-19) and diabetes. MATERIALS AND METHODS We carried out this retrospective cohort study from March 2022 to May 2022. The population was stratified by tertiles of SAA levels: low (<8.5 mg/L), medium (8.5-36 mg/L) and high (>36 mg/L). The primary outcome was whether the patient developed severe COVID-19, and secondary outcomes included the need for invasive mechanical ventilation and length of hospital stay. Logistic regression analyses were carried out to identify risk factors affecting the prognosis of patients with COVID-19 and diabetes. RESULTS We analyzed 910 diabetes patients with COVID-19. The median age of the patients was 69 years, and 52.3% were men. As SAA levels increased, the proportion of severe COVID-19 (6.3% vs 7.3% vs 22.8%, P < 0.001) and the proportion of invasive mechanical ventilation also increased among the three groups. Patients with high SAA levels had a longer length of hospital stay compared with patients with medium SAA and low SAA levels. Univariate logistic regression analysis showed that SAA >36 mg/L further increased the odds ratio to 4.423 (P < 0.001) for the development of severe COVID-19 compared with low SAA. Multivariate logistic regression analysis, adjusted for age and sex, confirmed that SAA >36 mg/L remained an independent risk factor for the development of severe COVID-19 (adjusted odds ratio 3.038, P < 0.001). CONCLUSIONS SAA levels are strongly associated with poor prognosis in patients with COVID-19 and diabetes.
Collapse
Affiliation(s)
- Duoduo Qu
- Department of EndocrinologyShanghai Public Health Clinical CenterShanghaiChina
| | - Jia Liu
- Department of EndocrinologyShanghai Public Health Clinical CenterShanghaiChina
| | - Lihua Zhou
- Department of EndocrinologyShanghai Public Health Clinical CenterShanghaiChina
| | - Yaling Yang
- Department of EndocrinologyShanghai Public Health Clinical CenterShanghaiChina
| | - Chenwei Wu
- Department of EndocrinologyShanghai Public Health Clinical CenterShanghaiChina
| | - Xinyue Xu
- Department of EndocrinologyShanghai Public Health Clinical CenterShanghaiChina
| | - Qin Zhu
- Department of EndocrinologyShanghai Public Health Clinical CenterShanghaiChina
| | - Chunhong Wang
- Department of EndocrinologyShanghai Public Health Clinical CenterShanghaiChina
| | - Xiaolong Zhao
- Department of EndocrinologyShanghai Public Health Clinical CenterShanghaiChina
| |
Collapse
|
165
|
Mendes-Filho SPDM, de Souza Pinheiro R, Martins FS, Giroldi PJ, e Melo RH, de Oliveira EL, dos Santos AB, Medeiros DCO, Lopes JA, Chaves YO, Zuliani JP, Nogueira PA. Kinetics of IL-6, C-reactive Protein and Fibrinogen Levels in COVID-19 Outpatients Who Evolved to Hypoxemia. CLINICAL PATHOLOGY (THOUSAND OAKS, VENTURA COUNTY, CALIF.) 2024; 17:2632010X231222795. [PMID: 38188270 PMCID: PMC10768631 DOI: 10.1177/2632010x231222795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024]
Abstract
Introduction Despite the efficacy of the COVID-19, the search for improvements in the management of severe/critical cases continues to be important. The aim is to demonstrate the kinetics of 4 serological markers in patients with COVID-19 who evolved in hypoxemia. Methods From June to December 2020, the Health Secretariat of Rondônia State, Brazil, established a home medical care service team (HMCS) that provided clinical follow-up for health professionals and military personnel with COVID-19. The clinical and laboratory monitoring was individualized at home by a nursing and medical team. In addition to laboratory parameters, C-reactive protein (CRP), interleukin-6 (IL-6), fibrinogen, and D-dimer levels were periodically taken to monitor the evolution of treatment. Results Of 218 patients telemonitored, 48 patients needed special care by the HMCS team due to shortness of breath. Chest tomography showed multiple ground-glass shadows and lung parenchymal condensations that was compatible with secondary bacterial infection associated with leukocytosis, for which antibiotics were prescribed. The symptoms were accompanied by increases of CRP and IL-6 levels followed by fibrinogen after a few days, for which an anticoagulant therapy was included. Thirty-three patients evolved to improvements in clinical signs and laboratory results. Between the sixth and eighth day of illness, 15 patients presented signs of hypoxemia with low O2 saturation accompanied with an increase in the respiratory rate, with some of them requiring oxygen therapy. As they did not present signs of clinical severity, but their laboratory markers showed an abrupt IL-6 peak that was higher than the increase in CRP and a new alteration in fibrinogen levels, they received a supplemental dose of anticoagulant and a high dose of corticosteroids, which resulted in clinical improvement. Conclusion Our study demonstrates that monitoring of IL-6 and CRP may identify precocious hypoxemia in COVID-19 patients and prevented the progressive deterioration of the lung injury.
Collapse
Affiliation(s)
| | - Rebeca de Souza Pinheiro
- Programa de Pós-graduação de Imunologia Básica e Aplicada, Universidade Federal do Amazonas, Manaus/AM, Brazil
| | - Fernanda Simão Martins
- Serviço de Assistência Médica Domiciliar (SAMD), Secretaria Estadual da Saúde (SESAU), Porto Velho/RO, Brazil
| | - Paulo Jose Giroldi
- Laboratório Estadual de Patologia e Análises Clínicas (LEPAC), Porto Velho/RO, Brazil
| | - Raul Honorato e Melo
- Serviço de Assistência Médica Domiciliar (SAMD), Secretaria Estadual da Saúde (SESAU), Porto Velho/RO, Brazil
- Hospital Cemetron, Porto Velho/RO, Brazil
| | | | - Anibal Borin dos Santos
- Serviço de Assistência Médica Domiciliar (SAMD), Secretaria Estadual da Saúde (SESAU), Porto Velho/RO, Brazil
| | | | | | - Yury Oliveira Chaves
- Instituto Leônidas e Maria Deane (ILMD), Fundação Oswaldo Cruz – Amazonas (FIOCRUZ – AMAZONAS), Manaus/AM, Brazil
| | | | - Paulo Afonso Nogueira
- Programa de Pós-graduação de Imunologia Básica e Aplicada, Universidade Federal do Amazonas, Manaus/AM, Brazil
- Instituto Leônidas e Maria Deane (ILMD), Fundação Oswaldo Cruz – Amazonas (FIOCRUZ – AMAZONAS), Manaus/AM, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus/AM, Brazil
| |
Collapse
|
166
|
Ganesh R, Yadav S, Hurt RT, Mueller MR, Aakre CA, Gilman EA, Grach SL, Overgaard J, Snyder MR, Collins NM, Croghan IT, Badley AD, Razonable RR, Salonen BR. Pro Inflammatory Cytokines Profiles of Patients With Long COVID Differ Between Variant Epochs. J Prim Care Community Health 2024; 15:21501319241254751. [PMID: 38808863 PMCID: PMC11138192 DOI: 10.1177/21501319241254751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Over 30% of patients with COVID-19 have persistent symptoms that last beyond 30 days and referred to as Long COVID. Long COVID has been associated with a persistent elevation in peripheral cytokines including interleukin-6, interleukin-1β, and tumor necrosis factor-α. This study reports cytokine profiles of patients in our clinic across SARS-COV-2 variant epochs. METHODS The clinical cytokine panel was analyzed in patients with Long COVID during periods that were stratified according to variant epoch. The 4 variant epochs were defined as: (1) wild-type through alpha, (2) alpha/beta/gamma, (3) delta, and (4) omicron variants. RESULTS A total of 390 patients had the clinical cytokine panel performed; the median age was 48 years (IQR 38-59) and 62% were female. Distribution by variant was wild-type and alpha, 50% (n = 196); alpha/beta/gamma, 7.9% (n = 31); delta, 18% (n = 72); and omicron, 23% (n = 91). Time to cytokine panel testing was significantly longer for the earlier epochs. Tumor necrosis factor-α (P < .001) and interleukin 1β (P < .001) were significantly more elevated in the earlier epochs (median of 558 days in wild-type through Alpha epoch vs 263 days in omicron epoch, P < .001)). Nucleocapsid antibodies were consistently detected across epochs. DISCUSSION When stratified by variant epoch, patients with early epoch Long COVID had persistently elevated peripheral pro-inflammatory cytokine levels when compared to later epoch Long COVID. Patients with Long COVID have similar clusters of symptoms across epochs, suggesting that the underlying pathology is independent of the peripheral cytokine signature.
Collapse
|
167
|
Priyandoko D, Widowati W, Lenny L, Novianti S, Revika R, Kusuma HSW, Sholihah IA. Green Tea Extract Reduced Lipopolysaccharide-Induced Inflammation in L2 Cells as Acute Respiratory Distress Syndrome Model Through Genes and Cytokine Pro-Inflammatory. Avicenna J Med Biotechnol 2024; 16:57-65. [PMID: 38605739 PMCID: PMC11005400 DOI: 10.18502/ajmb.v16i1.14172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/23/2023] [Indexed: 04/13/2024] Open
Abstract
Background Acute Respiratory Distress Syndrome (ARDS) is a severe lung inflammatory condition that has the capacity to impair gas exchange and lead to hypoxemia. This condition is found to have been one of the most prevalent in patients of COVID-19 with a more serious condition. Green tea (Camellia sinensis L.) contains polyphenols that possess many health benefits. The purpose of this study was to assess the anti-inflammatory activities of green tea extract in Lipopolysaccharide (LPS)-induced lung cells as ARDS cells model. Methods In this study, rat lung cells (L2) were induced by LPS to mimic the inflammation observed in ARDS and later treated with green tea extract. Pro-inflammatory cytokines such as Interleukin (IL)-12, C-Reactive Protein (CRP) as well as Tumor Necrosis Factor-α (TNF-α) were investigated using the ELISA method. Gene expression of NOD-Like Receptor Protein 3 (NLRP-3), Receptor for Advanced Glycation End-product (RAGE), Toll-like Receptor-4 (TLR-4), and Nuclear Factor-kappa B (NF-κB) were evaluated by qRTPCR. Apoptotic cells were measured using flow cytometry. Results The results showed that green tea extract treatment can reduce inflammation by suppressing gene expressions of NF-κB, NLRP-3, TLR-4, and RAGE, as well as pro-inflammatory cytokines such as IL-12, TNF-α, and CRP, an acute phase protein. Apoptosis levels of inflamed cells also found to be lowered when green tea extract was administered; thus, also increasing live cells compared to non-treated cells. Conclusion These findings could lead to the future development of supplements from green tea to help alleviate ARDS symptoms, especially during critical moments such as the current pandemic.
Collapse
Affiliation(s)
- Didik Priyandoko
- Biology Study Program, Faculty of Mathematics and Natural Sciences, Indonesia University of Education, Bandung 40154, Indonesia
| | - Wahyu Widowati
- Faculty of Medicine, Maranatha Christian University, Bandung 40164, Indonesia
| | - Lenny Lenny
- Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, BSD Campus, Tangerang 15345, Indonesia
| | - Sintya Novianti
- Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, BSD Campus, Tangerang 15345, Indonesia
| | - Revika Revika
- Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, BSD Campus, Tangerang 15345, Indonesia
| | | | - Ika Adhani Sholihah
- Biomolecular and Biomedical Research Center, Aretha Medika Utama, Bandung 40163, Indonesia
- School of Life Sciences and Technology, Bandung Institute of Technology, Bandung 40132, Indonesia
| |
Collapse
|
168
|
Sarmadi S, Rahbar MR, Najafi H, Chukwudozie OS, Morowvat MH. In Silico Design and Evaluation of a Novel Therapeutic Agent Against the Spike Protein as a Novel Treatment Strategy for COVID-19 Treatment. Recent Pat Biotechnol 2024; 18:162-176. [PMID: 37231757 DOI: 10.2174/1872208317666230523105759] [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: 10/18/2022] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is a viral respiratory disease that is associated with severe damage to other human organs. It causes by a novel coronavirus, and it is spreading all over the world. To date, there is some approved vaccine or therapeutic agent which could be effective against this disease. But their effectiveness against mutated strains is not studied completely. The spike glycoprotein on the surface of the coronaviruses gives the virus the ability to bind to host cell receptors and enter cells. Inhibition of attachment of these spikes can lead to virus neutralization by inhibiting viral entrance. AIMS In this study, we tried to use the virus entrance strategy against itself by utilizing virus receptor (ACE-2) in order to design an engineered protein consisting of a human Fc antibody fragment and a part of ACE-2, which reacts with virus RBD, and we also evaluated this interaction by computational methods and in silico methods. Subsequently, we have designed a new protein structure to bind with this site and inhibit the virus from attaching to its cell receptor, mechanically or chemically. METHODS Various in silico software, bioinformatics, and patent databases were used to retrieve the requested gene and protein sequences. The physicochemical properties and possibility of allergenicity were also examined. Three-dimensional structure prediction and molecular docking were also performed to develop the most suitable therapeutic protein. RESULTS The designed protein consisted of a total of 256 amino acids with a molecular weight of 28984.62 and 5.92 as a theoretical isoelectric point. Instability and aliphatic index and grand average of hydropathicity are 49.99, 69.57 and -0.594, respectively. CONCLUSIONS In silico studies can provide a good opportunity to study viral proteins and new drugs or compounds since they do not need direct exposure to infectious agents or equipped laboratories. The suggested therapeutic agent should be further characterized in vitro and in vivo.
Collapse
Affiliation(s)
- Soroush Sarmadi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P.O. Box 71468-64685, Shiraz, Iran
- Department of Pathobiology, Faculty of Veterinary Medicine, Shiraz University, P.O. Box 71441-11731, Shiraz, Iran
| | - Mohammad Reza Rahbar
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P.O. Box 71468-64685, Shiraz, Iran
| | - Hamideh Najafi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, P.O. Box 14199-63111, Tehran, Iran
| | - Onyeka S Chukwudozie
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Mohammad Hossein Morowvat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P.O. Box 71468-64685, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 71468-64685, Shiraz, Iran
| |
Collapse
|
169
|
Patel P, Bhattacharjee M. Microbiome and the COVID-19 pandemic. MICROBES, MICROBIAL METABOLISM, AND MUCOSAL IMMUNITY 2024:287-348. [DOI: 10.1016/b978-0-323-90144-4.00008-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
170
|
Scott C, Hall S, Zhou J, Lehmann C. Cannabinoids and the Endocannabinoid System in Early SARS-CoV-2 Infection and Long COVID-19-A Scoping Review. J Clin Med 2023; 13:227. [PMID: 38202234 PMCID: PMC10779964 DOI: 10.3390/jcm13010227] [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: 11/17/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Coronavirus disease-19 (COVID-19) is a highly contagious illness caused by the SARS-CoV-2 virus. The clinical presentation of COVID-19 is variable, often including symptoms such as fever, cough, headache, fatigue, and an altered sense of smell and taste. Recently, post-acute "long" COVID-19 has emerged as a concern, with symptoms persisting beyond the acute infection. Vaccinations remain one of the most effective preventative methods against severe COVID-19 outcomes and the development of long-term COVID-19. However, individuals with underlying health conditions may not mount an adequate protective response to COVID-19 vaccines, increasing the likelihood of severe symptoms, hospitalization, and the development of long-term COVID-19 in high-risk populations. This review explores the potential therapeutic role of cannabinoids in limiting the susceptibility and severity of infection, both pre- and post-SARS-CoV-19 infection. Early in the SARS-CoV-19 infection, cannabinoids have been shown to prevent viral entry, mitigate oxidative stress, and alleviate the associated cytokine storm. Post-SARS-CoV-2 infection, cannabinoids have shown promise in treating symptoms associated with post-acute long COVID-19, including depression, anxiety, post-traumatic stress injury, insomnia, pain, and decreased appetite. While current research primarily focuses on potential treatments for the acute phase of COVID-19, there is a gap in research addressing therapeutics for the early and post-infectious phases. This review highlights the potential for future research to bridge this gap by investigating cannabinoids and the endocannabinoid system as a potential treatment strategy for both early and post-SARS-CoV-19 infection.
Collapse
Affiliation(s)
- Cassidy Scott
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 1X5, Canada; (C.S.); (J.Z.)
| | - Stefan Hall
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS B3H 1X5, Canada;
| | - Juan Zhou
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 1X5, Canada; (C.S.); (J.Z.)
| | - Christian Lehmann
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 1X5, Canada; (C.S.); (J.Z.)
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS B3H 1X5, Canada;
| |
Collapse
|
171
|
Zhang Z, Wu S, Wang Z, Wang Y, Chen H, Wu C, Xiong L. Long-term oral ACEI/ARB therapy is associated with disease severity in elderly COVID-19 omicron BA.2 patients with hypertension. BMC Infect Dis 2023; 23:882. [PMID: 38110869 PMCID: PMC10726588 DOI: 10.1186/s12879-023-08913-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 12/14/2023] [Indexed: 12/20/2023] Open
Abstract
OBJECTIVE To explore the effects of long-term oral ACEIs/ARBs on the incidence of exacerbation and in-hospital mortality in elderly COVID-19 Omicron BA.2 patients with hypertension, especially patients aged 80 years or older. MATERIALS AND METHODS In this retrospective study, patients suffering mild and rcommon COVID-19 with hypertension who were hospitalized in the Shanghai Fourth People's Hospital between April 2022 and June 2022 were enrolled. Primary outcomes included the incidence of exacerbation and in-hospital mortality. Secondary outcomes included the incidence of respiratory failure of patients, use of mechanical ventilation, nucleic acid conversion time (NCT), hospitalization costs, and the temporal trend of the incidence of exacerbations and in-hospital mortality in different age groups. The data were analysed using propensity score weighting (PSW). RESULTS In the entire cohort, there were 298 ACEI/ARB users and 465 non-ACEI/ARB users. The ACEI/ARB group showed a lower incidence of exacerbation (OR = 0.64, 95% CI for OR: 0.46-0.89, P = 0.0082) and lower in-hospital mortality (OR = 0.49, 95% CI for OR: 0.27-0.89, P = 0.0201) after PSW. Sensitivity analysis obtained the same results. The results of the subgroup of patients aged 80 years and older obtained a similar conclusion as the whole cohort. Most of the study indicators did not differ statistically significantly in the subgroup of patients aged 60 to 79 years except for rates of mechanical ventilation and respiratory failure. CONCLUSION Antihypertensive therapy with ACEIs/ARBs might reduce the incidence of exacerbation and in-hospital mortality. The findings of this study support the use of ACEIs/ARBs in COVID-19 patients infected by Omicron BA.2, especially in patients aged 80 years or older with hypertension.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Anesthesiology and Perioperative medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Shengyong Wu
- Department of Military Health Statistics, Naval Medical University, Shanghai, 200433, China
| | - Zhiyong Wang
- Department of Information Management, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, 200434, China
| | - Yue Wang
- Department of Anesthesiology and Perioperative medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Hui Chen
- Department of Anesthesiology and Perioperative medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Cheng Wu
- Department of Military Health Statistics, Naval Medical University, Shanghai, 200433, China.
| | - Lize Xiong
- Department of Anesthesiology and Perioperative medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
| |
Collapse
|
172
|
Ding H, Zhang W, Wang SA, Li C, Li W, Liu J, Yu F, Tao Y, Cheng S, Xie H, Chen Y. A semi-quantitative upconversion nanoparticle-based immunochromatographic assay for SARS-CoV-2 antigen detection. Front Microbiol 2023; 14:1289682. [PMID: 38149276 PMCID: PMC10750388 DOI: 10.3389/fmicb.2023.1289682] [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: 09/06/2023] [Accepted: 11/20/2023] [Indexed: 12/28/2023] Open
Abstract
The unprecedented public health and economic impact of the coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been met with an equally unprecedented scientific response. Sensitive point-of-care methods to detect SARS-CoV-2 antigens in clinical specimens are urgently required for the rapid screening of individuals with viral infection. Here, we developed an upconversion nanoparticle-based lateral flow immunochromatographic assay (UCNP-LFIA) for the high-sensitivity detection of SARS-CoV-2 nucleocapsid (N) protein. A pair of rabbit SARS-CoV-2 N-specific monoclonal antibodies was conjugated to UCNPs, and the prepared UCNPs were then deposited into the LFIA test strips for detecting and capturing the N protein. Under the test conditions, the limit of detection (LOD) of UCNP-LFIA for the N protein was 3.59 pg/mL, with a linear range of 0.01-100 ng/mL. Compared with that of the current colloidal gold-based LFIA strips, the LOD of the UCNP-LFIA-based method was increased by 100-fold. The antigen recovery rate of the developed method in the simulated pharyngeal swab samples ranged from 91.1 to 117.3%. Furthermore, compared with the reverse transcription-polymerase chain reaction, the developed UCNP-LFIA method showed a sensitivity of 94.73% for 19 patients with COVID-19. Thus, the newly established platform could serve as a promising and convenient fluorescent immunological sensing approach for the efficient screening and diagnosis of COVID-19.
Collapse
Affiliation(s)
- Hai Ding
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wanying Zhang
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shu-an Wang
- Department of Clinic Nutrition, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Chuang Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wanting Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Liu
- Polariton Life Technologies Ltd., Soochow, Jiangsu, China
| | - Fang Yu
- Polariton Life Technologies Ltd., Soochow, Jiangsu, China
| | - Yanru Tao
- Polariton Life Technologies Ltd., Soochow, Jiangsu, China
| | - Siyun Cheng
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hui Xie
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
173
|
Triwongwaranat D, Thanomkitti K, Korviriyakamol T, Saengthong-Aram P, Varothai S, Thuangtong R. Characteristics of hair loss in COVID-19 patients in Thailand. Medicine (Baltimore) 2023; 102:e36539. [PMID: 38065902 PMCID: PMC10713103 DOI: 10.1097/md.0000000000036539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
There is still a scarcity of data on hair loss caused by coronavirus disease 2019 (COVID-19) infection. This study aims to determine the characteristics of hair loss in Thai individuals after COVID-19 infection and to identify associated factors. From March to June 2022, a retrospective review of medical records and telephone interviews was conducted to determine the details of hair loss, the severity of infection, and the associated treatments of patients with an abrupt onset of hair loss after the diagnosis of COVID-19 infection at Siriraj Hospital in Bangkok, Thailand. This study included 43 patients who experienced hair loss within 4 months after COVID-19 infection. The mean age was 46.5 ± 14.5 years, predominantly women. Most had mild COVID-19 symptoms (59.3%), and 59.1% experienced weight loss, with a mean weight loss of 4.3 ± 2.0 kg per month. Preexisting hair loss was reported in 31.0% of participants, with approximately 3-quarters diagnosed with androgenetic alopecia. The median onset of hair loss after COVID-19 infection was 30 days (interquartile range 30-60). Telogen effluvium was the most common acute hair loss diagnosis, and topical minoxidil was the predominant treatment (95.3%). Female gender was correlated with a more severe shedding scale (adjusted odd ratio 24.76, 95% CI 1.67-168.86). Patients with a history of androgenetic alopecia tended to have a lower hair shedding scale (adjusted odd ratio 0.03, 95% CI 0.01-0.38). This study reviewed the characteristics of hair loss after COVID-19 infection during Omicron outbreaks in Thailand. The COVID-19-associated telogen effluvium, which is the primary cause in our patients, manifested with earlier onset at approximately 30 days.
Collapse
Affiliation(s)
- Daranporn Triwongwaranat
- Department of Dermatology, Siriraj Hospital, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanchalit Thanomkitti
- Department of Dermatology, Siriraj Hospital, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tarinee Korviriyakamol
- Department of Dermatology, Siriraj Hospital, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Phuwakorn Saengthong-Aram
- Department of Dermatology, Siriraj Hospital, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Supenya Varothai
- Department of Dermatology, Siriraj Hospital, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rattapon Thuangtong
- Department of Dermatology, Siriraj Hospital, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
174
|
Kato CD, Nsubuga J, Niyonzima N, Kitibwa A, Matovu E, Othieno E, Ssebugere P, Tumwine AA, Namayanja M. Immunological and biochemical biomarker alterations among SARS-COV-2 patients with varying disease phenotypes in Uganda. BMC Infect Dis 2023; 23:857. [PMID: 38057707 DOI: 10.1186/s12879-023-08854-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023] Open
Abstract
Every novel infection requires an assessment of the host response coupled with identification of unique biomarkers for predicting disease pathogenesis, treatment targets and diagnostic utility. Studies have exposed dysregulated inflammatory response induced by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as significant predictor or cause of disease severity/prognosis and death. This study evaluated inflammatory biomarkers induced by SARS-CoV-2 in plasma of patients with varying disease phenotypes and healthy controls with prognostic or therapeutic potential. We stratified SARS-CoV-2 plasma samples based on disease status (asymptomatic, mild, severe, and healthy controls), as diagnosed by RT-PCR SARS-CoV-2. We used a solid phase sandwich and competitive Enzyme-Linked Immunosorbent Assay (ELISA) to measure levels of panels of immunological (IFN-γ, TNF-α, IL-6, and IL-10) and biochemical markers (Ferritin, Procalcitonin, C-Reactive Protein, Angiotensin II, Homocysteine, and D-dimer). Biomarker levels were compared across SARS-CoV-2 disease stratification. Plasma IFN-γ, TNF-α, IL-6, and IL-10 levels were significantly (P < 0.05) elevated in the severe SARS-CoV-2 patients as compared to mild, asymptomatic, and healthy controls. Ferritin, Homocysteine, and D-dimer plasma levels were significantly elevated in severe cases over asymptomatic and healthy controls. Plasma C-reactive protein and Angiotensin II levels were significantly (P < 0.05) higher in mild than severe cases and healthy controls. Plasma Procalcitonin levels were significantly higher in asymptomatic than in mild, severe cases and healthy controls. Our study demonstrates the role of host inflammatory biomarkers in modulating the pathogenesis of COVID-19. The study proposes a number of potential biomarkers that could be explored as SARS-CoV-2 treatment targets and possible prognostic predictors for a severe outcome. The comprehensive analysis of prognostic biomarkers may contribute to the evidence-based management of COVID-19 patients.
Collapse
Affiliation(s)
- Charles Drago Kato
- School of Bio-security, Biotechnical & Laboratory Sciences, College of Veterinary Medicine, Animal Resources & Bio-security, Makerere University, P.O Box 7062, Kampala, Uganda.
| | - Julius Nsubuga
- School of Bio-security, Biotechnical & Laboratory Sciences, College of Veterinary Medicine, Animal Resources & Bio-security, Makerere University, P.O Box 7062, Kampala, Uganda.
| | | | - Annah Kitibwa
- School of Bio-security, Biotechnical & Laboratory Sciences, College of Veterinary Medicine, Animal Resources & Bio-security, Makerere University, P.O Box 7062, Kampala, Uganda
| | - Enock Matovu
- School of Bio-security, Biotechnical & Laboratory Sciences, College of Veterinary Medicine, Animal Resources & Bio-security, Makerere University, P.O Box 7062, Kampala, Uganda
| | - Emmanuel Othieno
- Department of Pathology, Soroti University, P.O. Box 211, Soroti, Uganda
| | - Patrick Ssebugere
- Department of Chemistry, College of Natural Sciences, Makerere University, P.O Box 7062, Kampala, Uganda
| | - Amanda Agnes Tumwine
- School of Bio-security, Biotechnical & Laboratory Sciences, College of Veterinary Medicine, Animal Resources & Bio-security, Makerere University, P.O Box 7062, Kampala, Uganda
| | - Monica Namayanja
- School of Bio-security, Biotechnical & Laboratory Sciences, College of Veterinary Medicine, Animal Resources & Bio-security, Makerere University, P.O Box 7062, Kampala, Uganda
| |
Collapse
|
175
|
Verma M, Rawat N, Rani R, Singh M, Choudhary A, Abbasi S, Kumar M, Kumar S, Tanwar A, Misir BR, Khanna S, Agrawal A, Faruq M, Rai S, Tripathi R, Kumar A, Pujani M, Bhojani M, Pandey AK, Nesari T, Prasher B. Adhatoda vasica and Tinospora cordifolia extracts ameliorate clinical and molecular markers in mild COVID-19 patients: a randomized open-label three-armed study. Eur J Med Res 2023; 28:556. [PMID: 38049897 PMCID: PMC10696694 DOI: 10.1186/s40001-023-01507-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 11/04/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND SARS-CoV-2 infections caused mild-to-moderate illness. However, a sizable portion of infected people experience a rapid progression of hyper-inflammatory and hypoxic respiratory illness that necessitates an effective and safer remedy to combat COVID-19. METHODS A total of 150 COVID-19-positive patients with no to mild symptoms, between the age groups 19-65 years were enrolled in this randomized, open-labeled three-armed clinical trial. Among them, 136 patients completed the study with RT-PCR negative reports. The patients received herbal drugs orally (Group A (Adhatoda vasica; AV; 500 mg; n = 50); Group B (Tinospora cordifolia; TC; 500 mg; n = 43), and Group C (AV + TC; 250 mg each; n = 43)) for 14 days. Clinical symptoms, vital parameters, and viral clearance were taken as primary outcomes, and biochemical, hematological parameters, cytokines, and biomarkers were evaluated at three time points as secondary outcomes. RESULTS We found that the mean viral clearance time was 13.92 days (95% confidence interval [CI] 12.85-14.99) in Group A, 13.44 days (95% confidence interval [CI] 12.14-14.74) in Group B, and 11.86 days (95% confidence interval [CI] 10.62-13.11) days in Group C. Over a period of 14 days, the mean temperature in Groups A, and B significantly decreased linearly. In Group A, during the trial period, eosinophils, and PT/INR increased significantly, while monocytes, SGOT, globulin, serum ferritin, and HIF-1α, a marker of hypoxia reduced significantly. On the other hand, in Group B hsCRP decreased at mid-treatment. Eosinophil levels increased in Group C during the treatment, while MCP-3 levels were significantly reduced. CONCLUSIONS All the patients of the three-armed interventions recovered from COVID-19 and none of them reported any adverse effects from the drugs. Group C patients (AV + TC) resulted in a quicker viral clearance as compared to the other two groups. We provide the first clinical report of AV herbal extract acting as a modifier of HIF-1α in COVID-19 patients along with a reduction in levels of ferritin, VEGF, and PT/INR as the markers of hypoxia, inflammation, and thrombosis highlighting the potential use in progression stages, whereas the TC group showed immunomodulatory effects. Trial registration Clinical Trials Database -India (ICMR-NIMS), CTRI/2020/09/028043. Registered 24th September 2020, https://www.ctri.nic.in/Clinicaltrials/pdf_generate.php?trialid=47443&EncHid=&modid=&compid=%27,%2747443det%27.
Collapse
Grants
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
Collapse
Affiliation(s)
- Mukta Verma
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- All India Institute of Ayurveda, New Delhi, India
| | - Neha Rawat
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- All India Institute of Ayurveda, New Delhi, India
| | - Ritu Rani
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Manju Singh
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Aditi Choudhary
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Sarfaraz Abbasi
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Manish Kumar
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Sachin Kumar
- ESIC Medical College and Hospital, Faridabad, Haryana, India
| | - Ankur Tanwar
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- All India Institute of Ayurveda, New Delhi, India
| | - Bishnu Raman Misir
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Sangeeta Khanna
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Anurag Agrawal
- Trivedi School of Biosciences, Ashoka University, Sonipat, Haryana, India
| | - Mohammed Faruq
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Shalini Rai
- All India Institute of Ayurveda, New Delhi, India
| | | | - Anil Kumar
- All India Institute of Ayurveda, New Delhi, India
| | - Mukta Pujani
- ESIC Medical College and Hospital, Faridabad, Haryana, India
| | | | | | | | - Bhavana Prasher
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India.
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India.
| |
Collapse
|
176
|
Levinson T, Wasserman A, Shenhar-Tsarfaty S, Halutz O, Shapira I, Zeltser D, Rogowski O, Berliner S, Ziv-Baran T. Comparative analysis of CRP as a biomarker of the inflammatory response intensity among common viral infections affecting the lungs: COVID-19 versus influenza A, influenza B and respiratory syncytial virus. Clin Exp Med 2023; 23:5307-5313. [PMID: 37640989 DOI: 10.1007/s10238-023-01176-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
Severe acute respiratory syndrome-corona virus 2 (SARS-CoV-2) is associated with significant morbidity and mortality. C-reactive protein (CRP) is a useful inflammatory biomarker for patients admitted with an infection. This study aimed to compare CRP level as an indicator of inflammation severity between SARS-CoV-2 and common respiratory viral infections. A cross-sectional study of all adult patients hospitalized in the internal medicine department, geriatric department, or internal intensive care unit between 02/2012 and 06/2021 with laboratory-confirmed respiratory viral infection was performed. SARS-CoV-2, influenza A, influenza B, and respiratory syncytial virus (RSV) were studied. Patients with laboratory-confirmed concurrent viral or bacterial infections were excluded. Patients with malignancy were also excluded. Age, gender, comorbidities, and CRP level upon admission were compared between groups. Univariate and multivariable analyses were applied. Among 1124 patients, 18.2% had SARS‑CoV‑2, 48.3% influenza A, 18.9% RSV, and 14.6% influenza B. SARS‑CoV‑2 patients were significantly younger (median 69.4 vs. ≥ 76 years) and had lower Charlson score (median 3 vs. ≥ 4 in other groups) compared to patients with other viral pathogens. After adjustment for patients' age, gender and comorbidities, SARS‑CoV‑2 patients had a higher probability (OR = 1.84-2.02, p < 0.01) of having CRP values in the upper quartile (> 117 mg/L) compared to all other viral pathogens while between all others there was no significant difference. To conclude, a higher CRP level upon admission is approximately twice more common among SARS-CoV-2 patients compared to other widespread respiratory viruses which may demonstrate the higher intensity of inflammation caused by SARS-CoV-2.
Collapse
Affiliation(s)
- Tal Levinson
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
- Infectious Diseases Unit, Tel-Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Asaf Wasserman
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Shani Shenhar-Tsarfaty
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Ora Halutz
- Clinical Microbiology Laboratory, Tel-Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel-Aviv, Israel
| | - Itzhak Shapira
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - David Zeltser
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Ori Rogowski
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Shlomo Berliner
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Tomer Ziv-Baran
- Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medicine, Tel Aviv University, P.O.B. 39040, 6997801, Tel Aviv, Israel.
| |
Collapse
|
177
|
Valdes Angues R, Perea Bustos Y. SARS-CoV-2 Vaccination and the Multi-Hit Hypothesis of Oncogenesis. Cureus 2023; 15:e50703. [PMID: 38234925 PMCID: PMC10792266 DOI: 10.7759/cureus.50703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 01/19/2024] Open
Abstract
Cancer is a complex and dynamic disease. The "hallmarks of cancer" were proposed by Hanahan and Weinberg (2000) as a group of biological competencies that human cells attain as they progress from normalcy to neoplastic transformation. These competencies include self-sufficiency in proliferative signaling, insensitivity to growth-suppressive signals and immune surveillance, the ability to evade cell death, enabling replicative immortality, reprogramming energy metabolism, inducing angiogenesis, and activating tissue invasion and metastasis. Underlying these competencies are genome instability, which expedites their acquisition, and inflammation, which fosters their function(s). Additionally, cancer exhibits another dimension of complexity: a heterogeneous repertoire of infiltrating and resident host cells, secreted factors, and extracellular matrix, known as the tumor microenvironment, that through a dynamic and reciprocal relationship with cancer cells supports immortality, local invasion, and metastatic dissemination. This staggering intricacy calls for caution when advising all people with cancer (or a previous history of cancer) to receive the COVID-19 primary vaccine series plus additional booster doses. Moreover, because these patients were not included in the pivotal clinical trials, considerable uncertainty remains regarding vaccine efficacy, safety, and the risk of interactions with anticancer therapies, which could reduce the value and innocuity of either medical treatment. After reviewing the available literature, we are particularly concerned that certain COVID-19 vaccines may generate a pro-tumorigenic milieu (i.e., a specific environment that could lead to neoplastic transformation) that predisposes some (stable) oncologic patients and survivors to cancer progression, recurrence, and/or metastasis. This hypothesis is based on biological plausibility and fulfillment of the multi-hit hypothesis of oncogenesis (i.e., induction of lymphopenia and inflammation, downregulation of angiotensin-converting enzyme 2 (ACE2) expression, activation of oncogenic cascades, sequestration of tumor suppressor proteins, dysregulation of the RNA-G quadruplex-protein binding system, alteration of type I interferon responses, unsilencing of retrotransposable elements, etc.) together with growing evidence and safety reports filed to Vaccine Adverse Effects Report System (VAERS) suggesting that some cancer patients experienced disease exacerbation or recurrence following COVID-19 vaccination. In light of the above and because some of these concerns (i.e., alteration of oncogenic pathways, promotion of inflammatory cascades, and dysregulation of the renin-angiotensin system) also apply to cancer patients infected with SARS-CoV-2, we encourage the scientific and medical community to urgently evaluate the impact of both COVID-19 and COVID-19 vaccination on cancer biology and tumor registries, adjusting public health recommendations accordingly.
Collapse
Affiliation(s)
- Raquel Valdes Angues
- Neurology, Oregon Health and Science University School of Medicine, Portland, USA
| | | |
Collapse
|
178
|
Moradi Marjaneh M, Challenger JD, Salas A, Gómez-Carballa A, Sivananthan A, Rivero-Calle I, Barbeito-Castiñeiras G, Foo CY, Wu Y, Liew F, Jackson HR, Habgood-Coote D, D'Souza G, Nichols SJ, Wright VJ, Levin M, Kaforou M, Thwaites RS, Okell LC, Martinón-Torres F, Cunnington AJ. Analysis of blood and nasal epithelial transcriptomes to identify mechanisms associated with control of SARS-CoV-2 viral load in the upper respiratory tract. J Infect 2023; 87:538-550. [PMID: 37863321 DOI: 10.1016/j.jinf.2023.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
OBJECTIVES The amount of SARS-CoV-2 detected in the upper respiratory tract (URT viral load) is a key driver of transmission of infection. Current evidence suggests that mechanisms constraining URT viral load are different from those controlling lower respiratory tract viral load and disease severity. Understanding such mechanisms may help to develop treatments and vaccine strategies to reduce transmission. Combining mathematical modelling of URT viral load dynamics with transcriptome analyses we aimed to identify mechanisms controlling URT viral load. METHODS COVID-19 patients were recruited in Spain during the first wave of the pandemic. RNA sequencing of peripheral blood and targeted NanoString nCounter transcriptome analysis of nasal epithelium were performed and gene expression analysed in relation to paired URT viral load samples collected within 15 days of symptom onset. Proportions of major immune cells in blood were estimated from transcriptional data using computational differential estimation. Weighted correlation network analysis (adjusted for cell proportions) and fixed transcriptional repertoire analysis were used to identify associations with URT viral load, quantified as standard deviations (z-scores) from an expected trajectory over time. RESULTS Eighty-two subjects (50% female, median age 54 years (range 3-73)) with COVID-19 were recruited. Paired URT viral load samples were available for 16 blood transcriptome samples, and 17 respiratory epithelial transcriptome samples. Natural Killer (NK) cells were the only blood cell type significantly correlated with URT viral load z-scores (r = -0.62, P = 0.010). Twenty-four blood gene expression modules were significantly correlated with URT viral load z-score, the most significant being a module of genes connected around IFNA14 (Interferon Alpha-14) expression (r = -0.60, P = 1e-10). In fixed repertoire analysis, prostanoid-related gene expression was significantly associated with higher viral load. In nasal epithelium, only GNLY (granulysin) gene expression showed significant negative correlation with viral load. CONCLUSIONS Correlations between the transcriptional host response and inter-individual variations in SARS-CoV-2 URT viral load, revealed many molecular mechanisms plausibly favouring or constraining viral replication. Existing evidence corroborates many of these mechanisms, including likely roles for NK cells, granulysin, prostanoids and interferon alpha-14. Inhibition of prostanoid production and administration of interferon alpha-14 may be attractive transmission-blocking interventions.
Collapse
Affiliation(s)
- Mahdi Moradi Marjaneh
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK; Centre for Paediatrics and Child Health, Imperial College London, London, UK; Section of Virology, Department of Infectious Diseases, Imperial College London, London, UK.
| | - Joseph D Challenger
- Medical Research Council Centre for Global Infections Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Antonio Salas
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain; Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Alberto Gómez-Carballa
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain; Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Abilash Sivananthan
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| | - Irene Rivero-Calle
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain; Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Gema Barbeito-Castiñeiras
- Servicio de Microbiología y Parasitología, Complejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Cher Y Foo
- School of Medicine, Imperial College London, London, UK
| | - Yue Wu
- Department of Surgery and Cancer, Imperial College London, St. Mary's Hospital, London, UK
| | - Felicity Liew
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Heather R Jackson
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK; Centre for Paediatrics and Child Health, Imperial College London, London, UK
| | - Dominic Habgood-Coote
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK; Centre for Paediatrics and Child Health, Imperial College London, London, UK
| | - Giselle D'Souza
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK; Centre for Paediatrics and Child Health, Imperial College London, London, UK
| | - Samuel J Nichols
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK; Centre for Paediatrics and Child Health, Imperial College London, London, UK
| | - Victoria J Wright
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK; Centre for Paediatrics and Child Health, Imperial College London, London, UK
| | - Michael Levin
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK; Centre for Paediatrics and Child Health, Imperial College London, London, UK
| | - Myrsini Kaforou
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK; Centre for Paediatrics and Child Health, Imperial College London, London, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Lucy C Okell
- Medical Research Council Centre for Global Infections Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Federico Martinón-Torres
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain; Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Aubrey J Cunnington
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK; Centre for Paediatrics and Child Health, Imperial College London, London, UK.
| |
Collapse
|
179
|
Bukhari AE, AlDosari DA, AlDosari MA, Muqrin RF, Moazin OM, Almutlq MM, Altalhab SM, Alharithy RS, Alkhalifah AI. COVID-19-induced hair shedding and related risk factors: A Saudi perspective. J Family Med Prim Care 2023; 12:3304-3311. [PMID: 38361868 PMCID: PMC10866268 DOI: 10.4103/jfmpc.jfmpc_314_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 02/17/2024] Open
Abstract
Introduction Many cases of telogen effluvium (TE), acute hair shedding, following coronavirus disease 2019 (COVID-19) were reported during the pandemic. Methodology We conducted a cross-sectional study to assess the relationship between COVID-19 and TE in Saudi Arabia. Self-administered online questionnaires were distributed online between March and September 2022 in Saudi Arabia. A multivariate logistic regression model was used to determine risk factors associated with TE post-COVID-19 (significance at P < 0.05). Results Of the 703 responders, 392 were included in the study. 59.70% (n = 234) recognized hair shedding during or after COVID-19. The time taken to realize hair shedding (3 or 6 months) and the duration varied (3, 6, >6 months). The risk factors significantly related to TE post-COVID-19 were: female sex (P < 0.001, odds ratio [OR] = 2.98), COVID-19 antiviral treatment (P = 0.032, OR = 3.02), and TE history (P = 0.001, OR = 3.78). Conclusion Healthcare providers and physicians should be aware of the relationship between TE and COVID-19, to easily recognize, treat, and improve their patients' outcomes).
Collapse
Affiliation(s)
- Abrar E. Bukhari
- Department of Dermatology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Dalal A. AlDosari
- Department of Dermatology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Manal A. AlDosari
- Department of Dermatology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Reema F. Muqrin
- Department of Dermatology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Othillah M. Moazin
- Department of Dermatology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Malak M. Almutlq
- Department of Dermatology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Saad M. Altalhab
- Department of Dermatology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Ruaa S. Alharithy
- Department of Dermatology, Princess Nourah Bint Abdul Rahman University, Security Forces Hospital, Riyadh, Saudi Arabia
| | | |
Collapse
|
180
|
Satheesan A, Sharma S, Basu A. Sodium Butyrate Induced Neural Stem/Progenitor Cell Death in an Experimental Model of Japanese Encephalitis. Metab Brain Dis 2023; 38:2831-2847. [PMID: 37650987 DOI: 10.1007/s11011-023-01279-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
The anti-inflammatory and neuroprotective effects of short chain fatty acid (SCFA) butyrate have been explored in a wide array of neurological pathologies. It is a 4-carbon SCFA produced from the fermentation of dietary fibers by the gut-microbiota. As evident from previous literature, butyrate plays a wide array of functions in CNS and interestingly enhances the differentiation potential of Neural stem/Progenitor Cells (NSPCs). Japanese encephalitis virus (JEV) is a well-known member of the Flaviviridae family and has been shown to alter neural stem cell pool of the brain, causing devastating consequences. In this study, we administered sodium butyrate (NaB) post JEV infection in BALB/c mouse model to examine any possible amelioration of the viral infection in NSPCs. In addition, ex vivo neurospheres and in vitro model of NSPCs were also used to study the effect of sodium butyrate in JEV infection. As an unprecedented finding, butyrate treated infected animals presented early onset of symptoms, as compared to their respective JEV infected groups. Alongside, we observed an increased viral load in NSPCs isolated from these animals as well as in cell culture models upon sodium butyrate treatment. Cytometric bead array analysis also revealed an increase in inflammatory cytokines, particularly, MCP-1 and IL-6. Further, increased expression of the key members of the canonical NF-κB pathway, viz-a-viz p-NF-κB, p-Iκ-Bα and p-IKK was observed. Overall, the increased inflammation and cell death caused early symptom progression in NaB-treated JEV infected animal model, which is contradictory to the well documented protective nature of NaB and therefore a better understanding of SCFA-based modulation of the gut-brain axis in viral infections is required.
Collapse
Affiliation(s)
| | - Shivangi Sharma
- National Brain Research Centre, Manesar, Haryana, 122052, India
| | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana, 122052, India.
| |
Collapse
|
181
|
Cai QY, Yang Y, Ruan LL, Wang DD, Cui HL, Yang S, Liu WJ, Luo X, Liu TH. Effects of COVID-19 home quarantine on pregnancy outcomes of patients with gestational diabetes mellitus: a retrospective cohort study. J Matern Fetal Neonatal Med 2023; 36:2193284. [PMID: 36977601 DOI: 10.1080/14767058.2023.2193284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
OBJECTIVE This study aimed to evaluate the effects of the home quarantine on pregnancy outcomes of gestational diabetes mellitus (GDM) patients during the COVID-19 outbreak. METHODS The complete electronic medical records of patients with GDM with home quarantine history were collected and classified into the home quarantine group from 24 February 2020 to 24 November 2020. The same period of patients with GDM without home quarantine history were included in the control group from 2018 to 2019. The pregnant outcomes of the home quarantine and control groups were systematically compared, such as neonatal weight, head circumference, body length, one-minute Apgar score, fetal macrosomia, and pre-term delivery. RESULTS A total of 1358 patients with GDM were included in the analysis, including 484 in 2018, 468 in 2019, and 406 in 2020. Patients with GDM with home quarantine in 2020 had higher glycemic levels and adverse pregnancy outcomes than in 2018 and 2019, including higher cesarean section rates, lower Apgar scores, and higher incidence of macrosomia and umbilical cord around the neck. More importantly, the second trimester of home quarantine had brought a broader impact on pregnant women and fetuses. CONCLUSION Home quarantine has aggravated the condition of GDM pregnant women and brought more adverse pregnancy outcomes during the COVID-19 outbreak. Therefore, we suggested governments and hospitals strengthen lifestyle guidance, glucose management, and antenatal care for patients with GDM with home quarantine during public health emergencies.
Collapse
Affiliation(s)
- Qin-Yu Cai
- Department of Bioinformatics, The School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Yin Yang
- Department of Infection Controlling Section, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ling-Ling Ruan
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Dang-Dang Wang
- Department of Bioinformatics, The School of Basic Medical Science, Chongqing Medical University, Chongqing, China
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Han-Lin Cui
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shu Yang
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Wen-Jie Liu
- Department of Bioinformatics, The School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Xin Luo
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tai-Hang Liu
- Department of Bioinformatics, The School of Basic Medical Science, Chongqing Medical University, Chongqing, China
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|
182
|
Lee J, Jang J, Cha SR, Lee SB, Hong SH, Bae HS, Lee YJ, Yang SR. Recombinant Human Bone Morphogenetic Protein-2 Priming of Mesenchymal Stem Cells Ameliorate Acute Lung Injury by Inducing Regulatory T Cells. Immune Netw 2023; 23:e48. [PMID: 38188599 PMCID: PMC10767548 DOI: 10.4110/in.2023.23.e48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) possess immunoregulatory properties and their regulatory functions represent a potential therapy for acute lung injury (ALI). However, uncertainties remain with respect to defining MSCs-derived immunomodulatory pathways. Therefore, this study aimed to investigate the mechanism underlying the enhanced effect of human recombinant bone morphogenic protein-2 (rhBMP-2) primed ES-MSCs (MSCBMP2) in promoting Tregs in ALI mice. MSC were preconditioned with 100 ng/ml rhBMP-2 for 24 h, and then administrated to mice by intravenous injection after intratracheal injection of 1 mg/kg LPS. Treating MSCs with rhBMP-2 significantly increased cellular proliferation and migration, and cytokines array reveled that cytokines release by MSCBMP2 were associated with migration and growth. MSCBMP2 ameliorated LPS induced lung injury and reduced myeloperoxidase activity and permeability in mice exposed to LPS. Levels of inducible nitric oxide synthase were decreased while levels of total glutathione and superoxide dismutase activity were further increased via inhibition of phosphorylated STAT1 in ALI mice treated with MSCBMP2. MSCBMP2 treatment increased the protein level of IDO1, indicating an increase in Treg cells, and Foxp3+CD25+ Treg of CD4+ cells were further increased in ALI mice treated with MSCBMP2. In co-culture assays with MSCs and RAW264.7 cells, the protein level of IDO1 was further induced in MSCBMP2. Additionally, cytokine release of IL-10 was enhanced while both IL-6 and TNF-α were further inhibited. In conclusion, these findings suggest that MSCBMP2 has therapeutic potential to reduce massive inflammation of respiratory diseases by promoting Treg cells.
Collapse
Affiliation(s)
- Jooyeon Lee
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Jimin Jang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Sang-Ryul Cha
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Se Bi Lee
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Han-Sol Bae
- Cellular Therapeutics Team, Daewoong Pharmaceutical, Yongin 17028, Korea
| | - Young Jin Lee
- Cellular Therapeutics Team, Daewoong Pharmaceutical, Yongin 17028, Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
183
|
Dey A, Vaishak K, Deka D, Radhakrishnan AK, Paul S, Shanmugam P, Daniel AP, Pathak S, Duttaroy AK, Banerjee A. Epigenetic perspectives associated with COVID-19 infection and related cytokine storm: an updated review. Infection 2023; 51:1603-1618. [PMID: 36906872 PMCID: PMC10008189 DOI: 10.1007/s15010-023-02017-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 02/27/2023] [Indexed: 03/13/2023]
Abstract
PURPOSE The COVID-19 pandemic caused by the novel Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) has put the world in a medical crisis for the past three years; nearly 6.3 million lives have been diminished due to the virus outbreak. This review aims to update the recent findings on COVID-19 infections from an epigenetic scenario and develop future perspectives of epi-drugs to treat the disease. METHODS Original research articles and review studies related to COVID-19 were searched and analyzed from the Google Scholar/PubMed/Medline databases mainly between 2019 and 2022 to brief the recent work. RESULTS Numerous in-depth studies of the mechanisms used by SARS-CoV-2 have been going on to minimize the consequences of the viral outburst. Angiotensin-Converting Enzyme 2 receptors and Transmembrane serine protease 2 facilitate viral entry to the host cells. Upon internalization, it uses the host machinery to replicate viral copies and alter the downstream regulation of the normal cells, causing infection-related morbidities and mortalities. In addition, several epigenetic regulations such as DNA methylation, acetylation, histone modifications, microRNA, and other factors (age, sex, etc.) are responsible for the regulations of viral entry, its immune evasion, and cytokine responses also play a major modulatory role in COVID-19 severity, which has been discussed in detail in this review. CONCLUSION Findings of epigenetic regulation of viral pathogenicity open a new window for epi-drugs as a possible therapeutical approach against COVID-19.
Collapse
Affiliation(s)
- Amit Dey
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, TN, 603103, India
| | - K Vaishak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, TN, 603103, India
| | - Dikshita Deka
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, TN, 603103, India
| | - Arun Kumar Radhakrishnan
- Department of Pharmacology, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, TN, India
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No.500 Fracc., CP 76130, San Pablo, Querétaro, Mexico
| | - Priyadarshini Shanmugam
- Department of Microbiology, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, TN, 603103, India
| | - Alice Peace Daniel
- Department of Microbiology, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, TN, 603103, India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, TN, 603103, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, TN, 603103, India.
| |
Collapse
|
184
|
Biagini D, Oliveri P, Baj A, Gasperina DD, Ferrante FD, Lomonaco T, Ghimenti S, Lenzi A, Bonini A, Vivaldi F, Oger C, Galano JM, Balas L, Durand T, Maggi F, Di Francesco F. The effect of SARS-CoV-2 variants on the plasma oxylipins and PUFAs of COVID-19 patients. Prostaglandins Other Lipid Mediat 2023; 169:106770. [PMID: 37633481 DOI: 10.1016/j.prostaglandins.2023.106770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Oxylipins are important signalling compounds that are significantly involved in the regulation of the immune system and the resolution of inflammation. Lipid metabolism is strongly activated upon SARS-CoV-2 infection, however the modulating effects of oxylipins induced by different variants remain unexplored. Here, we compare the plasma profiles of thirty-seven oxylipins and four PUFAs in subjects infected with Wild-type, Alpha (B.1.1.7), Delta (B.1.617.2), and Omicron (B.1.1.529) variants. The results suggest that oxidative stress and inflammation resulting from COVID-19 were highly dependent on the SARS-CoV-2 variant, and that the Wild-type elicited the strongest inflammatory storm. The Alpha and Delta variants induced a comparable lipid profile alteration upon infection, which differed significantly from Omicron. The latter variant increased the levels of pro-inflammatory mediators and decreased the levels of omega-3 PUFA in infected patients. We speculate that changes in therapeutics, vaccination, and prior infections may have a role in the alteration of the oxylipin profile besides viral mutations. The results shed new light on the evolution of the inflammatory response in COVID-19.
Collapse
Affiliation(s)
- Denise Biagini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy.
| | | | - Andreina Baj
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | | | | | - Tommaso Lomonaco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Silvia Ghimenti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Alessio Lenzi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Andrea Bonini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Federico Vivaldi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Camille Oger
- Institut des Biomolécules Max Mousseron (IBMM), Pôle Chimie Balard Recherche, University of Montpellier, ENSCN, UMR 5247 CNRS, France
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron (IBMM), Pôle Chimie Balard Recherche, University of Montpellier, ENSCN, UMR 5247 CNRS, France
| | - Laurence Balas
- Institut des Biomolécules Max Mousseron (IBMM), Pôle Chimie Balard Recherche, University of Montpellier, ENSCN, UMR 5247 CNRS, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron (IBMM), Pôle Chimie Balard Recherche, University of Montpellier, ENSCN, UMR 5247 CNRS, France
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| | - Fabio Di Francesco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy.
| |
Collapse
|
185
|
Syage A, Pachow C, Cheng Y, Mangale V, Green KN, Lane TE. Microglia influence immune responses and restrict neurologic disease in response to central nervous system infection by a neurotropic murine coronavirus. Front Cell Neurosci 2023; 17:1291255. [PMID: 38099152 PMCID: PMC10719854 DOI: 10.3389/fncel.2023.1291255] [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: 09/08/2023] [Accepted: 11/09/2023] [Indexed: 12/17/2023] Open
Abstract
Intracranial (i.c.) inoculation of susceptible mice with a glial-tropic strain of mouse hepatitis virus (JHMV), a murine coronavirus, results in an acute encephalomyelitis followed by viral persistence in white matter tracts accompanied by chronic neuroinflammation and demyelination. Microglia serve numerous functions including maintenance of the healthy central nervous system (CNS) and are among the first responders to injury or infection. More recently, studies have demonstrated that microglia aid in tailoring innate and adaptive immune responses following infection by neurotropic viruses including flaviviruses, herpesviruses, and picornaviruses. These findings have emphasized an important role for microglia in host defense against these viral pathogens. In addition, microglia are also critical in optimizing immune-mediated control of JHMV replication within the CNS while restricting the severity of demyelination and enhancing remyelination. This review will highlight our current understanding of the molecular and cellular mechanisms by which microglia aid in host defense, limit neurologic disease, and promote repair following CNS infection by a neurotropic murine coronavirus.
Collapse
Affiliation(s)
- Amber Syage
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| | - Collin Pachow
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Yuting Cheng
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Vrushali Mangale
- Department of Pathology, University of Utah, Salt Lake City, UT, United States
| | - Kim N. Green
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| | - Thomas E. Lane
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Center for Virus Research, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
186
|
Cao Y, Han Y, Wu J, Sun J, Dai Y, Qiao G, Li K, Li A, Zhang Y, Ma Y, Song Q. During the Omicron Pandemic Wave, the Severe Systemic Inflammatory Status of COVID-19 Indicated a Higher Risk of In-Hospital Mortality and Mediated the Clinical Efficacy of Corticosteroids. Infect Drug Resist 2023; 16:7377-7387. [PMID: 38053579 PMCID: PMC10695125 DOI: 10.2147/idr.s432679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/16/2023] [Indexed: 12/07/2023] Open
Abstract
Background For the distinct immune/inflammatory responses from Omicron variant infection, this study aimed to investigate the diagnostic efficacy of systemic inflammatory indicators and the clinical efficacy of corticosteroids on the in-hospital mortality among COVID-19 patients. Methods Under a retrospective cohort study, 1081 COVID-19 patients were recruited from Beijing Youan Hospital, Capital Medical University between November 16, 2022 and January 30, 2023. We chose neutrophil-to-lymphocyte ratio (NLR), CRP-to-lymphocyte ratio (CLR), and CRP-to-albumin ratio (CAR) as the systemic inflammatory indicators. Receiver operating curve (ROC) and multivariate logistic regression analysis were used to determine the diagnostic efficacy of systemic inflammatory indicators and the association between systemic inflammatory indicators and in-hospital mortality. Results Among 684 patients included in analysis, 96 died during hospitalization. NLR, CLR and CAR performed well (with an area under the curve (AUC) greater than 0.75) in discriminating in-hospital mortality among COVID-19 patients. The severe status of systemic inflammation, with optimal cut-off value derived from ROC analysis, significantly associated higher risk of in-hospital mortality (OR = 3.81 for NLR ≥ 6.131; OR = 3.76 for CLR ≥ 45.455; OR = 5.10 for CAR ≥ 1.436). Corticosteroids use within 72 hours of admission increased the in-hospital mortality 2.88-fold for COVID-19 patients. In the subgroup of patients with severe systemic inflammation, corticosteroids increased the risk of in-hospital mortality (OR = 2.11 for NLR, p = 0.055; OR = 2.94 for CLR, p = 0.005; OR = 2.31 for CAR, p = 0.036). Conclusion Systemic inflammatory indicators had good diagnostic performance for in-hospital mortality. Patients with severe systemic inflammatory status should not receive corticosteroid treatment and further studies are warranted for confirmation.
Collapse
Affiliation(s)
- Yu Cao
- Department of Clinical Epidemiology, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Ying Han
- Center of Liver Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jiangping Wu
- Department of Clinical Epidemiology, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jianping Sun
- Center of Biobank, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yanchao Dai
- Center of Biobank, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Guifang Qiao
- Center of Biobank, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Kang Li
- Center of Biobank, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Ang Li
- Center of Biobank, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yonghong Zhang
- Department of Hepatic Intervention, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yingmin Ma
- Department of Respiratory and Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Qingkun Song
- Department of Clinical Epidemiology, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Center of Biobank, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
187
|
Xia W, Jiang T, Tan Y, Li C, Wu S, Mei B. Characteristics of hematological parameters on admission in COVID-19 Omicron variant infected in Chinese population: a large-scale retrospective study. BMC Infect Dis 2023; 23:835. [PMID: 38012548 PMCID: PMC10683119 DOI: 10.1186/s12879-023-08771-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND The Omicron variant of SARS-CoV-2, currently the most prevalent strain, has rapidly spread in Jingzhou, China, due to changes in the country's epidemic prevention policy, resulting in an unprecedented increase in cases. Previous studies reported hematological parameters' predictive value in COVID-19 severity and prognosis, but their relevance for early diagnosis in patients infected by the Omicron variant, particularly in high-risk pneumonia cases, remains unclear. Our study aimed to evaluate these parameters as early warning indicators for Omicron-infected patients in fever clinics and those with pulmonary infections (PI). METHODS A total of 2,021 COVID-19 patients admitted to the fever clinic and infectious disease department of Jingzhou Hospital Affiliated to Yangtze University from November 1, 2022, to December 31, 2022, were retrospectively recruited. Demographic and hematological parameters were obtained from the electronic medical records of eligible patients. These hematological parameters were analyzed by receiver operating characteristic (ROC) curves to determine whether they can be used for early diagnosis of COVID-19 patients in fever clinics and the presence of PI in COVID-19 patients. RESULTS Statistical differences in hematological parameters were observed between COVID-19 patients with fever and PI and control groups (P < 0.01). The ROC curve further demonstrated that lymphocyte (LYM) counts, neutrophil (NEU) counts, monocyte-to-lymphocyte ratios (MLR), platelet-to-lymphocyte ratios (PLR), white blood cell counts (WBC), and mean corpuscular hemoglobin concentration (MCHC) were the top 6 indicators in diagnosing Omicron infection with fever, with area under the curve (AUC) values of 0.738, 0.718, 0.713, 0.702, 0.700, and 0.687, respectively (P < 0.01). Furthermore, the mean platelet volume (MPV) with an AUC of 0.764, red blood cell count (RBC) with 0.753, hematocrit (HCT) with 0.698, MLR with 0.694, mean corpuscular hemoglobin (MCH) with 0.676, and systemic inflammation response indexes (SIRI) with 0.673 were the top 6 indicators for the diagnosis of COVID-19 patients with PI (P < 0.01). CONCLUSIONS LYM, NEU, MLR, PLR, WBC, and MCHC can serve as potential prescreening indicators for Omicron infection in fever clinics. Additionally, MPV, RBC, HCT, MLR, MCH, and SIRI can predict the presence of PI in COVID-19 patients infected by the Omicron variant.
Collapse
Affiliation(s)
- Wei Xia
- Department of Laboratory Medicine, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, 434020, China
| | - Tao Jiang
- Department of Laboratory Medicine, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, 434020, China
| | - Yafeng Tan
- Department of Laboratory Medicine, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, 434020, China
| | - Chengbin Li
- Department of Laboratory Medicine, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, 434020, China
| | - Song Wu
- Department of Laboratory Medicine, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, 434020, China
| | - Bing Mei
- Department of Laboratory Medicine, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, 434020, China.
| |
Collapse
|
188
|
Peng B, Li J, Chen M, Yang X, Hao M, Wu F, Yang Z, Liu D. Clinical value of glucocorticoids for severe community-acquired pneumonia: A systematic review and meta-analysis based on randomized controlled trials. Medicine (Baltimore) 2023; 102:e36047. [PMID: 37986401 PMCID: PMC10659673 DOI: 10.1097/md.0000000000036047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Severe community-acquired pneumonia (sCAP) is characterized by severe symptoms and a poor prognosis, especially with the recent global impact of novel coronavirus in recent years. The use of glucocorticoids in sCAP is currently a subject of debate. To evaluate the clinical efficacy and safety of glucocorticoids and provide guidance for their rational use in clinical practice, we conducted this study. METHODS We searched PubMed, Web of Science, and China National Knowledge Infrastructure using the following search terms: "pneumonia", "pneumonias", "Pulmonary Inflammation", "Pulmonary Inflammations", "Lung Inflammation", and "Lung Inflammations". The primary outcomes included mortality and the length of hospital stay. The secondary outcomes included the duration of mechanical ventilation, duration of vasoactive drug use, gastrointestinal bleeding, and multiple infections. The Cochrane Collaboration was used to assess the risk of bias of the included studies. Stata/MP14 was used for meta-analysis. RESULTS These studies contained information on 1252 patients who received glucocorticoids and 1280 patients who did not. Meta-analysis showed that there was no difference in terms of mortality [risk ratio (RR) = 0.93, 95% confidence interval (CI): 0.81-1.07, P > .05], gastrointestinal bleeding (RR = 1.38, 95% CI: 0.83-2.30, P < .05), multiple infections (RR = 1.17, 95% CI: 0.90-1.53, P > .05) and length of hospital stay (mean difference [MD] = -0.87, 95% CI: -2.35 to 0.61, P > .05) between the hormonal and nonhormonal groups. However, there was a significant difference in the duration of mechanical ventilation (MD = -1.54; 95% CI, -1.89 to -1.12, P < .05) and the duration of use of vasoactive drugs (MD = -14.09, 95% CI: -15.72 to -12.46, P < .05). CONCLUSION Glucocorticoids reduced the duration of mechanical ventilation duration and vasoactive drug use in sCAP patients without increasing the risk of adverse events including hyperglycemia and multiple infections. However, there was no significant difference in mortality or length of hospital stay in sCAP patients between glucocorticoid and non-glucocorticoid groups. Glucocorticoids could be recommended for patients with sCAP with respiratory failure or hemodynamic instability.
Collapse
Affiliation(s)
- Biao Peng
- Department of Respiratory and Critical Care Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Jin Li
- Medical College of Jishou University, Jishou, China
| | - Minwei Chen
- Department of Critical Care Medicine, Anhua County People’s Hospital, Anhua, China
| | - Xianghui Yang
- Department of Oncology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Meng Hao
- Department of Endocrinology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Feifei Wu
- Department of Respiratory and Critical Care Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - ZhiChao Yang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Da Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| |
Collapse
|
189
|
Mazurak VC, Rivas-Serna IM, Parsons SR, Monirujjaman M, Maybank KE, Woo SK, Rewa OG, Cave AJ, Richard C, Clandinin MT. Plasma essential fatty acid on hospital admission is a marker of COVID-19 disease severity. Sci Rep 2023; 13:18973. [PMID: 37923927 PMCID: PMC10624896 DOI: 10.1038/s41598-023-46247-0] [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: 06/15/2023] [Accepted: 10/30/2023] [Indexed: 11/06/2023] Open
Abstract
It is important for allocation of resources to predict those COVID patients at high risk of dying or organ failure. Early signals to initiate cellular events of host immunity can be derived from essential fatty acid metabolites preceding the cascade of proinflammatory signals. Much research has focused on understanding later proinflammatory responses. We assessed if remodelling of plasma phospholipid content of essential fatty acids by the COVID-19 virus provides early markers for potential death and disease severity. Here we show that, at hospital admission, COVID-19 infected subjects who survive exhibit higher proportions of C20:4n-6 in plasma phospholipids concurrent with marked proinflammatory cytokine elevation in plasma compared to healthy subjects. In contrast, more than half of subjects who die of this virus exhibit very low C18:2n-6 and C20:4n-6 content in plasma phospholipids on hospital admission compared with healthy control subjects. Moreover, in these subjects who die, the low level of primary inflammatory signals indicates limited or aberrant stimulation of host immunity. We conclude that COVID-19 infection results in early fundamental remodelling of essential fatty acid metabolism. In subjects with high mortality, it appears that plasma n-6 fatty acid content is too low to stimulate cellular events of host immunity.
Collapse
Affiliation(s)
- Vera C Mazurak
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Irma Magaly Rivas-Serna
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Sarah R Parsons
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Md Monirujjaman
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Krista E Maybank
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Stanley K Woo
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Oleksa G Rewa
- Department of Critical Care Medicine, Faculty of Medicine, University of Alberta, Edmonton, Canada
| | - Andrew J Cave
- Department of Family Medicine, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Caroline Richard
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - M Thomas Clandinin
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada.
- Department of Medicine, University of Alberta, Edmonton, T6G 2P5, Canada.
| |
Collapse
|
190
|
Chaubey GK, Dilawari R, Modanwal R, Talukdar S, Dhiman A, Raje CI, Raje M. Excess iron aggravates the severity of COVID-19 infection. Free Radic Biol Med 2023; 208:186-193. [PMID: 37553026 DOI: 10.1016/j.freeradbiomed.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/10/2023]
Abstract
Coronavirus disease-19 (COVID-19) can induce severe inflammation of the lungs and respiratory system. Severe COVID-19 is frequently associated with hyper inflammation and hyper-ferritinemia. High iron levels are known to trigger pro-inflammatory effects. Cumulative iron loading negatively impacts on a patients innate immune effector functions and increases the risk for infection related complications. Prognosis of severe acute respiratory SARS-CoV-2 patients may be impacted by iron excess. Iron is an essential co-factor for numerous essential cellular enzymes and vital cellular operations. Viruses hijack cells in order to replicate, and efficient replication requires an iron-replete host. Utilizing iron loaded cells in culture we evaluated their susceptibility to infection by pseudovirus expressing the SARS-CoV-2 spike protein and resultant cellular inflammatory response. We observed that, high levels of iron enhanced host cell ACE2 receptor expression contributing to higher infectivity of pseudovirus. In vitro Cellular iron overload also synergistically enhanced the levels of; reactive oxygen species, reactive nitrogen species, pro-inflammatory cytokines (IL-1β, IL-6, IL-8 & TNF-α) and chemokine (CXCL-1&CCL-4) production in response to inflammatory stimulation of cells with spike protein. These results were confirmed using an in vivo mouse model. In future, limiting iron levels may be a promising adjuvant strategy in treating viral infection.
Collapse
Affiliation(s)
| | - Rahul Dilawari
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Radheshyam Modanwal
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Sharmila Talukdar
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Asmita Dhiman
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Chaaya Iyengar Raje
- National Institute of Pharmaceutical Education & Research, Phase X, Sector 67, SAS Nagar, Punjab, 160062, India
| | - Manoj Raje
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India.
| |
Collapse
|
191
|
Kumar M. Hydrogen sulfide: From a toxic gas to a potential therapy for COVID-19 and inflammatory disorders. Nitric Oxide 2023; 140-141:8-15. [PMID: 37648016 DOI: 10.1016/j.niox.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/06/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023]
Abstract
COVID-19 has been shown to induce inflammatory disorders and CNS manifestations. Swift and efficient treatment strategies are urgently warranted for the management of COVID, inflammatory and neurological disorders. Hydrogen sulfide (H2S) has been associated with several clinical disorders due to its potential to influence a broad range of biological signalling pathways. According to recent clinical studies, COVID patients with lower physiological H2S had higher fatality rates. These findings clearly demonstrate an inverse correlation between H2S levels and the severity of COVID-19. H2S has been proposed as a protective molecule because of its antioxidant, anti-inflammatory, and antiviral properties. Various H2S-releasing prodrugs, hybrids and natural compounds have been tested for their therapeutic efficacy in viral infections and inflammatory disorders. In this review, I am highlighting the rationale for using H2S-based interventions for the management of COVID-19 and post-infection inflammatory disorders including neuroinflammation. I am also proposing therepurposing of existing H2S-releasing prodrugs, developing new NO-H2S-hybrids, targeting H2S metabolic pathways, and using H2S-producing dietary supplements as viable defensive strategies against SARS-CoV-2 infection and COVID-19 pathologies.
Collapse
Affiliation(s)
- Mohit Kumar
- Centre for Excellence in Functional Foods, Food and Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Punjab, 140306, India.
| |
Collapse
|
192
|
Zivancevic-Simonovic S, Minic R, Cupurdija V, Stanojevic-Pirkovic M, Milosevic-Djordjevic O, Jakovljevic V, Mihaljevic O. Transforming growth factor beta 1 (TGF-β1) in COVID-19 patients: relation to platelets and association with the disease outcome. Mol Cell Biochem 2023; 478:2461-2471. [PMID: 36869188 PMCID: PMC9984293 DOI: 10.1007/s11010-023-04674-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 02/07/2023] [Indexed: 03/05/2023]
Abstract
Transforming growth factor beta (TGF-β) is a ubiquitously distributed cytokine known to contribute to the pathogenesis of numerous pathological processes. The aim of this study was to measure serum concentrations of TGF-β1 in severely ill COVID-19 patients and to analyze its relationship with selected hematological and biochemical parameters and with the disease outcome. The study population included 53 COVID-19 patients with severe clinical expression of the disease and 15 control subjects. TGF-β1 was determined in serum samples and supernatants from PHA-stimulated whole blood cultures using ELISA assay. Biochemical and hematological parameters were analyzed using standard accepted methods. Our results showed that serum levels of TGF-β1 in COVID-19 patients and controls correlate with the platelet counts. Also, positive correlations of TGF-β1 with white blood cell and lymphocyte counts, platelet-to-lymphocyte (PLR) ratio, and fibrinogen level were shown, while negative correlations of this cytokine with platelet distribution width (PDW), D-dimer and activated partial thromboplastin time (a-PTT) values in COVID-19 patients were observed. The lower serum values of TGF-β1 were associated with the unfavorable outcome of COVID-19. In conclusion, TGF-β1 levels were strongly associated with platelet counts and unfavorable disease outcome of severely ill COVID-19 patients.
Collapse
Affiliation(s)
| | - Rajna Minic
- Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Vojislav Cupurdija
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Marijana Stanojevic-Pirkovic
- University Clinical Center Kragujevac, Kragujevac, Serbia
- Department of Biochemistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Olgica Mihaljevic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
193
|
Liu S, Zhao Y, Feng X, Xu H. SARS-CoV-2 infection threatening intestinal health: A review of potential mechanisms and treatment strategies. Crit Rev Food Sci Nutr 2023; 63:12578-12596. [PMID: 35894645 DOI: 10.1080/10408398.2022.2103090] [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] [Indexed: 01/08/2023]
Abstract
The outbreak of the COVID-19 pandemic has brought great problems to mankind, including economic recession and poor health. COVID-19 patients are frequently reported with gastrointestinal symptoms such as diarrhea and vomiting in clinical diagnosis. Maintaining intestinal health is the key guarantee to maintain the normal function of multiple organs, otherwise it will be a disaster. Therefore, the purpose of this review was deeply understanded the potential mechanism of SARS-CoV-2 infection threatening intestinal health and put forward reasonable treatment strategies. Combined with the existing researches, we summarized the mechanism of SARS-CoV-2 infection threatening intestinal health, including intestinal microbiome disruption, intestinal barrier dysfunction, intestinal oxidative stress and intestinal cytokine storm. These adverse intestinal events may affect other organs through the circulatory system or aggravate the course of the disease. Typically, intestinal disadvantage may promote the progression of SARS-CoV-2 through the gut-lung axis and increase the disease degree of COVID-19 patients. In view of the lack of specific drugs to inhibit SARS-CoV-2 replication, the current review described new strategies of probiotics, prebiotics, postbiotics and nutrients to combat SARS-CoV-2 infection and maintain intestinal health. To provide new insights for the prevention and treatment of gastrointestinal symptoms and pneumonia in patients with COVID-19.
Collapse
Affiliation(s)
- Shanji Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yu Zhao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Xiaoyan Feng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
194
|
Fazlollahi A, Zahmatyar M, Shamekh A, Motamedi A, Seyedi F, Seyedmirzaei H, Mousavi SE, Nejadghaderi SA, Sullman MJM, Kolahi AA, Arshi S, Safiri S. Electroencephalographic findings post-COVID-19 vaccination: A systematic review of case reports and case series. Rev Med Virol 2023; 33:e2484. [PMID: 37807809 DOI: 10.1002/rmv.2484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/14/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023]
Abstract
A number of different neurological complications have been reported following vaccination against the coronavirus disease 2019 (COVID-19). Electroencephalogram (EEG) is one of the modalities used to evaluate the neurological complications of diseases. The aim of the present study was to identify the EEG changes in participants vaccinated against COVID-19. PubMed, Scopus, Web of Science, medRxiv, and Google Scholar were searched up to 1 September 2022, with terms related to COVID-19 vaccines, EEG, neurological signs/symptoms, or neurological disorders. All case reports and case series were included if the participants had received at least one dose of a COVID-19 vaccine and a post vaccination EEG report was also reported. We used the Joanna Briggs Institute (JBI) Critical Appraisal Checklist for case reports and case series to appraise the methodological quality of the included studies. Thirty-one studies were included, which were comprised of 24 case reports and seven case series and a total of 36 participants. Generalised slowing and non-convulsive focal status epilepticus were the most common EEG findings post-COVID-19 vaccination. The most frequent symptoms were headache, fatigue, generalised weakness, and vomiting. In addition, the most common signs were encephalopathy, post-ictal phases, and confusion. Encephalitis, acute disseminated encephalomyelitis, and post-vaccinal encephalopathy were the most commonly diagnosed adverse events. Furthermore, most of the imaging studies appeared normal. The EEG reports mainly showed background slowing and epileptiform discharges, encephalitis, encephalopathies, and demyelinating disorders. Future studies with larger samples and more vaccine types may help to further unravel the potential neurological effects of COVID-19 vaccinations on recipients.
Collapse
Affiliation(s)
- Asra Fazlollahi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Zahmatyar
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Motamedi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Seyedi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Homa Seyedmirzaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Interdisciplinary Neuroscience Research Program (INRP), Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ehsan Mousavi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Tuberculosis and Lung Diseases Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Aria Nejadghaderi
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mark J M Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahnam Arshi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Safiri
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
195
|
Bryson TD, Harding P. Prostaglandin E 2 and myocarditis; friend or foe? Biochem Pharmacol 2023; 217:115813. [PMID: 37722627 DOI: 10.1016/j.bcp.2023.115813] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
This review article summarizes the role of prostaglandin E2 (PGE2) and its receptors (EP1-EP4) as it relates to the inflammatory cardiomyopathy, myocarditis. During the COVID-19 pandemic, the onset of myocarditis in a subset of patients prompted a debate on the use of nonsteroidal anti-inflammatory drugs (NSAIDs), like ibuprofen, which act to inhibit the actions of prostaglandins. This review aims to further understanding of the role of PGE2 in the pathogenesis or protection of the myocardium in myocarditis. Inflammatory cardiomyopathies encompass a broad spectrum of disorders, all characterized by cardiac inflammation. Therefore, for the purpose of this review, the authors have placed particular emphasis on etiologies of myocarditis where effects of PGE2 have been documented.
Collapse
Affiliation(s)
- Timothy D Bryson
- Hypertension & Vascular Research Division, Department of Internal Medicine, Henry Ford Health, Detroit, MI, USA
| | - Pamela Harding
- Hypertension & Vascular Research Division, Department of Internal Medicine, Henry Ford Health, Detroit, MI, USA; Department of Physiology, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
196
|
Vetrugno L, Castaldo N, Fantin A, Deana C, Cortegiani A, Longhini F, Forfori F, Cammarota G, Grieco DL, Isola M, Navalesi P, Maggiore SM, Bassetti M, Chetta A, Confalonieri M, De Martino M, Ferrari G, Francisi D, Luzzati R, Meini S, Scozzafava M, Sozio E, Tascini C, Bassi F, Patruno V, De Robertis E, Aldieri C, Ball L, Baratella E, Bartoletti M, Boscolo A, Burgazzi B, Catalanotti V, Confalonieri P, Corcione S, De Rosa FG, De Simoni A, Bono VD, Tria RD, Forlani S, Giacobbe DR, Granozzi B, Labate L, Lococo S, Lupia T, Matellon C, Mehrabi S, Morosi S, Mongodi S, Mura M, Nava S, Pol R, Pettenuzzo T, Quyen NH, Rescigno C, Righi E, Ruaro B, Salton F, Scabini S, Scarda A, Sibani M, Tacconelli E, Tartaglione G, Tazza B, Vania E, Viale P, Vianello A, Visentin A, Zuccon U, Meroi F, Buonsenso D. Ventilatory associated barotrauma in COVID-19 patients: A multicenter observational case control study (COVI-MIX-study). Pulmonology 2023; 29:457-468. [PMID: 36669936 PMCID: PMC9684110 DOI: 10.1016/j.pulmoe.2022.11.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/25/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The risk of barotrauma associated with different types of ventilatory support is unclear in COVID-19 patients. The primary aim of this study was to evaluate the effect of the different respiratory support strategies on barotrauma occurrence; we also sought to determine the frequency of barotrauma and the clinical characteristics of the patients who experienced this complication. METHODS This multicentre retrospective case-control study from 1 March 2020 to 28 February 2021 included COVID-19 patients who experienced barotrauma during hospital stay. They were matched with controls in a 1:1 ratio for the same admission period in the same ward of treatment. Univariable and multivariable logistic regression (OR) were performed to explore which factors were associated with barotrauma and in-hospital death. RESULTS We included 200 cases and 200 controls. Invasive mechanical ventilation was used in 39.3% of patients in the barotrauma group, and in 20.1% of controls (p<0.001). Receiving non-invasive ventilation (C-PAP/PSV) instead of conventional oxygen therapy (COT) increased the risk of barotrauma (OR 5.04, 95% CI 2.30 - 11.08, p<0.001), similarly for invasive mechanical ventilation (OR 6.24, 95% CI 2.86-13.60, p<0.001). High Flow Nasal Oxygen (HFNO), compared with COT, did not significantly increase the risk of barotrauma. Barotrauma frequency occurred in 1.00% [95% CI 0.88-1.16] of patients; these were older (p=0.022) and more frequently immunosuppressed (p=0.013). Barotrauma was shown to be an independent risk for death (OR 5.32, 95% CI 2.82-10.03, p<0.001). CONCLUSIONS C-PAP/PSV compared with COT or HFNO increased the risk of barotrauma; otherwise HFNO did not. Barotrauma was recorded in 1.00% of patients, affecting mainly patients with more severe COVID-19 disease. Barotrauma was independently associated with mortality. TRIAL REGISTRATION this case-control study was prospectively registered in clinicaltrial.gov as NCT04897152 (on 21 May 2021).
Collapse
Affiliation(s)
- Luigi Vetrugno
- Department of Anesthesiology, Critical Care Medicine and Emergency, SS. Annunziata Hospital, Chieti, Italy; Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy.
| | - Nadia Castaldo
- Pulmonology Unit, Department of Cardiothoracic Surgery, Health Integrated Agency of Friuli Centrale, Udine, Italy
| | - Alberto Fantin
- Pulmonology Unit, Department of Cardiothoracic Surgery, Health Integrated Agency of Friuli Centrale, Udine, Italy
| | - Cristian Deana
- Department of Anesthesia and Intensive Care, Health Integrated Agency of Friuli Centrale, Udine, Italy
| | - Andrea Cortegiani
- Department of Surgical, Oncological and Oral Science (Di.Chir.On.S.), University of Palermo, Palermo, Italy; Department of Anesthesia Intensive Care and Emergency, Policlinico Paolo Giaccone, Palermo, Italy
| | - Federico Longhini
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Francesco Forfori
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, AOUP-Pisa, Italy
| | - Gianmaria Cammarota
- Anesthesia and Intensive Care Service 2, University Hospital of Perugia, Perugia, Italy; Department of Medicine and Surgery, Universiy of Perugia, Perugia, Italy
| | - Domenico Luca Grieco
- Department of Anesthesiology and Intensive Care Medicine, Catholic University of The Sacred Heart, Rome, Italy; Department of Anesthesia, Emergency and Intensive Care Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Miriam Isola
- Department of Medicine, University of Udine, Udine, Italy
| | - Paolo Navalesi
- Institute of Anaesthesia and Intensive Care, Padua University Hospital, Padua, Italy; Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Salvatore Maurizio Maggiore
- Department of Anesthesiology, Critical Care Medicine and Emergency, SS. Annunziata Hospital, Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, Gabriele d'Annunzio University of Chieti Pescara, Chieti, Italy
| | - Matteo Bassetti
- Infectious Diseases Unit, Ospedale Policlinico San Martino - IRCCS, Genoa, Italy; Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Alfredo Chetta
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Marco Confalonieri
- Department of Pulmonology, University Hospital of Cattinara, Trieste, Italy; University of Trieste, Trieste, Italy
| | | | - Giovanni Ferrari
- Pneumologia e Unità di Terapia Semi Intensiva Respiratoria, AO Umberto I Mauriziano, Turin, Italy
| | - Daniela Francisi
- Department of Infectious Disease "S. Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy
| | - Roberto Luzzati
- Infectious Disease Unit, University of Trieste, Trieste, Italy
| | - Simone Meini
- U.O. Medicina Interna, Felice Lotti Hospital, Azienda USL Toscana Nord-Ovest, Pontedera, Italy
| | | | - Emanuela Sozio
- Infectious Diseases Division, Health Integrated Agency of Friuli Centrale, Udine, Italy
| | - Carlo Tascini
- Department of Medicine, University of Udine, Udine, Italy; Infectious Diseases Division, Health Integrated Agency of Friuli Centrale, Udine, Italy
| | - Flavio Bassi
- Department of Anesthesia and Intensive Care, Health Integrated Agency of Friuli Centrale, Udine, Italy
| | - Vincenzo Patruno
- Pulmonology Unit, Department of Cardiothoracic Surgery, Health Integrated Agency of Friuli Centrale, Udine, Italy
| | - Edoardo De Robertis
- Anesthesia and Intensive Care Service 2, University Hospital of Perugia, Perugia, Italy; Department of Medicine and Surgery, Universiy of Perugia, Perugia, Italy
| | - Chiara Aldieri
- Division of Infectious Diseases, Department of Medicine, Hospital Santa Croce e Carle, Cuneo, Italy
| | - Lorenzo Ball
- Anesthesia and Intensive Care, Ospedale Policlinico San Martino-IRCCS, Genoa, Italy; Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Elisa Baratella
- Department of Pulmonology, University Hospital of Cattinara, Trieste, Italy; University of Trieste, Trieste, Italy
| | - Michele Bartoletti
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, Policlinico Sant'Orsola, Bologna, Italy
| | - Annalisa Boscolo
- Institute of Anaesthesia and Intensive Care, Padua University Hospital, Padua, Italy
| | - Barbara Burgazzi
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Vito Catalanotti
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, Policlinico Sant'Orsola, Bologna, Italy
| | - Paola Confalonieri
- Department of Pulmonology, University Hospital of Cattinara, Trieste, Italy; University of Trieste, Trieste, Italy
| | - Silvia Corcione
- Department of Medical Sciences, University of Turin, Infectious Diseases, City of Health and Sciences, Turin, Italy
| | - Francesco Giuseppe De Rosa
- Infectious Diseases Unit, Cardinal Massaia Hospital, Asti, Italy; Infectious Diseases Unit, Cardinal Massaia Hospital, Asti, Italy
| | - Alessandro De Simoni
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Valerio Del Bono
- Division of Infectious Diseases, Department of Medicine, Hospital Santa Croce e Carle, Cuneo, Italy
| | - Roberta Di Tria
- Pneumologia e Unità di Terapia Semi Intensiva Respiratoria, AO Umberto I Mauriziano, Turin, Italy
| | - Sara Forlani
- Pulmonary Medicine Unit, Lodi General Hospital, Lodi, Italy
| | - Daniele Roberto Giacobbe
- Infectious Diseases Unit, Ospedale Policlinico San Martino - IRCCS, Genoa, Italy; Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Bianca Granozzi
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, Policlinico Sant'Orsola, Bologna, Italy
| | - Laura Labate
- Infectious Diseases Unit, Ospedale Policlinico San Martino - IRCCS, Genoa, Italy; Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Sara Lococo
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Tommaso Lupia
- Infectious Diseases Unit, Cardinal Massaia Hospital, Asti, Italy
| | - Carola Matellon
- Department of Anesthesia and Intensive Care, Health Integrated Agency of Friuli Centrale, Udine, Italy
| | - Sara Mehrabi
- Infectious Diseases Division, Diagnostics and Public Health Department, University of Verona, Verona, Italy
| | - Sabrina Morosi
- Department of Infectious Disease "S. Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy
| | - Silvia Mongodi
- Anaesthesia and Intensive Care, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maddalena Mura
- U.O. Medicina Interna, Felice Lotti Hospital, Azienda USL Toscana Nord-Ovest, Pontedera, Italy
| | - Stefano Nava
- Department of Clinical, Integrated and Experimental Medicine (DIMES), University of Bologna, Bologna, Italy; Respiratory and Critical Care Unit, Sant Orsola University Hospital, Bologna, Italy
| | - Riccardo Pol
- Infectious Disease Unit, University of Trieste, Trieste, Italy
| | - Tommaso Pettenuzzo
- Institute of Anaesthesia and Intensive Care, Padua University Hospital, Padua, Italy
| | - Nguyen Hoang Quyen
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Carolina Rescigno
- UOC Malattie Infettive ad Indirizzo Neurologico, AORN Ospedali dei Colli, P.O. "D. Cotugno", Naples, Italy
| | - Elda Righi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Barbara Ruaro
- Department of Pulmonology, University Hospital of Cattinara, Trieste, Italy; University of Trieste, Trieste, Italy
| | - Francesco Salton
- Department of Pulmonology, University Hospital of Cattinara, Trieste, Italy; University of Trieste, Trieste, Italy
| | - Silvia Scabini
- Department of Medical Sciences, University of Turin, Infectious Diseases, City of Health and Sciences, Turin, Italy
| | - Angelo Scarda
- Respiratory Disease Unit, "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | - Marcella Sibani
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Evelina Tacconelli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Gennaro Tartaglione
- Respiratory Disease Unit, "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | - Beatrice Tazza
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, Policlinico Sant'Orsola, Bologna, Italy
| | - Eleonora Vania
- Infectious Diseases Division, Health Integrated Agency of Friuli Centrale, Udine, Italy
| | - Pierluigi Viale
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, Policlinico Sant'Orsola, Bologna, Italy
| | - Andrea Vianello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Alessandro Visentin
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Umberto Zuccon
- Respiratory Disease Unit, "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | | | - Danilo Buonsenso
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
197
|
Tsilioni I, Theoharides TC. Recombinant SARS-CoV-2 Spike Protein and Its Receptor Binding Domain Stimulate Release of Different Pro-Inflammatory Mediators via Activation of Distinct Receptors on Human Microglia Cells. Mol Neurobiol 2023; 60:6704-6714. [PMID: 37477768 DOI: 10.1007/s12035-023-03493-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/10/2023] [Indexed: 07/22/2023]
Abstract
SARS-CoV-2 infects cells via its spike (S) protein binding to its surface receptor angiotensin converting enzyme 2 (ACE2) on target cells and results in acute symptoms involving especially the lungs known as COVID-19. However, increasing evidence indicates that SARS-CoV-2 infection produces neuroinflammation associated with neurological, neuropsychiatric, and cognitive symptoms persists well past the resolution of the infection, known as post-COVID-19 sequalae or long-COVID. The neuroimmune mechanism(s) involved in long-COVID have not been adequately characterized. In this study, we show that recombinant SARS-CoV-2 full-length S protein stimulates release of pro-inflammatory IL-1b, CXCL8, IL-6, and MMP-9 from cultured human microglia via TLR4 receptor activation. Instead, recombinant receptor-binding domain (RBD) stimulates release of TNF-α, IL-18, and S100B via ACE2 signaling. These results provide evidence that SARS-CoV-2 spike protein contributes to neuroinflammation through different mechanisms that may be involved in CNS pathologies associated with long-COVID.
Collapse
Affiliation(s)
- Irene Tsilioni
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, 136 Harrison Avenue, Suite 304, Boston, MA, 02111, USA.
| | - Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, 136 Harrison Avenue, Suite 304, Boston, MA, 02111, USA
- Institute of Neuro-Immune Medicine, Nova Southeastern University, Clearwater, FL, 33759, USA
| |
Collapse
|
198
|
Brunialti MKC, Leite GGF, Eburneo GS, de Araujo OR, Peçanha-Pietrobom PM, Ferreira PRA, Bellei NCJ, Arakaki JSO, Medina-Pestana J, Requião-Moura L, Salomao R. Patterns of Circulating Cytokines and Vascular Markers' Response in the Presence of COVID-19 in Kidney Transplant Recipients Compared with Non-Transplanted Patients. Viruses 2023; 15:2166. [PMID: 38005844 PMCID: PMC10675241 DOI: 10.3390/v15112166] [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: 08/13/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
COVID-19's severity has been associated with a possible imbalance in the cross-regulation of cytokines and vascular mediators. Since the beginning of the pandemic, kidney transplant recipients (KTRs) have been identified as patients of high vulnerability to more severe diseases. Thus, aiming to describe the patterns of cytokines and vascular mediators and to trace patients' differences according to their KTR status, this prospective study enrolled 67 COVID-19 patients (20 KTRs) and 29 non-COVID-19 controls before vaccination. A panel comprising 17 circulating cytokines and vascular mediators was run on samples collected at different time points. The cytokine and mediator patterns were investigated via principal component analysis (PCA) and correlation-based network (CBN). In both groups, compared to their respective controls, COVID-19 was associated with higher levels of cytokines and vascular mediators. Differentiating between the KTRs and non-KTRs, the number of correlations was much higher in the non-KTRs (44 vs. 14), and the node analysis showed the highest interactions of NGAL and sVCAM-1 in the non-KTRs and KTRs (9 vs. 4), respectively. In the PCA, while the non-KTRs with COVID-19 were differentiated from their controls in their IL-10, IFN-α, and TNF-α, this pattern was marked in the NGAL, sVCAM-1, and IL-8 of the KTRs.
Collapse
Affiliation(s)
- Milena Karina Coló Brunialti
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Giuseppe G. F. Leite
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Gabriela Strafolino Eburneo
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Orlei Ribeiro de Araujo
- Intensive Care Unit, GRAACC, Pediatric Institute of Oncology, Universidade Federal de São Paulo, São Paulo 04039-001, Brazil;
| | - Paula M. Peçanha-Pietrobom
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Paulo Roberto Abrão Ferreira
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Nancy C. Junqueira Bellei
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Jaquelina Sonoe Ota Arakaki
- Division of Respiratory Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04020-050, Brazil;
| | - José Medina-Pestana
- Division of Nephrology, Universidade Federal de São Paulo, São Paulo 04038-031, Brazil;
- Hospital do Rim, Fundação Oswalado Ramos, São Paulo 04038-002, Brazil
| | - Lúcio Requião-Moura
- Division of Nephrology, Universidade Federal de São Paulo, São Paulo 04038-031, Brazil;
- Hospital do Rim, Fundação Oswalado Ramos, São Paulo 04038-002, Brazil
| | - Reinaldo Salomao
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
- Hospital São Paulo, São Paulo 04024-002, Brazil
| |
Collapse
|
199
|
Tang M, Dong W, Yuan S, Chen J, Lin J, Wu J, Zhang J, Yin Y, Zhang L. Comparison of respiratory pathogens in children with community-acquired pneumonia before and during the COVID-19 pandemic. BMC Pediatr 2023; 23:535. [PMID: 37891511 PMCID: PMC10605329 DOI: 10.1186/s12887-023-04246-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 08/11/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Multifaceted non-pharmaceutical interventions during the COVID-19 pandemic have not only reduced the transmission of SARS-CoV2, but have had an effect on the prevalence of other pathogens. This retrospective study aimed to compare and analyze the changes of respiratory pathogens in hospitalized children with community-acquired pneumonia. METHODS From January 2019 to December 2020, children with community-acquired pneumonia were selected from the Department of Respiratory Medicine, Shanghai Children's Medical Center. On the first day of hospitalization, sputum, throat swabs, venous blood samples from them were collected for detection of pathogens. RESULTS A total of 2596 children with community-acquired pneumonia were enrolled, including 1871 patients in 2019 and 725 in 2020. The detection rate in 2020 was lower than in 2019, whether single or multiple pathogens. Compared with 2019, the detection rate of virus, especially parainfluenza virus, influenza virus and respiratory syncytial virus, all decreased in 2020. On the contrary, the prevalence of human rhinovirus was much higher than that in 2019. In addition, the positivity rate for bacteria did not change much over the two years, which seemed to be less affected by COVID-19. And Mycoplasma pneumoniae which broke out in 2019 has been in low prevalence since March 2020 even following the reopening of school. CONCLUSIONS Strict public health interventions for COVID-19 in China have effectively suppressed the spread of not only SARS-CoV2 but parainfluenza virus, influenza virus and Mycoplasma pneumonia as well. However, it had a much more limited effect on bacteria and rhinovirus. Therefore, more epidemiological surveillance of respiratory pathogens will help improve early preventive measures.
Collapse
Affiliation(s)
- Mingyu Tang
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Rd, Shanghai, 200127, China
| | - Wenfang Dong
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Rd, Shanghai, 200127, China
| | - Shuhua Yuan
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Rd, Shanghai, 200127, China
| | - Jiande Chen
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Rd, Shanghai, 200127, China
| | - Jie Lin
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Rd, Shanghai, 200127, China
| | - Jinhong Wu
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Rd, Shanghai, 200127, China
| | - Jing Zhang
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Rd, Shanghai, 200127, China
| | - Yong Yin
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Rd, Shanghai, 200127, China.
| | - Lei Zhang
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Rd, Shanghai, 200127, China.
| |
Collapse
|
200
|
Sánchez A, García-Pardo G, Gómez-Bertomeu F, López-Dupla M, Foguet-Romero E, Buzón MJ, Almirante B, Olona M, Fernández-Veledo S, Vidal F, Chafino S, Rull A, Peraire J, for the COVIDOMICS Study Group. Mitochondrial dysfunction, lipids metabolism, and amino acid biosynthesis are key pathways for COVID-19 recovery. iScience 2023; 26:107948. [PMID: 37810253 PMCID: PMC10551651 DOI: 10.1016/j.isci.2023.107948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/29/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023] Open
Abstract
The metabolic alterations caused by SARS-CoV-2 infection reflect disease progression. To analyze molecules involved in these metabolic changes, a multiomics study was performed using plasma from 103 patients with different degrees of COVID-19 severity during the evolution of the infection. With the increased severity of COVID-19, changes in circulating proteomic, metabolomic, and lipidomic profiles increased. Notably, the group of severe and critical patients with high HRG and ChoE (20:3) and low alpha-ketoglutaric acid levels had a high chance of unfavorable disease evolution (AUC = 0.925). Consequently, patients with the worst prognosis presented alterations in the TCA cycle (mitochondrial dysfunction), lipid metabolism, amino acid biosynthesis, and coagulation. Our findings increase knowledge regarding how SARS-CoV-2 infection affects different metabolic pathways and help in understanding the future consequences of COVID-19 to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Alba Sánchez
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Graciano García-Pardo
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Fréderic Gómez-Bertomeu
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Miguel López-Dupla
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Elisabet Foguet-Romero
- Eurecat, Centre Tecnologic de Catalunya, Centre for Omic Sciences (Joint Unit Eurecat - Universitat Rovira i Virgili), Unique Scientific and Technical Infrastructure (ICTS), Reus, Spain
| | - Maria José Buzón
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Infectious Diseases Department, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, (VHIR) Task Force COVID-19, Barcelona, Spain
| | - Benito Almirante
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Montserrat Olona
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Sonia Fernández-Veledo
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Francesc Vidal
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Silvia Chafino
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Anna Rull
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Joaquim Peraire
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - for the COVIDOMICS Study Group
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Eurecat, Centre Tecnologic de Catalunya, Centre for Omic Sciences (Joint Unit Eurecat - Universitat Rovira i Virgili), Unique Scientific and Technical Infrastructure (ICTS), Reus, Spain
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Infectious Diseases Department, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, (VHIR) Task Force COVID-19, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|