1
|
Donkers A, Seel W, Klümpen L, Simon MC. The Multiple Challenges of Nutritional Microbiome Research During COVID-19-A Perspective and Results of a Single-Case Study. Nutrients 2024; 16:3693. [PMID: 39519526 PMCID: PMC11547757 DOI: 10.3390/nu16213693] [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: 10/04/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has affected multiple aspects of people's lives, which may also influence the results of studies conducted during this period across diverse research domains. This particularly includes the field of nutritional science, investigating the gut microbiota as a potential mediator in the association between dietary intake and health-related outcomes. This article identifies the challenges currently facing this area of research, points out potential solutions, and highlights the necessity to consider a range of issues when interpreting trials conducted during this period. Some of these issues have arisen specifically because of the measures implemented to interrupt the spread of small acute respiratory syndrome coronavirus 2 (SARS-CoV-2), while others remain relevant beyond the pandemic.
Collapse
Affiliation(s)
| | | | | | - Marie-Christine Simon
- Nutrition and Microbiota, Institute of Nutrition and Food Science, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
2
|
He X, Zhang X, Zhong W. Emerging small-molecule antiviral agents in long COVID prevention. Front Pharmacol 2024; 15:1457672. [PMID: 39444602 PMCID: PMC11496125 DOI: 10.3389/fphar.2024.1457672] [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: 07/01/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Long COVID, or Post-Acute Sequelae of COVID-19 (PASC), was characterized by persistent symptoms such as fatigue, shortness of breath, and cognitive impairments. These symptoms, emerging one to 2 months post-infection and persisting for several months, cannot be attributed to other diagnoses. The pathophysiology of long COVID remained elusive; however, emerging studies suggested multiple potential mechanisms, including the reactivation of Epstein-Barr virus, persistent SARS-CoV-2 reservoirs, neuroinflammation, and vascular damage, which may contribute to its development. Long COVID affected multiple organ systems, including respiratory, circulatory, and nervous systems, leading to a range of functional impairments. Additionally, it showed a profound impact on mental health, manifesting as anxiety and depression, which significantly degraded the quality of life. The absence of definitive treatments underscored the importance of prevention. Recent evidence indicated that early antiviral intervention-particularly with small-molecule drugs such as Metformin, Ensitrelvir, Molnupiravir, and Nirmatrelvir-may effectively reduce the incidence of long COVID. This underscored the promising role of small-molecule compounds in mitigating long-term COVID-19 consequences, offering a novel preventive strategy against long COVID and its extensive impacts on patients.
Collapse
Affiliation(s)
- Xiaomeng He
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiang Zhang
- Department of Blood Transfusion Medicine, The 940th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Lanzhou, China
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
3
|
Monaco V, Iacobucci I, Canè L, Cipollone I, Ferrucci V, de Antonellis P, Quaranta M, Pascarella S, Zollo M, Monti M. SARS-CoV-2 uses Spike glycoprotein to control the host's anaerobic metabolism by inhibiting LDHB. Int J Biol Macromol 2024; 278:134638. [PMID: 39147351 DOI: 10.1016/j.ijbiomac.2024.134638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024]
Abstract
The SARS-CoV-2 pandemic, responsible for approximately 7 million deaths worldwide, highlights the urgent need to understand the molecular mechanisms of the virus in order to prevent future outbreaks. The Spike glycoprotein of SARS-CoV-2, which is critical for viral entry through its interaction with ACE2 and other host cell receptors, has been a focus of this study. The present research goes beyond receptor recognition to explore Spike's influence on cellular metabolism. AP-MS interactome analysis revealed an interaction between the Spike S1 domain and lactate dehydrogenase B (LDHB), which was further confirmed by co-immunoprecipitation and immunofluorescence, indicating colocalisation in cells expressing the S1 domain. The study showed that Spike inhibits the catalytic activity of LDHB, leading to increased lactate levels in HEK-293T cells overexpressing the S1 subunit. In the hypothesised mechanism, Spike deprives LDHB of NAD+, facilitating a metabolic switch from aerobic to anaerobic energy production during infection. The Spike-NAD+ interacting region was characterised and mainly involves the W436 within the RDB domain. This novel hypothesis suggests that the Spike protein may play a broader role in altering host cell metabolism, thereby contributing to the pathophysiology of viral infection.
Collapse
Affiliation(s)
- Vittoria Monaco
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
| | - Ilaria Iacobucci
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
| | - Luisa Canè
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; Department of Translational Medical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Irene Cipollone
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
| | - Veronica Ferrucci
- CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), "Federico II" University of Naples, Naples 80131, Italy
| | - Pasqualino de Antonellis
- CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), "Federico II" University of Naples, Naples 80131, Italy
| | - Miriana Quaranta
- Sapienza Università di Roma, Department of Biochemical Sciences "A. Rossi Fanelli", Rome 00185, Italy
| | - Stefano Pascarella
- Sapienza Università di Roma, Department of Biochemical Sciences "A. Rossi Fanelli", Rome 00185, Italy
| | - Massimo Zollo
- CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), "Federico II" University of Naples, Naples 80131, Italy
| | - Maria Monti
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy.
| |
Collapse
|
4
|
Oropeza-Valdez JJ, Padron-Manrique C, Vázquez-Jiménez A, Soberon X, Resendis-Antonio O. Exploring metabolic anomalies in COVID-19 and post-COVID-19: a machine learning approach with explainable artificial intelligence. Front Mol Biosci 2024; 11:1429281. [PMID: 39314212 PMCID: PMC11417410 DOI: 10.3389/fmolb.2024.1429281] [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: 05/07/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has led to significant challenges worldwide, including diverse clinical outcomes and prolonged post-recovery symptoms known as Long COVID or Post-COVID-19 syndrome. Emerging evidence suggests a crucial role of metabolic reprogramming in the infection's long-term consequences. This study employs a novel approach utilizing machine learning (ML) and explainable artificial intelligence (XAI) to analyze metabolic alterations in COVID-19 and Post-COVID-19 patients. Samples were taken from a cohort of 142 COVID-19, 48 Post-COVID-19, and 38 control patients, comprising 111 identified metabolites. Traditional analysis methods, like PCA and PLS-DA, were compared with ML techniques, particularly eXtreme Gradient Boosting (XGBoost) enhanced by SHAP (SHapley Additive exPlanations) values for explainability. XGBoost, combined with SHAP, outperformed traditional methods, demonstrating superior predictive performance and providing new insights into the metabolic basis of the disease's progression and aftermath. The analysis revealed metabolomic subgroups within the COVID-19 and Post-COVID-19 conditions, suggesting heterogeneous metabolic responses to the infection and its long-term impacts. Key metabolic signatures in Post-COVID-19 include taurine, glutamine, alpha-Ketoglutaric acid, and LysoPC a C16:0. This study highlights the potential of integrating ML and XAI for a fine-grained description in metabolomics research, offering a more detailed understanding of metabolic anomalies in COVID-19 and Post-COVID-19 conditions.
Collapse
Affiliation(s)
- Juan José Oropeza-Valdez
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Cristian Padron-Manrique
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Aarón Vázquez-Jiménez
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Xavier Soberon
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Colonia Chamilpa, Cuernavaca, México
| | - Osbaldo Resendis-Antonio
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Coordinación de la Investigación Científica – Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
5
|
Ristori MV, Guarrasi V, Soda P, Petrosillo N, Gurrieri F, Longo UG, Ciccozzi M, Riva E, Angeletti S. Emerging Microorganisms and Infectious Diseases: One Health Approach for Health Shared Vision. Genes (Basel) 2024; 15:908. [PMID: 39062687 PMCID: PMC11275270 DOI: 10.3390/genes15070908] [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: 06/14/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Emerging infectious diseases (EIDs) are newly emerging and reemerging infectious diseases. The National Institute of Allergy and Infectious Diseases identifies the following as emerging infectious diseases: SARS, MERS, COVID-19, influenza, fungal diseases, plague, schistosomiasis, smallpox, tick-borne diseases, and West Nile fever. The factors that should be taken into consideration are the genetic adaptation of microbial agents and the characteristics of the human host or environment. The new approach to identifying new possible pathogens will have to go through the One Health approach and omics integration data, which are capable of identifying high-priority microorganisms in a short period of time. New bioinformatics technologies enable global integration and sharing of surveillance data for rapid public health decision-making to detect and prevent epidemics and pandemics, ensuring timely response and effective prevention measures. Machine learning tools are being more frequently utilized in the realm of infectious diseases to predict sepsis in patients, diagnose infectious diseases early, and forecast the effectiveness of treatment or the appropriate choice of antibiotic regimen based on clinical data. We will discuss emerging microorganisms, omics techniques applied to infectious diseases, new computational solutions to evaluate biomarkers, and innovative tools that are useful for integrating omics data and electronic medical records data for the clinical management of emerging infectious diseases.
Collapse
Affiliation(s)
- Maria Vittoria Ristori
- Operative Research Unit of Laboratory, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (M.V.R.); (M.C.); (E.R.)
| | - Valerio Guarrasi
- Unit of Computer Systems and Bioinformatics, Department of Engineering, University Campus Bio-Medico of Rome, Via Alvaro del Portillo, 21, 00128 Rome, Italy; (V.G.); (P.S.)
| | - Paolo Soda
- Unit of Computer Systems and Bioinformatics, Department of Engineering, University Campus Bio-Medico of Rome, Via Alvaro del Portillo, 21, 00128 Rome, Italy; (V.G.); (P.S.)
- Department of Diagnostic and Intervention, Radiation Physics, Biomedical Engineering, Umeå University, 901 87 Umeå, Sweden
| | - Nicola Petrosillo
- Infection Prevention Control/Infectious Disease Service, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy;
| | - Fiorella Gurrieri
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy;
- Research Unit of Medical Genetics, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| | - Umile Giuseppe Longo
- Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy;
- Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| | - Massimo Ciccozzi
- Operative Research Unit of Laboratory, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (M.V.R.); (M.C.); (E.R.)
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| | - Elisabetta Riva
- Operative Research Unit of Laboratory, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (M.V.R.); (M.C.); (E.R.)
- Unit of Virology, University Campus Bio-Medico of Rome, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| | - Silvia Angeletti
- Operative Research Unit of Laboratory, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (M.V.R.); (M.C.); (E.R.)
- Research Unit of Clinical Laboratory Science, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| |
Collapse
|
6
|
Zhang Y, Chen S, Tian Y, Fu X. Host factors of SARS-CoV-2 in infection, pathogenesis, and long-term effects. Front Cell Infect Microbiol 2024; 14:1407261. [PMID: 38846354 PMCID: PMC11155306 DOI: 10.3389/fcimb.2024.1407261] [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: 03/26/2024] [Accepted: 05/08/2024] [Indexed: 06/09/2024] Open
Abstract
SARS-CoV-2 is the causative virus of the devastating COVID-19 pandemic that results in an unparalleled global health and economic crisis. Despite unprecedented scientific efforts and therapeutic interventions, the fight against COVID-19 continues as the rapid emergence of different SARS-CoV-2 variants of concern and the increasing challenge of long COVID-19, raising a vast demand to understand the pathomechanisms of COVID-19 and its long-term sequelae and develop therapeutic strategies beyond the virus per se. Notably, in addition to the virus itself, the replication cycle of SARS-CoV-2 and clinical severity of COVID-19 is also governed by host factors. In this review, we therefore comprehensively overview the replication cycle and pathogenesis of SARS-CoV-2 from the perspective of host factors and host-virus interactions. We sequentially outline the pathological implications of molecular interactions between host factors and SARS-CoV-2 in multi-organ and multi-system long COVID-19, and summarize current therapeutic strategies and agents targeting host factors for treating these diseases. This knowledge would be key for the identification of new pathophysiological aspects and mechanisms, and the development of actionable therapeutic targets and strategies for tackling COVID-19 and its sequelae.
Collapse
Affiliation(s)
| | | | - Yan Tian
- Department of Endocrinology and Metabolism, Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital and Cancer Center, Sichuan University and Collaborative Innovation Center of Biotherapy, Sichuan, Chengdu, China
| | - Xianghui Fu
- Department of Endocrinology and Metabolism, Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital and Cancer Center, Sichuan University and Collaborative Innovation Center of Biotherapy, Sichuan, Chengdu, China
| |
Collapse
|
7
|
O'Carroll SM, Henkel FDR, O'Neill LAJ. Metabolic regulation of type I interferon production. Immunol Rev 2024; 323:276-287. [PMID: 38465724 DOI: 10.1111/imr.13318] [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] [Indexed: 03/12/2024]
Abstract
Over the past decade, there has been a surge in discoveries of how metabolic pathways regulate immune cell function in health and disease, establishing the field of immunometabolism. Specifically, pathways such as glycolysis, the tricarboxylic acid (TCA) cycle, and those involving lipid metabolism have been implicated in regulating immune cell function. Viral infections cause immunometabolic changes which lead to antiviral immunity, but little is known about how metabolic changes regulate interferon responses. Interferons are critical cytokines in host defense, rapidly induced upon pathogen recognition, but are also involved in autoimmune diseases. This review summarizes how metabolic change impacts interferon production. We describe how glycolysis, lipid metabolism (specifically involving eicosanoids and cholesterol), and the TCA cycle-linked intermediates itaconate and fumarate impact type I interferons. Targeting these metabolic changes presents new therapeutic possibilities to modulate type I interferons during host defense or autoimmune disorders.
Collapse
Affiliation(s)
- Shane M O'Carroll
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Fiona D R Henkel
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
8
|
Morales-Rodriguez DP, González-Cantú A, Garza-Silva A, Rivera-Cavazos A, Fernández-Chau IF, Cepeda-Medina AB, Sanz-Sánchez MA, Del Rio-Parra GF, Torres-Fuentes MA, Rodriguez-Puente MA, Romero-Ibarguengoitia ME. Effect of the SARS-CoV-2 pandemic on metabolic control in patients with type 2 diabetes: a 5-year cohort follow-up managed by a dynamic multidisciplinary team in Northeastern Mexico. Diabetol Metab Syndr 2024; 16:94. [PMID: 38664823 PMCID: PMC11044561 DOI: 10.1186/s13098-024-01318-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The SARS-CoV-2 pandemic brought a radical shift in the healthcare system and suboptimal care for vulnerable patients, such as those with Type 2 Diabetes Mellitus (T2D). Therefore, we compared metabolic control and macro/microvascular complications of patients with T2D before and throughout the three-year SARS-CoV-2 pandemic. RESEARCH DESIGN AND METHODS A retrospective observational cohort of subjects with T2D studied from 2018 to 2022 in Northern Mexico was treated by a dynamic multidisciplinary team. Levels of Glycated hemoglobin (HbA1c), fasting serum glucose (FG), LDL-Cholesterol (LDL-C), blood pressure (BP), albuminuria, triglycerides, Body Mass Index (BMI), and FIB-4 score, micro and macrovascular complications were evaluated. RESULTS A total of 999 patients were studied, 51.7% males with a mean (SD) age of 60.1 (12.7) years. Adequate glycemic control based on HbA1c increased by 15.2% and 42.3% in FSG (p < 0.001) between the beginning 2018 and the end of 2022. LDL-C control decreased by 5.1% between 2018 and 2022 (p < 0.001). Systolic BP control decreased by 2.6% (p < 0.001), whereas diastolic BP control increased by 1.8% (p = 0.01) between 2018 and 2022. Albuminuria control increased by 8.5% (p = 0.002). When comparing the Area Under the Curve (AUC) of metabolic parameters between patients who developed SARS-CoV-2 vs. those who did not, AUC was statistically higher in those who developed SARS-CoV-2 (p < 0.05). Diabetic neuropathy was the most prevalent microvascular complication (n = 35; 3.6%); ischemic heart disease was the most frequent macrovascular complication (n = 11;1.1%). CONCLUSIONS A multidisciplinary dynamic team that adapts to the pandemic SARS-CoV-2 maintains and increases metabolic control in subjects with type 2 diabetes in Mexico. This represents a low percentage of chronic complications. The AUC of metabolic parameters of subjects with SARS-CoV-2 infection is higher, reflecting more variability in metabolic control.
Collapse
Affiliation(s)
- Devany Paola Morales-Rodriguez
- Reseach Deparment, Hospital Clinica Nova de Monterrey, San Nicolas de los Garza, Nuevo Leon, Mexico
- Medical School, Vicerrectoria de Ciencias de la Salud, Universidad de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Arnulfo González-Cantú
- Reseach Deparment, Hospital Clinica Nova de Monterrey, San Nicolas de los Garza, Nuevo Leon, Mexico
- Medical School, Vicerrectoria de Ciencias de la Salud, Universidad de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Arnulfo Garza-Silva
- Reseach Deparment, Hospital Clinica Nova de Monterrey, San Nicolas de los Garza, Nuevo Leon, Mexico
| | - Andrea Rivera-Cavazos
- Reseach Deparment, Hospital Clinica Nova de Monterrey, San Nicolas de los Garza, Nuevo Leon, Mexico
| | - Iván Francisco Fernández-Chau
- Reseach Deparment, Hospital Clinica Nova de Monterrey, San Nicolas de los Garza, Nuevo Leon, Mexico
- Medical School, Vicerrectoria de Ciencias de la Salud, Universidad de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Andrea Belinda Cepeda-Medina
- Reseach Deparment, Hospital Clinica Nova de Monterrey, San Nicolas de los Garza, Nuevo Leon, Mexico
- Medical School, Vicerrectoria de Ciencias de la Salud, Universidad de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Miguel Angel Sanz-Sánchez
- Reseach Deparment, Hospital Clinica Nova de Monterrey, San Nicolas de los Garza, Nuevo Leon, Mexico
- Medical School, Vicerrectoria de Ciencias de la Salud, Universidad de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | | | - María Angelina Torres-Fuentes
- Medical School, Vicerrectoria de Ciencias de la Salud, Universidad de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Miguel Assael Rodriguez-Puente
- Medical School, Vicerrectoria de Ciencias de la Salud, Universidad de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Maria Elena Romero-Ibarguengoitia
- Reseach Deparment, Hospital Clinica Nova de Monterrey, San Nicolas de los Garza, Nuevo Leon, Mexico.
- Medical School, Vicerrectoria de Ciencias de la Salud, Universidad de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico.
| |
Collapse
|
9
|
Nahalka J. 1-L Transcription of SARS-CoV-2 Spike Protein S1 Subunit. Int J Mol Sci 2024; 25:4440. [PMID: 38674024 PMCID: PMC11049929 DOI: 10.3390/ijms25084440] [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: 02/29/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
The COVID-19 pandemic prompted rapid research on SARS-CoV-2 pathogenicity. Consequently, new data can be used to advance the molecular understanding of SARS-CoV-2 infection. The present bioinformatics study discusses the "spikeopathy" at the molecular level and focuses on the possible post-transcriptional regulation of the SARS-CoV-2 spike protein S1 subunit in the host cell/tissue. A theoretical protein-RNA recognition code was used to check the compatibility of the SARS-CoV-2 spike protein S1 subunit with mRNAs in the human transcriptome (1-L transcription). The principle for this method is elucidated on the defined RNA binding protein GEMIN5 (gem nuclear organelle-associated protein 5) and RNU2-1 (U2 spliceosomal RNA). Using the method described here, it was shown that 45% of the genes/proteins identified by 1-L transcription of the SARS-CoV-2 spike protein S1 subunit are directly linked to COVID-19, 39% are indirectly linked to COVID-19, and 16% cannot currently be associated with COVID-19. The identified genes/proteins are associated with stroke, diabetes, and cardiac injury.
Collapse
Affiliation(s)
- Jozef Nahalka
- Institute of Chemistry, Centre for Glycomics, Slovak Academy of Sciences, Dubravska Cesta 9, SK-84538 Bratislava, Slovakia;
- Institute of Chemistry, Centre of Excellence for White-Green Biotechnology, Slovak Academy of Sciences, Trieda Andreja Hlinku 2, SK-94976 Nitra, Slovakia
| |
Collapse
|
10
|
Chatterjee S, Prashanth P, Rawat V, Ghosh Roy S. Regulation of lipid and serine metabolism by the oncogene c-Myc. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 389:236-256. [PMID: 39396848 DOI: 10.1016/bs.ircmb.2024.03.005] [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: 10/15/2024]
Abstract
Tumor formation is supported by metabolic reprogramming, characterized by increase nutrient uptake, glycolysis and glutaminolysis. The c-Myc proto-oncogene is a transcription factor, upregulated in most cancers and several reports showed the role of c-Myc in other metabolic pathways such as glucose, amino acid, and nucleotide metabolism. In this short report, we tried to summarize the existing takeaway points from studies conducted in different cancer types with respect to c-Myc and lipid and serine metabolism. Here, we report that c-Myc can activate both lipid and serine metabolism against the backdrop of tumor formation, and different therapies like aspirin and lomitapide target the links between c-Myc and metabolism to slow down tumor progression and invasion. We also report diverse upstream regulators that influence c-Myc in different cancers, and interestingly components of the lipid metabolism (like lipid phosphate phosphatase and leptin) and serine metabolism can also act upstream of c-Myc in certain occasions. Finally, we also summarize the existing knowledge on the involvement of epigenetic pathways and non-coding RNAs in regulating lipid and serine metabolism and c-Myc in tumor cells. Identification of non-coding factors and epigenetic mechanisms present a promising avenue of study that could empower researchers with novel anticancer treatment targeting c-Myc and lipid and serine metabolism pathways!
Collapse
Affiliation(s)
- Subhajit Chatterjee
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Prarthana Prashanth
- Department of Physiology and Biophysics, University of Illinois College of Medicine, University of Illinois Cancer Center, Chicago, IL, United States
| | - Vipin Rawat
- Department of Physiology and Biophysics, University of Illinois College of Medicine, University of Illinois Cancer Center, Chicago, IL, United States.
| | - Sounak Ghosh Roy
- Henry M Jackson Foundation for the Advancement of Military Medicine (In Support of Agile Vaccines & Therapeutics, Directorate for Defense Infectious Diseases Research, Naval Medical Research Command, Silver Spring, MD, United States.
| |
Collapse
|
11
|
Chen S, Fu Z, Chen K, Zheng X, Fu Z. Decoding HiPSC-CM's Response to SARS-CoV-2: mapping the molecular landscape of cardiac injury. BMC Genomics 2024; 25:271. [PMID: 38475718 DOI: 10.1186/s12864-024-10194-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 03/06/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Acute cardiac injury caused by coronavirus disease 2019 (COVID-19) increases mortality. Acute cardiac injury caused by COVID-19 requires understanding how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directly infects cardiomyocytes. This study provides a solid foundation for related studies by using a model of SARS-CoV-2 infection in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) at the transcriptome level, highlighting the relevance of this study to related studies. SARS-CoV-2 infection in hiPSC-CMs has previously been studied by bioinformatics without presenting the full molecular biological process. We present a unique bioinformatics view of the complete molecular biological process of SARS-CoV-2 infection in hiPSC-CMs. METHODS To validate the RNA-seq datasets, we used GSE184715 and GSE150392 for the analytical studies, GSE193722 for validation at the cellular level, and GSE169241 for validation in heart tissue samples. GeneCards and MsigDB databases were used to find genes associated with the phenotype. In addition to differential expression analysis and principal component analysis (PCA), we also performed protein-protein interaction (PPI) analysis, functional enrichment analysis, hub gene analysis, upstream transcription factor prediction, and drug prediction. RESULTS Differentially expressed genes (DEGs) were classified into four categories: cardiomyocyte cytoskeletal protein inhibition, proto-oncogene activation and inflammation, mitochondrial dysfunction, and intracellular cytoplasmic physiological function. Each of the hub genes showed good diagnostic prediction, which was well validated in other datasets. Inhibited biological functions included cardiomyocyte cytoskeletal proteins, adenosine triphosphate (ATP) synthesis and electron transport chain (ETC), glucose metabolism, amino acid metabolism, fatty acid metabolism, pyruvate metabolism, citric acid cycle, nucleic acid metabolism, replication, transcription, translation, ubiquitination, autophagy, and cellular transport. Proto-oncogenes, inflammation, nuclear factor-kappaB (NF-κB) pathways, and interferon signaling were activated, as well as inflammatory factors. Viral infection activates multiple pathways, including the interferon pathway, proto-oncogenes and mitochondrial oxidative stress, while inhibiting cardiomyocyte backbone proteins and energy metabolism. Infection limits intracellular synthesis and metabolism, as well as the raw materials for mitochondrial energy synthesis. Mitochondrial dysfunction and energy abnormalities are ultimately caused by proto-oncogene activation and SARS-CoV-2 infection. Activation of the interferon pathway, proto-oncogene up-regulation, and mitochondrial oxidative stress cause the inflammatory response and lead to diminished cardiomyocyte contraction. Replication, transcription, translation, ubiquitination, autophagy, and cellular transport are among the functions that decline physiologically. CONCLUSION SARS-CoV-2 infection in hiPSC-CMs is fundamentally mediated via mitochondrial dysfunction. Therapeutic interventions targeting mitochondrial dysfunction may alleviate the cardiovascular complications associated with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Sicheng Chen
- Department of Cardiology, Shantou Central Hospital, Shantou, 515031, China
| | - Zhenquan Fu
- School of Information Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Kaitong Chen
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Xinyao Zheng
- Shantou University Medical College, Shantou, 515041, China
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zhenyang Fu
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China.
| |
Collapse
|
12
|
Kumar S, Granados J, Aceves M, Peralta J, Leandro AC, Thomas J, Williams-Blangero S, Curran JE, Blangero J. Pre-Infection Innate Immunity Attenuates SARS-CoV-2 Infection and Viral Load in iPSC-Derived Alveolar Epithelial Type 2 Cells. Cells 2024; 13:369. [PMID: 38474333 PMCID: PMC10931100 DOI: 10.3390/cells13050369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/05/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
A large portion of the heterogeneity in coronavirus disease 2019 (COVID-19) susceptibility and severity of illness (SOI) remains poorly understood. Recent evidence suggests that SARS-CoV-2 infection-associated damage to alveolar epithelial type 2 cells (AT2s) in the distal lung may directly contribute to disease severity and poor prognosis in COVID-19 patients. Our in vitro modeling of SARS-CoV-2 infection in induced pluripotent stem cell (iPSC)-derived AT2s from 10 different individuals showed interindividual variability in infection susceptibility and the postinfection cellular viral load. To understand the underlying mechanism of the AT2's capacity to regulate SARS-CoV-2 infection and cellular viral load, a genome-wide differential gene expression analysis between the mock and SARS-CoV-2 infection-challenged AT2s was performed. The 1393 genes, which were significantly (one-way ANOVA FDR-corrected p ≤ 0.05; FC abs ≥ 2.0) differentially expressed (DE), suggest significant upregulation of viral infection-related cellular innate immune response pathways (p-value ≤ 0.05; activation z-score ≥ 3.5), and significant downregulation of the cholesterol- and xenobiotic-related metabolic pathways (p-value ≤ 0.05; activation z-score ≤ -3.5). Whilst the effect of post-SARS-CoV-2 infection response on the infection susceptibility and postinfection viral load in AT2s is not clear, interestingly, pre-infection (mock-challenged) expression of 238 DE genes showed a high correlation with the postinfection SARS-CoV-2 viral load (FDR-corrected p-value ≤ 0.05 and r2-absolute ≥ 0.57). The 85 genes whose expression was negatively correlated with the viral load showed significant enrichment in viral recognition and cytokine-mediated innate immune GO biological processes (p-value range: 4.65 × 10-10 to 2.24 × 10-6). The 153 genes whose expression was positively correlated with the viral load showed significant enrichment in cholesterol homeostasis, extracellular matrix, and MAPK/ERK pathway-related GO biological processes (p-value range: 5.06 × 10-5 to 6.53 × 10-4). Overall, our results strongly suggest that AT2s' pre-infection innate immunity and metabolic state affect their susceptibility to SARS-CoV-2 infection and viral load.
Collapse
Affiliation(s)
- Satish Kumar
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (J.G.); (M.A.); (J.T.)
| | - Jose Granados
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (J.G.); (M.A.); (J.T.)
| | - Miriam Aceves
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (J.G.); (M.A.); (J.T.)
| | - Juan Peralta
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.P.); (A.C.L.); (S.W.-B.); (J.E.C.); (J.B.)
| | - Ana C. Leandro
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.P.); (A.C.L.); (S.W.-B.); (J.E.C.); (J.B.)
| | - John Thomas
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (J.G.); (M.A.); (J.T.)
| | - Sarah Williams-Blangero
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.P.); (A.C.L.); (S.W.-B.); (J.E.C.); (J.B.)
| | - Joanne E. Curran
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.P.); (A.C.L.); (S.W.-B.); (J.E.C.); (J.B.)
| | - John Blangero
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.P.); (A.C.L.); (S.W.-B.); (J.E.C.); (J.B.)
| |
Collapse
|
13
|
Wu A, Shi K, Wang J, Zhang R, Wang Y. Targeting SARS-CoV-2 entry processes: The promising potential and future of host-targeted small-molecule inhibitors. Eur J Med Chem 2024; 263:115923. [PMID: 37981443 DOI: 10.1016/j.ejmech.2023.115923] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/16/2023] [Accepted: 10/28/2023] [Indexed: 11/21/2023]
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has had a huge impact on global health. To respond to rapidly mutating viruses and to prepare for the next pandemic, there is an urgent need to develop small molecule therapies that target critical stages of the SARS-CoV-2 life cycle. Inhibiting the entry process of the virus can effectively control viral infection and play a role in prevention and treatment. Host factors involved in this process, such as ACE2, TMPRSS2, ADAM17, furin, PIKfyve, TPC2, CTSL, AAK1, V-ATPase, HSPG, and NRP1, have been found to be potentially good targets with stability. Through further exploration of the cell entry process of SARS-CoV-2, small-molecule drugs targeting these host factors have been developed. This review focuses on the structural functions of potential host cell targets during the entry of SARS-CoV-2 into host cells. The research progress, chemical structure, structure-activity relationship, and clinical value of small-molecule inhibitors against COVID-19 are reviewed to provide a reference for the development of small-molecule drugs against COVID-19.
Collapse
Affiliation(s)
- Aijia Wu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, National Clinical Research Center for Geriatrics, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Kunyu Shi
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, National Clinical Research Center for Geriatrics, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Ruofei Zhang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, National Clinical Research Center for Geriatrics, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, National Clinical Research Center for Geriatrics, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
14
|
D’Avila H, Lima CNR, Rampinelli PG, Mateus LCO, de Sousa Silva RV, Correa JR, de Almeida PE. Lipid Metabolism Modulation during SARS-CoV-2 Infection: A Spotlight on Extracellular Vesicles and Therapeutic Prospects. Int J Mol Sci 2024; 25:640. [PMID: 38203811 PMCID: PMC10778989 DOI: 10.3390/ijms25010640] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) have a significant impact on the pathophysiological processes associated with various diseases such as tumors, inflammation, and infection. They exhibit molecular, biochemical, and entry control characteristics similar to viral infections. Viruses, on the other hand, depend on host metabolic machineries to fulfill their biosynthetic requirements. Due to potential advantages such as biocompatibility, biodegradation, and efficient immune activation, EVs have emerged as potential therapeutic targets against the SARS-CoV-2 infection. Studies on COVID-19 patients have shown that they frequently have dysregulated lipid profiles, which are associated with an increased risk of severe repercussions. Lipid droplets (LDs) serve as organelles with significant roles in lipid metabolism and energy homeostasis as well as having a wide range of functions in infections. The down-modulation of lipids, such as sphingolipid ceramide and eicosanoids, or of the transcriptional factors involved in lipogenesis seem to inhibit the viral multiplication, suggesting their involvement in the virus replication and pathogenesis as well as highlighting their potential as targets for drug development. Hence, this review focuses on the role of modulation of lipid metabolism and EVs in the mechanism of immune system evasion during SARS-CoV-2 infection and explores the therapeutic potential of EVs as well as application for delivering therapeutic substances to mitigate viral infections.
Collapse
Affiliation(s)
- Heloisa D’Avila
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | | | - Pollianne Garbero Rampinelli
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - Laiza Camila Oliveira Mateus
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - Renata Vieira de Sousa Silva
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - José Raimundo Correa
- Laboratory of Microscopy and Microanalysis, University of Brasília, Brasília 70910-900, Brazil;
| | - Patrícia Elaine de Almeida
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| |
Collapse
|
15
|
Chen G, Zhao X, Chen X, Liu C. Early decrease in blood lymphocyte count is associated with poor prognosis in COVID-19 patients: a retrospective cohort study. BMC Pulm Med 2023; 23:453. [PMID: 37986163 PMCID: PMC10662697 DOI: 10.1186/s12890-023-02767-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Previous studies have declared that baseline lymphocyte count is associated with COVID-19-related death. However, whether dynamic lymphocyte change over time affects prognosis in COVID-19 patients is unknown. This study aims to investigate the significance of lymphocyte count during the progression of the disease in COVID-19 patients. METHODS The retrospective cohort study recruited COVID-19 patients at the First People's Hospital of Jiangxia District in Wuhan from January 7, 2020, to February 28, 2020. The demographics, medical histories, results of the blood routine test, and patients' outcomes were collected. We utilized a generalized additive mixed model to compare trends in lymphocyte count over time among survivors and non-survivors, with an adjustment for potential confounders. The statistical analysis used R software and EmpowerStats. Significance was determined at a P-value of less than 0.05 (two-sided). RESULTS A total of 532 patients were included in the study. Overall, there were 29/532 in-hospital deaths (5.45%). Lymphocytes declined over time in the non-survivor group and increased in the survivor group in the first 10 days of hospitalization. Within 10 days after admission, lymphocyte count increased in the survivor group and decreased in the non-survivor group. The difference in lymphocyte counts between survivors and non-survivors increased by an average of 0.0732 × 109/L daily. After adjusting for several covariables, the increasing value remained at 0.0731 × 109/L per day. CONCLUSION In the early stage, lymphocyte count can dynamically reflect the pathophysiological changes in COVID-19 patients. An early decrease in lymphocyte count is associated with mortality in COVID-19 patients.
Collapse
Affiliation(s)
- Gong Chen
- Department of Anesthesiology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaofang Zhao
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinglin Chen
- Department of Epidemiology and Biostatistics, Empower U, X&Y Solutions Inc., Boston, MA, USA
| | - Chengyun Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- The First People's Hospital of Jiangxia District, Wuhan City & Union Jiangnan Hospital, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
16
|
Nair S, Nova-Lamperti E, Labarca G, Kulasinghe A, Short KR, Carrión F, Salomon C. Genomic communication via circulating extracellular vesicles and long-term health consequences of COVID-19. J Transl Med 2023; 21:709. [PMID: 37817137 PMCID: PMC10563316 DOI: 10.1186/s12967-023-04552-2] [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: 05/06/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
COVID-19 continues to affect an unprecedented number of people with the emergence of new variants posing a serious challenge to global health. There is an expansion of knowledge in understanding the pathogenesis of Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the impact of the acute disease on multiple organs. In addition, growing evidence reports that the impact of COVID-19 on different organs persists long after the recovery phase of the disease, leading to long-term consequences of COVID-19. These long-term consequences involve pulmonary as well as extra-pulmonary sequelae of the disease. Noteably, recent research has shown a potential association between COVID-19 and change in the molecular cargo of extracellular vesicles (EVs). EVs are vesicles released by cells and play an important role in cell communication by transfer of bioactive molecules between cells. Emerging evidence shows a strong link between EVs and their molecular cargo, and regulation of metabolism in health and disease. This review focuses on current knowledge about EVs and their potential role in COVID-19 pathogenesis, their current and future implications as tools for biomarker and therapeutic development and their possible effects on long-term impact of COVID-19.
Collapse
Affiliation(s)
- Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4072, Australia
| | - Estefania Nova-Lamperti
- Molecular and Translational Immunology Laboratory, Clinical Biochemistry and Immunology Department, Pharmacy Faculty, Universidad de Concepción, Concepción, Chile
| | - Gonzalo Labarca
- Molecular and Translational Immunology Laboratory, Clinical Biochemistry and Immunology Department, Pharmacy Faculty, Universidad de Concepción, Concepción, Chile
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4102, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Flavio Carrión
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4072, Australia.
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
17
|
Mink S, Saely CH, Frick M, Leiherer A, Drexel H, Fraunberger P. Association between Lipid Levels, Anti-SARS-CoV-2 Spike Antibodies and COVID-19 Mortality: A Prospective Cohort Study. J Clin Med 2023; 12:5068. [PMID: 37568470 PMCID: PMC10420155 DOI: 10.3390/jcm12155068] [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: 07/07/2023] [Revised: 07/21/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Recent studies suggest that both lipid levels and anti-severe-acute-respiratory-syndrome-coronavirus-2 (SARS-CoV-2) antibody levels are associated with outcome in coronavirus disease 2019 (COVID-19). While both parameters have separately been implicated in the neutralization and clearance of pathogens during severe infections, it is currently unclear whether the interplay of these parameters affects outcome in COVID-19. We therefore aimed to determine whether there was a relationship between lipoproteins, anti-SARS-CoV-2 antibodies, and COVID-19 mortality. METHODS In this prospective, multicenter cohort study, we recruited 1152 hospitalized patients with COVID-19 from five hospitals. Total cholesterol (TC), LDL-C, HDL-C, triglycerides, and anti-SARS-CoV-2 spike antibodies were measured on hospital admission. The investigated endpoint was in-hospital mortality. RESULTS LDL-C, HDL-C, and TC were significantly lower in non-survivors than in survivors (mg/dL, 95%CI; 56.1, 50.4-61.8 vs. 72.6, 70.2-75.0, p < 0.001; 34.2, 31.7-36.8 vs. 38.1, 37.2-39.1, p = 0.025; 139.3, 130.9-147.7 vs. 157.4, 54.1-160.6, p = 0.002). Mortality risk increased progressively with lower levels of LDL-C, HDL-C, and TC (aOR 1.73, 1.30-2.31, p < 0.001; 1.44, 1.10-1.88, p = 0.008; 1.49, 1.14-1.94, p < 0.001). Mortality rates varied between 2.1% for high levels of both LDL-C and anti-SARS-CoV-2 antibodies and 16.3% for low levels of LDL-C and anti-SARS-CoV-2 antibodies (aOR 9.14, 95%CI 3.17-26.34, p < 0.001). Accordingly, for total cholesterol and anti-SARS-CoV-2 antibodies, mortality rates varied between 2.1% and 15.0% (aOR 8.01, 95%CI 2.77-23.18, p < 0.001). CONCLUSION The combination of serum lipid levels and anti-SARS-CoV-2 antibodies is strongly associated with in-hospital mortality of patients with COVID-19. Patients with low levels of LDL-C and total cholesterol combined with low levels of anti-SARS-CoV-2 antibodies exhibited the highest mortality rates.
Collapse
Affiliation(s)
- Sylvia Mink
- Central Medical Laboratories, 6800 Feldkirch, Austria
- Medical-Scientific Faculty, Private University of the Principality of Liechtenstein, 9495 Triesen, Liechtenstein
| | - Christoph H. Saely
- Medical-Scientific Faculty, Private University of the Principality of Liechtenstein, 9495 Triesen, Liechtenstein
- VIVIT Institute, Academic Teaching Hospital Feldkirch, 6800 Feldkirch, Austria
| | - Matthias Frick
- Department of Internal Medicine, Academic Teaching Hospital Feldkirch, 6800 Feldkirch, Austria
| | - Andreas Leiherer
- Central Medical Laboratories, 6800 Feldkirch, Austria
- Medical-Scientific Faculty, Private University of the Principality of Liechtenstein, 9495 Triesen, Liechtenstein
- VIVIT Institute, Academic Teaching Hospital Feldkirch, 6800 Feldkirch, Austria
| | - Heinz Drexel
- Medical-Scientific Faculty, Private University of the Principality of Liechtenstein, 9495 Triesen, Liechtenstein
- VIVIT Institute, Academic Teaching Hospital Feldkirch, 6800 Feldkirch, Austria
- Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Peter Fraunberger
- Central Medical Laboratories, 6800 Feldkirch, Austria
- Medical-Scientific Faculty, Private University of the Principality of Liechtenstein, 9495 Triesen, Liechtenstein
| |
Collapse
|