1
|
Rosas-Murrieta NH, Rodríguez-Enríquez A, Herrera-Camacho I, Millán-Pérez-Peña L, Santos-López G, Rivera-Benítez JF. Comparative Review of the State of the Art in Research on the Porcine Epidemic Diarrhea Virus and SARS-CoV-2, Scope of Knowledge between Coronaviruses. Viruses 2024; 16:238. [PMID: 38400014 PMCID: PMC10892376 DOI: 10.3390/v16020238] [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: 12/13/2023] [Revised: 01/17/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
This review presents comparative information corresponding to the progress in knowledge of some aspects of infection by the porcine epidemic diarrhea virus (PEDV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) coronaviruses. PEDV is an alphacoronavirus of great economic importance due to the million-dollar losses it generates in the pig industry. PEDV has many similarities to the SARS-CoV-2 betacoronavirus that causes COVID-19 disease. This review presents possible scenarios for SARS-CoV-2 based on the collected literature on PEDV and the tools or strategies currently developed for SARS-CoV-2 that would be useful in PEDV research. The speed of the study of SARS-CoV-2 and the generation of strategies to control the pandemic was possible due to the knowledge derived from infections caused by other human coronaviruses such as severe acute respiratory syndrome (SARS) and middle east respiratory syndrome (MERS). Therefore, from the information obtained from several coronaviruses, the current and future behavior of SARS-CoV-2 could be inferred and, with the large amount of information on the virus that causes COVID-19, the study of PEDV could be improved and probably that of new emerging and re-emerging coronaviruses.
Collapse
Affiliation(s)
- Nora H. Rosas-Murrieta
- Centro de Química, Laboratorio de Bioquímica y Biología Molecular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (A.R.-E.); (I.H.-C.); (L.M.-P.-P.)
| | - Alan Rodríguez-Enríquez
- Centro de Química, Laboratorio de Bioquímica y Biología Molecular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (A.R.-E.); (I.H.-C.); (L.M.-P.-P.)
- Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico
| | - Irma Herrera-Camacho
- Centro de Química, Laboratorio de Bioquímica y Biología Molecular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (A.R.-E.); (I.H.-C.); (L.M.-P.-P.)
| | - Lourdes Millán-Pérez-Peña
- Centro de Química, Laboratorio de Bioquímica y Biología Molecular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (A.R.-E.); (I.H.-C.); (L.M.-P.-P.)
| | - Gerardo Santos-López
- Centro de Investigación Biomédica de Oriente, Laboratorio de Biología Molecular y Virología, Instituto Mexicano del Seguro Social (IMSS), Metepec 74360, Mexico;
| | - José F. Rivera-Benítez
- Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Ciudad de México 38110, Mexico;
| |
Collapse
|
2
|
Kakavandi S, Zare I, VaezJalali M, Dadashi M, Azarian M, Akbari A, Ramezani Farani M, Zalpoor H, Hajikhani B. Structural and non-structural proteins in SARS-CoV-2: potential aspects to COVID-19 treatment or prevention of progression of related diseases. Cell Commun Signal 2023; 21:110. [PMID: 37189112 PMCID: PMC10183699 DOI: 10.1186/s12964-023-01104-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/15/2023] [Indexed: 05/17/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by a new member of the Coronaviridae family known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). There are structural and non-structural proteins (NSPs) in the genome of this virus. S, M, H, and E proteins are structural proteins, and NSPs include accessory and replicase proteins. The structural and NSP components of SARS-CoV-2 play an important role in its infectivity, and some of them may be important in the pathogenesis of chronic diseases, including cancer, coagulation disorders, neurodegenerative disorders, and cardiovascular diseases. The SARS-CoV-2 proteins interact with targets such as angiotensin-converting enzyme 2 (ACE2) receptor. In addition, SARS-CoV-2 can stimulate pathological intracellular signaling pathways by triggering transcription factor hypoxia-inducible factor-1 (HIF-1), neuropilin-1 (NRP-1), CD147, and Eph receptors, which play important roles in the progression of neurodegenerative diseases like Alzheimer's disease, epilepsy, and multiple sclerosis, and multiple cancers such as glioblastoma, lung malignancies, and leukemias. Several compounds such as polyphenols, doxazosin, baricitinib, and ruxolitinib could inhibit these interactions. It has been demonstrated that the SARS-CoV-2 spike protein has a stronger affinity for human ACE2 than the spike protein of SARS-CoV, leading the current study to hypothesize that the newly produced variant Omicron receptor-binding domain (RBD) binds to human ACE2 more strongly than the primary strain. SARS and Middle East respiratory syndrome (MERS) viruses against structural and NSPs have become resistant to previous vaccines. Therefore, the review of recent studies and the performance of current vaccines and their effects on COVID-19 and related diseases has become a vital need to deal with the current conditions. This review examines the potential role of these SARS-CoV-2 proteins in the initiation of chronic diseases, and it is anticipated that these proteins could serve as components of an effective vaccine or treatment for COVID-19 and related diseases. Video Abstract.
Collapse
Affiliation(s)
- Sareh Kakavandi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz, 7178795844, Iran
| | - Maryam VaezJalali
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Dadashi
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Maryam Azarian
- Department of Radiology, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Abdullatif Akbari
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Marzieh Ramezani Farani
- Department of Biological Sciences and Bioengineering, Nano Bio High-Tech Materials Research Center, Inha University, Incheon, 22212, Republic of Korea
| | - Hamidreza Zalpoor
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Bahareh Hajikhani
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Bansal A, Kumar S, Rai N, Kumari S, Kumar V, Kumar A, Chandra NC. A Pilot Study on Blood Components in COVID-19 Affected Subjects: A Correlation to UPR Signalling and ER-Stress. Indian J Clin Biochem 2023; 38:374-384. [PMCID: PMC9997434 DOI: 10.1007/s12291-023-01121-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 02/20/2023] [Indexed: 03/12/2023]
Abstract
Abstract The endoplasmic reticulum (ER) is the site for protein synthesis, its folding and secretion. An intricate set of signalling pathways, called UPR pathways, have been evolved by ER in mammalian cells, to allow the cell to respond the presence of misfolded proteins within the ER. Breaching of these signalling systems by disease oriented accumulation of unfolded proteins may develop cellular stress. The aim of this study is to explore whether COVID-19 infection is responsible for developing this kind of endoplasmic reticulum related stress (ER-stress). ER-stress was evaluated by checking the expression of ER-stress markers e.g. PERK (adapting) and TRAF2 (alarming). ER-stress was correlated to several blood parameters viz. IgG, pro- and anti-inflammatory cytokines, leukocytes, lymphocytes, RBC, haemoglobin and PaO2/FiO2 ratio (ratio of arterial oxygen partial pressure to fractional inspired oxygen) in COVID-19 affected subjects. COVID-19 infection was found to be a state of protein homeostasis (proteostasis) collapse. Changes in IgG levels showed very poor immune response by the infected subjects. At the initial phase of the disease, pro-inflammatory cytokine levels were high and anti-inflammatory cytokines levels were low; though they were partly compromised at later phase of the disease. Total leukocyte concentration increased over the period of time; while percentage of lymphocytes were dropped. No significant changes were observed in cases of RBC counts and haemoglobin (Hb) levels. Both RBC and Hb were maintained at their normal range. In mildly stressed group, PaO2/FiO2 ratio (oxygenation status) was in the higher side of normal range; whereas in other two groups the ratio was in respiratory distress syndrome mode. Virus could induce mild to severe ER-stress, which could be the cause of cellular death and systemic dysfunction introducing fatal consequences. Graphical Abstract Schematic representation of SARS-CoV-2 infection and related consequences.![]()
Collapse
Affiliation(s)
- Akash Bansal
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, 801507 India
| | - Sushil Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, 801507 India
| | - Neha Rai
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, 801507 India
| | - Shilpi Kumari
- Department of Biochemistry, School of Basic Applied Sciences, Galgotias University, Greater Noida, Gautam Budh Nagar, Uttar Pradesh 201301 India
| | - Visesh Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, 801507 India
| | - Ajeet Kumar
- Department of Anesthesiology, All India Institute of Medical Sciences, Patna, 801507 India
| | - Nimai Chand Chandra
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, 801507 India ,Present Address: Department of Biochemistry, SGT University, Budhera, Gurugram, Haryana 122505 India
| |
Collapse
|
4
|
Yu S, Li X, Xin Z, Sun L, Shi J. Proteomic insights into SARS-CoV-2 infection mechanisms, diagnosis, therapies and prognostic monitoring methods. Front Immunol 2022; 13:923387. [PMID: 36203586 PMCID: PMC9530739 DOI: 10.3389/fimmu.2022.923387] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/05/2022] [Indexed: 01/08/2023] Open
Abstract
At the end of 2019, the COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection, seriously damaged world public health security. Several protein markers associated with virus infection have been extensively explored to combat the ever-increasing challenge posed by SARS-CoV-2. The proteomics of COVID-19 deepened our understanding of viral particles and their mechanisms of host invasion, providing us with information on protein changes in host tissues, cells and body fluids following infection in COVID-19 patients. In this review, we summarize the proteomic studies of SARS-CoV-2 infection and review the current understanding of COVID-19 in terms of the quantitative and qualitative proteomics of viral particles and host entry factors from the perspective of protein pathological changes in the organism following host infection.
Collapse
Affiliation(s)
- Shengman Yu
- Department of Laboratory Medicine Center, China-Japan Union Hospital, Jilin University, Changchun, China
- School of Laboratory Medicine, Beihua University, Jilin, China
| | - Xiaoyan Li
- Department of Infection Control Department, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhuoyuan Xin
- The Key Laboratory of Zoonosis Research, Chinese Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Liyuan Sun
- School of Laboratory Medicine, Beihua University, Jilin, China
| | - Jingwei Shi
- Department of Laboratory Medicine Center, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
5
|
Moin A, Huwaimel B, Alobaida A, Break MKB, Iqbal D, Unissa R, Jamal QMS, Hussain T, Sharma DC, Rizvi SMD. Dithymoquinone Analogues as Potential Candidate(s) for Neurological Manifestation Associated with COVID-19: A Therapeutic Strategy for Neuro-COVID. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071076. [PMID: 35888166 PMCID: PMC9323060 DOI: 10.3390/life12071076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022]
Abstract
The COVID-19 era has prompted several researchers to search for a linkage between COVID-19 and its associated neurological manifestation. Toll-like receptor 4 (TLR-4) acts as one such connecting link. spike protein of SARS-CoV-2 can bind either to ACE-2 receptors or to TLR-4 receptors, leading to aggregation of α-synuclein and neurodegeneration via the activation of various cascades in neurons. Recently, dithymoquinone has been reported as a potent multi-targeting candidate against SARS-CoV-2. Thus, in the present study, dithymoquinone and its six analogues were explored to target 3CLpro (main protease of SARS-CoV-2), TLR4 and PREP (Prolyl Oligopeptidases) by using the molecular docking and dynamics approach. Dithymoquinone (DTQ) analogues were designed in order to investigate the effect of different chemical groups on its bioactivity. It is noteworthy to mention that attention was given to the feasibility of synthesizing these analogues by a simple photo-dimerisation reaction. The DTQ analogue containing the 4-fluoroaniline moiety [Compound (4)] was selected for further analysis by molecular dynamics after screening via docking-interaction analyses. A YASARA structure tool built on the AMBER14 force field was used to analyze the 100 ns trajectory by taking 400 snapshots after every 250 ps. Moreover, RMSD, RoG, potential energy plots were successfully obtained for each interaction. Molecular docking results indicated strong interaction of compound (4) with 3CLpro, TLR4 and PREP with a binding energy of -8.5 kcal/mol, -10.8 kcal/mol and -9.5 kcal/mol, respectively, which is better than other DTQ-analogues and control compounds. In addition, compound (4) did not violate Lipinski's rule and showed no toxicity. Moreover, molecular dynamic analyses revealed that the complex of compound (4) with target proteins was stable during the 100 ns trajectory. Overall, the results predicted that compound (4) could be developed into a potent anti-COVID agent with the ability to mitigate neurological manifestations associated with COVID-19.
Collapse
Affiliation(s)
- Afrasim Moin
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia; (A.M.); (A.A.); (R.U.)
| | - Bader Huwaimel
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia; (B.H.); (M.K.B.B.)
| | - Ahmed Alobaida
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia; (A.M.); (A.A.); (R.U.)
| | - Mohammed Khaled Bin Break
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia; (B.H.); (M.K.B.B.)
| | - Danish Iqbal
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia;
| | - Rahamat Unissa
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia; (A.M.); (A.A.); (R.U.)
| | - Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al Bukayriyah 52741, Saudi Arabia;
| | - Talib Hussain
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
- Correspondence: (T.H.); (S.M.D.R.)
| | - Dinesh C. Sharma
- School of Life Sciences, The Glocal University, Saharanpur 247121, Uttar Pradesh, India;
- Department of Microbiology, School of Life Sciences, Starex University, Gurugram 122413, Haryana, India
| | - Syed Mohd Danish Rizvi
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia; (A.M.); (A.A.); (R.U.)
- Correspondence: (T.H.); (S.M.D.R.)
| |
Collapse
|
6
|
Oleuropein as a Potent Compound against Neurological Complications Linked with COVID-19: A Computational Biology Approach. ENTROPY 2022; 24:e24070881. [PMID: 35885104 PMCID: PMC9319675 DOI: 10.3390/e24070881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 12/04/2022]
Abstract
The association of COVID-19 with neurological complications is a well-known fact, and researchers are endeavoring to investigate the mechanistic perspectives behind it. SARS-CoV-2 can bind to Toll-like receptor 4 (TLR-4) that would eventually lead to α-synuclein aggregation in neurons and stimulation of neurodegeneration pathways. Olive leaves have been reported as a promising phytotherapy or co-therapy against COVID-19, and oleuropein is one of the major active components of olive leaves. In the current study, oleuropein was investigated against SARS-CoV-2 target (main protease 3CLpro), TLR-4 and Prolyl Oligopeptidases (POP), to explore oleuropein potency against the neurological complications associated with COVID-19. Docking experiments, docking validation, interaction analysis, and molecular dynamic simulation analysis were performed to provide insight into the binding pattern of oleuropein with the three target proteins. Interaction analysis revealed strong bonding between oleuropein and the active site amino acid residues of the target proteins. Results were further compared with positive control lopinavir (3CLpro), resatorvid (TLR-4), and berberine (POP). Moreover, molecular dynamic simulation was performed using YASARA structure tool, and AMBER14 force field was applied to examine an 100 ns trajectory run. For each target protein-oleuropein complex, RMSD, RoG, and total potential energy were estimated, and 400 snapshots were obtained after each 250 ps. Docking analyses showed binding energy as −7.8, −8.3, and −8.5 kcal/mol for oleuropein-3CLpro, oleuropein-TLR4, and oleuropein-POP interactions, respectively. Importantly, target protein-oleuropein complexes were stable during the 100 ns simulation run. However, an experimental in vitro study of the binding of oleuropein to the purified targets would be necessary to confirm the present study outcomes.
Collapse
|
7
|
Kitchen LC, Berman M, Halper J, Chazot P. Rationale for 1068 nm Photobiomodulation Therapy (PBMT) as a Novel, Non-Invasive Treatment for COVID-19 and Other Coronaviruses: Roles of NO and Hsp70. Int J Mol Sci 2022; 23:ijms23095221. [PMID: 35563611 PMCID: PMC9105035 DOI: 10.3390/ijms23095221] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 01/08/2023] Open
Abstract
Researchers from across the world are seeking to develop effective treatments for the ongoing coronavirus disease 2019 (COVID-19) outbreak, which arose as a major public health issue in 2019, and was declared a pandemic in early 2020. The pro-inflammatory cytokine storm, acute respiratory distress syndrome (ARDS), multiple-organ failure, neurological problems, and thrombosis have all been linked to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) fatalities. The purpose of this review is to explore the rationale for using photobiomodulation therapy (PBMT) of the particular wavelength 1068 nm as a therapy for COVID-19, investigating the cellular and molecular mechanisms involved. Our findings illustrate the efficacy of PBMT 1068 nm for cytoprotection, nitric oxide (NO) release, inflammation changes, improved blood flow, and the regulation of heat shock proteins (Hsp70). We propose, therefore, that PBMT 1068 is a potentially effective and innovative approach for avoiding severe and critical illness in COVID-19 patients, although further clinical evidence is required.
Collapse
Affiliation(s)
- Lydia C. Kitchen
- Department of Biosciences, Durham University, Durham DH1 3LE, UK;
| | - Marvin Berman
- Quietmind Foundation, Philadelphia, PA 19147, USA; (M.B.); (J.H.)
| | - James Halper
- Quietmind Foundation, Philadelphia, PA 19147, USA; (M.B.); (J.H.)
| | - Paul Chazot
- Department of Biosciences, Durham University, Durham DH1 3LE, UK;
- Correspondence:
| |
Collapse
|
8
|
The Potential of Dietary Bioactive Compounds against SARS-CoV-2 and COVID-19-Induced Endothelial Dysfunction. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27051623. [PMID: 35268723 PMCID: PMC8912066 DOI: 10.3390/molecules27051623] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 12/24/2022]
Abstract
COVID-19 is an endothelial disease. All the major comorbidities that increase the risk for severe SARS-CoV-2 infection and severe COVID-19 including old age, obesity, diabetes, hypertension, respiratory disease, compromised immune system, coronary artery disease or heart failure are associated with dysfunctional endothelium. Genetics and environmental factors (epigenetics) are major risk factors for endothelial dysfunction. Individuals with metabolic syndrome are at increased risk for severe SARS-CoV-2 infection and poor COVID-19 outcomes and higher risk of mortality. Old age is a non-modifiable risk factor. All other risk factors are modifiable. This review also identifies dietary risk factors for endothelial dysfunction. Potential dietary preventions that address endothelial dysfunction and its sequelae may have an important role in preventing SARS-CoV-2 infection severity and are key factors for future research to address. This review presents some dietary bioactives with demonstrated efficacy against dysfunctional endothelial cells. This review also covers dietary bioactives with efficacy against SARS-CoV-2 infection. Dietary bioactive compounds that prevent endothelial dysfunction and its sequelae, especially in the gastrointestinal tract, will result in more effective prevention of SARS-CoV-2 variant infection severity and are key factors for future food research to address.
Collapse
|
9
|
Jiang W, Majumder S, Kumar S, Subramaniam S, Li X, Khedri R, Mondal T, Abolghasemian M, Satia I, Deen MJ. A Wearable Tele-Health System towards Monitoring COVID-19 and Chronic Diseases. IEEE Rev Biomed Eng 2022; 15:61-84. [PMID: 33784625 PMCID: PMC8905615 DOI: 10.1109/rbme.2021.3069815] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/01/2021] [Accepted: 03/22/2021] [Indexed: 11/10/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a pandemic since early 2020. The coronavirus disease 2019 (COVID-19) has already caused more than three million deaths worldwide and affected people's physical and mental health. COVID-19 patients with mild symptoms are generally required to self-isolate and monitor for symptoms at least for 14 days in the case the disease turns towards severe complications. In this work, we overviewed the impact of COVID-19 on the patients' general health with a focus on their cardiovascular, respiratory and mental health, and investigated several existing patient monitoring systems. We addressed the limitations of these systems and proposed a wearable telehealth solution for monitoring a set of physiological parameters that are critical for COVID-19 patients such as body temperature, heart rate, heart rate variability, blood oxygen saturation, respiratory rate, blood pressure, and cough. This physiological information can be further combined to potentially estimate the lung function using artificial intelligence (AI) and sensor fusion techniques. The prototype, which includes the hardware and a smartphone app, showed promising results with performance comparable to or better than similar commercial devices, thus potentially making the proposed system an ideal wearable solution for long-term monitoring of COVID-19 patients and other chronic diseases.
Collapse
Affiliation(s)
- Wei Jiang
- McMaster School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4K1Canada
| | - Sumit Majumder
- Electrical and Computer Engineering DepartmentMcMaster UniversityHamiltonONL8S 4K1Canada
| | - Samarth Kumar
- Electrical and Computer Engineering DepartmentMcMaster UniversityHamiltonONL8S 4K1Canada
| | - Sophini Subramaniam
- McMaster School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4K1Canada
| | - Xiaohe Li
- The Third People's Hospital of ShenzhenGuangdong Province518112China
| | - Ridha Khedri
- Computing and Software DepartmentMcMaster UniversityHamiltonONL8S 4K1Canada
| | - Tapas Mondal
- PediatricsMcMaster UniversityHamiltonONL8S 4K1Canada
| | | | - Imran Satia
- Department of Medicine, Division of RespirologyMcMaster UniversityHamiltonONL8S 4K1Canada
- Firestone Institute for Respiratory Health, St Joseph's HealthcareHamiltonONL8S 4K1Canada
| | - M. Jamal Deen
- McMaster School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4K1Canada
- and also with the Electrical and Computer Engineering DepartmentMcMaster UniversityHamiltonONL8S 4K1Canada
| |
Collapse
|
10
|
Heubel AD, Viana AA, Linares SN, do Amaral VT, Schafauser NS, de Oliveira GYO, Ramírez PC, Martinelli B, da Silva Alexandre T, Borghi‐Silva A, Ciolac EG, Mendes RG. Determinants of endothelial dysfunction in noncritically ill hospitalized COVID-19 patients: A cross-sectional study. Obesity (Silver Spring) 2022; 30:165-171. [PMID: 34554646 PMCID: PMC8661847 DOI: 10.1002/oby.23311] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/24/2021] [Accepted: 09/21/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVE The aim of this study was to identify determinants of endothelial dysfunction in patients hospitalized with acute COVID-19. METHODS A total of 109 hospitalized COVID-19 patients in noncritical status were cross-sectionally studied. Clinical data (age, sex, comorbidities, and medications) and BMI were assessed. Laboratory tests included serum hemoglobin, leukocytes, lymphocytes, platelets, C-reactive protein, ferritin, D-dimer, and creatinine. Physical status was evaluated using a handgrip dynamometer. Endothelial function was assessed noninvasively using the flow-mediated dilation (FMD) method. RESULTS The sample average age was 51 years, 51% of patients were male, and the most frequent comorbidity was obesity (62%). Univariate analysis showed association of lower FMD with higher BMI, hypertension, use of oral antihypertensive, higher blood levels of creatinine, and larger baseline artery diameter. After adjusting for confounders, the multivariate analysis showed BMI (95% CI: -0.26 to -0.11; p < 0.001) as the major factor associated with FMD. Other factors associated with FMD were baseline artery diameter (95% CI: -1.77 to -0.29; p = 0.007) and blood levels of creatinine (95% CI: -1.99 to -0.16; p = 0.022). CONCLUSIONS Increased BMI was the major factor associated with endothelial dysfunction in noncritically hospitalized COVID-19 patients. This may explain one of the pathways in which obesity may increase the risk for severe COVID-19.
Collapse
Affiliation(s)
- Alessandro Domingues Heubel
- Cardiopulmonary Physiotherapy LaboratoryDepartment of Physical TherapyFederal University of São CarlosSão CarlosSão PauloBrazil
| | - Ariane Aparecida Viana
- Exercise and Chronic Disease Research LaboratoryDepartment of Physical EducationSchool of SciencesSão Paulo State UniversityBauruSão PauloBrazil
| | - Stephanie Nogueira Linares
- Cardiopulmonary Physiotherapy LaboratoryDepartment of Physical TherapyFederal University of São CarlosSão CarlosSão PauloBrazil
| | - Vanessa Teixeira do Amaral
- Exercise and Chronic Disease Research LaboratoryDepartment of Physical EducationSchool of SciencesSão Paulo State UniversityBauruSão PauloBrazil
| | - Nathany Souza Schafauser
- Cardiopulmonary Physiotherapy LaboratoryDepartment of Physical TherapyFederal University of São CarlosSão CarlosSão PauloBrazil
| | - Gustavo Yudi Orikassa de Oliveira
- Exercise and Chronic Disease Research LaboratoryDepartment of Physical EducationSchool of SciencesSão Paulo State UniversityBauruSão PauloBrazil
| | - Paula Camila Ramírez
- Department of Physical TherapyUniversidad Industrial de SantanderBucaramangaSantanderColombia
- Department of GerontologyFederal University of São CarlosSão CarlosSão PauloBrazil
| | - Bruno Martinelli
- Department of Physical TherapySacred Heart University CenterBauruSão PauloBrazil
| | | | - Audrey Borghi‐Silva
- Cardiopulmonary Physiotherapy LaboratoryDepartment of Physical TherapyFederal University of São CarlosSão CarlosSão PauloBrazil
| | - Emmanuel Gomes Ciolac
- Exercise and Chronic Disease Research LaboratoryDepartment of Physical EducationSchool of SciencesSão Paulo State UniversityBauruSão PauloBrazil
| | - Renata Gonçalves Mendes
- Cardiopulmonary Physiotherapy LaboratoryDepartment of Physical TherapyFederal University of São CarlosSão CarlosSão PauloBrazil
| |
Collapse
|
11
|
Schmelter F, Föh B, Mallagaray A, Rahmöller J, Ehlers M, Lehrian S, von Kopylow V, Künsting I, Lixenfeld AS, Martin E, Ragab M, Meyer-Saraei R, Kreutzmann F, Eitel I, Taube S, Käding N, Jantzen E, Graf T, Sina C, Günther UL. Metabolic and Lipidomic Markers Differentiate COVID-19 From Non-Hospitalized and Other Intensive Care Patients. Front Mol Biosci 2021; 8:737039. [PMID: 34938772 PMCID: PMC8686182 DOI: 10.3389/fmolb.2021.737039] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a viral infection affecting multiple organ systems of great significance for metabolic processes. Thus, there is increasing interest in metabolic and lipoprotein signatures of the disease, and early analyses have demonstrated a metabolic pattern typical for atherosclerotic and hepatic damage in COVID-19 patients. However, it remains unclear whether this is specific for COVID-19 and whether the observed signature is caused by the disease or rather represents an underlying risk factor. To answer this question, we have analyzed 482 serum samples using nuclear magnetic resonance metabolomics, including longitudinally collected samples from 12 COVID-19 and 20 cardiogenic shock intensive care patients, samples from 18 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibody-positive individuals, and single time point samples from 58 healthy controls. COVID-19 patients showed a distinct metabolic serum profile, including changes typical for severe dyslipidemia and a deeply altered metabolic status compared with healthy controls. Specifically, very-low-density lipoprotein and intermediate-density lipoprotein particles and associated apolipoprotein B and intermediate-density lipoprotein cholesterol were significantly increased, whereas cholesterol and apolipoprotein A2 were decreased. Moreover, a similarly perturbed profile was apparent when compared with other patients with cardiogenic shock who are in the intensive care unit when looking at a 1-week time course, highlighting close links between COVID-19 and lipid metabolism. The metabolic profile of COVID-19 patients distinguishes those from healthy controls and also from patients with cardiogenic shock. In contrast, anti-SARS-CoV-2 antibody-positive individuals without acute COVID-19 did not show a significantly perturbed metabolic profile compared with age- and sex-matched healthy controls, but SARS-CoV-2 antibody-titers correlated significantly with metabolic parameters, including levels of glycine, ApoA2, and small-sized low- and high-density lipoprotein subfractions. Our data suggest that COVID-19 is associated with dyslipidemia, which is not observed in anti-SARS-CoV-2 antibody-positive individuals who have not developed severe courses of the disease. This suggests that lipoprotein profiles may represent a confounding risk factor for COVID-19 with potential for patient stratification.
Collapse
Affiliation(s)
- Franziska Schmelter
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany.,Research and Development Department, GALAB Laboratories GmbH, Hamburg, Germany
| | - Bandik Föh
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany.,Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Alvaro Mallagaray
- Institute of Chemistry and Metabolomics, University of Lübeck, Lübeck, Germany
| | - Johann Rahmöller
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany.,Department of Anesthesiology and Intensive Care, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Marc Ehlers
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany
| | - Selina Lehrian
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany
| | - Vera von Kopylow
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany
| | - Inga Künsting
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany
| | | | - Emily Martin
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany
| | - Mohab Ragab
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany
| | - Roza Meyer-Saraei
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Lübeck, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Fabian Kreutzmann
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Lübeck, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Ingo Eitel
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Lübeck, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Stefan Taube
- Institute of Virology and Cell Biology, University of Lübeck, Lübeck, Germany
| | - Nadja Käding
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Eckard Jantzen
- Research and Development Department, GALAB Laboratories GmbH, Hamburg, Germany
| | - Tobias Graf
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Lübeck, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Christian Sina
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany.,Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Ulrich L Günther
- Institute of Chemistry and Metabolomics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
12
|
Teixeira AL, Krause TM, Ghosh L, Shahani L, Machado-Vieira R, Lane SD, Boerwinkle E, Soares JC. Analysis of COVID-19 Infection and Mortality Among Patients With Psychiatric Disorders, 2020. JAMA Netw Open 2021; 4:e2134969. [PMID: 34812848 PMCID: PMC8611476 DOI: 10.1001/jamanetworkopen.2021.34969] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
IMPORTANCE People with major psychiatric disorders are more likely to have comorbidities associated with worse outcomes of COVID-19. This fact alone could determine greater vulnerability of people with major psychiatric disorders to COVID-19. OBJECTIVE To assess the odds of testing positive for and mortality from COVID-19 among and between patients with schizophrenia, mood disorders, anxiety disorders and a reference group in a large national database. DESIGN, SETTING, AND PARTICIPANTS This cross-sectional study used an electronic health record data set aggregated from many national sources in the United States and licensed from Optum with current and historical data on patients tested for COVID-19 in 2020. Three psychiatric cohorts (patients with schizophrenia, mood disorders, or anxiety disorders) were compared with a reference group with no major psychiatric conditions. Statistical analysis was performed from March to April 2021. EXPOSURE The exposures observed include lab-confirmed positivity for COVID-19 and mortality. MAIN OUTCOMES AND MEASURES The odds of testing positive for COVID-19 in 2020 and the odds of death from COVID-19 were measured. RESULTS The population studied included 2 535 098 unique persons, 3350 with schizophrenia, 26 610 with mood disorders, and 18 550 with anxiety disorders. The mean (SD) age was 44 (23) years; 233 519 were non-Hispanic African American, 1 583 440 were non-Hispanic Caucasian; and 1 580 703 (62%) were female. The schizophrenia cohort (positivity rate: 9.86%; adjusted OR, 0.90 [95% CI, 0.84-0.97]) and the mood disorder cohort (positivity rate: 9.86%; adjusted OR, 0.93 [95% CI, 0.87-0.99]) had a significantly lower rate of positivity than the anxiety disorder cohort (positivity rate: 11.17%; adjusted OR, 1.05 [95% CI, 0.98-1.12) which was closer to the reference group (11.91%). After fully adjusting for demographic factors and comorbid conditions, patients with schizophrenia were nearly 4 times more likely to die from the disease than the reference group (OR, 3.74; 95% CI, 2.66-5.24). The mood disorders COVID-19 cohort had a 2.76 times greater odds of mortality than the reference group (OR, 2.76; 95% CI, 2.00-3.81), and the anxiety disorders cohort had a 2.39 times greater odds of mortality than the reference group (OR, 2.39; 95% CI, 1.68-3.27). CONCLUSIONS AND RELEVANCE By examining a large database while controlling for multiple confounding factors such as age, race and ethnicity, and comorbid medical conditions, the present study found that patients with schizophrenia had much increased odds of mortality by COVID-19.
Collapse
Affiliation(s)
- Antonio L. Teixeira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston
| | - Trudy Millard Krause
- School of Public Health, The University of Texas Health Science Center at Houston, Houston
| | - Lopita Ghosh
- School of Public Health, The University of Texas Health Science Center at Houston, Houston
| | - Lokesh Shahani
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston
| | - Rodrigo Machado-Vieira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston
| | - Scott D. Lane
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston
| | - Eric Boerwinkle
- School of Public Health, The University of Texas Health Science Center at Houston, Houston
| | - Jair C. Soares
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston
| |
Collapse
|
13
|
Tan HW, Xu YM, Lau ATY. Human bronchial-pulmonary proteomics in coronavirus disease 2019 (COVID-19) pandemic: applications and implications. Expert Rev Proteomics 2021; 18:925-938. [PMID: 34812694 DOI: 10.1080/14789450.2021.2010549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/22/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The outbreak of the newly discovered human coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has disrupted the normal life of almost every civilization worldwide. Studies have shown that the coronavirus disease 2019 (COVID-19) caused by the SARS-CoV-2 can affect multiple human organs and physiological systems, but the respiratory system remains the primary location for viral infection. AREAS COVERED We summarize how omics technologies are used in SARS-CoV-2 research and specifically review the current knowledge of COVID-19 from the aspect of human bronchial-pulmonary proteomics. Also, knowledge gaps in COVID-19 that can be fulfilled by proteomics are discussed. EXPERT OPINION Overall, human bronchial-pulmonary proteomics plays an important role in revealing the dynamics, functions, tropism, and pathogenicity of SARS-CoV-2, which is crucial for COVID-19 biomarker and therapeutic target discoveries. To more fully understand the impact of COVID-19, research from various angles using multi-omics approaches should also be conducted on the lungs as well as other organs.
Collapse
Affiliation(s)
- Heng Wee Tan
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, People's Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, People's Republic of China
| | - Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, People's Republic of China
| |
Collapse
|
14
|
Lopes LR. Functional and tissue enrichment analyses suggest that SARS-CoV-2 infection affects host metabolism and catabolism mediated by interference on host proteins. Braz J Microbiol 2021; 52:1151-1159. [PMID: 33956332 PMCID: PMC8099703 DOI: 10.1007/s42770-021-00497-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/08/2021] [Indexed: 12/24/2022] Open
Abstract
Infection by SARS-CoV-2, the causative agent of COVID-19, is critically connected with host metabolism. Through functional enrichment analysis, the present study aims to evaluate the biological processes involving host proteins interfered by SARS-CoV-2 to verify the potential metabolic impact of the infection. Furthermore, tissue enrichment analyses and differential gene expression of host proteins were applied to understand the interference by SARS-CoV-2 on tissue levels. Results based on functional and tissue-specific enrichment analyses, presented in this study, suggest that SARS-CoV-2, mediated interference on host proteins, can affect the metabolism and catabolism of molecular building blocks and control intracellular mechanisms, including gene expression in metabolism-related organs, to support viral demands. Thus, SARS-CoV-2 can broadly affect the host metabolism and catabolism at tissue and physiological levels contributing to a more severe disease.
Collapse
Affiliation(s)
- Luciano Rodrigo Lopes
- Bioinformatics and Bio-Data Science Division, Health Informatics Department, Universidade Federal de São Paulo-UNIFESP, Rua Botucatu 862 - Prédio Leal Prado (térreo), São Paulo, SP, CEP: 04023-062, Brazil.
| |
Collapse
|
15
|
Agarwal P, Kaushik A, Sarkar S, Rao D, Mukherjee N, Bharat V, Das S, Saha AK. Global survey-based assessment of lifestyle changes during the COVID-19 pandemic. PLoS One 2021; 16:e0255399. [PMID: 34388151 PMCID: PMC8362972 DOI: 10.1371/journal.pone.0255399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/15/2021] [Indexed: 12/24/2022] Open
Abstract
Along with the major impact on public health, the COVID-19 outbreak has caused unprecedented concerns ranging from sudden loss of employment to mental stress and anxiety. We implemented a survey-based data collection platform to characterize how the COVID-19 pandemic has affected the socio-economic, physical and mental health conditions of individuals. We focused on three broad areas, namely, changes in social interaction during home confinement, economic impact and their health status. We identified a substantial increase in virtual interaction among individuals, which might be a way to alleviate the sudden unprecedented mental health burden, exacerbated by general awareness about viral infections or other manifestations associated with them. The majority of participants (85%) lived with one or more companions and unemployment issues did not affect 91% of the total survey takers, which was one of the crucial consequences of the pandemic. Nevertheless, measures such as an increased frequency of technology-aided distant social interaction, focus on physical fitness and leisure activities were adopted as coping mechanisms during this period of home isolation. Collectively, these metrics provide a succinct and informative summary of the socio-economic and health impact of the COVID-19 pandemic on the individuals. Findings from our study reflect that continuous surveillance of the psychological consequences for outbreaks should become routine as part of preparedness efforts worldwide. Given the limitations of analyzing the large number of variables, we have made the raw data publicly available on the OMF ME/CFS Data Center server to facilitate further analyses (https://igenomed.stanford.edu/dataset/survey-study-on-lifestyle-changes-during-covid-19-pandemic).
Collapse
Affiliation(s)
- Poonam Agarwal
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Abhinav Kaushik
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Sutapa Sarkar
- Gastroenterology and Hepatology, Stanford University School of Medicine, VA Palo Alto, Palo Alto, CA, United States of America
| | - Deepti Rao
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Nilanjan Mukherjee
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Vinita Bharat
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Subhamoy Das
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Amit Kumar Saha
- Department of Biochemistry, Stanford University School of Medicine, Palo Alto, CA United States of America
| |
Collapse
|
16
|
Logette E, Lorin C, Favreau C, Oshurko E, Coggan JS, Casalegno F, Sy MF, Monney C, Bertschy M, Delattre E, Fonta PA, Krepl J, Schmidt S, Keller D, Kerrien S, Scantamburlo E, Kaufmann AK, Markram H. A Machine-Generated View of the Role of Blood Glucose Levels in the Severity of COVID-19. Front Public Health 2021; 9:695139. [PMID: 34395368 PMCID: PMC8356061 DOI: 10.3389/fpubh.2021.695139] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/30/2021] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 started spreading toward the end of 2019 causing COVID-19, a disease that reached pandemic proportions among the human population within months. The reasons for the spectrum of differences in the severity of the disease across the population, and in particular why the disease affects more severely the aging population and those with specific preconditions are unclear. We developed machine learning models to mine 240,000 scientific articles openly accessible in the CORD-19 database, and constructed knowledge graphs to synthesize the extracted information and navigate the collective knowledge in an attempt to search for a potential common underlying reason for disease severity. The machine-driven framework we developed repeatedly pointed to elevated blood glucose as a key facilitator in the progression of COVID-19. Indeed, when we systematically retraced the steps of the SARS-CoV-2 infection, we found evidence linking elevated glucose to each major step of the life-cycle of the virus, progression of the disease, and presentation of symptoms. Specifically, elevations of glucose provide ideal conditions for the virus to evade and weaken the first level of the immune defense system in the lungs, gain access to deep alveolar cells, bind to the ACE2 receptor and enter the pulmonary cells, accelerate replication of the virus within cells increasing cell death and inducing an pulmonary inflammatory response, which overwhelms an already weakened innate immune system to trigger an avalanche of systemic infections, inflammation and cell damage, a cytokine storm and thrombotic events. We tested the feasibility of the hypothesis by manually reviewing the literature referenced by the machine-generated synthesis, reconstructing atomistically the virus at the surface of the pulmonary airways, and performing quantitative computational modeling of the effects of glucose levels on the infection process. We conclude that elevation in glucose levels can facilitate the progression of the disease through multiple mechanisms and can explain much of the differences in disease severity seen across the population. The study provides diagnostic considerations, new areas of research and potential treatments, and cautions on treatment strategies and critical care conditions that induce elevations in blood glucose levels.
Collapse
Affiliation(s)
- Emmanuelle Logette
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Henry Markram
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| |
Collapse
|
17
|
Triana S, Metz‐Zumaran C, Ramirez C, Kee C, Doldan P, Shahraz M, Schraivogel D, Gschwind AR, Sharma AK, Steinmetz LM, Herrmann C, Alexandrov T, Boulant S, Stanifer ML. Single-cell analyses reveal SARS-CoV-2 interference with intrinsic immune response in the human gut. Mol Syst Biol 2021; 17:e10232. [PMID: 33904651 PMCID: PMC8077299 DOI: 10.15252/msb.202110232] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/31/2021] [Accepted: 03/31/2021] [Indexed: 12/26/2022] Open
Abstract
Exacerbated pro-inflammatory immune response contributes to COVID-19 pathology. However, despite the mounting evidence about SARS-CoV-2 infecting the human gut, little is known about the antiviral programs triggered in this organ. To address this gap, we performed single-cell transcriptomics of SARS-CoV-2-infected intestinal organoids. We identified a subpopulation of enterocytes as the prime target of SARS-CoV-2 and, interestingly, found the lack of positive correlation between susceptibility to infection and the expression of ACE2. Infected cells activated strong pro-inflammatory programs and produced interferon, while expression of interferon-stimulated genes was limited to bystander cells due to SARS-CoV-2 suppressing the autocrine action of interferon. These findings reveal that SARS-CoV-2 curtails the immune response and highlights the gut as a pro-inflammatory reservoir that should be considered to fully understand SARS-CoV-2 pathogenesis.
Collapse
Affiliation(s)
- Sergio Triana
- Structural and Computational Biology Unit, European Molecular Biology LaboratoryHeidelbergGermany
- Faculty of BiosciencesCollaboration for Joint PhD Degree between EMBL and Heidelberg UniversityHeidelbergGermany
| | - Camila Metz‐Zumaran
- Department of Infectious Diseases, VirologyHeidelberg University HospitalHeidelbergGermany
| | - Carlos Ramirez
- Health Data Science UnitMedical Faculty University Heidelberg and BioQuantHeidelbergGermany
| | - Carmon Kee
- Department of Infectious Diseases, VirologyHeidelberg University HospitalHeidelbergGermany
- Research Group “Cellular Polarity and Viral Infection”German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Patricio Doldan
- Department of Infectious Diseases, VirologyHeidelberg University HospitalHeidelbergGermany
- Research Group “Cellular Polarity and Viral Infection”German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Mohammed Shahraz
- Structural and Computational Biology Unit, European Molecular Biology LaboratoryHeidelbergGermany
| | - Daniel Schraivogel
- Genome Biology Unit, European Molecular Biology LaboratoryHeidelbergGermany
| | - Andreas R Gschwind
- Department of GeneticsStanford University School of MedicineStanfordCAUSA
| | - Ashwini K Sharma
- Department of Infectious Diseases, VirologyHeidelberg University HospitalHeidelbergGermany
- Health Data Science UnitMedical Faculty University Heidelberg and BioQuantHeidelbergGermany
| | - Lars M Steinmetz
- Genome Biology Unit, European Molecular Biology LaboratoryHeidelbergGermany
- Department of GeneticsStanford University School of MedicineStanfordCAUSA
- Stanford Genome Technology CenterPalo AltoCAUSA
| | - Carl Herrmann
- Health Data Science UnitMedical Faculty University Heidelberg and BioQuantHeidelbergGermany
| | - Theodore Alexandrov
- Structural and Computational Biology Unit, European Molecular Biology LaboratoryHeidelbergGermany
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCAUSA
- Molecular Medicine Partnership Unit (MMPU)European Molecular Biology LaboratoryHeidelbergGermany
| | - Steeve Boulant
- Department of Infectious Diseases, VirologyHeidelberg University HospitalHeidelbergGermany
- Research Group “Cellular Polarity and Viral Infection”German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Megan L Stanifer
- Department of Infectious DiseasesMolecular VirologyHeidelberg University HospitalHeidelbergGermany
| |
Collapse
|
18
|
Host-virus interactions mediated by long non-coding RNAs. Virus Res 2021; 298:198402. [PMID: 33771610 DOI: 10.1016/j.virusres.2021.198402] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 11/21/2022]
Abstract
Viruses are obligate pathogens that cause a wide range of diseases across all kingdoms of life. They have a colossal impact on the economy and healthcare infrastructure world-wide. Plants and animals have developed sophisticated molecular mechanisms to defend themselves against viruses and viruses in turn hijack host mechanisms to ensure their survival inside their hosts. Long non-coding (lnc) RNAs have emerged as important macromolecules that regulate plant-virus and animal-virus interactions. Both pro-viral and anti-viral lncRNAs have been reported and they show immense potential to be used as markers and in therapeutics. The current review is focussed on the recent developments that have been made in viral interactions of animals and plants.
Collapse
|
19
|
Yu L. Restoring Good Health in Elderly with Diverse Gut Microbiome and Food Intake Restriction to Combat COVID-19. Indian J Microbiol 2021; 61:104-107. [PMID: 33424043 PMCID: PMC7778869 DOI: 10.1007/s12088-020-00913-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
COVID-19 continues to be an ongoing global threat. The elderly with underlying health conditions like cardiovascular and lung diseases, diabetes, obesity, are the most vulnerable to this disease. Curing the pre-existing health conditions will greatly increase a person's resilience to COVID-19 and lower the death rate of the old people. Digestion and immunity form an integrated nutrition acquisition process, especially in obtaining essential amino acids and essential fatty acids from living microbial cells. A mature strong immunity coupled with gut dysbiosis in adults is the main cause of nutritional disorders like morbid obesity, diabetes mellitus, cardiovascular and pulmonary diseases. Nutrition disorders in return worsen dysbiosis. Human microbiome has an intrinsic duality. While a diverse microbiome provides a full spectrum of essential nutrients to our body, nutrition disorders fuel overgrowth of microbiota (dysbiosis) at many sites on or inside our body, and are the main causes of chronic inflammation at these sites. In the case of COVID-19, nutritional disorder impairs the immunity, causes hyperinflammation, and leads to the protracted overload of cytokines by the immune system, i.e., the cytokine storm. Autophagy induced by restrictive eating is an ideal inhibitor of microbiota overgrowth, as autophagy deprives microbiota of excessive nutrition for replication. Autophagy also attenuates inflammation. Therefore, as a precaution, the author suggests restoring good health in the elderly with the support from a diverse gut microbiome and daily regular food intake restriction, so as to lower the risk of developing into severe case even if they are infected by COVID-19.
Collapse
Affiliation(s)
- Ligen Yu
- grid.59025.3b0000 0001 2224 0361Talent Recruitment and Career Support (TRACS) Office and Bibliometrics Analysis, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
20
|
Rosa RB, Dantas WM, do Nascimento JCF, da Silva MV, de Oliveira RN, Pena LJ. In Vitro and In Vivo Models for Studying SARS-CoV-2, the Etiological Agent Responsible for COVID-19 Pandemic. Viruses 2021; 13:379. [PMID: 33673614 PMCID: PMC7997194 DOI: 10.3390/v13030379] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/19/2021] [Accepted: 01/31/2021] [Indexed: 02/07/2023] Open
Abstract
The emergence and rapid worldwide spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has prompted the scientific community to rapidly develop in vitro and in vivo models that could be applied in COVID-19 research. In vitro models include two-dimensional (2D) cultures of immortalized cell lines or primary cells and three-dimensional (3D) cultures derived from lung, alveoli, bronchi, and other organs. Although cell-based systems are economic and allow strict control of experimental variables, they do not always resemble physiological conditions. Thus, several in vivo models are being developed, including different strains of mice, hamsters, ferrets, dogs, cats, and non-human primates. In this review, we summarize the main models of SARS-CoV-2 infection developed so far and discuss their advantages, drawbacks and main uses.
Collapse
Affiliation(s)
- Rafael B. Rosa
- Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife 50740-465, Brazil; (R.B.R.); (J.C.F.d.N.)
- Rodents Animal Facilities Complex, Federal University of Uberlandia, Uberlandia 38400-902, Brazil;
| | - Willyenne M. Dantas
- Department of Chemistry, Federal Rural University of Pernambuco (UFRPE), Recife 52171-900, Brazil; (W.M.D.); (R.N.d.O.)
| | - Jessica C. F. do Nascimento
- Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife 50740-465, Brazil; (R.B.R.); (J.C.F.d.N.)
| | - Murilo V. da Silva
- Rodents Animal Facilities Complex, Federal University of Uberlandia, Uberlandia 38400-902, Brazil;
| | - Ronaldo N. de Oliveira
- Department of Chemistry, Federal Rural University of Pernambuco (UFRPE), Recife 52171-900, Brazil; (W.M.D.); (R.N.d.O.)
| | - Lindomar J. Pena
- Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife 50740-465, Brazil; (R.B.R.); (J.C.F.d.N.)
| |
Collapse
|
21
|
Hong S, Chang J, Jeong K, Lee W. Raloxifene as a treatment option for viral infections. J Microbiol 2021; 59:124-131. [PMID: 33527314 PMCID: PMC7849956 DOI: 10.1007/s12275-021-0617-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 01/31/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused corona virus disease 2019 (COVID-19) pandemic and led to mass casualty. Even though much effort has been put into development of vaccine and treatment methods to combat COVID-19, no safe and efficient cure has been discovered. Drug repurposing or drug repositioning which is a process of investigating pre-existing drug candidates for novel applications outside their original medical indication can speed up the drug development process. Raloxifene is a selective estrogen receptor modulator (SERM) that has been approved by FDA in 1997 for treatment and prevention of postmenopausal osteoporosis and cancer. Recently, raloxifene demonstrates efficacy in treating viral infections by Ebola, influenza A, and hepatitis C viruses and shows potential for drug repurposing for the treatment of SARS-CoV-2 infection. This review will provide an overview of raloxifene's mechanism of action as a SERM and present proposed mechanisms of action in treatment of viral infections.
Collapse
Affiliation(s)
- Subin Hong
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| | - JuOae Chang
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| | - Kwiwan Jeong
- Bio-center, Gyeonggido Business & Science Accelerator, Suwon, 16229 Republic of Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| |
Collapse
|
22
|
Lenze EJ, Mattar C, Zorumski CF, Stevens A, Schweiger J, Nicol GE, Miller JP, Yang L, Yingling M, Avidan MS, Reiersen AM. Fluvoxamine vs Placebo and Clinical Deterioration in Outpatients With Symptomatic COVID-19: A Randomized Clinical Trial. JAMA 2020; 324:2292-2300. [PMID: 33180097 PMCID: PMC7662481 DOI: 10.1001/jama.2020.22760] [Citation(s) in RCA: 372] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
IMPORTANCE Coronavirus disease 2019 (COVID-19) may lead to serious illness as a result of an excessive immune response. Fluvoxamine may prevent clinical deterioration by stimulating the σ-1 receptor, which regulates cytokine production. OBJECTIVE To determine whether fluvoxamine, given during mild COVID-19 illness, prevents clinical deterioration and decreases the severity of disease. DESIGN, SETTING, AND PARTICIPANTS Double-blind, randomized, fully remote (contactless) clinical trial of fluvoxamine vs placebo. Participants were community-living, nonhospitalized adults with confirmed severe acute respiratory syndrome coronavirus 2 infection, with COVID-19 symptom onset within 7 days and oxygen saturation of 92% or greater. One hundred fifty-two participants were enrolled from the St Louis metropolitan area (Missouri and Illinois) from April 10, 2020, to August 5, 2020. The final date of follow-up was September 19, 2020. INTERVENTIONS Participants were randomly assigned to receive 100 mg of fluvoxamine (n = 80) or placebo (n = 72) 3 times daily for 15 days. MAIN OUTCOMES AND MEASURES The primary outcome was clinical deterioration within 15 days of randomization defined by meeting both criteria of (1) shortness of breath or hospitalization for shortness of breath or pneumonia and (2) oxygen saturation less than 92% on room air or need for supplemental oxygen to achieve oxygen saturation of 92% or greater. RESULTS Of 152 patients who were randomized (mean [SD] age, 46 [13] years; 109 [72%] women), 115 (76%) completed the trial. Clinical deterioration occurred in 0 of 80 patients in the fluvoxamine group and in 6 of 72 patients in the placebo group (absolute difference, 8.7% [95% CI, 1.8%-16.4%] from survival analysis; log-rank P = .009). The fluvoxamine group had 1 serious adverse event and 11 other adverse events, whereas the placebo group had 6 serious adverse events and 12 other adverse events. CONCLUSIONS AND RELEVANCE In this preliminary study of adult outpatients with symptomatic COVID-19, patients treated with fluvoxamine, compared with placebo, had a lower likelihood of clinical deterioration over 15 days. However, the study is limited by a small sample size and short follow-up duration, and determination of clinical efficacy would require larger randomized trials with more definitive outcome measures. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04342663.
Collapse
Affiliation(s)
- Eric J. Lenze
- Department of Psychiatry, School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Caline Mattar
- Division of Infectious Diseases, Department of Internal Medicine, School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Charles F. Zorumski
- Department of Psychiatry, School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Angela Stevens
- Department of Psychiatry, School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Julie Schweiger
- Department of Psychiatry, School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Ginger E. Nicol
- Department of Psychiatry, School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - J. Philip Miller
- Division of Biostatistics, Informatics Institute, School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Lei Yang
- Department of Psychiatry, School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Michael Yingling
- Department of Psychiatry, School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Michael S. Avidan
- Department of Anesthesiology, School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Angela M. Reiersen
- Department of Psychiatry, School of Medicine, Washington University in St Louis, St Louis, Missouri
| |
Collapse
|
23
|
Xu Y, Kang L, Shen Z, Li X, Wu W, Ma W, Fang C, Yang F, Jiang X, Gong S, Zhang L, Li M. Dynamics of severe acute respiratory syndrome coronavirus 2 genome variants in the feces during convalescence. J Genet Genomics 2020; 47:610-617. [PMID: 33388272 PMCID: PMC7649052 DOI: 10.1016/j.jgg.2020.10.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/23/2020] [Accepted: 10/23/2020] [Indexed: 01/22/2023]
Abstract
In response to the current coronavirus disease 2019 (COVID-19) pandemic, it is crucial to understand the origin, transmission, and evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which relies on close surveillance of genomic diversity in clinical samples. Although the mutation at the population level had been extensively investigated, how the mutations evolve at the individual level is largely unknown. Eighteen time-series fecal samples were collected from nine patients with COVID-19 during the convalescent phase. The nucleic acids of SARS-CoV-2 were enriched by the hybrid capture method. First, we demonstrated the outstanding performance of the hybrid capture method in detecting intra-host variants. We identified 229 intra-host variants at 182 sites in 18 fecal samples. Among them, nineteen variants presented frequency changes > 0.3 within 1-5 days, reflecting highly dynamic intra-host viral populations. Moreover, the evolution of the viral genome demonstrated that the virus was probably viable in the gastrointestinal tract during the convalescent period. Meanwhile, we also found that the same mutation showed a distinct pattern of frequency changes in different individuals, indicating a strong random drift. In summary, dramatic changes of the SARS-CoV-2 genome were detected in fecal samples during the convalescent period; whether the viral load in feces is sufficient to establish an infection warranted further investigation.
Collapse
Affiliation(s)
- Yi Xu
- Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Lu Kang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 101300, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zijie Shen
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 101300, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xufang Li
- Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Weili Wu
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 101300, China
| | - Wentai Ma
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 101300, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunxiao Fang
- Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Fengxia Yang
- Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xuan Jiang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 101300, China
| | - Sitang Gong
- Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Li Zhang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 101300, China.
| | - Mingkun Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 101300, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| |
Collapse
|