151
|
Li S, Yang S, Zhou Y, Disoma C, Dong Z, Du A, Zhang Y, Chen Y, Huang W, Chen J, Song D, Chen Z, Liu P, Li S, Zheng R, Liu S, Razzaq A, Chen X, Tao S, Yu C, Feng T, Liao W, Peng Y, Jiang T, Huang J, Wu W, Hu L, Wang L, Li S, Xia Z. Microbiome Profiling Using Shotgun Metagenomic Sequencing Identified Unique Microorganisms in COVID-19 Patients With Altered Gut Microbiota. Front Microbiol 2021; 12:712081. [PMID: 34707577 PMCID: PMC8542975 DOI: 10.3389/fmicb.2021.712081] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/10/2021] [Indexed: 12/23/2022] Open
Abstract
COVID-19 is mainly associated with respiratory distress syndrome, but a subset of patients often present gastrointestinal (GI) symptoms. Imbalances of gut microbiota have been previously linked to respiratory virus infection. Understanding how the gut–lung axis affects the progression of COVID-19 can provide a novel framework for therapies and management. In this study, we examined the gut microbiota of patients with COVID-19 (n = 47) and compared it to healthy controls (n = 19). Using shotgun metagenomic sequencing, we have identified four microorganisms unique in COVID-19 patients, namely Streptococcus thermophilus, Bacteroides oleiciplenus, Fusobacterium ulcerans, and Prevotella bivia. The abundances of Bacteroides stercoris, B. vulgatus, B. massiliensis, Bifidobacterium longum, Streptococcus thermophilus, Lachnospiraceae bacterium 5163FAA, Prevotella bivia, Erysipelotrichaceae bacterium 6145, and Erysipelotrichaceae bacterium 2244A were enriched in COVID-19 patients, whereas the abundances of Clostridium nexile, Streptococcus salivarius, Coprococcus catus, Eubacterium hallii, Enterobacter aerogenes, and Adlercreutzia equolifaciens were decreased (p < 0.05). The relative abundance of butyrate-producing Roseburia inulinivorans is evidently depleted in COVID-19 patients, while the relative abundances of Paraprevotella sp. and the probiotic Streptococcus thermophilus were increased. We further identified 30 KEGG orthology (KO) modules overrepresented, with 7 increasing and 23 decreasing modules. Notably, 15 optimal microbial markers were identified using the random forest model to have strong diagnostic potential in distinguishing COVID-19. Based on Spearman’s correlation, eight species were associated with eight clinical indices. Moreover, the increased abundance of Bacteroidetes and decreased abundance of Firmicutes were also found across clinical types of COVID-19. Our findings suggest that the alterations of gut microbiota in patients with COVID-19 may influence disease severity. Our COVID-19 classifier, which was cross-regionally verified, provides a proof of concept that a set of microbial species markers can distinguish the presence of COVID-19.
Collapse
Affiliation(s)
- Sijia Li
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Siyuan Yang
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yuzheng Zhou
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Cyrollah Disoma
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Zijun Dong
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Ashuai Du
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Yongxing Zhang
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Yong Chen
- The First Hospital of Changsha, Changsha, China
| | | | - Junru Chen
- Suzhou Geneworks Technology Co., Ltd., Suzhou, China
| | - Deqiang Song
- Suzhou Geneworks Technology Co., Ltd., Suzhou, China
| | - Zongpeng Chen
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Pinjia Liu
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Shiqin Li
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Rong Zheng
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Sixu Liu
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Aroona Razzaq
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Xuan Chen
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Siyi Tao
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Chengping Yu
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Tianxu Feng
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Wenyan Liao
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Yousong Peng
- Hunan Provincial Key Laboratory of Medical Virology, Bioinformatics Center, College of Biology, Hunan University, Changsha, China
| | - Taijiao Jiang
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jufang Huang
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Wei Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liqiang Hu
- The First Hospital of Changsha, Changsha, China
| | - Linghang Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Shanni Li
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Zanxian Xia
- Hunan Key Laboratory of Animal Models for Human Diseases, Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
152
|
Zhu Z, Zhang S, Wang P, Chen X, Bi J, Cheng L, Zhang X. A comprehensive review of the analysis and integration of omics data for SARS-CoV-2 and COVID-19. Brief Bioinform 2021; 23:6412396. [PMID: 34718395 PMCID: PMC8574485 DOI: 10.1093/bib/bbab446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/06/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022] Open
Abstract
Since the first report of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in December 2019, over 100 million people have been infected by COVID-19, millions of whom have died. In the latest year, a large number of omics data have sprung up and helped researchers broadly study the sequence, chemical structure and function of SARS-CoV-2, as well as molecular abnormal mechanisms of COVID-19 patients. Though some successes have been achieved in these areas, it is necessary to analyze and mine omics data for comprehensively understanding SARS-CoV-2 and COVID-19. Hence, we reviewed the current advantages and limitations of the integration of omics data herein. Firstly, we sorted out the sequence resources and database resources of SARS-CoV-2, including protein chemical structure, potential drug information and research literature resources. Next, we collected omics data of the COVID-19 hosts, including genomics, transcriptomics, microbiology and potential drug information data. And subsequently, based on the integration of omics data, we summarized the existing data analysis methods and the related research results of COVID-19 multi-omics data in recent years. Finally, we put forward SARS-CoV-2 (COVID-19) multi-omics data integration research direction and gave a case study to mine deeper for the disease mechanisms of COVID-19.
Collapse
Affiliation(s)
- Zijun Zhu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China, 150081
| | - Sainan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China, 150081
| | - Ping Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China, 150081
| | - Xinyu Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China, 150081
| | - Jianxing Bi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China, 150081
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China, 150081.,NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang, China, 150028
| | - Xue Zhang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang, China, 150028.,McKusick-Zhang Center for Genetic Medicine, Peking Union Medical College, Beijing, China, 100005
| |
Collapse
|
153
|
Haran JP, Bradley E, Zeamer AL, Cincotta L, Salive MC, Dutta P, Mutaawe S, Anya O, Meza-Segura M, Moormann AM, Ward DV, McCormick BA, Bucci V. Inflammation-type dysbiosis of the oral microbiome associates with the duration of COVID-19 symptoms and long COVID. JCI Insight 2021; 6:e152346. [PMID: 34403368 PMCID: PMC8564890 DOI: 10.1172/jci.insight.152346] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
In the COVID-19 pandemic, caused by SARS-CoV-2, many individuals experience prolonged symptoms, termed long-lasting COVID-19 symptoms (long COVID). Long COVID is thought to be linked to immune dysregulation due to harmful inflammation, with the exact causes being unknown. Given the role of the microbiome in mediating inflammation, we aimed to examine the relationship between the oral microbiome and the duration of long COVID symptoms. Tongue swabs were collected from patients presenting with COVID-19 symptoms. Confirmed infections were followed until resolution of all symptoms. Bacterial composition was determined by metagenomic sequencing. We used random forest modeling to identify microbiota and clinical covariates that are associated with long COVID symptoms. Of the patients followed, 63% developed ongoing symptomatic COVID-19 and 37% went on to long COVID. Patients with prolonged symptoms had significantly higher abundances of microbiota that induced inflammation, such as members of the genera Prevotella and Veillonella, which, of note, are species that produce LPS. The oral microbiome of patients with long COVID was similar to that of patients with chronic fatigue syndrome. Altogether, our findings suggest an association with the oral microbiome and long COVID, revealing the possibility that dysfunction of the oral microbiome may have contributed to this draining disease.
Collapse
Affiliation(s)
- John P Haran
- Department of Emergency Medicine.,Department of Microbiology and Physiological Systems.,Program in Microbiome Dynamics, and
| | - Evan Bradley
- Department of Emergency Medicine.,Program in Microbiome Dynamics, and
| | - Abigail L Zeamer
- Department of Microbiology and Physiological Systems.,Program in Microbiome Dynamics, and
| | | | | | | | | | | | | | - Ann M Moormann
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Doyle V Ward
- Department of Microbiology and Physiological Systems.,Program in Microbiome Dynamics, and
| | - Beth A McCormick
- Department of Microbiology and Physiological Systems.,Program in Microbiome Dynamics, and
| | - Vanni Bucci
- Department of Microbiology and Physiological Systems.,Program in Microbiome Dynamics, and
| |
Collapse
|
154
|
Hussain I, Cher GLY, Abid MA, Abid MB. Role of Gut Microbiome in COVID-19: An Insight Into Pathogenesis and Therapeutic Potential. Front Immunol 2021; 12:765965. [PMID: 34721437 PMCID: PMC8551858 DOI: 10.3389/fimmu.2021.765965] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), resulted in an unprecedented global crisis. Although primarily a respiratory illness, dysregulated immune responses may lead to multi-organ dysfunction. Prior data showed that the resident microbial communities of gastrointestinal and respiratory tracts act as modulators of local and systemic inflammatory activity (the gut-lung axis). Evolving evidence now signals an alteration in the gut microbiome, brought upon either by cytokines from the infected respiratory tract or from direct infection of the gut, or both. Dysbiosis leads to a "leaky gut". The intestinal permeability then allows access to bacterial products and toxins into the circulatory system and further exacerbates the systemic inflammatory response. In this review, we discuss the available data related to the role of the gut microbiome in the development and progression of COVID-19. We provide mechanistic insights into early data with a focus on immunological crosstalk and the microbiome's potential as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Ikram Hussain
- Department of Gastroenterology, Khoo Teck Puat Hospital, Singapore, Singapore
| | | | - Muhammad Abbas Abid
- Department of Hematopathology and Microbiology, Aga Khan University Hospital, Karachi, Pakistan
| | - Muhammad Bilal Abid
- Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
155
|
Hilpert K, Mikut R. Is There a Connection Between Gut Microbiome Dysbiosis Occurring in COVID-19 Patients and Post-COVID-19 Symptoms? Front Microbiol 2021; 12:732838. [PMID: 34603261 PMCID: PMC8485028 DOI: 10.3389/fmicb.2021.732838] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/13/2021] [Indexed: 01/07/2023] Open
Affiliation(s)
- Kai Hilpert
- Institute of Infection and Immunology, St George's, University of London, London, United Kingdom
| | - Ralf Mikut
- Institute for Automation and Applied Informatics (IAI), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
156
|
Das G, Heredia JB, de Lourdes Pereira M, Coy-Barrera E, Rodrigues Oliveira SM, Gutiérrez-Grijalva EP, Cabanillas-Bojórquez LA, Shin HS, Patra JK. Korean traditional foods as antiviral and respiratory disease prevention and treatments: A detailed review. Trends Food Sci Technol 2021; 116:415-433. [PMID: 34345117 PMCID: PMC8321624 DOI: 10.1016/j.tifs.2021.07.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Korean traditional food (KTF), originated from ancestral agriculture and the nomadic traditions of the Korean peninsula and southern Manchuria, is based on healthy food that balances disease prevention and treatment. Fermented foods that include grains, herbs, fruits, and mushrooms are also an important practice in KTF, providing high levels of Lactobacilli, which confer relevant health benefits, including antiviral properties. Some of these probiotics may also protect against the Influenza virus through the modulation of innate immunity. SCOPE AND APPROACH The emerging of the COVID-19 pandemic, in addition to other diseases of viral origin, and the problems associated with other respiratory disorders, highlight how essential is a healthy eating pattern to strengthen our immune system.Key Findings and Conclusions: The present review covers the information available on edible plants, herbs, mushrooms, and preparations used in KTF to outline their multiple medicinal effects (e.g., antidiabetic, chemopreventive, antioxidative, anti-inflammatory, antibacterial), emphasizing their role and effects on the immune system with an emphasis on modulating properties of the gut microbiota that further support strong respiratory immunity. Potential functional foods commonly used in Korean cuisine such as Kimchi (a mixture of fermented vegetables), Meju, Doenjang, Jeotgal, and Mekgeolli and fermented sauces, among others, are highlighted for their great potential to improve gut-lung immunity. The traditional Korean diet and dietary mechanisms that may target viruses ACE-2 receptors or affect any step of a virus infection pathway that can determine a patient's prognosis are also highlighted. The regular oral intake of bioactive ingredients used in Korean foods can offer protection for some viral diseases, through protective and immunomodulatory effects, as evidenced in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Gitishree Das
- Research Institute of Biotechnology & Medical Converged Science, Dongguk University-Seoul, Goyangsi, South Korea
| | - J Basilio Heredia
- Centro de Investigación en Alimentación y Desarrollo, A.C., Carretera a Eldorado Km. 5.5, Col. Campo El Diez, CP. 80110, Culiacán, Sinaloa, Mexico
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ericsson Coy-Barrera
- Bioorganic Chemistry Laboratory, Facultad de Ciencias Básicas y Aplicadas, Universidad Militar Nueva Granada, Campus Nueva Granada, 250247, Cajicá, Colombia
| | - Sonia Marlene Rodrigues Oliveira
- CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
- HMRI and Hunter Cancer Research Alliance Centres, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Erick Paul Gutiérrez-Grijalva
- Catedras CONACYT-Centro de Investigación en Alimentación y Desarrollo, A.C., Carretera a Eldorado Km. 5.5, Col. Campo El Diez, CP. 80110 Culiacán, Sinaloa, Mexico
| | - Luis Angel Cabanillas-Bojórquez
- Centro de Investigación en Alimentación y Desarrollo, A.C., Carretera a Eldorado Km. 5.5, Col. Campo El Diez, CP. 80110, Culiacán, Sinaloa, Mexico
| | - Han-Seung Shin
- Department of Food Science & Biotechnology, Dongguk University-Seoul, Goyangsi, South Korea
| | - Jayanta Kumar Patra
- Research Institute of Biotechnology & Medical Converged Science, Dongguk University-Seoul, Goyangsi, South Korea
| |
Collapse
|
157
|
Wang X, Xu G, Liu X, Liu Y, Zhang S, Zhang Z. Multiomics: unraveling the panoramic landscapes of SARS-CoV-2 infection. Cell Mol Immunol 2021; 18:2313-2324. [PMID: 34471261 PMCID: PMC8408367 DOI: 10.1038/s41423-021-00754-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/30/2021] [Indexed: 02/07/2023] Open
Abstract
In response to emerging infectious diseases, such as the recent pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), it is critical to quickly identify and understand responsible pathogens, risk factors, host immune responses, and pathogenic mechanisms at both the molecular and cellular levels. The recent development of multiomic technologies, including genomics, proteomics, metabolomics, and single-cell transcriptomics, has enabled a fast and panoramic grasp of the pathogen and the disease. Here, we systematically reviewed the major advances in the virology, immunology, and pathogenic mechanisms of SARS-CoV-2 infection that have been achieved via multiomic technologies. Based on well-established cohorts, omics-based methods can greatly enhance the mechanistic understanding of diseases, contributing to the development of new diagnostics, drugs, and vaccines for emerging infectious diseases, such as COVID-19.
Collapse
Affiliation(s)
- Xin Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Gang Xu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Xiaoju Liu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Yang Liu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Shuye Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China.
- Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, Guangdong Province, China.
- Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong Province, China.
- Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China.
| |
Collapse
|
158
|
Natarajan A, Han A, Zlitni S, Brooks EF, Vance SE, Wolfe M, Singh U, Jagannathan P, Pinsky BA, Boehm A, Bhatt AS. Standardized preservation, extraction and quantification techniques for detection of fecal SARS-CoV-2 RNA. Nat Commun 2021; 12:5753. [PMID: 34599164 PMCID: PMC8486790 DOI: 10.1038/s41467-021-25576-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/18/2021] [Indexed: 12/28/2022] Open
Abstract
Patients with COVID-19 shed SARS-CoV-2 RNA in stool, sometimes well after their respiratory infection has cleared. This may be significant for patient health, epidemiology, and diagnosis. However, methods to preserve stool, and to extract and quantify viral RNA are not standardized. We test the performance of three preservative approaches at yielding detectable SARS-CoV-2 RNA: the OMNIgene-GUT kit, Zymo DNA/RNA shield kit, and the most commonly applied, storage without preservative. We test these in combination with three extraction kits: QIAamp Viral RNA Mini Kit, Zymo Quick-RNA Viral Kit, and MagMAX Viral/Pathogen Kit. We also test the utility of ddPCR and RT-qPCR for the reliable quantification of SARS-CoV-2 RNA from stool. We identify that the Zymo DNA/RNA preservative and the QiaAMP extraction kit yield more detectable RNA than the others, using both ddPCR and RT-qPCR. Taken together, we recommend a comprehensive methodology for preservation, extraction and detection of RNA from SARS-CoV-2 and other coronaviruses in stool.
Collapse
Affiliation(s)
- Aravind Natarajan
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Medicine (Hematology, Blood and Marrow Transplantation), Stanford University, Stanford, CA, USA
| | - Alvin Han
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Soumaya Zlitni
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Medicine (Hematology, Blood and Marrow Transplantation), Stanford University, Stanford, CA, USA
| | - Erin F Brooks
- Department of Medicine (Hematology, Blood and Marrow Transplantation), Stanford University, Stanford, CA, USA
| | - Summer E Vance
- Department of Medicine (Hematology, Blood and Marrow Transplantation), Stanford University, Stanford, CA, USA
| | - Marlene Wolfe
- Department of Civil and Environmental Engineering, Stanford University, Stanford, CA, USA
| | - Upinder Singh
- Department of Medicine (Infectious Diseases and Geographic Medicine), Stanford University, Stanford, CA, USA
| | - Prasanna Jagannathan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Department of Medicine (Infectious Diseases and Geographic Medicine), Stanford University, Stanford, CA, USA
| | - Benjamin A Pinsky
- Department of Medicine (Infectious Diseases and Geographic Medicine), Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Alexandria Boehm
- Department of Civil and Environmental Engineering, Stanford University, Stanford, CA, USA
| | - Ami S Bhatt
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Department of Medicine (Hematology, Blood and Marrow Transplantation), Stanford University, Stanford, CA, USA.
| |
Collapse
|
159
|
Microbiota and Its Impact on the Immune System in COVID-19-A Narrative Review. J Clin Med 2021; 10:jcm10194537. [PMID: 34640553 PMCID: PMC8509181 DOI: 10.3390/jcm10194537] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 01/08/2023] Open
Abstract
The microbiota is of interest for the development of a therapeutic strategy against SARS-CoV-2 coronavirus disease 2019 (COVID-19) due to its impact on the host immune system. Proven communications of the gut microbiota with the pulmonary microbiota (gut-lung axis) and the pathway of neural connections between the gut and brain (gut-brain axis) may be important in the face of the pandemic. SARS-CoV-2 was shown to affect almost all organs because of the presence of a host receptor known as angiotensin converting enzyme 2 (ACE2). The ACE2 receptor is mainly present in the brush border of intestinal enterocytes, ciliary cells, and type II alveolar epithelial cells in the lungs. The transport function of ACE2 has been linked to the ecology of gut microbes in the digestive tract, suggesting that COVID-19 may be related to the gut microbiota. The severity of COVID-19 may be associated with a number of comorbidities, such as hypertension, diabetes, obesity, and/or old age; therefore, attention is also paid to multiple morbidities and the modulation of microbiota through comorbidities and medications. This paper reviews the research in the context of the state of the intestinal microbiota and its impact on the cells of the immune system during the SARS-CoV-2 pandemic.
Collapse
|
160
|
Hawryłkowicz V, Lietz-Kijak D, Kaźmierczak-Siedlecka K, Sołek-Pastuszka J, Stachowska L, Folwarski M, Parczewski M, Stachowska E. Patient Nutrition and Probiotic Therapy in COVID-19: What Do We Know in 2021? Nutrients 2021; 13:3385. [PMID: 34684384 PMCID: PMC8538178 DOI: 10.3390/nu13103385] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The main nutritional consequences of COVID-19 include reduced food intake, hypercatabolism, and rapid muscle wasting. Some studies showed that malnutrition is a significant problem among patients hospitalized due to COVID-19 infection, and the outcome of patients with SARS-CoV-2 is strongly associated with their nutritional status. The purpose of this study was to collect useful information about the possible elements of nutritional and probiotic therapy in patients infected with the SARS-CoV-2 virus. METHODS A narrative review of the literature, including studies published up to 13 September 2021. RESULTS Probiotics may support patients by inhibiting the ACE2 receptor, i.e., the passage of the virus into the cell, and may also be effective in suppressing the immune response caused by the proinflammatory cytokine cascade. In patients' diet, it is crucial to ensure an adequate intake of micronutrients, such as omega-3 fatty acids (at 2-4 g/d), selenium (300-450 μg/d) and zinc (30-50 mg/d), and vitamins A (900-700 µg/d), E (135 mg/d), D (20,000-50,000 IU), C (1-2 g/d), B6, and B12. Moreover, the daily calorie intake should amount to ≥1500-2000 with 75-100 g of protein. CONCLUSION In conclusion, the treatment of gut dysbiosis involving an adequate intake of prebiotic dietary fiber and probiotics could turn out to be an immensely helpful instrument for immunomodulation, both in COVID-19 patients and prophylactically in individuals with no history of infection.
Collapse
Affiliation(s)
- Viktoria Hawryłkowicz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (L.S.)
| | - Danuta Lietz-Kijak
- Department of Propedeutics, Physiodiagnostics and Dental Physiotherapy, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | | | - Joanna Sołek-Pastuszka
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University, 71-242 Szczecin, Poland;
| | - Laura Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (L.S.)
| | - Marcin Folwarski
- Department of Clinical Nutrition, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Miłosz Parczewski
- Department of Infectious, Tropical and Acquired Immunological Diseases, Pomeranian Medical University, 71-455 Szczecin, Poland;
| | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (L.S.)
| |
Collapse
|
161
|
Cruz CS, Ricci MF, Vieira AT. Gut Microbiota Modulation as a Potential Target for the Treatment of Lung Infections. Front Pharmacol 2021; 12:724033. [PMID: 34557097 PMCID: PMC8453009 DOI: 10.3389/fphar.2021.724033] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal and respiratory systems are colonized by a complex ecosystem of microorganisms called the microbiota. These microorganisms co-evolved over millions of years with the host, creating a symbiotic relationship that is fundamental for promoting host homeostasis by producing bioactive metabolites and antimicrobial molecules, and regulating the immune and inflammatory responses. Imbalance in the abundance, diversity, and function of the gut microbiota (known as dysbiosis) have been shown to increase host susceptibility to infections in the lungs, suggesting crosstalk between these organs. This crosstalk is now referred to as the gut-lung axis. Hence, the use of probiotics, prebiotics, and synbiotics for modulation of gut microbiota has been studied based on their effectiveness in reducing the duration and severity of respiratory tract infections, mainly owing to their effects on preventing pathogen colonization and modulating the immune system. This review discusses the role and responses of probiotics, prebiotics, and synbiotics in the gut-lung axis in the face of lung infections.
Collapse
Affiliation(s)
- Clênio Silva Cruz
- Laboratory of Microbiota and Immunomodulation (LMI), Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mayra Fernanda Ricci
- Laboratory of Microbiota and Immunomodulation (LMI), Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Angélica Thomaz Vieira
- Laboratory of Microbiota and Immunomodulation (LMI), Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
162
|
Molnar T, Varnai R, Schranz D, Zavori L, Peterfi Z, Sipos D, Tőkés-Füzesi M, Illes Z, Buki A, Csecsei P. Severe Fatigue and Memory Impairment Are Associated with Lower Serum Level of Anti-SARS-CoV-2 Antibodies in Patients with Post-COVID Symptoms. J Clin Med 2021; 10:jcm10194337. [PMID: 34640355 PMCID: PMC8509483 DOI: 10.3390/jcm10194337] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Post-COVID manifestation is defined as persistent symptoms or long-term complications beyond 4 weeks from disease onset. Fatigue and memory impairment are common post-COVID symptoms. We aimed to explore associations between the timeline and severity of post-COVID fatigue and anti-SARS-CoV-2 antibodies. Methods: Fatigue and memory impairment were assessed in a total of 101 post-COVID subjects using the Chalder fatigue scale (CFQ-11) and a visual analogue scale. Using the bimodal scoring system generated from CFQ-11, a score ≥4 was defined as severe fatigue. Serum anti-SARS-CoV-2 spike (anti-S-Ig) and nucleocapsid (anti-NC-Ig) antibodies were examined at two time points: 4–12 weeks after onset of symptoms, and beyond 12 weeks. Results: The serum level of anti-S-Ig was significantly higher in patients with non-severe fatigue compared to those with severe fatigue at 4–12 weeks (p = 0.006) and beyond 12 weeks (p = 0.016). The serum level of anti-NC-Ig remained high in patients with non-severe fatigue at both time points. In contrast, anti-NC-Ig decreased significantly in severe fatigue cases regardless of the elapsed time (4–12 weeks: p = 0.024; beyond 12 weeks: p = 0.005). The incidence of memory impairment was significantly correlated with lower anti-S-Ig levels (−0.359, p < 0.001). Conclusion: The systemic immune response reflected by antibodies to SARS-CoV-2 is strongly correlated with the severity of post-COVID fatigue.
Collapse
Affiliation(s)
- Tihamer Molnar
- Department of Anaesthesiology and Intensive Care, University of Pecs, Medical School, H7632 Pecs, Hungary;
| | - Reka Varnai
- Department of Primary Health Care, University of Pecs, Medical School, H7632 Pecs, Hungary
- Correspondence: ; Tel.: +36-72535900
| | - Daniel Schranz
- Department of Neurology, University of Pecs, Medical School, H7632 Pecs, Hungary;
| | - Laszlo Zavori
- Salisbury NHS Foundation Trust, Salisbury SP2 8BJ, UK;
| | - Zoltan Peterfi
- 1st Department of Internal Medicine, Division of Infectology, University of Pecs, Medical School, H7632 Pecs, Hungary; (Z.P.); (D.S.)
| | - David Sipos
- 1st Department of Internal Medicine, Division of Infectology, University of Pecs, Medical School, H7632 Pecs, Hungary; (Z.P.); (D.S.)
| | - Margit Tőkés-Füzesi
- Department of Laboratory Medicine, University of Pecs, Medical School, H7632 Pecs, Hungary;
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, University of Southern Denmark, 5230 Odense, Denmark;
| | - Andras Buki
- Department of Neurosurgery, University of Pecs, Medical School, H7632 Pecs, Hungary; (A.B.); (P.C.)
| | - Peter Csecsei
- Department of Neurosurgery, University of Pecs, Medical School, H7632 Pecs, Hungary; (A.B.); (P.C.)
| |
Collapse
|
163
|
Zuo T, Wu X, Wen W, Lan P. Gut Microbiome Alterations in COVID-19. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:679-688. [PMID: 34560321 PMCID: PMC8478109 DOI: 10.1016/j.gpb.2021.09.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022]
Abstract
Since the outset of the coronavirus disease 2019 (COVID-19) pandemic, the gut microbiome in COVID-19 has garnered substantial interest, given its significant roles in human health and pathophysiology. Accumulating evidence is unveiling that the gut microbiome is broadly altered in COVID-19, including the bacterial microbiome, mycobiome, and virome. Overall, the gut microbial ecological network is significantly weakened and becomes sparse in patients with COVID-19, together with a decrease in gut microbiome diversity. Beyond the existence of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), the gut microbiome of patients with COVID-19 is also characterized by enrichment of opportunistic bacteria, fungi, and eukaryotic viruses, which are also associated with disease severity and presentation. Meanwhile, a multitude of symbiotic bacteria and bacteriophages are decreased in abundance in patients with COVID-19. Such gut microbiome features persist in a significant subset of patients with COVID-19 even after disease resolution, coinciding with ‘long COVID’ (also known as post-acute sequelae of COVID-19). The broadly-altered gut microbiome is largely a consequence of SARS-CoV-2infection and its downstream detrimental effects on the systemic host immunity and the gut milieu. The impaired host immunity and distorted gut microbial ecology, particularly loss of low-abundance beneficial bacteria and blooms of opportunistic fungi including Candida, may hinder the reassembly of the gut microbiome post COVID-19. Future investigation is necessary to fully understand the role of the gut microbiome in host immunity against SARS-CoV-2 infection, as well as the long-term effect of COVID-19 on the gut microbiome in relation to the host health after the pandemic.
Collapse
Affiliation(s)
- Tao Zuo
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China; Center for Fecal Microbiota Transplantation Research, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China.
| | - Xiaojian Wu
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China; Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China.
| | - Weiping Wen
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, China; Guangzhou Key Laboratory of Otorhinolaryngology, Guangzhou 510080, China; Department of Otorhinolaryngology, Head and Neck Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China.
| | - Ping Lan
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China; Center for Fecal Microbiota Transplantation Research, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China; Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China.
| |
Collapse
|
164
|
Piccioni A, Saviano A, Cicchinelli S, Franza L, Rosa F, Zanza C, Santoro MC, Candelli M, Covino M, Nannini G, Amedei A, Franceschi F. Microbiota and Myopericarditis: The New Frontier in the Car-Diological Field to Prevent or Treat Inflammatory Cardiomyo-Pathies in COVID-19 Outbreak. Biomedicines 2021; 9:1234. [PMID: 34572420 PMCID: PMC8468627 DOI: 10.3390/biomedicines9091234] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 02/05/2023] Open
Abstract
Myopericarditis is an inflammatory heart condition involving the pericardium and myocardium. It can lead to heart failure, dilated cardiomyopathy, arrhythmia and sudden death. Its pathogenesis is mainly mediated by viral infections but also can be induced by bacterial infections, toxic substances and immune mediated disorders. All these conditions can produce severe inflammation and myocardial injury, often associated with a poor prognosis. The specific roles of these different pathogens (in particular viruses), the interaction with the host, the interplay with gut microbiota, and the immune system responses to them are still not completely clear and under investigation. Interestingly, some research has demonstrated the contribution of the gut microbiota, and its related metabolites (some of which can mimic the cardiac myosin), in cardiac inflammation and in the progression of this disease. They can stimulate a continuous and inadequate immune response, with a subsequent myocardial inflammatory damage. The aim of our review is to investigate the role of gut microbiota in myopericarditis, especially for the cardiovascular implications of COVID-19 viral infection, based on the idea that the modulation of gut microbiota can be a new frontier in the cardiological field to prevent or treat inflammatory cardiomyopathies.
Collapse
Affiliation(s)
- Andrea Piccioni
- Department of Emergency Medicine, Fondazione Policlinico Universitario, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (A.P.); (S.C.); (L.F.); (F.R.); (M.C.S.); (M.C.); (M.C.); (F.F.)
| | - Angela Saviano
- Department of Emergency Medicine, Fondazione Policlinico Universitario, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (A.P.); (S.C.); (L.F.); (F.R.); (M.C.S.); (M.C.); (M.C.); (F.F.)
| | - Sara Cicchinelli
- Department of Emergency Medicine, Fondazione Policlinico Universitario, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (A.P.); (S.C.); (L.F.); (F.R.); (M.C.S.); (M.C.); (M.C.); (F.F.)
| | - Laura Franza
- Department of Emergency Medicine, Fondazione Policlinico Universitario, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (A.P.); (S.C.); (L.F.); (F.R.); (M.C.S.); (M.C.); (M.C.); (F.F.)
| | - Federico Rosa
- Department of Emergency Medicine, Fondazione Policlinico Universitario, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (A.P.); (S.C.); (L.F.); (F.R.); (M.C.S.); (M.C.); (M.C.); (F.F.)
| | - Christian Zanza
- Department of Anesthesia, Critical Care and Emergency Medicine, Pietro and Michele Ferrero Hospital, Foundation Nuovo-Ospedale Alba-Bra, 12060 Verduno, Italy;
| | - Michele Cosimo Santoro
- Department of Emergency Medicine, Fondazione Policlinico Universitario, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (A.P.); (S.C.); (L.F.); (F.R.); (M.C.S.); (M.C.); (M.C.); (F.F.)
| | - Marcello Candelli
- Department of Emergency Medicine, Fondazione Policlinico Universitario, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (A.P.); (S.C.); (L.F.); (F.R.); (M.C.S.); (M.C.); (M.C.); (F.F.)
| | - Marcello Covino
- Department of Emergency Medicine, Fondazione Policlinico Universitario, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (A.P.); (S.C.); (L.F.); (F.R.); (M.C.S.); (M.C.); (M.C.); (F.F.)
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Firenze, 50134 Firenze, Italy; (G.N.); (A.A.)
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, 50134 Firenze, Italy; (G.N.); (A.A.)
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (A.P.); (S.C.); (L.F.); (F.R.); (M.C.S.); (M.C.); (M.C.); (F.F.)
| |
Collapse
|
165
|
Chen F, Dai Z, Huang C, Chen H, Wang X, Li X. Gastrointestinal Disease and COVID-19: A Review of Current Evidence. Dig Dis 2021; 40:506-514. [PMID: 34510032 PMCID: PMC8678221 DOI: 10.1159/000519412] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/30/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has had an unprecedented and catastrophic impact on humanity and continues to progress. In addition to typical respiratory symptoms such as fever, cough, and dyspnea, a large percentage of COVID-19 patients experience gastrointestinal (GI) complaints, with the most common symptoms being diarrhea, nausea, vomiting, and abdominal discomfort. SUMMARY We comprehensively summarize the latest knowledge of the adverse effects of COVID-19 and therapeutic drugs on the GI system, as well as related disease pathogenesis, and then provide a discussion focusing on the management and vaccination of patients who have inflammatory bowel disease (IBD) and GI cancer. The virus can affect the digestive system via binding to ACE2 receptors and subsequent gut microbiome dysbiosis. Through a variety of molecular pathways and mechanisms, numerous drugs for the treatment of COVID-19 could interfere with GI function and lead to multiple clinical manifestations, which may further intensify the risk and severity of GI symptoms of COVID-19 infection, such as nausea, vomiting, gastroparesis, and gastric ulcers. KEY MESSAGES We should monitor GI manifestations closely while managing COVID-19 patients and take timely measures to reduce the incidence of SARS-CoV-2 infections in GI cancer patients. IBD patients should receive vaccination timely, but corticosteroid use should be minimized when they are vaccinated. Simultaneously, for persons with IBD who have known or suspected COVID-19, immunosuppressive agents, especially thiopurines, should be avoided/minimized if possible.
Collapse
|
166
|
Zhang S, Luo P, Xu J, Yang L, Ma P, Tan X, Chen Q, Zhou M, Song S, Xia H, Wang S, Ma Y, Yang F, Liu Y, Li Y, Ma G, Wang Z, Duan Y, Jin Y. Plasma Metabolomic Profiles in Recovered COVID-19 Patients without Previous Underlying Diseases 3 Months After Discharge. J Inflamm Res 2021; 14:4485-4501. [PMID: 34522117 PMCID: PMC8434912 DOI: 10.2147/jir.s325853] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND It remains unclear whether discharged COVID-19 patients have fully recovered from severe complications, including the differences in the post-infection metabolomic profiles of patients with different disease severities. METHODS COVID-19-recovered patients, who had no previous underlying diseases and were discharged from Wuhan Union Hospital for 3 months, and matched healthy controls (HCs) were recruited in this prospective cohort study. We examined the blood biochemical indicators, cytokines, lung computed tomography scans, including 39 HCs, 18 recovered asymptomatic (RAs), 34 recovered moderate (RMs), and 44 recovered severe/ critical patients (RCs). A liquid chromatography-mass spectrometry-based metabolomics approach was employed to profile the global metabolites of fasting plasma of these participants. RESULTS Clinical data and metabolomic profiles suggested that RAs recovered well, but some clinical indicators and plasma metabolites in RMs and RCs were still abnormal as compared with HCs, such as decreased taurine, succinic acid, hippuric acid, some indoles, and lipid species. The disturbed metabolic pathway mainly involved the tricarboxylic cycle, purine, and glycerophospholipid metabolism. Moreover, metabolite alterations differ between RMs and RCs when compared with HCs. Correlation analysis revealed that many differential metabolites were closely associated with inflammation and the renal, pulmonary, heart, hepatic, and coagulation system functions. CONCLUSION We uncovered metabolite clusters pathologically relevant to the recovery state in discharged COVID-19 patients which may provide new insights into the pathogenesis of potential organ damage in recovered patients.
Collapse
Affiliation(s)
- Shujing Zhang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Ping Luo
- Department of Translational Medicine Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Juanjuan Xu
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Pei Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Xueyun Tan
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Qing Chen
- Health Checkup Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Mei Zhou
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Siwei Song
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Hui Xia
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Sufei Wang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Yanling Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Fan Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Yu Liu
- Health Checkup Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Yumei Li
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Guanzhou Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Zhihui Wang
- Department of Scientific Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Yanran Duan
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People’s Republic of China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| |
Collapse
|
167
|
Li T, Garcia-Gutierrez E, Yara DA, Scadden J, Davies J, Hutchins C, Aydin A, O'Grady J, Narbad A, Romano S, Sayavedra L. An optimised protocol for detection of SARS-CoV-2 in stool. BMC Microbiol 2021; 21:242. [PMID: 34488633 PMCID: PMC8419809 DOI: 10.1186/s12866-021-02297-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/18/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND SARS-CoV-2 has been detected in stool samples of COVID-19 patients, with potential implications for faecal-oral transmission. Compared to nasopharyngeal swab samples, the complexity of the stool matrix poses a challenge in the detection of the virus that has not yet been solved. However, robust and reliable methods are needed to estimate the prevalence and persistence of SARS-CoV-2 in the gut and to ensure the safety of microbiome-based procedures such as faecal microbiota transplant (FMT). The aim of this study was to establish a sensitive and reliable method for detecting SARS-CoV-2 in stool samples. RESULTS Stool samples from individuals free of SARS-CoV-2 were homogenised in saline buffer and spiked with a known titre of inactivated virus ranging from 50 to 750 viral particles per 100 mg stool. Viral particles were concentrated by ultrafiltration, RNA was extracted, and SARS-CoV-2 was detected via real-time reverse-transcription polymerase chain reaction (RT-qPCR) using the CDC primers and probes. The RNA extraction procedure we used allowed for the detection of SARS-CoV-2 via RT-qPCR in most of the stool samples tested. We could detect as few as 50 viral particles per 100 mg of stool. However, high variability was observed across samples at low viral titres. The primer set targeting the N1 region provided more reliable and precise results and for this primer set our method had a limit of detection of 1 viral particle per mg of stool. CONCLUSIONS Here we describe a sensitive method for detecting SARS-CoV-2 in stool samples. This method can be used to establish the persistence of SARS-CoV-2 in stool and ensure the safety of clinical practices such as FMT.
Collapse
Affiliation(s)
- Tianqi Li
- Gut Health and Microbes, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | | | - Daniel A Yara
- Gut Health and Microbes, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Jacob Scadden
- Gut Health and Microbes, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Jade Davies
- Gut Health and Microbes, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Chloe Hutchins
- Gut Health and Microbes, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Alp Aydin
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Justin O'Grady
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Arjan Narbad
- Gut Health and Microbes, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Stefano Romano
- Gut Health and Microbes, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
| | - Lizbeth Sayavedra
- Gut Health and Microbes, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
| |
Collapse
|
168
|
Seibert B, Cáceres CJ, Cardenas-Garcia S, Carnaccini S, Geiger G, Rajao DS, Ottesen E, Perez DR. Mild and Severe SARS-CoV-2 Infection Induces Respiratory and Intestinal Microbiome Changes in the K18-hACE2 Transgenic Mouse Model. Microbiol Spectr 2021; 9:e0053621. [PMID: 34378965 PMCID: PMC8455067 DOI: 10.1128/spectrum.00536-21] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/15/2021] [Indexed: 01/27/2023] Open
Abstract
Transmission of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in millions of deaths and declining economies around the world. K18-hACE2 mice develop disease resembling severe SARS-CoV-2 infection in a virus dose-dependent manner. The relationship between SARS-CoV-2 and the intestinal or respiratory microbiome is not fully understood. In this context, we characterized the cecal and lung microbiomes of SARS-CoV-2-challenged K18-hACE2 transgenic mice in the presence or absence of treatment with the Mpro inhibitor GC-376. Cecum microbiome showed decreased Shannon and inverse (Inv) Simpson diversity indexes correlating with SARS-CoV-2 infection dosage and a difference of Bray-Curtis dissimilarity distances among control and infected mice. Bacterial phyla such as Firmicutes, particularly, Lachnospiraceae and Oscillospiraceae, were significantly less abundant, while Verrucomicrobia, particularly, the family Akkermansiaceae, were increasingly more prevalent during peak infection in mice challenged with a high virus dose. In contrast to the cecal microbiome, the lung microbiome showed similar microbial diversity among the control, low-, and high-dose challenge virus groups, independent of antiviral treatment. Bacterial phyla in the lungs such as Bacteroidetes decreased, while Firmicutes and Proteobacteria were significantly enriched in mice challenged with a high dose of SARS-CoV-2. In summary, we identified changes in the cecal and lung microbiomes of K18-hACE2 mice with severe clinical signs of SARS-CoV-2 infection. IMPORTANCE The COVID-19 pandemic has resulted in millions of deaths. The host's respiratory and intestinal microbiome can affect directly or indirectly the immune system during viral infections. We characterized the cecal and lung microbiomes in a relevant mouse model challenged with a low or high dose of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the presence or absence of an antiviral Mpro inhibitor, GC-376. Decreased microbial diversity and taxonomic abundances of the phyla Firmicutes, particularly, Lachnospiraceae, correlating with infection dosage were observed in the cecum. In addition, microbes within the family Akkermansiaceae were increasingly more prevalent during peak infection, which is observed in other viral infections. The lung microbiome showed similar microbial diversity to that of the control, independent of antiviral treatment. Decreased Bacteroidetes and increased Firmicutes and Proteobacteria were observed in the lungs in a virus dose-dependent manner. These studies add to a better understanding of the complexities associated with the intestinal microbiome during respiratory infections.
Collapse
Affiliation(s)
- Brittany Seibert
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - C. Joaquín Cáceres
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Stivalis Cardenas-Garcia
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Silvia Carnaccini
- Tifton Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, Tifton, Georgia, USA
| | - Ginger Geiger
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Daniela S. Rajao
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Elizabeth Ottesen
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Daniel R. Perez
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
169
|
AKTAS B, ASLIM B. Neuropathy in COVID-19 associated with dysbiosis-related inflammation. Turk J Biol 2021; 45:390-403. [PMID: 34803442 PMCID: PMC8573843 DOI: 10.3906/biy-2105-53] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/30/2021] [Indexed: 01/08/2023] Open
Abstract
Although COVID-19 affects mainly lungs with a hyperactive and imbalanced immune response, gastrointestinal and neurological symptoms such as diarrhea and neuropathic pains have been described as well in patients with COVID-19. Studies indicate that gut-lung axis maintains host homeostasis and disease development with the association of immune system, and gut microbiota is involved in the COVID-19 severity in patients with extrapulmonary conditions. Gut microbiota dysbiosis impairs the gut permeability resulting in translocation of gut microbes and their metabolites into the circulatory system and induce systemic inflammation which, in turn, can affect distal organs such as the brain. Moreover, gut microbiota maintains the availability of tryptophan for kynurenine pathway, which is important for both central nervous and gastrointestinal system in regulating inflammation. SARS-CoV-2 infection disturbs the gut microbiota and leads to immune dysfunction with generalized inflammation. It has been known that cytokines and microbial products crossing the blood-brain barrier induce the neuroinflammation, which contributes to the pathophysiology of neurodegenerative diseases including neuropathies. Therefore, we believe that both gut-lung and gut-brain axes are involved in COVID-19 severity and extrapulmonary complications. Furthermore, gut microbial dysbiosis could be the reason of the neurologic complications seen in severe COVID-19 patients with the association of dysbiosis-related neuroinflammation. This review will provide valuable insights into the role of gut microbiota dysbiosis and dysbiosis-related inflammation on the neuropathy in COVID-19 patients and the disease severity.
Collapse
Affiliation(s)
- Busra AKTAS
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Burdur Mehmet Akif Ersoy University, BurdurTurkey
| | - Belma ASLIM
- Department of Biology, Faculty of Sciences, Gazi University, AnkaraTurkey
| |
Collapse
|
170
|
Skrajnowska D, Brumer M, Kankowska S, Matysek M, Miazio N, Bobrowska-Korczak B. Covid 19: Diet Composition and Health. Nutrients 2021; 13:2980. [PMID: 34578858 PMCID: PMC8472186 DOI: 10.3390/nu13092980] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/09/2021] [Accepted: 08/26/2021] [Indexed: 12/29/2022] Open
Abstract
The virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the disease coronavirus disease 2019 (COVID-19). The cumulative number of cases reported globally is now nearly 197 million and the number of cumulative deaths is 4.2 million (26 July to 1 August 2021). Currently we are focusing primarily on keeping a safe distance from others, washing our hands, and wearing masks, and the question of the effects of diet and diet-dependent risk factors remains outside the center of attention. Nevertheless, numerous studies indicate that diet can play an important role in the course of COVID-19. In this paper, based on select scientific reports, we discuss the structure and replication cycle of SARS-CoV-2, risk factors, dietary standards for sick patients, and the roles of the microbiome and dietary components supporting the immune system in preventing COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | - Barbara Bobrowska-Korczak
- Department of Bromatology, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland; (D.S.); (M.B.); (S.K.); (M.M.); (N.M.)
| |
Collapse
|
171
|
Ghannoum MA, Ford M, Bonomo RA, Gamal A, McCormick TS. A Microbiome-Driven Approach to Combating Depression During the COVID-19 Pandemic. Front Nutr 2021; 8:672390. [PMID: 34504858 PMCID: PMC8421528 DOI: 10.3389/fnut.2021.672390] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/30/2021] [Indexed: 12/18/2022] Open
Abstract
The significant stressors brought about and exacerbated by COVID-19 are associated with startling surges in mental health illnesses, specifically those related to depressive disorders. Given the huge impact of depression on society, and an incomplete understanding of impactful therapeutics, we have examined the current literature surrounding the microbiome and gut-brain axis to advance a potential complementary approach to address depression and depressive disorders that have increased during the COVID-19 pandemic. While we understand that the impact of the human gut microbiome on emotional health is a newly emerging field and more research needs to be conducted, the current evidence is extremely promising and suggests at least part of the answer to understanding depression in more depth may lie within the microbiome. As a result of these findings, we propose that a microbiome-based holistic approach, which involves carefully annotating the microbiome and potential modification through diet, probiotics, and lifestyle changes, may address depression. This paper's primary purpose is to shed light on the link between the gut microbiome and depression, including the gut-brain axis and propose a holistic approach to microbiome modification, with the ultimate goal of assisting individuals to manage their battle with depression through diet, probiotics, and lifestyle changes, in addition to offering a semblance of hope during these challenging times.
Collapse
Affiliation(s)
- Mahmoud A. Ghannoum
- Integrated Microbiome Core, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
- University Hospitals Cleveland Medical Center, Cleveland, OH, United States
- BIOHM Health LLC, Cleveland, OH, United States
| | | | - Robert A. Bonomo
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Ahmed Gamal
- Integrated Microbiome Core, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Thomas S. McCormick
- Integrated Microbiome Core, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
172
|
Gamon E, Tammena D, Wattenberg M, Augenstein T. [Rare superinfection in a COVID-19 patient-A chronology]. Anaesthesist 2021; 71:38-49. [PMID: 34427689 PMCID: PMC8383027 DOI: 10.1007/s00101-021-01018-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023]
Abstract
Bei einem 55-jährigen Mann, der sich zur elektiven Ablation bei Vorhofflimmern in der Klinik befand, wurde nach einer Reanimationssituation auf der peripheren Station ein SARS-CoV-2-Abstrich positiv getestet. Anamnestisch gab es im Vorwege gesicherten Kontakt zu einem COVID-19-positiven Patienten. Im Verlauf entwickelte sich das Vollbild einer COVID-19-Pneumonie mit umfangreicher intensivmedizinischer Behandlung. Nach rund 2‑wöchiger Therapie musste das Weaning bei erneuter Verschlechterung abgebrochen werden, und es ergaben sich bei dem wiederholt hochseptischen Patienten mikrobiologische Nachweise einer Superinfektion mit Cryptococcus neoformans und später Leclercia adecarboxylata. Der Patient wurde erfolgreich behandelt und überlebte die Erkrankung.
Collapse
Affiliation(s)
- E Gamon
- Klinik für Anästhesiologie, Intensivmedizin und Notfallmedizin, Klinikum Links der Weser, Bremen, Deutschland.
| | - D Tammena
- Klinik für Anästhesiologie, Intensivmedizin und Notfallmedizin, Klinikum Links der Weser, Bremen, Deutschland
| | - M Wattenberg
- Klinik für Anästhesiologie, Intensivmedizin und Notfallmedizin, Klinikum Links der Weser, Bremen, Deutschland
| | - T Augenstein
- Klinik für Anästhesiologie, Intensivmedizin und Notfallmedizin, Klinikum Links der Weser, Bremen, Deutschland
| |
Collapse
|
173
|
Evaluation of Hematological Parameters in Predicting Short-Term Mortality for COVID 19 Patients with Gastrointestinal Symptoms: A Case-Control Study. JOURNAL OF CONTEMPORARY MEDICINE 2021. [DOI: 10.16899/jcm.972664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
174
|
Yaneske E, Zampieri G, Bertoldi L, Benvenuto G, Angione C. Genome-scale metabolic modelling of SARS-CoV-2 in cancer cells reveals an increased shift to glycolytic energy production. FEBS Lett 2021; 595:2350-2365. [PMID: 34409594 PMCID: PMC8427129 DOI: 10.1002/1873-3468.14180] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/02/2021] [Accepted: 08/15/2021] [Indexed: 01/08/2023]
Abstract
Cancer is considered a high‐risk condition for severe illness resulting from COVID‐19. The interaction between severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2) and human metabolism is key to elucidating the risk posed by COVID‐19 for cancer patients and identifying effective treatments, yet it is largely uncharacterised on a mechanistic level. We present a genome‐scale map of short‐term metabolic alterations triggered by SARS‐CoV‐2 infection of cancer cells. Through transcriptomic‐ and proteomic‐informed genome‐scale metabolic modelling, we characterise the role of RNA and fatty acid biosynthesis in conjunction with a rewiring in energy production pathways and enhanced cytokine secretion. These findings link together complementary aspects of viral invasion of cancer cells, while providing mechanistic insights that can inform the development of treatment strategies.
Collapse
Affiliation(s)
- Elisabeth Yaneske
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK
| | - Guido Zampieri
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK.,Department of Biology, University of Padua, Italy
| | | | | | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK.,Healthcare Innovation Centre, Teesside University, Middlesbrough, UK.,Centre for Digital Innovation, Teesside University, Middlesbrough, UK
| |
Collapse
|
175
|
Machado MG, Sencio V, Trottein F. Short-Chain Fatty Acids as a Potential Treatment for Infections: a Closer Look at the Lungs. Infect Immun 2021; 89:e0018821. [PMID: 34097474 PMCID: PMC8370681 DOI: 10.1128/iai.00188-21] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are the main metabolites produced by the gut microbiota via the fermentation of complex carbohydrates and fibers. Evidence suggests that SCFAs play a role in the control of infections through direct action both on microorganisms and on host signaling. This review summarizes the main microbicidal effects of SCFAs and discusses studies highlighting the effect of SCFAs in the virulence and viability of microorganisms. We also describe the diverse and complex modes of action of the SCFAs on the immune system in the face of infections with a specific focus on bacterial and viral respiratory infections. A growing body of evidence suggests that SCFAs protect against lung infections. Finally, we present potential strategies that may be leveraged to exploit the biological properties of SCFAs for increasing effectiveness and optimizing patient benefits.
Collapse
Affiliation(s)
- Marina Gomes Machado
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, ICB, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Valentin Sencio
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - François Trottein
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
176
|
Linking the gut microbiota to persistent symptoms in survivors of COVID-19 after discharge. J Microbiol 2021; 59:941-948. [PMID: 34382150 PMCID: PMC8356893 DOI: 10.1007/s12275-021-1206-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/22/2021] [Accepted: 07/12/2021] [Indexed: 12/23/2022]
Abstract
Several follow-up studies have found that COVID-19 (coronavirus disease 2019) patients had persistent symptoms after discharge. Gut microbiota play an important role in human health and immune responses. Therefore, this study investigated the gut microbiota of recovered COVID-19 patients and the correlations between gut microbiota and persistent symptoms after discharge. Stool samples were collected from 15 recovered healthcare workers (HCWs) with COVID-19 at three months after discharge, in addition, stool samples were collected from 14 healthy controls (HCs) to perform 16S rRNA gene sequencing between May and July 2020. Compared with HCs, recovered HCWs had reduced bacterial diversity at three months after discharge, with a significantly higher relative abundance of opportunistic pathogens, and a significantly lower relative abundance of beneficial bacteria. In addition, Escherichia unclassified was positively correlated with persistent symptoms at three months after discharge, including fatigue (r = 0.567, p = 0.028), chest tightness after activity (r = 0.687, p = 0.005), and myalgia (r = 0.523, p = 0.045). Intestinibacter bartlettii was positively correlated with anorexia (r = 0.629, p = 0.012) and fatigue (r = 0.545, p = 0.036). However, Faecalibacterium prausnitzii was negatively correlated with chest tightness after activity (r = -0.591, p = 0.02), and Intestinimonas butyriciproducens was negatively correlated with cough (r = -0.635, p = 0.011). In conclusion, the gut microbiota of recovered HCWs with COVID-19 at three months after discharge was different from that of HCs, and altered gut microbiota was correlated with persistent symptoms after discharge, highlighting that gut microbiota may play an important role in the recovery of patients with COVID-19.
Collapse
|
177
|
Johnson SD, Olwenyi OA, Bhyravbhatla N, Thurman M, Pandey K, Klug EA, Johnston M, Dyavar SR, Acharya A, Podany AT, Fletcher CV, Mohan M, Singh K, Byrareddy SN. Therapeutic implications of SARS-CoV-2 dysregulation of the gut-brain-lung axis. World J Gastroenterol 2021; 27:4763-4783. [PMID: 34447225 PMCID: PMC8371510 DOI: 10.3748/wjg.v27.i29.4763] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/10/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
The emergence and rapid spread of novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused over 180 million confirmed cases resulting in over 4 million deaths worldwide with no clear end in sight for the coronavirus disease 19 (COVID-19) pandemic. Most SARS-CoV-2 exposed individuals experience mild to moderate symptoms, including fever, cough, fatigue, and loss of smell and taste. However, many individuals develop pneumonia, acute respiratory distress syndrome, septic shock, and multiorgan dysfunction. In addition to these primarily respiratory symptoms, SARS-CoV-2 can also infiltrate the central nervous system, which may damage the blood-brain barrier and the neuron's synapses. Resultant inflammation and neurodegeneration in the brain stem can further prevent efferent signaling to cranial nerves, leading to the loss of anti-inflammatory signaling and normal respiratory and gastrointestinal functions. Additionally, SARS-CoV-2 can infect enterocytes resulting in gut damage followed by microbial dysbiosis and translocation of bacteria and their byproducts across the damaged epithelial barrier. As a result, this exacerbates pro-inflammatory responses both locally and systemically, resulting in impaired clinical outcomes. Recent evidence has highlighted the complex interactions that mutually modulate respiratory, neurological, and gastrointestinal function. In this review, we discuss the ways SARS-CoV-2 potentially disrupts the gut-brain-lung axis. We further highlight targeting specific responses to SARS-CoV-2 for the development of novel, urgently needed therapeutic interventions. Finally, we propose a prospective related to the individuals from Low- and Middle-Income countries. Here, the underlying propensity for heightened gut damage/microbial translocation is likely to result in worse clinical outcomes during this COVID-19 pandemic.
Collapse
Affiliation(s)
- Samuel D Johnson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Omalla A Olwenyi
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Namita Bhyravbhatla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Michellie Thurman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Kabita Pandey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Elizabeth A Klug
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Morgan Johnston
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Shetty Ravi Dyavar
- Antiviral Pharmacology Laboratory, University of Nebraska Medical Center (UNMC) Center for Drug Discovery, Omaha, NE 68198, United States
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Anthony T Podany
- Antiviral Pharmacology Laboratory, University of Nebraska Medical Center (UNMC) Center for Drug Discovery, Omaha, NE 68198, United States
| | - Courtney V Fletcher
- Antiviral Pharmacology Laboratory, University of Nebraska Medical Center (UNMC) Center for Drug Discovery, Omaha, NE 68198, United States
| | - Mahesh Mohan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, United States
| | - Kamal Singh
- Department of Molecular Microbiology and Immunology and Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, United States
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| |
Collapse
|
178
|
Manosso LM, Arent CO, Borba LA, Ceretta LB, Quevedo J, Réus GZ. Microbiota-Gut-Brain Communication in the SARS-CoV-2 Infection. Cells 2021; 10:1993. [PMID: 34440767 PMCID: PMC8391332 DOI: 10.3390/cells10081993] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease of 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome 2 (SARS-CoV-2). In addition to pneumonia, individuals affected by the disease have neurological symptoms. Indeed, SARS-CoV-2 has a neuroinvasive capacity. It is known that the infection caused by SARS-CoV-2 leads to a cytokine storm. An exacerbated inflammatory state can lead to the blood-brain barrier (BBB) damage as well as to intestinal dysbiosis. These changes, in turn, are associated with microglial activation and reactivity of astrocytes that can promote the degeneration of neurons and be associated with the development of psychiatric disorders and neurodegenerative diseases. Studies also have been shown that SARS-CoV-2 alters the composition and functional activity of the gut microbiota. The microbiota-gut-brain axis provides a bidirectional homeostatic communication pathway. Thus, this review focuses on studies that show the relationship between inflammation and the gut microbiota-brain axis in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Luana M. Manosso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma 77054-000, SC, Brazil; (L.M.M.); (C.O.A.); (L.A.B.); (J.Q.)
| | - Camila O. Arent
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma 77054-000, SC, Brazil; (L.M.M.); (C.O.A.); (L.A.B.); (J.Q.)
| | - Laura A. Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma 77054-000, SC, Brazil; (L.M.M.); (C.O.A.); (L.A.B.); (J.Q.)
| | - Luciane B. Ceretta
- Programa de Pós-Graduação em Saúde Coletiva, Universidade do Extremo Sul Catarinense, Criciúma 88806-000, SC, Brazil;
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma 77054-000, SC, Brazil; (L.M.M.); (C.O.A.); (L.A.B.); (J.Q.)
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
- Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA
| | - Gislaine Z. Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma 77054-000, SC, Brazil; (L.M.M.); (C.O.A.); (L.A.B.); (J.Q.)
| |
Collapse
|
179
|
Rouchka EC, Chariker JH, Alejandro B, Adcock RS, Singhal R, Ramirez J, Palmer KE, Lasnik AB, Carrico R, Arnold FW, Furmanek S, Zhang M, Wolf LA, Waigel S, Zacharias W, Bordon J, Chung D. Induction of interferon response by high viral loads at early stage infection may protect against severe outcomes in COVID-19 patients. Sci Rep 2021; 11:15715. [PMID: 34344959 PMCID: PMC8333042 DOI: 10.1038/s41598-021-95197-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/21/2021] [Indexed: 12/13/2022] Open
Abstract
Key elements for viral pathogenesis include viral strains, viral load, co-infection, and host responses. Several studies analyzing these factors in the function of disease severity of have been published; however, no studies have shown how all of these factors interplay within a defined cohort. To address this important question, we sought to understand how these four key components interplay in a cohort of COVID-19 patients. We determined the viral loads and gene expression using high throughput sequencing and various virological methods. We found that viral loads in the upper respiratory tract in COVID-19 patients at an early phase of infection vary widely. While the majority of nasopharyngeal (NP) samples have a viral load lower than the limit of detection of infectious viruses, there are samples with an extraordinary amount of SARS-CoV-2 RNA and a high viral titer. No specific viral factors were identified that are associated with high viral loads. Host gene expression analysis showed that viral loads were strongly correlated with cellular antiviral responses. Interestingly, however, COVID-19 patients who experience mild symptoms have a higher viral load than those with severe complications, indicating that naso-pharyngeal viral load may not be a key factor of the clinical outcomes of COVID-19. The metagenomics analysis revealed that the microflora in the upper respiratory tract of COVID-19 patients with high viral loads were dominated by SARS-CoV-2, with a high degree of dysbiosis. Finally, we found a strong inverse correlation between upregulation of interferon responses and disease severity. Overall our study suggests that a high viral load in the upper respiratory tract may not be a critical factor for severe symptoms; rather, dampened antiviral responses may be a critical factor for a severe outcome from the infection.
Collapse
Affiliation(s)
- Eric C Rouchka
- Department of Computer Science and Engineering, University of Louisville, Louisville, KY, USA
- Kentucky IDeA Network of Biomedical Research Excellence (KY-INBRE) Bioinformatics Core, Louisville, KY, USA
| | - Julia H Chariker
- Kentucky IDeA Network of Biomedical Research Excellence (KY-INBRE) Bioinformatics Core, Louisville, KY, USA
- Department of Neuroscience Training, University of Louisville, Louisville, KY, USA
| | - Brian Alejandro
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Robert S Adcock
- Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Richa Singhal
- Kentucky IDeA Network of Biomedical Research Excellence (KY-INBRE) Bioinformatics Core, Louisville, KY, USA
- Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Julio Ramirez
- Department of Medicine, University of Louisville, Louisville, KY, USA
- Division of Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - Kenneth E Palmer
- Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Amanda B Lasnik
- Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Ruth Carrico
- Department of Medicine, University of Louisville, Louisville, KY, USA
- Division of Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - Forest W Arnold
- Department of Medicine, University of Louisville, Louisville, KY, USA
- Division of Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - Stephen Furmanek
- Department of Medicine, University of Louisville, Louisville, KY, USA
- Division of Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - Mei Zhang
- Department of Medicine, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
- Genomics Core Facility, University of Louisville, Louisville, KY, USA
| | - Leslie A Wolf
- Division of Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - Sabine Waigel
- Department of Medicine, University of Louisville, Louisville, KY, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Genomics Core Facility, University of Louisville, Louisville, KY, USA
| | - Wolfgang Zacharias
- Department of Medicine, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Genomics Core Facility, University of Louisville, Louisville, KY, USA
| | - Jose Bordon
- Washington Health Institute, George Washington University School of Medicine, Washington, D.C, USA
- Department of Medicine, George Washington University School of Medicine, Washington, D.C, USA
| | - Donghoon Chung
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, USA.
- Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
180
|
Boutin S, Hildebrand D, Boulant S, Kreuter M, Rüter J, Pallerla SR, Velavan TP, Nurjadi D. Host factors facilitating SARS-CoV-2 virus infection and replication in the lungs. Cell Mol Life Sci 2021; 78:5953-5976. [PMID: 34223911 PMCID: PMC8256233 DOI: 10.1007/s00018-021-03889-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 06/01/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2 is the virus causing the major pandemic facing the world today. Although, SARS-CoV-2 primarily causes lung infection, a variety of symptoms have proven a systemic impact on the body. SARS-CoV-2 has spread in the community quickly infecting humans from all age, ethnicities and gender. However, fatal outcomes have been linked to specific host factors and co-morbidities such as age, hypertension, immuno-deficiencies, chronic lung diseases or metabolic disorders. A major shift in the microbiome of patients suffering of the coronavirus disease 2019 (COVID-19) have also been observed and is linked to a worst outcome of the disease. As many co-morbidities are already known to be associated with a dysbiosis of the microbiome such as hypertension, diabetes and metabolic disorders. Host factors and microbiome changes are believed to be involved as a network in the acquisition of the infection and the development of the diseases. We will review in detail in this manuscript, the immune response toward SARS-CoV-2 infection as well as the host factors involved in the facilitation and worsening of the infection. We will also address the impact of COVID-19 on the host's microbiome and secondary infection which also worsen the disease.
Collapse
Affiliation(s)
- Sébastien Boutin
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.
| | - Dagmar Hildebrand
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Steeve Boulant
- Division of Cellular Polarity and Viral Infection, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Kreuter
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
- Center for Interstitial and Rare Lung Diseases, Pneumology, Thoraxklinik, University of Heidelberg, Heidelberg, Germany
| | - Jule Rüter
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Tübingen, Germany
| | | | - Thirumalaisamy P Velavan
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Tübingen, Germany
- Vietnamese-German Center for Medical Research, Hanoi, Vietnam
| | - Dennis Nurjadi
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| |
Collapse
|
181
|
Iebba V, Zanotta N, Campisciano G, Zerbato V, Di Bella S, Cason C, Luzzati R, Confalonieri M, Palamara AT, Comar M. Profiling of Oral Microbiota and Cytokines in COVID-19 Patients. Front Microbiol 2021; 12:671813. [PMID: 34394024 PMCID: PMC8361794 DOI: 10.3389/fmicb.2021.671813] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
The presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been recently demonstrated in the sputum or saliva, suggesting how the shedding of viral RNA outlasts the end of symptoms. Recent data from transcriptome analysis show that the oral cavity mucosa harbors high levels of angiotensin-converting enzyme 2 (ACE2) and transmembrane protease, serine 2 (TMPRSS2), highlighting its role as a double-edged sword for SARS-CoV-2 body entrance or interpersonal transmission. Here, we studied the oral microbiota structure and inflammatory profile of 26 naive severe coronavirus disease 2019 (COVID-19) patients and 15 controls by 16S rRNA V2 automated targeted sequencing and magnetic bead-based multiplex immunoassays, respectively. A significant diminution in species richness was observed in COVID-19 patients, along with a marked difference in beta-diversity. Species such as Prevotella salivae and Veillonella infantium were distinctive for COVID-19 patients, while Neisseria perflava and Rothia mucilaginosa were predominant in controls. Interestingly, these two groups of oral species oppositely clustered within the bacterial network, defining two distinct Species Interacting Groups (SIGs). COVID-19-related pro-inflammatory cytokines were found in both oral and serum samples, along with a specific bacterial consortium able to counteract them. We introduced a new parameter, named CytoCOV, able to predict COVID-19 susceptibility for an unknown subject at 71% of power with an Area Under Curve (AUC) equal to 0.995. This pilot study evidenced a distinctive oral microbiota composition in COVID-19 subjects, with a definite structural network in relation to secreted cytokines. Our results would be usable in clinics against COVID-19, using bacterial consortia as biomarkers or to reduce local inflammation.
Collapse
Affiliation(s)
- Valerio Iebba
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy
| | - Nunzia Zanotta
- Laboratory of Advanced Microbiology Diagnosis and Translational Research, Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| | - Giuseppina Campisciano
- Laboratory of Advanced Microbiology Diagnosis and Translational Research, Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| | - Verena Zerbato
- Infectious Diseases Department, University of Udine, Udine, Italy
| | - Stefano Di Bella
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy
| | - Carolina Cason
- Laboratory of Advanced Microbiology Diagnosis and Translational Research, Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| | - Roberto Luzzati
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy
| | - Marco Confalonieri
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy
- Pulmonology Department, University Hospital of Cattinara, Trieste, Italy
| | - Anna Teresa Palamara
- IRCCS San Raffaele Pisana, Rome, Italy
- Laboratory Affiliated to Institute Pasteur Italia- Cenci Bolognetti Foundation, Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Manola Comar
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy
- Laboratory of Advanced Microbiology Diagnosis and Translational Research, Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
182
|
Hung YP, Lee CC, Lee JC, Tsai PJ, Ko WC. Gut Dysbiosis during COVID-19 and Potential Effect of Probiotics. Microorganisms 2021; 9:1605. [PMID: 34442684 PMCID: PMC8402052 DOI: 10.3390/microorganisms9081605] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), an RNA virus of the family Coronaviridae, causes coronavirus disease 2019 (COVID-19), an influenza-like disease that chiefly infects the lungs through respiratory transmission. The spike protein of SARS-CoV-2, a transmembrane protein in its outer portion, targets angiotensin-converting enzyme 2 (ACE2) as the binding receptor for the cell entry. As ACE2 is highly expressed in the gut and pulmonary tissues, SARS-CoV-2 infections frequently result in gastrointestinal inflammation, with presentations ordinarily ranging from intestinal cramps to complications with intestinal perforations. However, the evidence detailing successful therapy for gastrointestinal involvement in COVID-19 patients is currently limited. A significant change in fecal microbiomes, namely dysbiosis, was characterized by the enrichment of opportunistic pathogens and the depletion of beneficial commensals and their crucial association to COVID-19 severity has been evidenced. Oral probiotics had been evidenced to improve gut health in achieving homeostasis by exhibiting their antiviral effects via the gut-lung axis. Although numerous commercial probiotics have been effective against coronavirus, their efficacies in treating COVID-19 patients remain debated. In ClinicalTrials.gov, 19 clinical trials regarding the dietary supplement of probiotics, in terms of Lactobacillus and mixtures of Bifidobacteria and Lactobacillus, for treating COVID-19 cases are ongoing. Accordingly, the preventive or therapeutic role of probiotics for COVID-19 patients can be elucidated in the near future.
Collapse
Affiliation(s)
- Yuan-Pin Hung
- Department of Internal Medicine, Tainan Hospital, Ministry of Health and Welfare, Tainan 700, Taiwan;
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
| | - Ching-Chi Lee
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
- Clinical Medicine Research Center, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 705, Taiwan
- Graduate Institute of Medical Sciences, College of Health Sciences, Chang Jung Christian University, Tainan 711, Taiwan
| | - Jen-Chieh Lee
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan;
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan
- Department of Pathology, National Cheng Kung University Hospital, Tainan 705, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan
| |
Collapse
|
183
|
Walton GE, Gibson GR, Hunter KA. Mechanisms linking the human gut microbiome to prophylactic and treatment strategies for COVID-19. Br J Nutr 2021; 126:219-227. [PMID: 33032673 PMCID: PMC7684010 DOI: 10.1017/s0007114520003980] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/18/2020] [Accepted: 09/28/2020] [Indexed: 12/22/2022]
Abstract
The recent COVID-19 pandemic has altered the face of biology, social interaction and public health worldwide. It has had a destructive effect upon millions of people and is approaching a devastating one million fatalities. Emerging evidence has suggested a link between the infection and gut microbiome status. This is one of the several factors that may contribute towards severity of infection. Given the fact that the gut is heavily linked to immunity, inflammatory status and the ability to challenge pathogens, it is worthwhile to consider dietary intervention of the gut microbiota as means of potentially challenging the viral outcome. In this context, probiotics and prebiotics have been used to mitigate similar respiratory infections. Here, we summarise links between the gut microbiome and COVID-19 infection, as well as propose mechanisms whereby probiotic and prebiotic interventions may act.
Collapse
Affiliation(s)
- Gemma E. Walton
- Food Microbial Sciences Unit, Department of Food and Nutritional Sciences, University of Reading, ReadingRG6 6AP, UK
| | - Glenn R. Gibson
- Food Microbial Sciences Unit, Department of Food and Nutritional Sciences, University of Reading, ReadingRG6 6AP, UK
| | - Kirsty A. Hunter
- Exercise and Health Research Group, Department of Sport Science, Sport, Health and Performance Enhancement (SHAPE) Research Centre, Nottingham Trent University, NottinghamNG11 8NS, UK
| |
Collapse
|
184
|
Moreira-Rosário A, Marques C, Pinheiro H, Araújo JR, Ribeiro P, Rocha R, Mota I, Pestana D, Ribeiro R, Pereira A, de Sousa MJ, Pereira-Leal J, de Sousa J, Morais J, Teixeira D, Rocha JC, Silvestre M, Príncipe N, Gatta N, Amado J, Santos L, Maltez F, Boquinhas A, de Sousa G, Germano N, Sarmento G, Granja C, Póvoa P, Faria A, Calhau C. Gut Microbiota Diversity and C-Reactive Protein Are Predictors of Disease Severity in COVID-19 Patients. Front Microbiol 2021; 12:705020. [PMID: 34349747 PMCID: PMC8326578 DOI: 10.3389/fmicb.2021.705020] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/10/2021] [Indexed: 12/15/2022] Open
Abstract
The risk factors for coronavirus disease 2019 (COVID-19) severity are still poorly understood. Considering the pivotal role of the gut microbiota on host immune and inflammatory functions, we investigated the association between changes in the gut microbiota composition and the clinical severity of COVID-19. We conducted a multicenter cross-sectional study prospectively enrolling 115 COVID-19 patients categorized according to: (1) the WHO Clinical Progression Scale-mild, 19 (16.5%); moderate, 37 (32.2%); or severe, 59 (51.3%), and (2) the location of recovery from COVID-19-ambulatory, 14 (household isolation, 12.2%); hospitalized in ward, 40 (34.8%); or hospitalized in the intensive care unit, 61 (53.0%). Gut microbiota analysis was performed through 16S rRNA gene sequencing, and the data obtained were further related to the clinical parameters of COVID-19 patients. The risk factors for COVID-19 severity were identified by univariate and multivariable logistic regression models. In comparison to mild COVID-19 patients, the gut microbiota of moderate and severe patients have: (a) lower Firmicutes/Bacteroidetes ratio; (b) higher abundance of Proteobacteria; and (c) lower abundance of beneficial butyrate-producing bacteria such as the genera Roseburia and Lachnospira. Multivariable regression analysis showed that the Shannon diversity index [odds ratio (OR) = 2.85, 95% CI = 1.09-7.41, p = 0.032) and C-reactive protein (OR = 3.45, 95% CI = 1.33-8.91, p = 0.011) are risk factors for severe COVID-19 (a score of 6 or higher in the WHO Clinical Progression Scale). In conclusion, our results demonstrated that hospitalized patients with moderate and severe COVID-19 have microbial signatures of gut dysbiosis; for the first time, the gut microbiota diversity is pointed out as a prognostic biomarker of COVID-19 severity.
Collapse
Affiliation(s)
- André Moreira-Rosário
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Cláudia Marques
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Hélder Pinheiro
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Department of Infectious Diseases, Hospital Curry Cabral, Centro Hospitalar Universitário de Lisboa Central, Lisboa, Portugal
| | - João Ricardo Araújo
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Pedro Ribeiro
- Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | - Rita Rocha
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Inês Mota
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Diogo Pestana
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Rita Ribeiro
- Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | - Ana Pereira
- Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | - Maria José de Sousa
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | | | - José de Sousa
- Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | - Juliana Morais
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal.,Faculdade de Ciências Médicas, Comprehensive Health Research Centre (CHRC), NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Diana Teixeira
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, Comprehensive Health Research Centre (CHRC), NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Júlio César Rocha
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Marta Silvestre
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Nuno Príncipe
- Department of Emergency and Intensive Care Medicine, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Nuno Gatta
- Department of Emergency and Intensive Care Medicine, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - José Amado
- Department of Emergency and Intensive Care Medicine, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Lurdes Santos
- Infectious Diseases Service, ID Intensive Care Unit, Faculdade de Medicina, Centro Hospitalar Universitário de São João, Universidade do Porto, Porto, Portugal
| | - Fernando Maltez
- Department of Infectious Diseases, Hospital Curry Cabral, Centro Hospitalar Universitário de Lisboa Central, Lisboa, Portugal
| | - Ana Boquinhas
- Department of Emergency, CUF Infante Santo Hospital, Lisboa, Portugal
| | - Germano de Sousa
- Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | - Nuno Germano
- Polyvalent Intensive Care Unit, Hospital Curry Cabral, Centro Hospitalar Universitário de Lisboa Central, Lisboa, Portugal
| | - Gonçalo Sarmento
- Department of Internal Medicine, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira, Portugal
| | - Cristina Granja
- Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal.,Department of Anesthesiology, Centro Hospitalar Universitário de São João, Porto, Portugal.,Department of Surgery and Physiology, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Pedro Póvoa
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Polyvalent Intensive Care Unit, Hospital São Francisco Xavier, Centro Hospitalar Lisboa Ocidental, Lisboa, Portugal.,Center for Clinical Epidemiology, Research Unit of Clinical Epidemiology, OUH Odense University Hospital, Odense, Denmark
| | - Ana Faria
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, Comprehensive Health Research Centre (CHRC), NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Conceição Calhau
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| |
Collapse
|
185
|
Zhang Z, Zhang G, Guo M, Tao W, Liu X, Wei H, Jin T, Zhang Y, Zhu S. The Potential Role of an Aberrant Mucosal Immune Response to SARS-CoV-2 in the Pathogenesis of IgA Nephropathy. Pathogens 2021; 10:pathogens10070881. [PMID: 34358031 PMCID: PMC8308514 DOI: 10.3390/pathogens10070881] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/23/2021] [Accepted: 07/07/2021] [Indexed: 12/20/2022] Open
Abstract
The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global concern. Immunoglobin A (IgA) contributes to virus neutralization at the early stage of infection. Longitudinal studies are needed to assess whether SARS-CoV-2-specific IgA production persists for a longer time in patients recovered from severe COVID-19 and its lasting symptoms that can have disabling consequences should also be alerted to susceptible hosts. Here, we tracked the anti-SARS-CoV-2 spike protein receptor-binding domain (RBD) antibody levels in a cohort of 88 COVID-19 patients. We found that 52.3% of the patients produced more anti-SARS-CoV-2 RBD IgA than IgG or IgM, and the levels of IgA remained stable during 4–41 days of infection. One of these IgA-dominant COVID-19 patients, concurrently with IgA nephropathy (IgAN), presented with elevated serum creatinine and worse proteinuria during the infection, which continued until seven months post-infection. The serum levels of anti-SARS-CoV-2 RBD and total IgA were higher in this patient than in healthy controls. Changes in the composition of the intestinal microbiota, increased IgA highly coated bacteria, and elevated concentration of the proinflammatory cytokine IL-18 were indicative of potential involvement of intestinal dysbiosis and inflammation to the systemic IgA level and, consequently, the disease progression. Collectively, our work highlighted the potential adverse effect of the mucosal immune response to SARS-CoV-2 infection, and that additional care should be taken with COVID-19 patients presenting with chronic diseases such as IgAN.
Collapse
Affiliation(s)
- Zhao Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing 100034, China; (Z.Z.); (X.L.)
| | - Guorong Zhang
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (G.Z.); (M.G.); (W.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China;
| | - Meng Guo
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (G.Z.); (M.G.); (W.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China;
| | - Wanyin Tao
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (G.Z.); (M.G.); (W.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China;
| | - Xingzi Liu
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing 100034, China; (Z.Z.); (X.L.)
| | - Haiming Wei
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China;
| | - Tengchuan Jin
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China;
- Correspondence: (T.J.); (Y.Z.); (S.Z.)
| | - Yuemiao Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing 100034, China; (Z.Z.); (X.L.)
- Correspondence: (T.J.); (Y.Z.); (S.Z.)
| | - Shu Zhu
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (G.Z.); (M.G.); (W.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China;
- School of Data Science, University of Science and Technology of China, Hefei 230026, China
- CAS Centre for Excellence in Cell and Molecular Biology, University of Science and Technology of China, Hefei 230026, China
- Correspondence: (T.J.); (Y.Z.); (S.Z.)
| |
Collapse
|
186
|
Chen ZR, Liu J, Liao ZG, Zhou J, Peng HW, Gong F, Hu JF, Zhou Y. COVID-19 and gastroenteric manifestations. World J Clin Cases 2021; 9:4990-4997. [PMID: 34307549 PMCID: PMC8283602 DOI: 10.12998/wjcc.v9.i19.4990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/14/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the infection of a novel coronavirus [severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)], has become a pandemic. The infection has resulted in about one hundred million COVID-19 cases and millions of deaths. Although SARS-CoV-2 mainly spreads through the air and impairs the function of the respiratory system, it also attacks the gastrointestinal epithelial cells through the same receptor, angiotensin converting enzyme 2 receptor, which results in gastroenteric symptoms and potential fecal-oral transmission. Besides the infection of SARS-CoV-2, the treatments of COVID-19 also contribute to the gastroenteric manifestations due to the adverse drug reactions of anti-COVID-19 drugs. In this review, we update the clinical features, basic studies, and clinical practices of COVID-19-associated gastroenteric manifestations.
Collapse
Affiliation(s)
- Zhang-Ren Chen
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Jing Liu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Zhi-Guo Liao
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Jian Zhou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Hong-Wei Peng
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Fei Gong
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Jin-Fang Hu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Ying Zhou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| |
Collapse
|
187
|
Zhang J, Garrett S, Sun J. Gastrointestinal symptoms, pathophysiology, and treatment in COVID-19. Genes Dis 2021; 8:385-400. [PMID: 33521210 PMCID: PMC7836435 DOI: 10.1016/j.gendis.2020.08.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/18/2020] [Accepted: 08/28/2020] [Indexed: 01/08/2023] Open
Abstract
The novel coronavirus Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has emerged and is responsible for the Coronavirus Disease 2019 global pandemic. Coronaviruses, including SARS-CoV-2, are strongly associated with respiratory symptoms during infection, but gastrointestinal symptoms, such as diarrhea, vomiting, nausea, and abdominal pain, have been identified in subsets of COVID-19 patients. This article focuses on gastrointestinal symptoms and pathophysiology in COVID-19 disease. Evidence suggests that the gastrointestinal tract could be a viral target for SARS-CoV-2 infection. Not only is the SARS-CoV-2 receptor ACE2 highly expressed in the GI tract and is associated with digestive symptoms, but bleeding and inflammation are observed in the intestine of COVID-19 patients. We further systemically summarize the correlation between COVID-19 disease, gastrointestinal symptoms and intestinal microbiota. The potential oral-fecal transmission of COVID-19 was supported by viral RNA and live virus detection in the feces of COVID-19 patients. Additionally, the viral balance in the GI tract could be disordered during SARS-CoV-2 infection which could further impact the homeostasis of the gut microbial flora. Finally, we discuss the clinical and ongoing trials of treatments/therapies, including antiviral drugs, plasma transfusion and immunoglobulins, and diet supplementations for COVID-19. By reviewing the pathogenesis of SARS-CoV-2 virus, and understanding the correlation among COVID-19, inflammation, intestinal microbiota, and lung microbiota, we provide perspective in prevention and control, as well as diagnosis and treatment of the COVID-19 disease.
Collapse
Affiliation(s)
- Jilei Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Shari Garrett
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- UIC Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
188
|
Yu Z, Yang Z, Wang Y, Zhou F, Li S, Li C, Li L, Zhang W, Li X. Recent advance of ACE2 and microbiota dysfunction in COVID-19 pathogenesis. Heliyon 2021; 7:e07548. [PMID: 34296023 PMCID: PMC8270732 DOI: 10.1016/j.heliyon.2021.e07548] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/08/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the coronavirus disease 2019 (COVID-19) and has become the world's most pressing public health threat. Although not as common as respiratory symptoms, a substantial proportion of patients with COVID-19 presented the gastrointestinal symptoms. ACE2, as the receptor of SARS-CoV and SARS-CoV-2, is highly expressed in the epithelia of the epithelium cells in lung and intestine. In addition, ACE2 is essential for the innate immunity, amino acid transportation and the homeostasis of intestinal microecology. The composition of gut microbiota in COVID-19 patients was altered and concordant with inflammatory, which may explain the gastrointestinal symptoms in patients. Here we reviewed and discussed the evolving role for ACE2 and gut microbiota in SARS-CoV-2 infection which might provide innovative approaches to targeting ACE2 and gut microbiota for the COVID-19 therapy.
Collapse
Affiliation(s)
- Zhidan Yu
- Department of Gastroenterology, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Zhaojie Yang
- The Sixth People's Hospital of Zhengzhou, Zhengzhou 450000, China
| | - Yuesheng Wang
- Department of Gastroenterology, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Fang Zhou
- Department of Gastroenterology, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Suli Li
- Department of Gastroenterology, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Chan Li
- Department of Biostatistics, University at Buffalo, 208 Norton Hall, Buffalo, NY 14260-1800, USA
| | - Lifeng Li
- Department of Gastroenterology, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Wancun Zhang
- Department of Gastroenterology, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Xiaoqin Li
- Department of Gastroenterology, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| |
Collapse
|
189
|
Rajput S, Paliwal D, Naithani M, Kothari A, Meena K, Rana S. COVID-19 and Gut Microbiota: A Potential Connection. Indian J Clin Biochem 2021; 36:266-277. [PMID: 33495676 PMCID: PMC7818076 DOI: 10.1007/s12291-020-00948-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
Currently, world is facing a global outbreak causing a pandemic threat known as COVID-19. This infectious disease is triggered by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Gut microbiota harbours multi species community with a strong impact on host immune homeostasis. However, our knowledge about this gut microbiota and its symbiotic relationship with immune activation in association with SARS-CoV-2 is limited. Unbalanced bacterial flora with too many opportunistic infections can shift immune system towards a cascade of inflammatory responses leading to multi organ damage. This review will highlight immune-regulation via various mechanisms in SARS-CoV-2 infection. Diet has an unbelievable influence on gut microbiome that allows a new state of homeostasis to be reached through timing, frequency and duration of intake. This review article focuses on gut, lung microbiota and immunomodulation with specific attention on immune activation by gut microbiota.
Collapse
Affiliation(s)
- Swati Rajput
- Department of Biochemistry, AIIMS Rishikesh, Uttarakhand, 249203 India
| | - Deepanshu Paliwal
- Department of Biochemistry, AIIMS Rishikesh, Uttarakhand, 249203 India
| | - Manisha Naithani
- Department of Biochemistry, AIIMS Rishikesh, Uttarakhand, 249203 India
| | - Aashish Kothari
- Department of Microbiology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand India
| | - Kiran Meena
- Department of Biochemistry, AIIMS Rishikesh, Uttarakhand, 249203 India
| | - Satyavati Rana
- Department of Biochemistry, AIIMS Rishikesh, Uttarakhand, 249203 India
| |
Collapse
|
190
|
Ren Z, Wang H, Cui G, Lu H, Wang L, Luo H, Chen X, Ren H, Sun R, Liu W, Liu X, Liu C, Li A, Wang X, Rao B, Yuan C, Zhang H, Sun J, Chen X, Li B, Hu C, Wu Z, Yu Z, Kan Q, Li L. Alterations in the human oral and gut microbiomes and lipidomics in COVID-19. Gut 2021; 70:1253-1265. [PMID: 33789966 PMCID: PMC8042598 DOI: 10.1136/gutjnl-2020-323826] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/04/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To characterise the oral microbiome, gut microbiome and serum lipid profiles in patients with active COVID-19 and recovered patients; evaluate the potential of the microbiome as a non-invasive biomarker for COVID-19; and explore correlations between the microbiome and lipid profile. DESIGN We collected and sequenced 392 tongue-coating samples, 172 faecal samples and 155 serum samples from Central China and East China. We characterised microbiome and lipid molecules, constructed microbial classifiers in discovery cohort and verified their diagnostic potential in 74 confirmed patients (CPs) from East China and 37 suspected patients (SPs) with IgG positivity. RESULTS Oral and faecal microbial diversity was significantly decreased in CPs versus healthy controls (HCs). Compared with HCs, butyric acid-producing bacteria were decreased and lipopolysaccharide-producing bacteria were increased in CPs in oral cavity. The classifiers based on 8 optimal oral microbial markers (7 faecal microbial markers) achieved good diagnostic efficiency in different cohorts. Importantly, diagnostic efficacy reached 87.24% in the cross-regional cohort. Moreover, the classifiers successfully diagnosed SPs with IgG antibody positivity as CPs, and diagnostic efficacy reached 92.11% (98.01% of faecal microbiome). Compared with CPs, 47 lipid molecules, including sphingomyelin (SM)(d40:4), SM(d38:5) and monoglyceride(33:5), were depleted, and 122 lipid molecules, including phosphatidylcholine(36:4p), phosphatidylethanolamine (PE)(16:0p/20:5) and diglyceride(20:1/18:2), were enriched in confirmed patients recovery. CONCLUSION This study is the first to characterise the oral microbiome in COVID-19, and oral microbiomes and lipid alterations in recovered patients, to explore their correlations and to report the successful establishment and validation of a diagnostic model for COVID-19.
Collapse
Affiliation(s)
- Zhigang Ren
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyu Wang
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guangying Cui
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haifeng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ling Wang
- Department of Clinical Laboratory, Henan Provincial Chest Hospital, Zhengzhou, Henan, China
| | - Hong Luo
- Department of General Surgery, Guangshan County People's Hospital, Xinyang, Henan, China
| | - Xinhua Chen
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongyan Ren
- Shanghai Mobio Biomedical Technology Co, Ltd, Shanghai, Shanghai, China
| | - Ranran Sun
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenli Liu
- Clinical Laboratory Diagnostics, Medical Technology College, Beihua University, Jilin, Jilin, China
| | - Xiaorui Liu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Chao Liu
- Shanghai Mobio Biomedical Technology Co, Ltd, Shanghai, Shanghai, China
| | - Ang Li
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuemei Wang
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Benchen Rao
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chengyu Yuan
- Department of General Surgery, Guangshan County People's Hospital, Xinyang, Henan, China
| | - Hua Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiarui Sun
- Shanghai Mobio Biomedical Technology Co, Ltd, Shanghai, Shanghai, China
| | - Xiaolong Chen
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bingjie Li
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chuansong Hu
- Department of General Surgery, Guangshan County People's Hospital, Xinyang, Henan, China
| | - Zhongwen Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zujiang Yu
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Quancheng Kan
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Shulan (Hangzhou) Hospital, Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
191
|
Ramakrishnan RK, Kashour T, Hamid Q, Halwani R, Tleyjeh IM. Unraveling the Mystery Surrounding Post-Acute Sequelae of COVID-19. Front Immunol 2021; 12:686029. [PMID: 34276671 PMCID: PMC8278217 DOI: 10.3389/fimmu.2021.686029] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022] Open
Abstract
More than one year since its emergence, corona virus disease 2019 (COVID-19) is still looming large with a paucity of treatment options. To add to this burden, a sizeable subset of patients who have recovered from acute COVID-19 infection have reported lingering symptoms, leading to significant disability and impairment of their daily life activities. These patients are considered to suffer from what has been termed as “chronic” or “long” COVID-19 or a form of post-acute sequelae of COVID-19, and patients experiencing this syndrome have been termed COVID-19 long-haulers. Despite recovery from infection, the persistence of atypical chronic symptoms, including extreme fatigue, shortness of breath, joint pains, brain fogs, anxiety and depression, that could last for months implies an underlying disease pathology that persist beyond the acute presentation of the disease. As opposed to the direct effects of the virus itself, the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is believed to be largely responsible for the appearance of these lasting symptoms, possibly through facilitating an ongoing inflammatory process. In this review, we hypothesize potential immunological mechanisms underlying these persistent and prolonged effects, and describe the multi-organ long-term manifestations of COVID-19.
Collapse
Affiliation(s)
- Rakhee K Ramakrishnan
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Tarek Kashour
- Department of Cardiac Sciences, King Fahad Cardiac Center, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Qutayba Hamid
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Meakins-Christie Laboratories, Research Institute of the McGill University Healthy Center, McGill University, Montreal, QC, Canada
| | - Rabih Halwani
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Prince Abdullah Ben Khaled Celiac Disease Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Imad M Tleyjeh
- Infectious Diseases Section, Department of Medical Specialties, King Fahad Medical City, Riyadh, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.,Division of Infectious Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN, United States.,Division of Epidemiology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|
192
|
Gaibani P, D’Amico F, Bartoletti M, Lombardo D, Rampelli S, Fornaro G, Coladonato S, Siniscalchi A, Re MC, Viale P, Brigidi P, Turroni S, Giannella M. The Gut Microbiota of Critically Ill Patients With COVID-19. Front Cell Infect Microbiol 2021; 11:670424. [PMID: 34268136 PMCID: PMC8276076 DOI: 10.3389/fcimb.2021.670424] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/03/2021] [Indexed: 01/04/2023] Open
Abstract
The SARS-CoV-2-associated COVID-19 pandemic has shaken the global healthcare system. Although the best-known symptoms are dry cough and pneumonia, viral RNA has been detected in the stool and about half of COVID-19 patients exhibit gastrointestinal upset. In this scenario, special attention is being paid to the possible role of the gut microbiota (GM). Fecal samples from 69 COVID-19 patients from three different hospitals of Bologna (Italy) were analyzed by 16S rRNA gene-based sequencing. The GM profile was compared with the publicly available one of healthy age- and gender-matched Italians, as well as with that of other critically ill non-COVID-19 patients. The GM of COVID-19 patients appeared severely dysbiotic, with reduced diversity, loss of health-associated microorganisms and enrichment of potential pathogens, particularly Enterococcus. This genus was far overrepresented in patients developing bloodstream infections (BSI) and admitted to the intensive care unit, while almost absent in other critically ill non-COVID-19 patients. Interestingly, the percentage of patients with BSI due to Enterococcus spp. was significantly higher during the COVID-19 pandemic than in the previous 3 years. Monitoring the GM of critically ill COVID-19 patients could help clinical management, by predicting the onset of medical complications such as difficult-to-treat secondary infections.
Collapse
Affiliation(s)
- Paolo Gaibani
- Microbiology Unit, Department of Experimental, Diagnostic and Specialty Medicine, IRCCS St. Orsola Hospital and University of Bologna, Bologna, Italy
| | - Federica D’Amico
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Michele Bartoletti
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, IRCCS St. Orsola Hospital, Bologna, Italy
| | - Donatella Lombardo
- Microbiology Unit, Department of Experimental, Diagnostic and Specialty Medicine, IRCCS St. Orsola Hospital and University of Bologna, Bologna, Italy
| | - Simone Rampelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giacomo Fornaro
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, IRCCS St. Orsola Hospital, Bologna, Italy
| | - Simona Coladonato
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, IRCCS St. Orsola Hospital, Bologna, Italy
| | - Antonio Siniscalchi
- Intensive Care Unit, Department of Medical and Surgical Sciences, IRCCS St. Orsola Hospital and University of Bologna, Bologna, Italy
| | - Maria Carla Re
- Microbiology Unit, Department of Experimental, Diagnostic and Specialty Medicine, IRCCS St. Orsola Hospital and University of Bologna, Bologna, Italy
| | - Pierluigi Viale
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, IRCCS St. Orsola Hospital, Bologna, Italy
| | - Patrizia Brigidi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Maddalena Giannella
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, IRCCS St. Orsola Hospital, Bologna, Italy
| |
Collapse
|
193
|
Multidrug-resistant Acinetobacter baumannii infections in COVID-19 patients hospitalized in intensive care unit. Infection 2021; 50:83-92. [PMID: 34176088 PMCID: PMC8236000 DOI: 10.1007/s15010-021-01643-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/15/2021] [Indexed: 02/08/2023]
Abstract
Objectives Superinfections in patients hospitalized in intensive care unit (ICU) are an important and challenging complication, also in COVID-19. However, no definitive data are available about the role of multidrug-resistant Acinetobacter baumannii (MDR-AB) in COVID-19. Methods This was a single-center, cross-sectional study including patients with MDR-AB infections admitted to ICU with or without COVID-19, between January 2019 and January 2021. The primary objective of the study was to evaluate risk factor for MDR-AB infections in ICU patients hospitalized for COVID-19 or other etiology. The secondary endpoints were 30-days mortality in all study population and risk factors associated with development of bloodstream infection (BSI). Results During the study period 32 adults with COVID-19 were enrolled and compared with 115 patients admitted in the same ICU for other reasons. We observed a total of 114 deaths, with a survival rate of 29.3%: 18.8% in COVID-19 and 32.2% in control group. Relative risk for MDR-AB infection in COVID-19 showed that serum lactate levels mmol/l > 2, Acinetobacter baumannii colonization, BSI and steroid therapy were observed more frequently in COVID-19 patients. Cox regression analysis showed that serum lactate levels > 2 mmol/l, Acinetobacter baumannii colonization, BSI, and steroid therapy were associated with 30-days mortality. Finally, patients with COVID-19, white blood cells count > 11,000 mm3, serum lactate levels > 2 mmol/l, infections at time of ICU admission, Acinetobacter baumannii colonization, and steroid therapy were independently associated with development of BSI. Conclusions Our data highlight the impact of BSI on outcome, the role of Acinetobacter baumannii colonization and the use of steroids on the risk to develop MDR-AB infections also during COVID-19.
Collapse
|
194
|
Yamamoto S, Saito M, Tamura A, Prawisuda D, Mizutani T, Yotsuyanagi H. The human microbiome and COVID-19: A systematic review. PLoS One 2021; 16:e0253293. [PMID: 34161373 PMCID: PMC8221462 DOI: 10.1371/journal.pone.0253293] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/02/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Human microbiotas are communities of microorganisms living in symbiosis with humans. They play an important role in the host immune response to respiratory viral infection. However, evidence on the human microbiome and coronavirus disease (COVID-19) relationship is insufficient. The aim of this systematic literature review was to evaluate existing evidence on the association between the microbiome and COVID-19 in humans and summarize these data in the pandemic era. METHODS We conducted a systematic literature review on the association between the microbiome and COVID-19 in humans by searching PubMed, Embase, and the Cochrane Library, CINAHL, and Web of Science databases for articles in English published up to October 31, 2020. The results were analyzed qualitatively. This study is registered with PROSPERO (CRD42020195982). RESULTS Of the 543 articles identified by searching databases, 16 in line with the research objectives were eligible for qualitative review: eight sampled the microbiome using stool, four using nasopharyngeal or throat swab, three using bronchoalveolar lavage fluid, and one using lung tissue. Fecal microbiome dysbiosis and increased opportunistic pathogens were reported in COVID-19 patients. Several studies suggested the dysbiosis in the lung microbiome of COVID-19 patients with an abundance of opportunistic pathogens using lower respiratory tract samples. The association between COVID-19 severity and the human microbiome remains uncertain. CONCLUSION The human fecal and respiratory tract microbiome changed in COVID-19 patients with opportunistic pathogen abundance. Further research to elucidate the effect of alternation of the human microbiome in disease pathogenesis is warranted.
Collapse
Affiliation(s)
- Shinya Yamamoto
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Makoto Saito
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Azumi Tamura
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Diki Prawisuda
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Taketoshi Mizutani
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan,* E-mail:
| | - Hiroshi Yotsuyanagi
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
195
|
Li J, Richards EM, Handberg EM, Pepine CJ, Raizada MK. Distinct Gene Expression Profiles in Colonic Organoids from Normotensive and the Spontaneously Hypertensive Rats. Cells 2021; 10:cells10061523. [PMID: 34204247 PMCID: PMC8234507 DOI: 10.3390/cells10061523] [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: 04/29/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Hypertension is associated with gut bacterial dysbiosis and gut pathology in animal models and people. Butyrate-producing gut bacteria are decreased in hypertension. RNA-seq analysis of gut colonic organoids prepared from spontaneously hypertensive rats (SHR) and normotensive Wistar Kyoto (WKY) rats was used to test the hypothesis that impaired interactions between the gut microbiome and gut epithelium are involved and that these would be remediated with butyrate supplementation. Gene expressions in immune responses including antigen presentation and antiviral pathways were decreased in the gut epithelium of the SHR in organoids and confirmed in vivo; these deficits were corrected by butyrate supplementation. Deficits in gene expression driving epithelial proliferation and differentiation were also observed in SHR. These findings highlight the importance of aligned interactions of the gut microbiome and gut immune responses to blood pressure homeostasis.
Collapse
Affiliation(s)
- Jing Li
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, USA; (J.L.); (E.M.R.)
| | - Elaine M. Richards
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, USA; (J.L.); (E.M.R.)
| | - Eileen M. Handberg
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA; (E.M.H.); (C.J.P.)
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA; (E.M.H.); (C.J.P.)
| | - Mohan K. Raizada
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, USA; (J.L.); (E.M.R.)
- Correspondence: ; Tel.: +1-352-392-9299
| |
Collapse
|
196
|
Badu K, Oyebola K, Zahouli JZB, Fagbamigbe AF, de Souza DK, Dukhi N, Amankwaa EF, Tolba MF, Sylverken AA, Mosi L, Mante PK, Matoke-Muhia D, Goonoo N. SARS-CoV-2 Viral Shedding and Transmission Dynamics: Implications of WHO COVID-19 Discharge Guidelines. Front Med (Lausanne) 2021; 8:648660. [PMID: 34239886 PMCID: PMC8259580 DOI: 10.3389/fmed.2021.648660] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 05/14/2021] [Indexed: 12/20/2022] Open
Abstract
The evolving nature of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has necessitated periodic revisions of COVID-19 patient treatment and discharge guidelines. Since the identification of the first COVID-19 cases in November 2019, the World Health Organization (WHO) has played a crucial role in tackling the country-level pandemic preparedness and patient management protocols. Among others, the WHO provided a guideline on the clinical management of COVID-19 patients according to which patients can be released from isolation centers on the 10th day following clinical symptom manifestation, with a minimum of 72 additional hours following the resolution of symptoms. However, emerging direct evidence indicating the possibility of viral shedding 14 days after the onset of symptoms called for evaluation of the current WHO discharge recommendations. In this review article, we carried out comprehensive literature analysis of viral shedding with specific focus on the duration of viral shedding and infectivity in asymptomatic and symptomatic (mild, moderate, and severe forms) COVID-19 patients. Our literature search indicates that even though, there are specific instances where the current protocols may not be applicable ( such as in immune-compromised patients there is no strong evidence to contradict the current WHO discharge criteria.
Collapse
Affiliation(s)
- Kingsley Badu
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Theoretical and Applied Biology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Kolapo Oyebola
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Biochemistry and Nutrition Department, Nigerian Institute of Medical Research, Lagos, Nigeria
- Department of Zoology, Faculty of Science, University of Lagos, Lagos, Nigeria
| | - Julien Z. B. Zahouli
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, Abidjan, Côte d'Ivoire
- Centre d'Entomologie Médicale et Vétérinaire, Université Alassane Ouattara, Bouaké, Côte d'Ivoire
| | - Adeniyi Francis Fagbamigbe
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Epidemiology and Medical Statistics, Faculty of Public Health, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Division of Population and Behavioral Sciences, School of Medicine, St. Andrews University, St. Andrews, United Kingdom
| | - Dziedzom K. de Souza
- African Academy of Sciences Affiliates, Nairobi, Kenya
- College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Natisha Dukhi
- African Academy of Sciences Affiliates, Nairobi, Kenya
- College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
- Human and Social Capabilities Division, Human Sciences Research Council, Cape Town, South Africa
| | - Ebenezer F. Amankwaa
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Geography and Resource Development, University of Ghana, Accra, Ghana
| | - Mai F. Tolba
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- The Center of Drug Discovery Research and Development, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Egypt
| | - Augustina A. Sylverken
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Theoretical and Applied Biology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Lydia Mosi
- African Academy of Sciences Affiliates, Nairobi, Kenya
- West African Centre for Cell Biology of Infectious Diseases, University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | - Priscilla Kolibea Mante
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Pharmacology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Damaris Matoke-Muhia
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya
| | - Nowsheen Goonoo
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Biomaterials, Drug Delivery and Nanotechnology Unit, Center for Biomedical and Biomaterials Research (CBBR), University of Mauritius, Reduit, Mauritius
| |
Collapse
|
197
|
Peng J, Zhang M, Yao G, Kwok LY, Zhang W. Probiotics as Adjunctive Treatment for Patients Contracted COVID-19: Current Understanding and Future Needs. Front Nutr 2021; 8:669808. [PMID: 34179059 PMCID: PMC8222530 DOI: 10.3389/fnut.2021.669808] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/17/2021] [Indexed: 01/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) is caused by a novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which rages all over the world and seriously threatens human life and health. Currently, there is no optimal treatment for COVID-19, and emerging evidence found that COVID-19 infection results in gut microbiota dysbiosis. The intestinal microbial richness of patients of COVID-19 does not return to normal levels even six months after recovery, but probiotic adjunctive treatment has been found to restore gut homeostasis. An updated PubMed search returned four finished clinical trials that supported the use of probiotics as adjunctive treatment for COVID-19, while at least six clinical trials aiming to investigate beneficial effects of probiotic intake in managing COVID-19 are currently in progress worldwide. Here in we tentatively summarized the understanding of the actions and potential mechanisms of probiotics in the management of COVID-19. We also highlighted some future needs for probiotic researchers in the field. The success in using probiotics as adjunctive treatment for COVID-19 has expanded the scope of application of probiotics, meanwhile deepening our knowledge in the physiological function of probiotics in modulating the gut-lung axis.
Collapse
Affiliation(s)
- Jiangying Peng
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
| | - Meng Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
| | - Guoqiang Yao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
| | - Wenyi Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China,*Correspondence: Wenyi Zhang
| |
Collapse
|
198
|
Behl T, Kaur I, Sehgal A, Singh S, Bhatia S, Al-Harrasi A, Zengin G, Babes EE, Brisc C, Stoicescu M, Toma MM, Sava C, Bungau SG. Bioinformatics Accelerates the Major Tetrad: A Real Boost for the Pharmaceutical Industry. Int J Mol Sci 2021; 22:6184. [PMID: 34201152 PMCID: PMC8227524 DOI: 10.3390/ijms22126184] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/03/2021] [Accepted: 06/05/2021] [Indexed: 02/01/2023] Open
Abstract
With advanced technology and its development, bioinformatics is one of the avant-garde fields that has managed to make amazing progress in the pharmaceutical-medical field by modeling the infrastructural dimensions of healthcare and integrating computing tools in drug innovation, facilitating prevention, detection/more accurate diagnosis, and treatment of disorders, while saving time and money. By association, bioinformatics and pharmacovigilance promoted both sample analyzes and interpretation of drug side effects, also focusing on drug discovery and development (DDD), in which systems biology, a personalized approach, and drug repositioning were considered together with translational medicine. The role of bioinformatics has been highlighted in DDD, proteomics, genetics, modeling, miRNA discovery and assessment, and clinical genome sequencing. The authors have collated significant data from the most known online databases and publishers, also narrowing the diversified applications, in order to target four major areas (tetrad): DDD, anti-microbial research, genomic sequencing, and miRNA research and its significance in the management of current pandemic context. Our analysis aims to provide optimal data in the field by stratification of the information related to the published data in key sectors and to capture the attention of researchers interested in bioinformatics, a field that has succeeded in advancing the healthcare paradigm by introducing developing techniques and multiple database platforms, addressed in the manuscript.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
| | - Ishnoor Kaur
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
| | - Aayush Sehgal
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
| | - Sukhbir Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University, Gurugram 122413, India;
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa 616, Oman;
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa 616, Oman;
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, 42130 Konya, Turkey;
| | - Elena Emilia Babes
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (E.E.B.); (C.B.); (M.S.); (C.S.)
| | - Ciprian Brisc
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (E.E.B.); (C.B.); (M.S.); (C.S.)
| | - Manuela Stoicescu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (E.E.B.); (C.B.); (M.S.); (C.S.)
| | - Mirela Marioara Toma
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| | - Cristian Sava
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (E.E.B.); (C.B.); (M.S.); (C.S.)
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
199
|
Battaglini D, Robba C, Fedele A, Trancǎ S, Sukkar SG, Di Pilato V, Bassetti M, Giacobbe DR, Vena A, Patroniti N, Ball L, Brunetti I, Torres Martí A, Rocco PRM, Pelosi P. The Role of Dysbiosis in Critically Ill Patients With COVID-19 and Acute Respiratory Distress Syndrome. Front Med (Lausanne) 2021; 8:671714. [PMID: 34150807 PMCID: PMC8211890 DOI: 10.3389/fmed.2021.671714] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
In late December 2019, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) quickly spread worldwide, and the syndrome it causes, coronavirus disease 2019 (COVID-19), has reached pandemic proportions. Around 30% of patients with COVID-19 experience severe respiratory distress and are admitted to the intensive care unit for comprehensive critical care. Patients with COVID-19 often present an enhanced immune response with a hyperinflammatory state characterized by a "cytokine storm," which may reflect changes in the microbiota composition. Moreover, the evolution to acute respiratory distress syndrome (ARDS) may increase the severity of COVID-19 and related dysbiosis. During critical illness, the multitude of therapies administered, including antibiotics, sedatives, analgesics, body position, invasive mechanical ventilation, and nutritional support, may enhance the inflammatory response and alter the balance of patients' microbiota. This status of dysbiosis may lead to hyper vulnerability in patients and an inappropriate response to critical circumstances. In this context, the aim of our narrative review is to provide an overview of possible interaction between patients' microbiota dysbiosis and clinical status of severe COVID-19 with ARDS, taking into consideration the characteristic hyperinflammatory state of this condition, respiratory distress, and provide an overview on possible nutritional strategies for critically ill patients with COVID-19-ARDS.
Collapse
Affiliation(s)
- Denise Battaglini
- Anesthesia and Intensive Care, Ospedale Policlinico San Martino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) per l'Oncologia e le Neuroscienze, Genova, Italy
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Chiara Robba
- Anesthesia and Intensive Care, Ospedale Policlinico San Martino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) per l'Oncologia e le Neuroscienze, Genova, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Università degli Studi di Genova, Genova, Italy
| | - Andrea Fedele
- Anesthesia and Intensive Care, Ospedale Policlinico San Martino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) per l'Oncologia e le Neuroscienze, Genova, Italy
| | - Sebastian Trancǎ
- Department of Anesthesia and Intensive Care II, Clinical Emergency County Hospital of Cluj, Iuliu Hatieganu, University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Anaesthesia and Intensive Care 1, Clinical Emergency County Hospital Cluj-Napoca, Cluj-Napoca, Romania
| | - Samir Giuseppe Sukkar
- Dietetics and Clinical Nutrition Unit, Ospedale Policlinico San Martino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) per l'Oncologia e le Neuroscienze, Genova, Italy
| | - Vincenzo Di Pilato
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Università degli Studi di Genova, Genova, Italy
| | - Matteo Bassetti
- Clinica Malattie Infettive, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) per l'Oncologia e le Neuroscienze, Genova, Italy
- Dipartimento di Scienze della Salute (DISSAL), Università degli Studi di Genova, Genova, Italy
| | - Daniele Roberto Giacobbe
- Clinica Malattie Infettive, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) per l'Oncologia e le Neuroscienze, Genova, Italy
- Dipartimento di Scienze della Salute (DISSAL), Università degli Studi di Genova, Genova, Italy
| | - Antonio Vena
- Clinica Malattie Infettive, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) per l'Oncologia e le Neuroscienze, Genova, Italy
| | - Nicolò Patroniti
- Anesthesia and Intensive Care, Ospedale Policlinico San Martino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) per l'Oncologia e le Neuroscienze, Genova, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Università degli Studi di Genova, Genova, Italy
| | - Lorenzo Ball
- Anesthesia and Intensive Care, Ospedale Policlinico San Martino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) per l'Oncologia e le Neuroscienze, Genova, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Università degli Studi di Genova, Genova, Italy
| | - Iole Brunetti
- Anesthesia and Intensive Care, Ospedale Policlinico San Martino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) per l'Oncologia e le Neuroscienze, Genova, Italy
| | - Antoni Torres Martí
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Division of Animal Experimentation, Department of Pulmonology, Hospital Clinic, Barcelona, Spain
- Centro de Investigacion en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Institut d'investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- COVID-19-Network, Ministry of Science, Technology, Innovation and Communication, Brasilia, Brazil
| | - Paolo Pelosi
- Anesthesia and Intensive Care, Ospedale Policlinico San Martino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) per l'Oncologia e le Neuroscienze, Genova, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Università degli Studi di Genova, Genova, Italy
| |
Collapse
|
200
|
Luo J, Liang S, Jin F. Gut microbiota in antiviral strategy from bats to humans: a missing link in COVID-19. SCIENCE CHINA. LIFE SCIENCES 2021; 64:942-956. [PMID: 33521857 PMCID: PMC7847806 DOI: 10.1007/s11427-020-1847-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/21/2020] [Indexed: 01/31/2023]
Abstract
Bats are a potential natural reservoir for SARS-CoV-2 virus and other viruses detrimental to humans. Accumulated evidence has shown that, in their adaptation to a flight-based lifestyle, remodeling of the gut microbiota in bats may have contributed to immune tolerance to viruses. This evidence from bats provides profound insights into the potential influence of gut microbiota in COVID-19 disease in humans. Here, we highlight recent advances in our understanding of the mechanisms by which the gut microbiota helps bats tolerate deadly viruses, and summarize the current clinical evidence on the influence of gut microbiota on the susceptibility to SARS-CoV-2 infection and risk of COVID-19 leading to a fatal outcome. In addition, we discuss the implications of gut microbiota-targeted approaches for preventing infection and reducing disease severity in COVID-19 patients.
Collapse
Affiliation(s)
- Jia Luo
- Department of Psychology, Sichuan Normal University, Chengdu, 610068, China
| | - Shan Liang
- Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Feng Jin
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|